Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Mol Cell Endocrinol ; 365(1): 95-107, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23063623

ABSTRACT

Prostate cancer growth depends on androgens. Synthetic antiandrogens are used in the cancer treatment. However, antiandrogens, such as bicalutamide (BIC), have a mixed agonist/antagonist activity. Here we compare the antiandrogenic capacity of BIC to a new antiandrogen, MDV3100 (MDV) or Enzalutamide™. By reconstitution of a hormone-regulated enhancer in Xenopus oocytes we show that both antagonists trigger the androgen receptor (AR) translocation to the nucleus, albeit with a reduced efficiency for MDV. Once in the nucleus, both AR-antagonist complexes can bind sequence specifically to DNA in vivo. The forkhead box transcription factor A (FoxA1) is a negative prognostic indicator for prostate cancer disease. FoxA1 expression presets the enhancer chromatin and makes the DNA more accessible for AR binding. In this context the BIC-AR antiandrogenic effect is seriously compromised as demonstrated by a significant chromatin remodeling and induction of a robust MMTV transcription whereas the MDV-AR complex displays a more persistent antagonistic character.


Subject(s)
Anilides/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Hepatocyte Nuclear Factor 3-alpha/metabolism , Neoplasm Proteins/metabolism , Nitriles/pharmacology , Nonsteroidal Anti-Androgens/pharmacology , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Tosyl Compounds/pharmacology , Anilides/adverse effects , Anilides/metabolism , Animals , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/metabolism , Benzamides , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatin Assembly and Disassembly/drug effects , Female , HEK293 Cells , Hepatocyte Nuclear Factor 3-alpha/antagonists & inhibitors , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nitriles/adverse effects , Nitriles/metabolism , Nonsteroidal Anti-Androgens/adverse effects , Nonsteroidal Anti-Androgens/metabolism , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Phenylthiohydantoin/metabolism , Phenylthiohydantoin/pharmacology , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Transport/drug effects , RNA Interference , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Tosyl Compounds/adverse effects , Tosyl Compounds/metabolism , Xenopus laevis
2.
Oncogene ; 27(36): 4865-76, 2008 Aug 21.
Article in English | MEDLINE | ID: mdl-18469865

ABSTRACT

Transcriptional control by androgens via androgen receptor (AR) is strongly involved in prostate cancer development, but the critical target genes have remained elusive. We have characterized E twenty-six-like transcription factor 4 (ELK4) (also known as serum response factor accessory protein 1) as a novel AR target in human prostate cancer cells. In-silico screening identified three putative AR response elements (AREs) within -10 kb from the transcription start site of ELK4. Both ARE1 at -167/-153 and ARE2 at -481/-467 bound AR in vitro and mediated androgen induction as isolated elements in transcription assays in non-prostate cells. However, merely the ARE2 that cooperates with a proximal forkhead box A1-binding site was critical for the AR-dependent activation of ELK4 promoter in prostate cancer cells. Preferential loading of holo-AR onto the ARE2 and concomitant recruitment of RNA polymerase II onto the ELK4 promoter was confirmed in prostate cancer cells by chromatin immunoprecipitation. Database searches indicated that the expression of ELK4 is markedly increased in prostate cancers relative to normal prostates. Moreover, prostate cancer tissue immunostainings showed that nuclear ELK4 levels are significantly increased in androgen-refractory prostate cancers compared to untreated tumours. Reduction of the amount of ELK4 in LNCaP cells by RNAi retarded cell growth. In conclusion, ELK4 is a direct AR target in prostate cancer cells. Androgens may thus contribute to the growth of prostate cancer via influencing ELK4 levels.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , ets-Domain Protein Elk-4/metabolism , Androgens/physiology , Cell Line, Tumor , Humans , Male , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , ets-Domain Protein Elk-4/biosynthesis , ets-Domain Protein Elk-4/genetics
3.
Biochem Soc Trans ; 35(Pt 6): 1405-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18031232

ABSTRACT

Transcriptional activity of signal-dependent transcription factors, including nuclear receptors, relies on interacting co-regulator proteins, many of which possess protein-modifying activity. SUMOs (small ubiquitin-related modifiers) and their conjugation pathway components act as co-regulator proteins for numerous transcription factors that also are often targets for SUMO modification. PIAS [protein inhibitor of activated STAT (signal transducer and activator of transcription)] proteins promote SUMOylation in a manner that resembles the action of RING-type ubiquitin E3 ligases. PIAS proteins were initially named for their ability to interact with STAT proteins and inhibit their activity, but their interactions and functions are not restricted to the STATs. Moreover, PIAS proteins do not operate merely as SUMO E3s, since their co-regulator effects are often independent of their RING finger but dependent on their SIM (SUMO-interacting motif) or SAP (scaffold attachment factor-A/B/acinus/PIAS) domain capable of interacting with DNA. The modulator activity imparted by the PIAS/SUMO system involves altered subnuclear targeting and/or assembly of transcription complexes. PIAS proteins may act as platforms that facilitate both removal and recruitment of other regulatory proteins in the transcription complexes.


