Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Nat Genet ; 19(2): 148-54, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9620770

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is an autosomal dominant neurodegenerative disorder caused by expansion of a polyglutamine tract in ataxin-1. In affected neurons of SCA1 patients and transgenic mice, mutant ataxin-1 accumulates in a single, ubiquitin-positive nuclear inclusion. In this study, we show that these inclusions stain positively for the 20S proteasome and the molecular chaperone HDJ-2/HSDJ. Similarly, HeLa cells transfected with mutant ataxin-1 develop nuclear aggregates which colocalize with the 20S proteasome and endogenous HDJ-2/HSDJ. Overexpression of wild-type HDJ-2/HSDJ in HeLa cells decreases the frequency of ataxin-1 aggregation. These data suggest that protein misfolding is responsible for the nuclear aggregates seen in SCA1, and that overexpression of a DnaJ chaperone promotes the recognition of a misfolded polyglutamine repeat protein, allowing its refolding and/or ubiquitin-dependent degradation.


Subject(s)
Cysteine Endopeptidases/metabolism , Molecular Chaperones/physiology , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Folding , Spinocerebellar Degenerations/pathology , Animals , Ataxin-1 , Ataxins , Carrier Proteins/metabolism , Cells, Cultured , HSC70 Heat-Shock Proteins , HSP40 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Proteasome Endopeptidase Complex , Protein Conformation , Purkinje Cells/metabolism , Purkinje Cells/pathology , Spinocerebellar Degenerations/genetics , Transfection
2.
J Cell Biol ; 137(3): 523-38, 1997 May 05.
Article in English | MEDLINE | ID: mdl-9151662

ABSTRACT

We have used digitonin-permeabilized cells to examine in vitro nuclear export of glucocorticoid receptors (GRs). In situ biochemical extractions in this system revealed a distinct subnuclear compartment, which collects GRs that have been released from chromatin and serves as a nuclear export staging area. Unliganded nuclear GRs within this compartment are not restricted in their subnuclear trafficking as they have the capacity to recycle to chromatin upon rebinding hormone. Thus, GRs that release from chromatin do not require transit through the cytoplasm to regain functionality. In addition, chromatin-released receptors export from nuclei of permeabilized cells in an ATP- and cytosol-independent process that is stimulated by sodium molybdate, other group VI-A transition metal oxyanions, and some tyrosine phosphatase inhibitors. The stimulation of in vitro nuclear export by these compounds is not unique to GR, but is restricted to other proteins such as the 70- and 90-kD heat shock proteins, hsp70 and hsp90, respectively, and heterogeneous nuclear RNP (hnRNP) A1. Under analogous conditions, the 56-kD heat shock protein, hsp56, and hnRNP C do not export from nuclei of permeabilized cells. If tyrosine kinase inhibitors genistein and tyrphostin AG126 are included to prevent increased tyrosine phosphorylation, in vitro nuclear export of GR is inhibited. Thus, our results are consistent with the involvement of a phosphotyrosine system in the general regulation of nuclear protein export, even for proteins such as GR and hnRNP A1 that use distinct nuclear export pathways.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Receptors, Glucocorticoid/metabolism , Adenosine Triphosphate/pharmacology , Animals , Biological Transport/drug effects , Cell Compartmentation , Cell Membrane Permeability , Corticosterone/metabolism , Corticosterone/pharmacology , Digitonin/chemistry , Enzyme Inhibitors/pharmacology , Genistein , HSP90 Heat-Shock Proteins/metabolism , Heparin/pharmacology , Isoflavones/pharmacology , Liver/metabolism , Liver Neoplasms, Experimental , Microcystins , Molybdenum/pharmacology , Peptides, Cyclic/pharmacology , Phosphotyrosine/metabolism , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Rats , Tumor Cells, Cultured , Tungsten Compounds/pharmacology , Vanadates/pharmacology
3.
Curr Biol ; 11(24): R1036-7, 2001 Dec 11.
Article in English | MEDLINE | ID: mdl-11747843

ABSTRACT

Calreticulin, a calcium-binding protein of the endoplasmic reticulum, has been found to function as a nuclear export factor for a large family of nuclear receptors. Atypical nuclear export pathways may thus exist that regulate the compartmentalization and activity of a distinct set of transcription factors.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Calreticulin , Protein Transport
4.
Neuroscience ; 145(1): 248-55, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17207579

ABSTRACT

Oxidative stress after cerebral ischemia and reperfusion activates extracellular signal-regulated kinases (ERK) in brain. However, the mechanism of this activation has not been elucidated. We have previously reported that in an in vitro model of oxidative stress in immature cortical neuronal cultures, the inhibition of ERK phosphatase activity contributes to ERK1/2 activation and subsequent neuronal toxicity. This study examined whether ERK activation was associated with altered activity of ERK phosphatases in a rat cardiac arrest model. Rats in experimental groups were subjected to asphyxial cardiac arrest for 8 min and then resuscitated for 30 min. Significant ERK activation was detected in both cortex and hippocampus following ischemia/reperfusion by immunoblotting. ERK phosphatase activity was reversibly inhibited in cerebral cortex but not affected in hippocampus following ischemia/reperfusion. MEK1/2 was activated in both cerebral cortex and hippocampus following ischemia/reperfusion. Using a specific inhibitor of protein phosphatase 2A (PP2A), okadaic acid (OA), we have identified PP2A to be the major ERK phosphatase that is responsible for regulating ERK activation in ischemic brain tissues. Orthovanadate inhibited ERK phosphatase activity in brain tissues, suggesting that tyrosine phosphatases and dual specificity phosphatases may also contribute to the ERK phosphatase activity in brain tissues. Together, these data implicate ERK phosphatase in the regulation of ERK activation in distinct brain regions following global ischemia.


Subject(s)
Brain/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Ischemia/enzymology , Ischemia/pathology , Reperfusion , Animals , Blotting, Western/methods , Brain/drug effects , Brain/pathology , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Male , Okadaic Acid/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Rats , Rats, Sprague-Dawley
5.
Mol Cell Biol ; 16(5): 1989-2001, 1996 May.
Article in English | MEDLINE | ID: mdl-8628265

ABSTRACT

Glucocorticoid receptors (GRs) have the capacity to shuttle between the nuclear and cytoplasmic compartments, sharing that trait with other steroid receptors and unrelated nuclear proteins of diverse function. Although nuclear import of steroid receptors, like that of nearly all other karyophilic proteins examined to date, requires ATP, there appear to be different energetic requirements for export of proteins, including steroid receptors, from nuclei. In an attempt to reveal which steps, if any, in the nuclear export pathway utilized by steroid receptors require ATP, we have used indirect immunofluorescence to visualize GRs within cells subjected to a reversible ATP depletion. Under conditions which lead to >95% depletion of cellular ATP levels within 90 min, GRs remain localized within nuclei and do not efflux into the cytoplasm. Under analogous conditions of ATP depletion, transfected progesterone receptors are also retained within nuclei. Importantly, GRs which accumulate within nuclei of ATP-depleted cells are distinguished from nuclear receptors in metabolically active cells by their resistance to in situ extraction with a hypotonic, detergent-containing buffer. GRs in ATP-depleted cells are not permanently trapped in this nuclear compartment, as nuclear receptors rapidly regain their capacity to be extracted upon restoration of cellular ATP, even in the absence of de novo protein synthesis. More extensive extraction of cells with high salt and detergent, coupled with DNase I digestion, established that a significant fraction of GRs in ATP-depleted cells are associated with an RNA-containing nuclear matrix. Quantitative Western blot (immunoblot) analysis confirmed the dramatic increase in GR binding to the nuclear matrix of ATP-depleted cells, while confocal microscopy revealed that GRs are bound to the matrix throughout all planes of the nucleus. ATP depletion does not lead to wholesale collapse of nuclear proteins onto the matrix, as the interaction of a subpopulation of simian virus 40 large tumor antigen with the nuclear matrix is not quantitatively altered in ATP-depleted Cos-1 cells. Nuclear GRs which are not bound to the nuclear matrix of metabolically active cells (i.e., a DNA-binding domain deletion mutant and a beta-galactosidase chimera possessing the GR nuclear localization signal sequence) are not recruited to the matrix upon depletion of cellular ATP. Thus, it appears that ATP depletion does not expose the GR to nuclear matrix interactions which are not normally encountered in cells but merely alters the dynamics of such interactions. The dynamic association of steroid receptors with the nuclear matrix may provide a mechanism which is utilized by these regulable transcription factors to facilitate their efficient scanning of the genome.


Subject(s)
Adenosine Triphosphate/metabolism , Nuclear Matrix/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Animals , Antigens, Polyomavirus Transforming/metabolism , Blotting, Western , CHO Cells , Chickens , Cricetinae , Dexamethasone/pharmacology , Fluorescent Antibody Technique, Indirect , Kinetics , Liver Neoplasms , Liver Neoplasms, Experimental , Rats , Receptors, Progesterone/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured
6.
Mol Cell Biol ; 14(8): 5088-98, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8035791