Subject(s)
Protein Inhibitors of Activated STAT/physiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Protein Inhibitors of Activated STAT/chemistry , STAT Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/physiology , Ubiquitin-Protein Ligases/metabolism
4.
J Mol Endocrinol ; 34(3): 645-54, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15956336

ABSTRACT

PIASx belongs to the PIAS protein family, the members of which modulate activities of several transcription factors and act as E3 ligases in the sumoylation pathway. The PIASx gene is highly expressed in testis, suggesting a role in spermatogenesis. To investigate the function of PIASx in vivo, we have disrupted the PIASx gene in mice. Interestingly, the knockout mice were viable and fertile. Despite the normal fertility, the testis weight of the mutant animals was reduced and their number of apoptotic testicular cells was increased. Also, the sperm count of mutant mice tended to be reduced, but the quality of their sperm cells was normal. No significant changes were observed in the serum levels of LH and FSH or in the intratesticular testosterone concentration between the knockout animals and their wild-type littermates. Compensatory increases in other PIAS protein mRNAs were not observed in the knockout mice. These results imply that PIASx is required quantitatively rather than qualitatively for normal spermatogenesis.


Subject(s)
Ligases/genetics , Organ Size , Small Ubiquitin-Related Modifier Proteins/genetics , Testis/metabolism , Animals , Apoptosis , Base Sequence , DNA Primers , Immunoblotting , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Protein Inhibitors of Activated STAT , RNA, Messenger/genetics , Testis/pathology , Ubiquitin-Protein Ligases
5.
FEBS Lett ; 560(1-3): 56-62, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-14987998

ABSTRACT

SNURF/RNF4 has been implicated in transcriptional regulation and growth inhibition in a RING finger-dependent fashion. In this work, we show that SNURF mediates its own ubiquitination in vitro in a ubiquitin-conjugating enzyme (E2)-selective manner: SNURF acts as an E3 ligase with UbcH5A and B, HHR6B (RAD6B), E2-25K, MmUbc7 and UbcH13, but not with UbcH3, UbcM4, MmUbc6 or E2-20K. In contrast, the well-characterized RING E3, AO7, functions only with members of the UbcH5 family. Furthermore, depending on the E2 used, the ubiquitin modification manifests as mono- or multi-ubiquitination. Mutation of conserved cysteine residues within the RING finger motif of SNURF abolishes the ubiquitination in vitro and in intact cells. Size fractionation of murine embryonal carcinoma F9 cell proteins shows that the majority of endogenous SNURF resides in salt-resistant > or =500-kDa complexes, suggesting that SNURF functions as a RING component in a multiprotein complex. Taken together, SNURF/RNF4 functions as an E3 ligase and this activity is closely linked to its transcription regulatory functions.


Subject(s)
Ligases/genetics , Ligases/metabolism , Transcription, Genetic , Ubiquitin/metabolism , Amino Acid Motifs , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Chromatography, Gel , Cysteine/metabolism , Glutathione/metabolism , Mice , Point Mutation , Precipitin Tests , Recombinant Fusion Proteins/metabolism , Teratocarcinoma , Transfection , Ubiquitin-Protein Ligases
6.
FEBS Lett ; 523(1-3): 43-7, 2002 Jul 17.
Article in English | MEDLINE | ID: mdl-12123801

ABSTRACT

We have used confocal microscopy to elucidate the effects of antiandrogens on nuclear localization of the androgen receptor (AR) with its transcriptional coactivator GRIP1. We show that the agonist-activated AR recruits GRIP1 to colocalize with the receptor in the nucleoplasm. By contrast, AR complexed to the antiandrogens hydroxyflutamide and bicalutamide fails to influence nuclear distribution of GRIP1. Likewise, the non-steroidal antiandrogens prevent the agonist-induced AR-GRIP1 colocalization from occurring. Androgen antagonists affect nuclear redistribution of AR-GRIP1 in a fashion that parallels their effects on the transcriptional activity of AR, in that the pure antagonists block GRIP1-dependent activation of AR function, whereas the mixed antagonist/agonist cyproterone acetate promotes both AR-driven redistribution of GRIP1 and activation of AR by GRIP1.