ABSTRACT

Nuclear import of glucocorticoid receptors (GRs) was analyzed in vitro with digitonin-permeabilized cells (S. A. Adam, R. Sterne-Marr, and L. Gerace, J. Cell Biol. 111:807-816, 1990). Indirect immunofluorescence methods were used to monitor the transport of GRs from rat hepatoma and fibroblast cell cytosol into HeLa nuclei. In vitro nuclear import of GRs was shown to be hormone dependent and to require ATP and incubation at ambient temperatures (i.e., 30 degrees C). Hormone-dependent dissociation of GR-bound proteins, such as the 90-kDa heat shock protein, hsp90, is part of an activation process that is obligatory for the expression of the receptor's DNA-binding activity. Inhibition of in vitro GR activation by Na2MoO4 blocked hormone-dependent nuclear import, demonstrating that receptor activation is required for nuclear import. The addition to GR-containing cytosol of antiserum directed against the cytosolic 70-kDa heat shock protein, hsp70, while effective in blocking the nuclear import of simian virus 40 large tumor antigen (SV40 TAg), did not affect hormone-dependent nuclear import of endogenous, full-length GRs or an exogenously added truncated GR protein (i.e., XGR556) that lacks a hormone-binding domain but possesses a constitutively active nuclear localization signal sequence (NLS). Depletion of hsp70 from HeLa cell cytosol did not affect the nuclear import of exogenously added XGR556 but led to inhibition of SV40 TAg nuclear import. Thus, two closely related NLSs, one contained within GRs and the other contained within SV40 TAg, are distinguished by their differential requirements for hsp70 in vitro.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Cell Nucleus/metabolism , Heat-Shock Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Amino Acid Sequence , Animals , Biological Transport , Cell Compartmentation , HeLa Cells , Humans , In Vitro Techniques , Molecular Sequence Data , Rats , Sequence Alignment , Sequence Homology, Amino Acid
7.
Mol Endocrinol ; 20(1): 56-64, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16141357

ABSTRACT

Hic-5/androgen receptor (AR) coactivator 55 (ARA55) is a group III LIM domain protein that functions as a nuclear receptor coactivator. In the present study, we examined the mechanism by which Hic-5/ARA55 potentiates glucocorticoid receptor (GR) transactivation in the A1-2 derivative of T47D breast cancer cells. Hic-5/ARA55 is an important component of GR-coactivator complexes in A1-2 cells because ablation of Hic-5/ARA55 expression by RNA interference-mediated silencing reduced GR transactivation. As shown by chromatin immunoprecipitation (ChIP) assays, Hic-5/ARA55 is recruited to glucocorticoid-responsive promoters of the mouse mammary tumor virus, c-fos, and p21 genes in response to glucocorticoid treatment. Results from sequential ChIPs established that Hic-5/ARA55 associates with GR-containing complexes at these promoters. We also used sequential ChIPs to examine Hic-5/ARA55 interactions with other well-characterized nuclear receptor coactivators and detected transcriptional intermediary factor 2, receptor-associated coactivator 3, cAMP response element binding protein-binding protein, and p300 within Hic-5/ARA55 complexes on the mouse mammary tumor virus promoter in hormone-treated cells. Ablation of Hic-5/ARA55 expression resulted in reduction of both transcriptional intermediary factor 2 and p300 recruitment to glucocorticoid-responsive promoters. Hic-5/ARA55 is also associated with the corepressor, nuclear receptor corepressor, on glucocorticoid-responsive promoters in cells not exposed to glucocorticoids. These results suggest that Hic-5/ARA55 is required for optimal GR-mediated gene expression possibly by providing a scaffold that organizes or stabilizes coactivator complexes at some hormone-responsive promoters.


Subject(s)
Cell Nucleus/metabolism , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Transcriptional Activation , Cell Line, Tumor , Chromatin Immunoprecipitation , Cyclic AMP Response Element-Binding Protein/metabolism , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Genes, fos/physiology , Glucocorticoids/physiology , Humans , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Mammary Tumor Virus, Mouse/genetics , Membrane Proteins/genetics , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , Receptors, Virus/genetics , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism
8.
Mol Biol Cell ; 8(5): 795-809, 1997 May.
Article in English | MEDLINE | ID: mdl-9168467

ABSTRACT

All steroid receptors possess a bipartite nuclear localization signal sequence (NLS) that localizes within the second zinc finger of their DNA-binding domain. Fine-structure mapping of the rat glucocorticoid receptor (rGS) NLS identified a composite signal composed of three distinct proto-NLSs that function effectively when present in unique pairs. At least one of the rGR proto-NLSs appears to influence receptor trafficking within the nucleus, as revealed by a unique nuclear staining pattern of receptors possessing a point mutation (i.e., arginine at position 496; R496), at proto-NLS, pNLS-2. Specifically, carboxyl-terminal-truncated rGRs possessing various point mutations at R496 localized within a limited number of large foci in nuclei of transiently transfected COS-1 cells. R496 mutations did not affect subnuclear targeting when present in full-length rGR, reflecting a protective effect of the receptor's ligand-binding domain that can be exerted in cis and in trans. The effects of rGR R496 mutations on subnuclear targeting were not autonomous because we also observed a coincident localization of hsp70, the 70-kDa heat shock protein, within nuclear foci that include r496 mutant receptors. The elimination of R496 mistargeting by overexpression of an hsp70 partner (i.e., the DnaJ homologue, HDJ-2/HSDJ) suggests that the hsp70/DnaJ chaperone system is mobilized to specific sites within the nucleus in response to inappropriate targeting or folding of specific mutant receptors. HDJ-2/HSDJ overexpression also corrects defective transactivation and transrepression activity of R496 mutant GRs. Thus, molecular chaperones, such as members of the hsp70 and DnaJ families, may survey the nucleus for misfolded proteins and actively participate in their refolding into biologically active conformational states.


Subject(s)
Carrier Proteins , Heat-Shock Proteins , Nuclear Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Transcriptional Activation , Zinc Fingers , Amino Acid Sequence , Animals , Arginine , Binding Sites , Biological Transport , COS Cells , Cell Nucleus/metabolism , Gene Expression Regulation , HSP40 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/metabolism , Humans , Ligands , Molecular Sequence Data , Mutation , Nuclear Localization Signals , Nuclear Proteins/genetics , Rats , Receptors, Glucocorticoid/genetics , Transfection
9.
Mol Biol Cell ; 11(6): 2007-18, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10848625

ABSTRACT

Hic-5 (hydrogen peroxide-inducible clone-5) is a focal adhesion protein that is involved in cellular senescence. In the present study, a yeast two-hybrid screen identified Hic-5 as a protein that interacts with a region of the glucocorticoid receptor that includes a nuclear matrix-targeting signal and the tau2 transcriptional activation domain. In transiently transfected mammalian cells, overexpression of Hic-5 potentiated the activation of reporter genes by all steroid receptors, excluding the estrogen receptor. The activity of the estrogen receptor and the thyroid hormone receptor was stimulated by Hic-5 in the presence but not in the absence of coexpressed coactivator GRIP1. In biochemical fractionations and indirect immunofluorescence assays, a fraction of endogenous Hic-5 in REF-52 cells and transiently expressed Hic-5 in Cos-1 cells was associated with the nuclear matrix. The C-terminal region of Hic-5, which contains seven zinc fingers arranged in four LIM domains, was required for interaction with focal adhesions, the nuclear matrix, steroid receptors, and the tau2 domain of glucocorticoid receptor. The N-terminal region of Hic-5 possesses a transcriptional activation domain and was essential for the coactivator activity of Hic-5. Given the coexisting cytoplasmic and nuclear distributions of Hic-5 and its role in steroid receptor-mediated transcriptional activation, it is proposed that Hic-5 might transmit signals that emanate at cell attachment sites and regulate transcription factors, such as steroid receptors.


Subject(s)
Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Transcriptional Activation , Zinc Fingers , Animals , Binding Sites , COS Cells , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytoplasm/metabolism , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Mice , Nuclear Matrix/metabolism , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Receptors, Progesterone/metabolism
10.
Transl Psychiatry ; 7(6): e1153, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28608856

ABSTRACT

Antenatal administration of synthetic glucocorticoids (sGC) is the standard of care for women at risk for preterm labor before 34 gestational weeks. Despite their widespread use, the type of sGC used and their dose or the dosing regimens are not standardized in the United States of America or worldwide. Several studies have identified neural deficits and the increased risk for cognitive and psychiatric disease later in life for children administered sGC prenatally. However, the precise molecular and cellular targets of GC action in the developing brain remain largely undefined. In this study, we demonstrate that a single dose of glucocorticoid during mid-gestation in mice leads to enhanced proliferation in select cerebral cortical neural stem/progenitor cell populations. These alterations are mediated by dose-dependent changes in the expression of cell cycle inhibitors and in genes that promote cell cycle re-entry. This leads to changes in neuronal number and density in the cerebral cortex at birth, coupled to long-term alterations in neurite complexity in the prefrontal cortex and hippocampus in adolescents, and changes in anxiety and depressive-like behaviors in adults.