Subject(s)
Androgen Antagonists/pharmacology , Cell Nucleus/drug effects , Flutamide/analogs & derivatives , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Androgens/metabolism , Anilides/pharmacology , Animals , COS Cells/metabolism , Cell Nucleus/metabolism , Chlorocebus aethiops , Cyproterone Acetate/pharmacology , Flutamide/pharmacology , Nitriles , Nuclear Receptor Coactivator 2 , Subcellular Fractions/metabolism , Tosyl Compounds , Transcription Factors/physiology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology
7.
Mol Endocrinol ; 15(5): 797-811, 2001 May.
Article in English | MEDLINE | ID: mdl-11328859

ABSTRACT

The androgen receptor (AR) is a ligand-regulated member of the nuclear receptor superfamily. The cyclin D1 gene product, which encodes the regulatory subunit of holoenzymes that phosphorylate the retinoblastoma protein (pRB), promotes cellular proliferation and inhibits cellular differentiation in several different cell types. Herein the cyclin D1 gene product inhibited ligand-induced AR- enhancer function through a pRB-independent mechanism requiring the cyclin D1 carboxyl terminus. The histone acetyltransferase activity of P/CAF (p300/CBP associated factor) rescued cyclin D1-mediated AR trans-repression. Cyclin D1 and the AR both bound to similar domains of P/CAF, and cyclin D1 displaced binding of the AR to P/CAF in vitro. These studies suggest cyclin D1 binding to the AR may repress ligand-dependent AR activity by directly competing for P/CAF binding.


Subject(s)
Acetyltransferases/physiology , Androgen Receptor Antagonists , Cell Cycle Proteins/physiology , Cyclin D1/physiology , Signal Transduction/physiology , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/metabolism , Amino Acid Sequence , Blotting, Western , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , Histone Acetyltransferases , Humans , Ligands , Male , Molecular Sequence Data , Mutation , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Receptors, Androgen/physiology , Sequence Alignment , Transcription Factors , Tumor Cells, Cultured , p300-CBP Transcription Factors
8.
Mol Endocrinol ; 15(6): 923-35, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11376111

ABSTRACT

We have compared the functional consequences of seven single-point mutations in the ligand-binding domain (LBD) of the androgen receptor (AR). The mutations span helices 3 to 11 and are present in patients suffering from androgen insensitivity syndromes (AIS) and other male-specific disorders. The mutants, except M742V, bound to androgen response elements in vivo and in vitro and showed a testosterone-dependent conformational change. With regard to functional activity, the mutant M742V had severely blunted ability to transactivate or exhibit the androgen-dependent amino/carboxyl-terminal (N/C) interaction; mutants F725L, G743V, and F754L showed reduced transactivation potential and attenuated N/C interaction; and mutants V715M, R726L, and M886V had minor functional impairments. The mutants belonging to the first two groups also displayed reduced response to coexpressed GRIP1. In addition, mutations of amino acids M894 and A896 in the putative core activation domain 2 (AF2) in helix 12 confirmed that this helix is important for N/C interactions. Thus, amino acids located between helices 3 and 4 (F725 and R726), in helix 5 (M742, G743, and F754), and in helix 12 (M894 and A896) play critical roles in mediating the N/C interaction of AR. The data also show that disrupted N/C interaction is a potential molecular abnormality in AIS cases in which LBD mutations have not resulted in markedly impaired ability to bind androgen.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Androgens/pharmacology , Point Mutation , Receptors, Androgen/metabolism , Transcriptional Activation , Amino Acid Sequence , Androgens/metabolism , Animals , Binding Sites , COS Cells , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genes, Reporter , Humans , Immunoblotting , Male , Molecular Sequence Data , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Receptor Coactivator 2 , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Transcription Factor RelA , Transcription Factors/metabolism , Transfection
9.
J Biol Chem ; 276(26): 23653-60, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11319220

ABSTRACT

The small nuclear C(3)HC(4) finger protein (SNURF), RNF4, acts as transcriptional coactivator for both steroid-dependent and -independent promoters such as those driven by androgen response elements and GC boxes, respectively. However, SNURF does not possess intrinsic transcription activation function, and the precise molecular mechanism of its action is unknown. We have studied herein the interaction of SNURF with DNA in vitro. SNURF binds to linear double-stranded DNA with no apparent sequence specificity in a cooperative fashion that is highly dependent on the length of the DNA fragment used. SNURF interacts efficiently with both supercoiled circular and four-way junction DNA, and importantly, it also recognizes nucleosomes. An intact RING structure of SNURF is not mandatory for DNA binding, whereas mutations of specific positively charged residues in the N terminus (amino acids 8-11) abolish DNA binding. Interestingly, the ability of SNURF to interact with DNA is associated with its capability to enhance transcription from promoters containing GC box elements. Because SNURF can interact with both DNA and protein (transcription) factors, it may promote assembly of nucleoprotein structures.


Subject(s)
Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , Amino Acid Sequence , Animals , COS Cells , DNA/metabolism , DNA, Superhelical/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Nucleosomes/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics , Transcriptional Activation , Transfection
10.
J Clin Endocrinol Metab ; 86(4): 1539-44, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11297580

ABSTRACT

We have developed a mammalian cell (COS-1) bioassay, which can measure androgen bioactivity directly from a small amount (10 microL) of human serum. The recombinant assay is based on androgen-dependent interaction between the ligand-binding domain and the N-terminal region of the androgen receptor (AR), which were fused to Gal4 DNA-binding domain of Saccharomyces cerevisiae and transcriptional activation domain of herpes simplex VP16 protein, respectively. The interaction is amplified by coexpression of AR-interacting protein 3 in the cells. The reporter plasmid contains 5 Gal4-binding sites upstream of the luciferase gene; luciferase activity in cell lysates is derived from androgen bioactivity in human serum. Saturating concentration of testosterone in FCS induced more than 700-fold induction in relative luciferase activity. The sensitivity was less than 1.0 nmol/L testosterone in FCS. The intra- and interassay coefficients of variation were 8.3% and 21%, respectively. Interaction between the AR termini was blocked by nonsteroidal antiandrogens, and the assay exhibited minimal cross-reactivity with 17 beta-estradiol. Serum androgen bioactivity was studied in 23 boys (13.9--16.8 yr old) with constitutional delay of puberty and in 9 prepubertal boys with cryptorchidism (1.0--6.4 yr old). Androgen bioactivity was detectable in 15 boys with constitutional delay of puberty and in all boys with cryptorchidism during treatment with human CG (range, 1.0-14.5 nmol/L testosterone equivalents). Serum androgen bioactivity measured by the bioassay correlated strongly with serum testosterone concentration (r = 0.93, P < 0.0001, n = 22) but not to 5 alpha-dihydrotestosterone, dehydroepiandrosterone, or androstenedione levels. We conclude that our novel bioassay enables quantitation of mammalian cell response to bioactive androgens in human serum, even in pediatric patients with relatively low androgen levels.


Subject(s)
Androgens/blood , Biological Assay/methods , Adolescent , Androgen Antagonists/pharmacology , Androgens/physiology , Animals , COS Cells , Child , Child, Preschool , Cryptorchidism/blood , Estrogens/physiology , Ether , Gonadal Steroid Hormones/pharmacology , Humans , Infant , Male , Sensitivity and Specificity , Testosterone/pharmacology
11.
Mol Endocrinol ; 14(12): 1986-2000, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11117529

ABSTRACT

Steroid receptors mediate their actions by using various coregulatory proteins. We have recently characterized ARIP3/PIASx alpha as an androgen receptor (AR)-interacting protein (ARIP) that belongs to the PIAS [protein inhibitor of activated STAT (signal transducer and activator of transcription)] protein family implicated in the inhibition of cytokine signaling. We have analyzed herein the roles that four different PIAS proteins (ARIP3/PIASx alpha, Miz1/PIASx beta, GBP/PIAS1, and PIAS3) play in the regulation of steroid receptor- or STAT-mediated transcriptional activation. All PIAS proteins are able to coactivate steroid receptor-dependent transcription but to a differential degree, depending on the receptor, the promoter, and the cell type. Miz1 and PIAS1 are more potent than ARIP3 in activating AR function on minimal promoters. With the natural probasin promoter, PIAS proteins influence AR function more divergently, in that ARIP3 represses, but Miz1 and PIAS1 activate it. Miz1 and PIAS1 possess inherent transcription activating function, whereas ARIP3 and PIAS3 are devoid of this feature. ARIP3 enhances glucocorticoid receptor-dependent transcription more efficiently than Miz1 or PIAS1, and all PIAS proteins also activate estrogen receptor- and progesterone receptor-dependent transcription but to a dissimilar degree. The same amounts of PIAS proteins that modulate steroid receptor-dependent transcription influence only marginally transactivation mediated by various STAT proteins. It remains to be established whether the PIAS proteins play a more significant physiological role in steroid receptor than in cytokine signaling.