Subject(s)
Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Dexamethasone/pharmacology , Neural Stem Cells/drug effects , Neurons/drug effects , Prenatal Exposure Delayed Effects/pathology , Animals , Anxiety/pathology , Anxiety/psychology , Cell Count , Cell Shape/drug effects , Cerebral Cortex/pathology , Depression/pathology , Depression/psychology , Female , Hippocampus/drug effects , Hippocampus/pathology , Mice , Neural Stem Cells/pathology , Neurons/pathology , Pregnancy , Prenatal Exposure Delayed Effects/psychology
11.
Sci STKE ; 2000(42): pe1, 2000 Jul 25.
Article in English | MEDLINE | ID: mdl-11752599

ABSTRACT

Steroid hormone receptors interact with several different molecular chaperones. DeFranco and Csermely discuss how the molecular chaperones p23 and Hsp90 may serve to regulate the activity of the ligand-bound steroid receptors within the nucleus. The authors hypothesize that these chaperone proteins may have a proactive role in promoting recycling of receptors once they have interacted with chromatin and in allowing rebinding of ligand once the receptors have been recycled.


Subject(s)
Cell Nucleus/metabolism , Molecular Chaperones/physiology , Receptors, Steroid/physiology , Cell Nucleus/physiology , Humans
12.
Mol Endocrinol ; 6(1): 26-34, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1310797

ABSTRACT

The effects of okadaic acid (OA), a protein phosphatase inhibitor, on transcriptional enhancement activity of rat glucocorticoid receptor (GR) were examined in transiently transfected cells. In the absence of hormone, GRs expressed in CV-1 and COS-1 fibroblasts were capable of enhancing transcription from cotransfected chloramphenicol acetyltransferase reporter plasmids in response to OA treatment. Synergistic enhancement resulted from combined hormone and OA treatment. The effects of OA on GR-mediated enhancement required the presence of linked glucocorticoid response elements and were observed with reporter plasmids that contained different promoters and glucocorticoid response elements. Since OA did not affect nuclear translocation of the receptor, enhancement mediated by unliganded GR was most likely accounted for by the accumulation of some unliganded GRs within nuclei of transfected CV-1 and COS-1 cells. Deletion of individual GR transactivation domains and point mutations within DNA- and hormone-binding domains severely reduced the response of receptors to OA, although some mutant receptors retained the capacity to elicit a synergistic response when exposed to OA and hormone. The effects of OA on transcriptional enhancement did not appear to correlate with major changes in GR phosphorylation, as visualized by two-dimensional tryptic mapping of in vivo 32P-labeled GRs. Thus, phosphorylation of various components of the GR signal transduction pathway, and not necessarily the receptor itself, may influence its transcriptional enhancement activity.


Subject(s)
Enhancer Elements, Genetic/physiology , Phosphoprotein Phosphatases/physiology , Receptors, Glucocorticoid/physiology , Animals , Cell Line , Enhancer Elements, Genetic/drug effects , Enhancer Elements, Genetic/genetics , Ethers, Cyclic/pharmacology , Mutagenesis, Site-Directed/genetics , Mutagenesis, Site-Directed/physiology , Okadaic Acid , Phosphoprotein Phosphatases/antagonists & inhibitors , Promoter Regions, Genetic/physiology , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/genetics , Transcription Factors/physiology , Transcriptional Activation/genetics , Transcriptional Activation/physiology
13.
Mol Endocrinol ; 6(5): 837-44, 1992 May.
Article in English | MEDLINE | ID: mdl-1603089

ABSTRACT

The chicken progesterone receptor (PR) is a transcriptional regulatory protein that localizes predominantly within the nucleus of hormone-treated and untreated cells. Transient heterokaryons were generated between PR-expressing Cos-1 cells and PR-negative NIH3T3 cells to examine whether PRs are confined to the nucleus or are capable of bidirectionally traversing the nuclear envelope. Migration of PR from Cos-1 to NIH3T3 nuclei was observed in both the presence and absence of hormone. Since de novo PR synthesis was inhibited in heterokaryons with cycloheximide treatment, PRs that localize within NIH3T3 nuclei of heterokaryons must derive from preexisting receptors that were exported from Cos-1 nuclei. Thus, PR, like some nucleolar and heat shock proteins, appears to be capable of shuttling between the nuclear and cytoplasmic compartments. Not all proteins that enter the nucleus exhibit this trait, since simian virus-40 large tumor antigen, endogenously expressed in Cos-1 cells, does not efficiently translocate to NIH3T3 nuclei of heterokaryons, which support internuclear migration of PR. Thus, proteins that may use analogous or identical mechanisms for nuclear import may differentially interact with the nuclear export machinery. Furthermore, the fact that PR and simian virus-40 large tumor antigen localization within nuclei is not identical, as revealed by laser scanning confocal microscopy, supports the notion that nuclear export may be influenced by subnuclear compartmentalization.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Cell Nucleus/metabolism , Receptors, Progesterone/metabolism , Animals , Biological Transport , Chickens , Cycloheximide/pharmacology , Cytoplasm/metabolism , Half-Life , Hybrid Cells
14.
Mol Endocrinol ; 10(1): 3-13, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8838140