Subject(s)
Carrier Proteins/physiology , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Proteins/physiology , Receptors, Steroid/metabolism , Transcription Factors/physiology , Transcriptional Activation , Amino Acid Sequence , Carrier Proteins/genetics , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Inhibitors of Activated STAT , Proteins/genetics , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/physiology , STAT1 Transcription Factor , Sequence Homology, Amino Acid , Trans-Activators/genetics , Trans-Activators/metabolism , Trans-Activators/physiology , Transcription Factors/genetics , Ubiquitin-Protein Ligases
12.
Proc Natl Acad Sci U S A ; 97(26): 14145-50, 2000 Dec 19.
Article in English | MEDLINE | ID: mdl-11121022

ABSTRACT

Modification by SUMO-1 is proposed to play a role in protein targeting and/or stability. The SUMO-1-conjugating enzyme Ubc9 interacts with androgen receptor (AR), a ligand-activated transcription factor belonging to the steroid receptor superfamily. We show here that AR is covalently modified by SUMO-1 (sumoylated) in an androgen-enhanced fashion and identify the principal acceptor site in the N-terminal domain of AR. Substitutions of sumoylated Lys residues enhanced transcriptional activity of AR without influencing its transrepressing activity. Interestingly, the same Lys residues form the cores of the recently described transcriptional synergy control motifs in AR [Iñiguez-Lluhi, J. A. & Pearce, D. (2000) Mol. Cell. Biol. 20, 6040-6050]. These motifs, which match perfectly with the sumoylation consensus sequence, are also present in the N-terminal domains of glucocorticoid, mineralocorticoid, and progesterone receptor. Taken together, our data suggest that reversible sumoylation is a mechanism for regulation of steroid receptor function.


Subject(s)
Receptors, Androgen/metabolism , Ubiquitin-Conjugating Enzymes , Ubiquitins/metabolism , Animals , Binding Sites , COS Cells , Catalysis , Chlorocebus aethiops , HeLa Cells , Humans , Ligases/metabolism , Receptors, Androgen/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SUMO-1 Protein , Transcription, Genetic , Ubiquitins/genetics
13.
FEBS Lett ; 480(2-3): 84-8, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-11034305

ABSTRACT

To study the regulation of the human xanthine oxidoreductase (XOR) gene, we cloned 1.9 kb of the promoter region. In reporter gene assays, a construct encompassing nucleotides between 142 to +42 conferred maximal basal activity of the XOR promoter in 293T cells, in comparison with shorter (-92 to +42) or longer (up to -1937 to +42) constructs. The promoter activity was low in NIH-3T3 cells. The most active construct contained a putative CCAAT motif at -119 to -123. Electrophoretic mobility shift assays showed that this sequence binds the ubiquitous nuclear factor Y (NF-Y). Mutation of the CCAAT motif (CTGAT) abolished the NF-Y binding and considerably reduced the promoter activity. Our data suggest an important functional role for NF-Y in the transcriptional activation of the human XOR gene.


Subject(s)
CCAAT-Binding Factor/metabolism , Gene Expression Regulation, Enzymologic , Promoter Regions, Genetic , Transcription Factors/metabolism , Xanthine Dehydrogenase/genetics , Xanthine Oxidase/genetics , 3T3 Cells , Animals , Base Sequence , Binding Sites , Cell Line, Transformed , DNA/metabolism , Humans , Mice , Molecular Sequence Data , Rats
14.
Biochem Soc Trans ; 28(4): 401-5, 2000.
Article in English | MEDLINE | ID: mdl-10961928

ABSTRACT

Androgen receptor (AR) belongs to the superfamily of nuclear hormone receptors that employ complex molecular mechanisms to guide the development and physiological functions of their target tissues. Our recent work has led to the identification of four novel proteins that recognize AR zinc-finger region (ZFR) both in vivo and in vitro. One is a small nuclear RING-finger protein that possesses separate interaction interfaces for AR and for other transcription activators such as Sp1. The second is a nuclear serine/threonine protein kinase (androgen-receptor-interacting nuclear protein kinase; ANPK); however, the receptor itself does not seem to be a substrate for this kinase. The third one is dubbed androgen-receptor-interacting protein 3 (ARIP3) and is a novel member of the PIAS (protein inhibitor of activated STAT) protein family. The fourth protein, termed ARIP4, is a nuclear ATPase that belongs to the SNF2-like family of chromatin remodelling proteins. All four proteins exhibit a punctate nuclear pattern when expressed in cultured cells. Each protein modulates AR-dependent transactivation in co-transfection experiments; their activating functions are not restricted to AR. Current work is aimed at elucidating the biochemical and functional properties of these AR-interacting proteins and at finding the partner proteins that form complexes with them in vivo.


Subject(s)
Cell Nucleus/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Adenosine Triphosphatases/metabolism , Animals , Carrier Proteins/metabolism , Chromatin/metabolism , DNA Helicases , DNA-Binding Proteins/metabolism , Down-Regulation , Gene Expression Regulation , Humans , Male , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Protein Inhibitors of Activated STAT , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Ubiquitin-Protein Ligases , X Chromosome , Zinc Fingers
15.
J Cell Sci ; 113 ( Pt 17): 2991-3001, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10934038

ABSTRACT

The androgen receptor (AR) is a transcription factor that mediates androgen action. We have used the green fluorescent protein (GFP) technique to investigate dynamics of nuclear trafficking of human AR in living cells. In the absence of ligand, the GFP-AR fusion protein is distributed between cytoplasm and nuclei. Androgen exposure leads to a rapid and complete import of GFP-AR to nuclei of CV-1 cells (>=90% nuclear in 30 minutes), whereas a pure antiandrogen, Casodex, elicits a slower (<40% nuclear in 30 minutes) and incomplete transfer. Unliganded ARs with mutations in the basic amino acids of the bipartite nuclear localization signal (NLS) within the second zinc finger and the hinge region are predominantly cytoplasmic and their androgen-dependent nuclear import is severely compromised ((3/4)20% nuclear in 30 minutes). Interestingly, substitutions of the Leu residues flanking the bipartite NLS lead to inefficient nuclear transfer in response to androgen ((3/4)20% nuclear in 30 minutes). The ligand-binding domain of AR, which represses bipartite NLS activity, contains an agonist-specific NLS. The small nuclear RING finger protein SNURF, which interacts with AR through a region overlapping with the bipartite NLS, facilitates AR import to nuclei and retards its export on hormone withdrawal. More AR is associated with the nuclear matrix in the presence than absence of coexpressed SNURF. We suggest that the SNURF-mediated tethering of AR in nuclei represents a novel mechanism for activating steroid receptor functions.


Subject(s)
Cell Nucleus/metabolism , Nuclear Proteins/physiology , Receptors, Androgen/metabolism , Transcription Factors/physiology , Amino Acid Sequence , Androgen Receptor Antagonists , Androgens/metabolism , Animals , Biological Transport , COS Cells , Cell Line , Green Fluorescent Proteins , Humans , Indicators and Reagents/pharmacology , Leucine/genetics , Luminescent Proteins/genetics , Luminescent Proteins/pharmacology , Molecular Sequence Data , Nuclear Localization Signals/genetics , Nuclear Matrix/metabolism , Nuclear Proteins/genetics , Point Mutation , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Transcription Factors/genetics , Transcription, Genetic , Transfection , Zinc Fingers/genetics
16.
J Immunol ; 164(11): 5689-97, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10820245

ABSTRACT

IFN-gamma and glucocorticoids regulate inflammatory and immune responses through Stat1 and glucocorticoid receptor (GR) transcription factors, respectively. The biological responses to these polypeptides are determined by integration of various signaling pathways in a cell-type and promoter-dependent manner. In this study we have characterized the molecular basis for the functional cooperation between IFN-gamma and dexamethasone (Dex) in the induction of the high-affinity Fc gamma receptor I (Fc gamma RI) in monocytes. Dex did not affect IFN-gamma-induced Stat1 DNA binding activity or induce novel DNA-binding complexes to the Fc gamma RI promoter. By using cell systems lacking functional GR or Stat1, we showed that GR stimulated Stat1-dependent transcription in a ligand-dependent manner, while Stat1 did not influence GR-dependent transcription. The cooperation required phosphorylation of Tyr701, DNA binding, and the trans-activation domain of Stat1, but did not involve Ser727 phosphorylation of Stat1 or physical interaction between GR and Stat1. The costimulatory effect of Dex was not dependent on a consensus glucocorticoid response element in the Stat1-responsive promoters, but required the DNA-binding and trans-activation functions of GR, and Dex-induced protein synthesis. GR activated the natural Fc gamma RI promoter construct, and this response required both Stat1 and the Ets family transcription factor PU.1. Previously, physical association between GR and Stat5 has been shown to enhance Stat5-dependent and suppress GR-dependent transcription. The results shown here demonstrate a distinct, indirect mechanism of cross-modulation between cytokine and steroid receptor signaling that integrates Stat1 and GR pathways with cell type-specific PU.1 transcription factor in the regulation of Fc gamma RI gene transcription.