ABSTRACT

Unliganded glucocorticoid receptors (GRs) that reside in the cytoplasm exist as heteromeric complexes comprised minimally of 90-kDa heat shock protein (hsp90) hsp70 and p56, a 56-kDa immunophilin. The binding of hsp90 to the GR occurs primarily through its carboxy-terminal, ligand-binding domain. Dissociation of GR-associated proteins accompanies hormone binding and leads to the exposure of its various functional domains. Although an association with hsp90 presumably masks the GR nuclear localization signal sequence, the recent demonstration of the coimport of GR and hsp90 into nuclei has led to the hypothesis that hsp90 facilitates GR interactions with the nuclear transport machinery. In this report we examined whether the dynamics of GR/hsp90 interactions in vivo influences its trafficking both into and out of the nucleus. GR/hsp90 complexes were stabilized in vivo by the introduction of sodium molybdate to cultured cells using a liposome-mediated delivery system. In agreement with previous in vitro studies, we found that stabilization of GR/hsp90 complexes in live cells severely restricts hormone-dependent nuclear import of GR. Constitutive nuclear import of a GR deletion derivative that does not bind hsp90 is unaffected by intracellular administration of molybdate, demonstrating that the inhibitory effects of molybdate require the coupling of the nuclear localization signal sequence to the GR ligand-binding domain. Interestingly, molybdate treatment traps both GR and progesterone receptor in the cytoplasm of cells chronically exposed to hormone, indicating that shuttling GRs and progesterone receptors can export, but not reimport into nuclei in the presence of molybdate. This result implies that the reassociation of recycled receptors with hsp90 must be an obligatory step for receptors that exit the nucleus to reacquire the capacity for nuclear import.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , HSP90 Heat-Shock Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cell Line , Dexamethasone/pharmacology , Haplorhini , Kidney , Liver Neoplasms, Experimental , Molybdenum/pharmacology , Rats , Receptors, Glucocorticoid/genetics , Transfection , Tumor Cells, Cultured
15.
Mol Endocrinol ; 14(1): 40-51, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10628746

ABSTRACT

Glucocorticoid receptors (GRs) are shuttling proteins, yet they preferentially accumulate within either the cytoplasmic or nuclear compartment when overall rates of nuclear import or export, respectively, are limiting. Hormone binding releases receptors from stable heteromeric complexes that restrict their interactions with soluble nuclear import factors and contribute to their cytoplasmic retention. Although hormone dissociation leads to the rapid release of GRs from chromatin, unliganded nuclear receptors are delayed in their export. We have used a chimeric GR that contains a heterologous, leucine-rich nuclear export signal sequence (NES) to assess the consequences of accelerated receptor nuclear export. Leucine-rich NESs utilize the exportin 1/CRM1-dependent nuclear export pathway, which can be blocked by leptomycin B (LMB). The fact that rapid nuclear export of the NES-GR chimera, but not the protracted export of wild-type GR, is sensitive to LMB, suggests that GR does not require the exportin 1/CRM1 pathway to exit the nucleus. Despite its more rapid export, the NES-GR chimera appears indistinguishable from wild-type GR in its transactivation activity in transiently transfected cells. However, accelerated nuclear export of the NES-GR chimera is associated with an increased rate of hormone-dependent down-regulation. The increase in NES-GR down-regulation is overcome by LMB treatment, thereby confirming the connection between receptor nuclear export and down-regulation. Given the presence of a nuclear recycling pathway for GR, the protracted rate of receptor nuclear export may increase the efficiency of biological responses to secondary hormone challenges by limiting receptor down-regulation and hormone desensitization.


Subject(s)
Carrier Proteins/metabolism , Karyopherins , Receptors, Cytoplasmic and Nuclear , Receptors, Glucocorticoid/metabolism , Animals , Blotting, Western , COS Cells , Cycloheximide/pharmacology , Dexamethasone/pharmacology , Down-Regulation , Fatty Acids, Unsaturated/pharmacology , Glucocorticoids/pharmacology , Kinetics , Leucine/metabolism , Luciferases/metabolism , Nuclear Proteins/metabolism , Plasmids/metabolism , Protein Synthesis Inhibitors/pharmacology , Receptors, Glucocorticoid/genetics , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Exportin 1 Protein
16.
Mol Endocrinol ; 11(9): 1365-74, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9259326