Subject(s)
DNA-Binding Proteins/physiology , Milk Proteins , Monocytes/metabolism , Proto-Oncogene Proteins/physiology , Receptors, Glucocorticoid/physiology , Receptors, IgG/metabolism , Trans-Activators/physiology , Transcriptional Activation/immunology , DNA/metabolism , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Drug Synergism , Humans , Interferon-gamma/physiology , Monocytes/drug effects , Promoter Regions, Genetic/immunology , Protein Binding/drug effects , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Glucocorticoid/metabolism , Receptors, IgG/biosynthesis , Receptors, IgG/genetics , STAT1 Transcription Factor , STAT5 Transcription Factor , Signal Transduction/immunology , Trans-Activators/metabolism , Tumor Cells, Cultured
17.
Am J Hum Genet ; 66(2): 378-92, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10677297

ABSTRACT

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is a monogenic autosomal disease with recessive inheritance. It is characterized by multiple autoimmune endocrinopathies, chronic mucocutaneous candidiasis, and ectodermal dystrophies. The defective gene responsible for this disease was recently isolated, and several different mutations in the novel gene, AIRE, have been identified, by us and by others, in patients with APECED. We have shown that the APECED protein is mainly localized, both in vitro and in vivo, to the cell nucleus, where it forms distinct speckles. This accords with the predicted structural features of the protein, which suggest involvement of AIRE in the regulation of gene transcription. Here, we report the results of mutational analyses of a series of 112 patients with APECED who were from various ethnic backgrounds. A total of 16 different mutations, covering 91% of disease alleles, were observed; of these, 8 were novel. The mutations are spread throughout the coding region of AIRE, yet four evident mutational hotspots were observed. In vitro expression of four different naturally occurring nonsense and missense mutations revealed a dramatically altered subcellular location of the protein in cultured cells. Interestingly, the wild-type APECED protein tethered to the Gal4 DNA-binding domain acted as a strong transcriptional activator of reporter genes in mammalian cells, whereas most of the analyzed mutant polypeptides had lost this capacity.


Subject(s)
Mutation/genetics , Polyendocrinopathies, Autoimmune/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Alleles , Animals , Biological Transport , Cell Line , Codon, Nonsense/genetics , Cytoplasm/metabolism , Ethnicity/genetics , Exons/genetics , Female , Genes, Reporter/genetics , Haplotypes/genetics , Humans , Immunohistochemistry , Male , Molecular Sequence Data , Mutation, Missense/genetics , Polyendocrinopathies, Autoimmune/metabolism , Polymorphism, Single-Stranded Conformational , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/chemistry , AIRE Protein
18.
Gene ; 242(1-2): 65-73, 2000 Jan 25.
Article in English | MEDLINE | ID: mdl-10721698

ABSTRACT

Mutations in the gene encoding cystatin B (CSTB) are responsible for the primary defect in progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1). A novel and unique type of disease-causing mutation, an unstable dodecamer repeat expansion, accounts for the majority of EPM1 patients world-wide. This minisatellite repeat expansion, located in the putative promoter of CSTB 175 bp upstream from the translation initiation codon, appears to downregulate CSTB gene expression in vivo. We report here the characterization of the CSTB promoter using different promoter-luciferase gene constructs. Transient transfections of cultured mammalian cells suggest that the region from -670 to -1 bp from the translation initiation codon functions as the CSTB promoter. Active binding to five Sp1 and four AP1 sites as well as weak binding to an androgen response element (ARE) half site was demonstrated by electrophoretic mobility shift assays. The effect of the minisatellite expansion on the promoter activity was evaluated by comparing the activity of constructs containing wild-type and expanded alleles. An increase in the number of dodecamer units from three to 19 repeats lowered transcription in vitro by 10-fold. Northern analysis of lymphoblastoid RNA from individuals with 'premutation' length dodecamer repeat (12-17 copies) expansions showed decreased levels of CSTB mRNA expression. These data indicate that expansion of the dodecamer repeat located in the proximal promoter of CSTB severely disrupts the function of the promoter and thereby reduces transcription of CSTB.


Subject(s)
Cystatins/genetics , Myoclonic Epilepsies, Progressive/genetics , Promoter Regions, Genetic/genetics , Animals , Base Sequence , COS Cells , Cystatin B , DNA/chemistry , DNA/genetics , DNA/metabolism , Gene Expression Regulation , Humans , Luciferases/genetics , Luciferases/metabolism , Minisatellite Repeats , Molecular Sequence Data , Protein Binding , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Repetitive Sequences, Nucleic Acid , Sequence Analysis, DNA , Sp1 Transcription Factor/metabolism
19.
J Biol Chem ; 275(1): 571-9, 2000 Jan 07.
Article in English | MEDLINE | ID: mdl-10617653

ABSTRACT

The small nuclear RING finger protein SNURF is not only a coactivator in steroid receptor-dependent transcription but also activates transcription from steroid-independent promoters. In this work, we show that SNURF, via the RING finger domain, enhances protein binding to Sp1 elements/GC boxes and interacts and cooperates with Sp1 in transcriptional activation. The activation of androgen receptor (AR) function requires regions other than the RING finger of SNURF, and SNURF does not influence binding of AR to cognate DNA elements. The zinc finger region (ZFR) together with the hinge region of AR are sufficient for contacting SNURF. The nuclear localization signal in the boundary between ZFR and the hinge region participates in the association of AR with SNURF, and a receptor mutant lacking the C-terminal part of the bipartite nuclear localization signal shows attenuated response to coexpressed SNURF. Some AR ZFR point mutations observed in patients with partial androgen insensitivity syndrome or male breast cancer impair the interaction of AR with SNURF and also render AR refractory to the transcription-activating effect of SNURF. Collectively, SNURF modulates the transcriptional activities of androgen receptor and Sp1 via different domains, and it may act as a functional link between steroid- and Sp1-regulated transcription.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Androgen/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Zinc Fingers , Amino Acid Sequence , Androgen-Insensitivity Syndrome/genetics , Breast Neoplasms, Male/genetics , Gene Expression Regulation , Humans , Male , Molecular Sequence Data , Nuclear Localization Signals , Nuclear Proteins/genetics , Point Mutation , Promoter Regions, Genetic , Protein Binding , Recombinant Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic , Zinc Fingers/genetics
20.
J Biol Chem ; 274(27): 19441-6, 1999 Jul 02.
Article in English | MEDLINE | ID: mdl-10383460

ABSTRACT

Ubc9, a homologue of the class E2 ubiquitin-conjugating enzymes, has recently been shown to catalyze conjugation of a small ubiquitin-like molecule-1 (SUMO-1) to a variety of target proteins. SUMO-1 modifications have been implicated in the targeting of proteins to the nuclear envelope and certain intranuclear structures and in converting proteins resistant to ubiquitin-mediated degradation. In the present work, we find that Ubc9 interacts with the androgen receptor (AR), a member of the steroid receptor family of ligand-activated transcription factors. In transiently transfected COS-1 cells, AR-dependent but not basal transcription is enhanced by the coexpression of Ubc9. The N-terminal half of the AR hinge region containing the C-terminal part of the bipartite nuclear localization signal is essential for the interaction with Ubc9. Deletion of this part of the nuclear localization signal, which does not completely prevent the transfer of AR to the nucleus, abolishes the AR-Ubc9 interaction and attenuates the transcriptional response to cotransfected Ubc9. The C93S substitution of Ubc9, which prevents SUMO-1 conjugation by abrogating the formation of a thiolester bond between SUMO-1 and Ubc9, does not influence the capability of Ubc9 to stimulate AR-dependent transactivation, implying that Ubc9 is able to act as an AR coregulator in a fashion independent of its ability to catalyze SUMO-1 conjugation.


Subject(s)
Ligases/metabolism , Receptors, Androgen/metabolism , Transcription, Genetic , Ubiquitin-Conjugating Enzymes , Amino Acid Sequence , Animals , Binding Sites , COS Cells , DNA/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Receptors, Androgen/genetics , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , SUMO-1 Protein , Ubiquitins/metabolism , Yeasts
SELECTION OF CITATIONS
SEARCH DETAIL
...