ABSTRACT

The synthesis of a number of heat shock proteins is induced in response to various environmental stresses. The resultant induction of heat shock protein gene transcription is brought about by the activation of specific transcription factors termed heat shock factors (HSFs) that exist in a latent form in nonstressed cells. Multiple mechanisms are likely to contribute to negative regulation of HSF activity. One model, which remains controversial, proposes the existence of a negative feedback loop by which one of the products of HSF activation, the 70-kDa heat shock protein (hsp70), acts as one of its negative regulators. Accordingly, HSF activation would proceed upon sequestration of hsp70 by substrates (i.e. unfolded proteins) that may accumulate to relatively high levels in stressed cells. To examine whether putative native substrates of hsp70 (e.g. steroid receptors) could impact the regulation of HSF activity, we have examined whether steroid receptors could activate endogenous HSF. We have found that overexpression of androgen (AR), glucocorticoid (GR), mineralocorticoid, and progesterone receptors in transiently transfected COS-1 cells induced HSF activity. With the exception of AR, which was competent to activate HSF when either liganded or unliganded, all other steroid receptors tested only activated HSF when unliganded. This activity was mapped to the ligand-binding domain of rat GR, making it unlikely that HSF activation results from the induction of a novel gene product by unliganded receptors. As overexpression of hsp70 can eliminate HSF activation by AR, GR, and progesterone receptors, we favor the view that HSF activation can result from the sequestration, by steroid receptor ligand-binding domains, of a negative regulator of HSF, such as hsp70 or an hsp70-associated protein.


Subject(s)
Heat-Shock Proteins/metabolism , Receptors, Steroid/metabolism , Animals , Binding Sites , COS Cells/drug effects , COS Cells/metabolism , Dexamethasone/pharmacology , HSP70 Heat-Shock Proteins/drug effects , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/genetics , Mifepristone/pharmacology , Rats , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/drug effects , Receptors, Steroid/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Sequence Deletion , Signal Transduction
17.
Mol Endocrinol ; 13(3): 355-65, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10076993

ABSTRACT

Unliganded glucocorticoid receptors (GRs) released from chromatin after hormone withdrawal remain associated with the nucleus within a novel subnuclear compartment that serves as a nuclear export staging area. We set out to examine whether unliganded nuclear receptors cycle between distinct subnuclear compartments or require cytoplasmic transit to regain hormone and chromatin-binding capacity. Hormone-withdrawn rat GrH2 hepatoma cells were permeabilized with digitonin to deplete cytoplasmic factors, and then hormone-binding and chromatin-binding properties of the recycled nuclear GRs were measured. We found that recycled nuclear GRs do not require cytosolic factors or ATP to rebind hormone. Nuclear GRs that rebind hormone in permeabilized cells target to high-affinity chromatin-binding sites at 30 C, but not 0 C, in the presence of ATP. Since geldanamycin, a heat shock protein-90 (hsp90)-binding drug, inhibits hormone binding to recycled nuclear GRs, hsp90 may be required to reassemble the receptor into a form capable of productive interactions with hormone. Geldanamycin also inhibits GR release from chromatin during hormone withdrawal, suggesting that hsp90 chaperone function may play multiple roles to facilitate chromatin recycling of GR.


Subject(s)
Chromatin/metabolism , HSP90 Heat-Shock Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Adenosine Triphosphate/metabolism , Animals , Benzoquinones , Biological Transport , Carcinoma, Hepatocellular/metabolism , Cell Membrane Permeability/drug effects , Cell Nucleus/metabolism , Corticosterone/metabolism , Corticosterone/pharmacology , Cytosol/metabolism , Digitonin/pharmacology , Enzyme Inhibitors/pharmacology , Lactams, Macrocyclic , Nuclear Proteins/metabolism , Quinones/pharmacology , Rats , Temperature , Tumor Cells, Cultured , Wheat Germ Agglutinins/pharmacology
18.
Mol Endocrinol ; 4(3): 455-64, 1990 Mar.
Article in English | MEDLINE | ID: mdl-2160602

ABSTRACT

In v-mos transformed cells, glucocorticoid receptor (GR) proteins that bind hormone agonist are not efficiently retained within nuclei and redistribute to the cytoplasmic compartment. These cytoplasmic desensitized receptors cannot be reutilized and may represent trapped intermediates derived from GR recycling. We have used the glucocorticoid antagonist RU486 to examine whether v-mos effects can be exerted on any ligand-bound GR. In the rat 6m2 cell line that expresses a temperature-sensitive p85gag-mos oncoprotein, RU486 is a complete antagonist and suppresses dexamethasone induction of metallothionein-1 mRNA at equimolar concentrations. Using indirect immunofluorescence, we observe efficient nuclear translocation of GR in response to RU486 treatment in either the presence or absence of v-mos oncoproteins. However, in contrast to the redistribution of agonist-bound nuclear receptors to the cytoplasm of v-mos-transformed cells, RU486-bound GRs are efficiently retained within nuclei. Interestingly, withdrawal of RU486 does not lead to efficient depletion of nuclear GR in either nontransformed or v-mos transformed cells. It is only after the addition of hormone agonist to RU486 withdrawn v-mos-transformed cells that GRs are depleted from nuclei and subsequently redistributed to the cytoplasm. Thus, only nuclear GRs that are agonist-bound and capable of modulating gene activity can be subsequently processed and recycled into the cytoplasm.


Subject(s)
Gene Expression/drug effects , Receptors, Cell Surface/genetics , Receptors, Glucocorticoid/metabolism , Retroviridae Proteins, Oncogenic/physiology , Animals , Cell Line, Transformed , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Dexamethasone/pharmacology , Fluorescent Antibody Technique , Mifepristone/metabolism , Mifepristone/pharmacology , Oncogene Proteins v-mos , Rats , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/physiology , Translocation, Genetic
19.
Mol Endocrinol ; 5(9): 1215-28, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1663212

ABSTRACT

We have used okadaic acid (OA), a cell-permeable inhibitor of serine/threonine protein phosphatase types 1 (PP-1) and 2A (PP-2A), to demonstrate that the subcellular distribution of glucocorticoid receptor (GR) in rat fibroblasts is influenced by its phosphorylation state. Nuclear GRs in OA-treated cells retain transcriptional enhancement activity. Nuclear import or export of hormone agonist-bound GRs is not affected by OA. However, a dose of OA that fully inhibits PP-2A and partially inhibits PP-1, but not a lower dose that only partially inhibits PP-2A, leads to inefficient nuclear retention of agonist-bound GRs, and their redistribution into the cytoplasm. These receptors appear to be trapped in the cytoplasmic compartment and are unable to recycle (i.e. reenter the nucleus). Addition of OA during different steps of GR recycling demonstrates that OA must be present during nuclear export of GRs to block GR recycling. A direct role for PP-1 and/or PP-2A in GR recycling is suggested by site-specific hyperphosphorylation of GRs in vivo during OA inhibition of recycling. These are the same sites that undergo in vitro site-specific dephosphorylation by PP-1 and PP-2A. The block in GR recycling that results from inhibition of PP-1 and/or PP-2A resembles effects previously observed in v-mos-transformed rat fibroblasts. Interestingly, OA inhibition of PP-2A in v-mos-transformed cells leads to the reversal of oncoprotein effects on GR recycling and retention of receptors within the nuclear compartment. We propose that GR recycling is influenced by the activities of distinct protein phosphatases (PP-1 and/or PP-2A), and that the interference of this pathway observed in v-mos-transformed cells may be the result of effects of the oncoprotein on the phosphatases or a specific subset of their targets.


Subject(s)
Ethers, Cyclic/pharmacology , Phosphoprotein Phosphatases/metabolism , RNA, Messenger/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Biological Transport , Cell Nucleus/metabolism , Cell Transformation, Neoplastic , Cytoplasm/metabolism , Dexamethasone/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation , Okadaic Acid , Oncogenes/physiology , Peptide Mapping , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation , Rats
20.
Mol Endocrinol ; 12(9): 1420-31, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9731709

ABSTRACT

Using an ATP-depletion paradigm to augment glucocorticoid receptor (GR) binding to the nuclear matrix, we have identified a minimal segment of the receptor that constitutes a nuclear matrix targeting signal (NMTS). While previous studies implicated a role for the receptor's DNA-binding domain in nuclear matrix targeting, we show here that this domain of rat GR is necessary, but not sufficient, for matrix targeting. A minimal NMTS can be generated by linking the rat GR DNA-binding domain to either its tau2 transactivation domain in its natural context, or a heterologous transactivation domain derived from the Herpes simplex virus VP16 protein. The transactivation and nuclear matrix-targeting activities of tau2 are separable, as transactivation mutants were identified that either inhibited or had no apparent effect on matrix targeting of tau2. A functional interaction between the NMTS of rat GR and the RNA-binding nuclear matrix protein hnRNP U was revealed in cotransfection experiments in which hnRNP U overexpression was found to interfere with the transactivation activity of GR derivatives that possess nuclear matrix-binding capacity. We have therefore ascribed a novel function to a steroid hormone transactivation domain that could be an important component of the mechanism used by steroid hormone receptors to regulate genes in their native configuration within the nucleus.


Subject(s)
DNA/metabolism , Nuclear Localization Signals , Receptors, Glucocorticoid/metabolism , Transcriptional Activation , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , CHO Cells , Cricetinae , Heterogeneous-Nuclear Ribonucleoprotein U , Heterogeneous-Nuclear Ribonucleoproteins , RNA/metabolism , Rats , Ribonucleoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL