Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Mol Cell Endocrinol ; 265-266: 157-61, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17207919

ABSTRACT

Steroid receptors (SR), which are ligand activated transcription factors, and their coactivators are phosphoproteins whose activities are regulated by cell signaling pathways. Many of the identified phosphorylation sites in these proteins contain Ser/Thr-Pro motifs suggesting that they are substrates for cyclin dependent kinases and/or for mitogen activated protein kinases. An analysis of the roles of cyclins and their kinases in regulating receptor action has revealed that there are both stimulatory and inhibitory actions of cyclins, that some of the actions are independent of the partner kinases and that these activities are receptor specific. Consistent with this finding, the limited analyses of receptor activity as a function of cell cycle reveal distinct patterns of activation. SR often regulate cell proliferation. Thus, the cross-talk between cyclins and their kinases and the SR provides a means for integrating the actions of the SR with the cell cycle status of cells.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Receptors, Steroid/metabolism , Cell Cycle , Cell Proliferation , Humans
2.
Bone ; 31(3): 381-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12231410

ABSTRACT

Skeletal unloading in an animal hindlimb suspension model and microgravity experienced by astronauts or as a result of prolonged bed rest causes site-specific losses in bone mineral density of 1%-2% per month. This is accompanied by reductions in circulating levels of 1,25-(OH)(2)D(3), the active metabolite of vitamin D. 1,25-(OH)(2)D(3), the ligand for the vitamin D receptor (VDR), is important for calcium absorption and plays a role in differentiation of osteoblasts and osteoclasts. To examine the responses of cells to activators of the VDR in a simulated microgravity environment, we used slow-turning lateral vessels (STLVs) in a rotating cell culture system. We found that, similar to cells grown in microgravity, MG-63 cells grown in the STLVs produce less osteocalcin, alkaline phosphatase, and collagen Ialpha1 mRNA and are less responsive to 1,25-(OH)(2)D(3). In addition, expression of VDR was reduced. Moreover, growth in the STLV caused activation of the stress-activated protein kinase pathway (SAPK), a kinase that inhibits VDR activity. In contrast, the 1,25-(OH)(2)D(3) analog, EB1089, was able to compensate for some of the STLV-associated responses by reducing SAPK activity, elevating VDR levels, and increasing expression of osteocalcin and alkaline phosphatase. These studies suggest that, not only does simulated microgravity reduce differentiation of MG-63 cells, but the activity of the VDR, an important regulator of bone metabolism, is reduced. Use of potent, less calcemic analogs of 1,25-(OH)(2)D(3) may aid in overcoming this defect.


Subject(s)
Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Gravitation , Receptors, Calcitriol/metabolism , Signal Transduction/drug effects , Cell Culture Techniques/methods , Humans , Signal Transduction/physiology , Tumor Cells, Cultured
3.
Prostate ; 49(3): 224-33, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11746268

ABSTRACT

BACKGROUND: We recently reported on novel vitamin D receptor (VDR) modulators that are structurally distinct from the secosteroid 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)), the endogenous activator of VDR. One of these compounds, LG190119, was tested for the ability to inhibit the growth of LNCaP human prostate cancer cell-derived tumors in athymic mice. METHODS: In one study, athymic mice with established LNCaP xenograft tumors were dosed orally every day with LG190119 (3 or 10 mg/kg) or with a synthetic analog of 1,25(OH)(2)D(3), EB1089 (1 microg/kg), for 15 days. In another study ("prevention mode"), oral administration (every other day) of 10 mg/kg LG190119 or a non-hypercalcemic dose of 1,25(OH)(2)D(3) (0.5 microg/kg) was initiated prior to tumor development and continued for 84 days. In both studies, tumor volumes, mouse weights, and serum calcium levels were measured. RESULTS: In the established tumor study, LG190119 at each dose resulted in significant tumor growth inhibition without hypercalcemia at both 10 and 15 days. EB1089 treatment resulted in significant tumor growth inhibition only at Day 10 and resulted in hypercalcemia at Day 15. In the prevention-mode study, LG190119 markedly slowed tumor growth without increased serum calcium in comparison with either vehicle or 1,25(OH)(2)D(3) treatment (P < 0.001). CONCLUSIONS: LG190119 effectively inhibited LNCaP xenograft tumor growth without increased serum calcium levels or any other apparent side effects. Compounds of this class may represent promising new therapeutics for treatment of prostate cancer and other cancers with fewer undesirable side effects than currently used drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Calcium/blood , Ketones/pharmacology , Prostatic Neoplasms/drug therapy , Receptors, Calcitriol/agonists , Animals , Body Weight , Cell Division/drug effects , Growth Inhibitors/pharmacology , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Prostatic Neoplasms/prevention & control , Random Allocation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
J Natl Cancer Inst ; 93(17): 1314-24, 2001 Sep 05.
Article in English | MEDLINE | ID: mdl-11535706

ABSTRACT

BACKGROUND: Adenovirus-mediated overexpression of the apoptosis-inducing protein Bax can induce apoptosis in prostate cancer cell lines. Constitutive overexpression of Bax could result in unwanted apoptosis in every site of accidental Bax accumulation in vivo. Therefore, we developed an adenoviral construct (Av-ARR2PB-Bax) in which the probasin promoter, modified to contain two androgen response elements, drives Bax expression. This promoter would be expected to limit expression of Bax to cells expressing the androgen receptor. METHODS: A variety of androgen receptor (AR)-positive and -negative cell lines of prostatic or nonprostatic origin were infected with Av-ARR2PB-Bax or a control virus, Av-ARR2PB-CAT, in which the same promoter drives expression of the chloramphenicol acetyl transferase-reporter gene. Bax expression and apoptosis in vitro were assessed by western blot analysis. Tumor size and apoptosis in vivo were assessed after four weekly injections of Av-ARR2PB-Bax or Av-ARR2PB-CAT into subcutaneous LNCaP xenografts growing in uncastrated male mice. All statistical tests were two-sided. RESULTS: Bax was overexpressed in an androgen-dependent way in AR-positive cell lines of prostatic origin but not in AR-positive cells of nonprostatic origin or in AR-negative cell lines of either prostatic or nonprostatic origin. The androgen dihydrotestosterone activated apoptosis in LNCaP cells infected with Av-ARR2PB-Bax but not in those infected with Av-ARR2PB-CAT. Av-ARR2PB-Bax-injected LNCaP xenograft tumors decreased in tumor size from 34.1 mm3 (95% confidence interval [CI] = 25.1 mm3 to 43.1 mm3) to 24.6 mm3 (95% CI = -2.5 mm3 to 51.7 mm3), but the difference was not statistically significant (P =.5). Tumors injected with Av-ARR2PB-CAT increased in size, from 28.9 mm3 (95% CI = 12.7 mm3 to 45.1 mm3) to 206 mm3 (95% CI = 122 mm3 to 290 mm3) (P =.002) and contained statistically significant more apoptotic cells (23.3% [95% CI = 21.1% to 25.6%] versus 9.5% [95% CI = 8.0% to 11.1]) (P<.001). CONCLUSIONS: Av-ARR2PB-Bax induces androgen-dependent therapeutic apoptosis in vitro and in vivo by activating apoptosis in AR-positive cells derived specifically from prostatic epithelium and does not affect nonprostatic cells.


Subject(s)
Androgen-Binding Protein/metabolism , Arabidopsis Proteins , DNA-Binding Proteins/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Adenoviridae , Androgen Antagonists/pharmacology , Androgen-Binding Protein/genetics , Animals , Apoptosis , Blotting, Western , Flutamide/analogs & derivatives , Flutamide/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Transplantation, Heterologous , Tumor Cells, Cultured , Up-Regulation , bcl-2-Associated X Protein
6.
J Biol Chem ; 276(11): 8475-83, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11110801

ABSTRACT

We have previously reported the identification of seven in vivo phosphorylation sites in the amino-terminal region of the human progesterone receptor (PR). From our previous in vivo studies, it was evident that several phosphopeptides remained unidentified. In particular, we wished to determine whether human PR contains a phosphorylation site in the hinge region, as do other steroid receptors including chicken PR, human androgen receptor, and mouse estrogen receptor. Previously, problematic trypsin cleavage sites hampered our ability to detect phosphorylation sites in large incomplete tryptic peptides. Using a combination of mass spectrometry and in vitro phosphorylation, we have identified six previously unidentified phosphorylation sites in human PR. Using nanoelectrospray ionization mass spectrometry, we have identified two new in vivo phosphorylation sites, Ser(20) and Ser(676), in baculovirus-expressed human PR. Ser(676) is analogous to the hinge site identified in other steroid receptors. Additionally, precursor ion scans identified another phosphopeptide that contains Ser(130)-Pro(131), a likely candidate for phosphorylation. In vitro phosphorylation of PR with Cdk2 has revealed five additional in vitro Cdk2 phosphorylation sites: Ser(25), Ser(213), Thr(430), Ser(554), and Ser(676). At least two of these, Ser(213) and Ser(676), are authentic in vivo sites. We confirmed the presence of the Cdk2-phosphorylated peptide containing Ser(213) in PR from in vivo labeled T47D cells, indicating that this is an in vivo site. Our combined studies indicate that most, if not all, of the Ser-Pro motifs in human PR are sites for phosphorylation. Taken together, these data indicate that the phosphorylation of PR is highly complex, with at least 14 phosphorylation sites.


Subject(s)
CDC2-CDC28 Kinases , Receptors, Progesterone/chemistry , Amino Acid Sequence , Animals , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/physiology , Humans , Mass Spectrometry , Molecular Sequence Data , Phosphopeptides/analysis , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Receptors, Progesterone/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spodoptera
7.
Mol Cell Biol ; 20(23): 8720-30, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11073973

ABSTRACT

Elevation of intracellular 8-bromo-cyclic AMP (cAMP) can activate certain steroid receptors and enhance the ligand-dependent activation of most receptors. During ligand-independent activation of the chicken progesterone receptor (cPR(A)) with the protein kinase A (PKA) activator, 8-bromo-cAMP, we found no alteration in cPR(A) phosphorylation (W. Bai, B. G. Rowan, V. E. Allgood, B. W. O'Malley, and N. L. Weigel, J. Biol. Chem. 272:10457-10463, 1997). To determine if other receptor-associated cofactors were targets of cAMP-dependent signaling pathways, we examined the phosphorylation of steroid receptor coactivator 1 (SRC-1). We detected a 1.8-fold increase in SRC-1 phosphorylation in transfected COS-1 cells incubated with 8-bromo-cAMP. Phosphorylation was increased on two mitogen-activated protein kinase (MAPK) sites, threonine 1179 and serine 1185. PKA did not phosphorylate these sites in vitro. However, blockage of PKA activity in COS-1 cells with the PKA inhibitor (PKI) prevented the 8-bromo-cAMP-mediated phosphorylation of these sites. Incubation of COS-1 cells with 8-bromo-cAMP resulted in activation of the MAPK pathway, as determined by Western blotting with antibodies to the phosphorylated (active) form of Erk-1/2, suggesting an indirect pathway to SRC-1 phosphorylation. Mutation of threonine 1179 and serine 1185 to alanine in COS-1 cells coexpressing cPR(A) and the GRE(2)E1bCAT reporter resulted in up to a 50% decrease in coactivation during both ligand-independent activation and ligand-dependent activation. This was due, in part, to loss of functional cooperation between SRC-1 and CREB binding protein for coactivation of cPR(A). This is the first demonstration of cross talk between a signaling pathway and specific phosphorylation sites in a nuclear receptor coactivator that can regulate steroid receptor activation.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Nuclear Proteins/metabolism , Receptors, Progesterone/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , CREB-Binding Protein , Chickens , Cyclic AMP-Dependent Protein Kinases/metabolism , Histone Acetyltransferases , Mitogen-Activated Protein Kinases/metabolism , Models, Genetic , Nuclear Receptor Coactivator 1 , Peptide Mapping , Phosphopeptides , Phosphorylation , Progesterone/pharmacology , Protein Binding , Recombinant Proteins/metabolism , Serine/metabolism , Signal Transduction , Threonine/metabolism , Transcription Factors/genetics , Transcriptional Activation
8.
Cancer Res ; 60(4): 779-82, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706079

ABSTRACT

Limited options for the treatment of prostate cancer have spurred the search for new therapies. One innovative approach is the use of 1alpha,25-dihydroxyvitamin D3 (calcitriol) analogues to inhibit cancer growth. We demonstrate here that the calcitriol analogue, EB1089, extensively inhibits the growth of LNCaP prostate cancer cells in culture and causes the cells to both accumulate in G0-G1 and undergo apoptosis. Importantly, we found that EB1089 inhibits the growth of LNCaP tumor xenografts in nude mice. Because of these antiproliferative properties in vivo, EB1089 is a potential new therapeutic agent for the treatment of prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Calcitriol/analogs & derivatives , Prostatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Calcitriol/therapeutic use , Calcium/blood , Cell Cycle/drug effects , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
9.
Cancer Res ; 60(4): 944-9, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706109

ABSTRACT

We analyzed the frequency and relevance of mutations in the coding region of the androgen receptor (AR) in genomic DNA extracted from 137 specimens of prostate cancer. The specimens were obtained from the primary tumors of patients affected by stage B disease [15 nonmicrodissected (group 1A) and 84 microdissected (group 1B)] and from the metastatic deposits of individuals with stage D1 disease [8 nonmicrodissected (group 2A) and 30 microdissected (group 2B)] who had not undergone androgen ablation therapy. The study was conducted by PCR-single strand conformational polymorphism (SSCP) analysis of exons 2-8 in the four groups and direct sequence analysis of exon 1 in group 1B. As positive and negative controls, we used genomic DNA extracted from genital skin fibroblasts of patients affected by various forms of androgen resistance with known mutations in the AR. To control for genetic instability, PCR-SSCP analysis of exon 2 of the human progesterone receptor was carried out on each specimen. The overall number of mutations detected was 11 (8%). No mutations were detected in any of the 99 patients with stage B disease. Eleven mutations were detected in exons 2-8 in 8 of the 38 patients with stage D1 disease (all in group 2B). Simultaneous analysis of exon 2 of the progesterone receptor was carried out, and no SSCP changes were identified. These data suggest that AR mutations are rare and presumably do not play a role in the initial phase of prostatic carcinogenesis. The presence of a significant number of AR mutations in metastatic disease indicates that mutations of this molecule may play a role in the most advanced phases of the natural history of this disease, either by facilitating growth or acquisition of the metastatic phenotype.


Subject(s)
Mutation , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Humans , Male , Neoplasm Metastasis , Neoplasm Staging , Polymorphism, Single-Stranded Conformational , Prostatic Neoplasms/pathology
10.
J Biol Chem ; 275(6): 4475-83, 2000 Feb 11.
Article in English | MEDLINE | ID: mdl-10660621

ABSTRACT

Steroid receptor coactivator-1 (SRC-1) is a member of a coactivator family that enhance the activation of the steroid/nuclear receptor superfamily of ligand-stimulated transcription factors. To study the regulation of SRC-1 by signaling pathways in the cell, the major phosphorylation sites of SRC-1 were identified in COS-1 cells using a combination of in vivo labeling with [(32)P]H(3)PO(4), modified manual Edman degradation, phosphoamino acid analysis, endoproteinase digestion, and mutagenesis of the SRC-1 phosphorylation sites. Seven phosphorylation sites were identified in SRC-1: serine 372, serine 395, serine 517, serine 569, serine 1033, threonine 1179, and serine 1185. All the sites contained consensus sequences for the serine/threonine-proline-directed family of protein kinases, and two sites (serine 395 and threonine 1179) contained a perfect consensus sequence for the mitogen-activated protein kinase family (Erk-1 and Erk-2). Furthermore, Erk-2 phosphorylated threonine 1179 and serine 1185 (and to a lesser extent, serine 395) in vitro, suggesting the importance of this pathway for SRC-1 regulation. Treatment of cells expressing SRC-1 with epidermal growth factor enhanced the ligand-dependent, progesterone receptor-mediated activation of a target reporter gene. These results identify phosphorylation as a regulatory modification of SRC-1 and provide a basis upon which to identify signaling pathways that regulate SRC-1 function and, consequently, modify steroid/nuclear receptor action.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Electrophoresis, Gel, Two-Dimensional , Epidermal Growth Factor/pharmacology , Genes, Reporter , Histone Acetyltransferases , Molecular Sequence Data , Nuclear Receptor Coactivator 1 , Phosphopeptides/chemistry , Phosphorylation , Progesterone/pharmacology , Receptors, Progesterone/metabolism , Signal Transduction , Transcription Factors/chemistry
11.
Mol Endocrinol ; 14(1): 52-65, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10628747

ABSTRACT

Human progesterone receptor (PR) is phosphorylated on multiple serine residues (at least seven sites) in a manner that involves distinct groups of sites coordinately regulated by hormone and different kinases. Progress on defining a functional role for PR phosphorylation has been hampered both by the complexity of phosphorylation and the lack of simple, nonradioactive methods to detect the influence of ligands and other signaling pathways on specific PR phosphorylation sites in vivo. Toward this end, we have produced monoclonal antibodies (MAbs) that recognize specific phosphorylation sites within human PR including a basal site at Ser 190 (MAb P190) and a hormone-induced site at Ser 294 (MAb P294). Biochemical experiments showed the differential reactivity of the P190 and P294 MAbs for phosphorylated and unphosphorylated forms of PR. Both MAbs recognize specific phosphorylated forms of PR under different experimental conditions including denatured PR protein by Western blots and PR in its native conformation in solution or complexed to specific target DNA. As detected by Western blot of T47D cells treated with hormone for different times, hormone-dependent down-regulation of total PR and the Ser 190 phosphorylation site occurred in parallel, whereas the Ser 294 phosphorylation site was down-regulated more rapidly. This difference in kinetics suggests that the Ser 294 site is more labile than basal sites and is acted upon by distinct phosphatases. A strong preferential hormone-dependent phosphorylation of Ser 294 was observed on PR-B as compared with the amino-terminal truncated A form of PR. This was unexpected because Ser 294 and flanking sequences are identical on both proteins, suggesting that a distinct conformation of the N-terminal domain of PR-A inhibits phosphorylation of this site. That Ser 294 lies within an inhibitory domain that mediates the unique repressive functions of PR-A raises the possibility that differential phosphorylation of Ser 294 is involved in the distinct functional properties of PR-A and PR-B.


Subject(s)
Antibodies, Monoclonal/metabolism , Phosphoserine/metabolism , Receptors, Progesterone/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , COS Cells , Humans , Kinetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phosphorylation , Precipitin Tests , Progesterone/antagonists & inhibitors , Progesterone Congeners/pharmacology , Promegestone/pharmacology , Protein Binding , Protein Conformation , Receptors, Progesterone/chemistry , Receptors, Progesterone/immunology , Serine/metabolism , Tumor Cells, Cultured
12.
Endocrinology ; 141(1): 10-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10614618

ABSTRACT

While the role of vitamin D in bone and mineral metabolism has been investigated extensively, the role of the vitamin D receptor in other tissues is less well understood. 1,25-Dihydroxyvitamin D3 (calcitriol) can act as a differentiating agent in normal tissues and can inhibit the growth of many cancer cell lines including LNCaP prostate cancer cells. We have shown previously that calcitriol causes LNCaP cell accumulation in the G0/G1 phase of the cell cycle. In this study, we demonstrate that calcitriol also induces apoptosis of LNCaP cells. The calcitriol-induced apoptosis is accompanied by a down-regulation of Bcl-2 and Bcl-X(L) proteins, both of which protect cells from undergoing apoptosis. Other proteins important in apoptotic control, Bax, Mcl-1, and Bcl-X(S), are unaffected by calcitriol treatment. We find that overexpression of Bcl-2 blocks calcitriol-induced apoptosis and reduces, but does not eliminate, calcitriol-induced growth inhibition. We conclude that both regulation of cell cycle and the apoptotic pathway are involved in calcitriol action in prostate cancer cells.


Subject(s)
Apoptosis , Calcitriol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Blotting, Western , Cell Cycle/drug effects , Cell Division/drug effects , DNA Fragmentation/drug effects , Down-Regulation/drug effects , Flow Cytometry , Fluorescent Antibody Technique, Direct , Humans , In Situ Nick-End Labeling , Ki-67 Antigen/biosynthesis , Male , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Cells, Cultured
13.
Hum Mol Genet ; 9(1): 133-44, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10587588

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is associated with an abnormal expansion of the (CAG)(n)repeat in the androgen receptor (AR) gene. Similar mutations have been reported in other proteins that cause neurodegenerative disorders. The CAG-coded elongated polyglutamine (polyGln) tracts induce the formation of neuronal intracellular aggregates. We have produced a model to study the effects of potentially 'neurotoxic' aggregates in SBMA using immortalized motoneuronal cells (NSC34) transfected with AR containing polyGln repeats of different sizes [(AR.Q(n = 0, 23 or 46)]. Using chimeras of AR.Q(n) and the green fluorescent protein (GFP), we have shown that aggregate formation occurs when the polyGln tract is elongated and AR is activated by androgens. In NSC34 cells co-expressing the AR with the polyGln of pathological length (AR.Q46) and the GFP we have noted the presence of several dystrophic neurites. Cell viability analyses have shown a reduced growth/survival rate in NSC34 expressing the AR.Q46, whereas testosterone treatment partially counteracted both cell death and the formation of dystrophic neurites. These observations indicate the lack of correlation between aggregate formation and cell survival, and suggest that neuronal degeneration in SBMA might be secondary to axonal/dendritic insults.


Subject(s)
Motor Neurons/pathology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Animals , Base Sequence , Cell Death/physiology , Cell Line , Cell Survival/genetics , Green Fluorescent Proteins , Hybrid Cells , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Molecular Sequence Data , Motor Neurons/cytology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Nerve Degeneration/genetics , Neuroblastoma/embryology , Neuroblastoma/pathology , Peptides/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spinal Cord/cytology , Spinal Cord/embryology
14.
Mol Endocrinol ; 13(12): 2065-75, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10598582

ABSTRACT

Androgen ablation therapy is a primary treatment for advanced prostate cancer, but tumors become refractive to therapy. Consequently, the role of the androgen receptors (ARs) and of mutations in the AR in prostate cancer has been a subject of much concern. In the course of analyzing tumors for mutations, we identified a somatic mutation that substitutes tyrosine for a cysteine at amino acid 619 (C619Y), which is near the cysteines that coordinate zinc in the DNA binding domain in the AR. The mutation was re-created in a wild-type expression vector and functional analyses carried out using transfection assays with androgen-responsive reporters. The mutant is transcriptionally inactive and unable to bind DNA. In response to ligand treatment, AR619Y localizes abnormally in numerous, well circumscribed predominantly nuclear aggregates in the nucleus and cytoplasm. Interestingly, these aggregates also contain the bulk of the coexpressed steroid receptor coactivator SRC-1, suggesting, in analogy to AR in spinal bulbar muscular atrophy, that this mutant may alter cellular physiology through sequestration of critical proteins. Although many inactivating mutations have been identified in androgen insensitivity syndrome patients, to our knowledge, this is the first characterization of an inactivating mutation identified in human prostate cancer.


Subject(s)
Mutation , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Transcription Factors/metabolism , Cell Nucleus/metabolism , Chromosome Mapping , Cysteine , Cytoplasm/metabolism , DNA/metabolism , HeLa Cells/metabolism , HeLa Cells/ultrastructure , Histone Acetyltransferases , Humans , Male , Middle Aged , Nuclear Receptor Coactivator 1 , Receptors, Androgen/analysis , Receptors, Androgen/metabolism , Response Elements , Transfection , Tyrosine , X Chromosome
15.
J Med Chem ; 42(11): 2021-34, 1999 Jun 03.
Article in English | MEDLINE | ID: mdl-10354410

ABSTRACT

We have synthesized several 7alpha-fluoro (F) and 7alpha-iodo (I) analogues of 5alpha-dihydrotestosterone (5alpha-DHT) and 19-nor-5alpha-dihydrotestosterone (5alpha-NDHT) and tested them for binding to the androgen receptor and for their biological activity in an in vitro assay with cells that have been engineered to respond to androgens. The relative binding affinity to the androgen receptor determined in competition assays showed that in the androstane series the fluoro steroids have the highest affinity and that F-17alpha-CH3-DHT (4) has a higher affinity than 5alpha-DHT. All other steroids were somewhat less potent than 5alpha-DHT with F-DHT (2) = I-17alpha-CH3-DHT (3) >/= F-NDHT (6) > F-17alpha-CH3-NDHT (8) = I-DHT (1) >/= I-NDHT (5) > I-17alpha-CH3-NDHT (7). The relative biological activity in cells transfected with the androgen receptor and an androgen responsive reporter gene is 4 >> 5alpha-DHT > 2 > 6 > 3 >/= 1 >/= 8 >/= 5 > 7. The iodinated compound, I-17alpha-CH3-DHT (3), with the highest binding activity was synthesized labeled with 125I and was shown to bind with high affinity, Ka = 1.9 x 10(10) L/mol, and low nonspecific binding to the androgen receptor in rat prostatic cytosol. However, when radiolabeled [125I]-17alpha-CH3-DHT ([125I]3) was injected into castrated male rats, it showed very poor androgen receptor-mediated uptake into the rat prostate. This was unexpected in light of its superior receptor binding properties and its protection by the 17alpha-methyl group from metabolic oxidation at C-17. However, the biological potency of I-17alpha-CH3-DHT (3) was not as high as would have been expected. When I-DHT (1) and I-17alpha-CH3-DHT (3) were incubated in aqueous media at 37 degrees C they rapidly decomposed, but they were stable at 0 degrees C. The fluorinated analogue 4 treated similarly at 37 degrees C was completely stable. The products of the decomposition reaction of I-DHT (1) at 37 degrees C were identified as iodide and principally 17beta-hydroxy-5alpha-androst-7-en-3-one. The temperature dependence of this elimination reaction explains the inconsistency between the high binding to the androgen receptor (measured at 0 degrees C) and the low biological activity, as well as the poor androgen receptor mediated concentration in vivo. The fluorinated analogue F-17alpha-CH3-DHT (4) has both high affinity for the androgen receptor and high stability in aqueous media. Of the compounds tested, 4 has the highest affinity for the androgen receptor as well as the highest androgenic activity. Thus it is likely that F-17alpha-CH3-DHT 4 labeled with 18F will be an excellent receptor-mediated diagnostic imaging agent.


Subject(s)
Dihydrotestosterone/analogs & derivatives , Radiopharmaceuticals/chemical synthesis , Receptors, Androgen/metabolism , Animals , Binding, Competitive , Cell Line , Dihydrotestosterone/chemical synthesis , Dihydrotestosterone/chemistry , Dihydrotestosterone/metabolism , Dihydrotestosterone/pharmacology , Drug Stability , Fluorine Radioisotopes , Haplorhini , Humans , In Vitro Techniques , Iodine Radioisotopes , Ligands , Male , Prostate/cytology , Prostate/metabolism , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacology , Rats , Rats, Sprague-Dawley , Solutions , Tissue Distribution , Transfection
16.
Proc Soc Exp Biol Med ; 221(2): 89-98, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10352118

ABSTRACT

Classically, the actions of vitamin D have been associated with bone and mineral metabolism. More recent studies have shown that vitamin D metabolites induce differentiation and/or inhibit cell proliferation of a number of malignant and nonmalignant cell types including prostate cancer cells. Epidemiological studies show correlations between the risk factors for prostate cancer and conditions that can result in decreased vitamin D levels. The active metabolite of vitamin D, 1,25-dihydroxyvitamin D3 (calcitriol), inhibits growth of both primary cultures of human prostate cancer cells and cancer cell lines, but the mechanism by which the cells are growth-inhibited has not been clearly defined. Initial studies suggest that calcitriol alters cell cycle progression and may also initiate apoptosis. One of the disadvantages of using vitamin D in vivo is side-effects such as hypercalcemia at doses above physiological levels. Analogs of calcitriol have been developed that have comparable or more potent antiproliferative effects but are less calcemic. Further research into the mechanisms of vitamin D action in prostate and identification of suitable analogs for use in vivo may lead to its use in the treatment or prevention of prostate cancer.


Subject(s)
Nutritional Status , Prostatic Neoplasms/prevention & control , Vitamin D/adverse effects , Calcitriol/analogs & derivatives , Humans , Hypercalcemia/chemically induced , Male
17.
Mol Cell ; 3(4): 521-6, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10230405

ABSTRACT

Steroidogenic factor 1 (SF-1) is an orphan nuclear receptor that serves as an essential regulator of many hormone-induced genes in the vertebrate endocrine system. The apparent absence of a SF-1 ligand prompted speculation that this receptor is regulated by alternative mechanisms involving signal transduction pathways. Here we show that maximal SF-1-mediated transcription and interaction with general nuclear receptor cofactors depends on phosphorylation of a single serine residue (Ser-203) located in a major activation domain (AF-1) of the protein. Moreover, phosphorylation-dependent SF-1 activation is likely mediated by the mitogen-activated protein kinase (MAPK) signaling pathway. We propose that this single modification of SF-1 and the subsequent recruitment of nuclear receptor cofactors couple extracellular signals to steroid and peptide hormone synthesis, thereby maintaining dynamic homeostatic responses in stress and reproduction.


Subject(s)
DNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/genetics , Epidermal Growth Factor/pharmacology , Fushi Tarazu Transcription Factors , Genes, Reporter , Homeodomain Proteins , Humans , Mutation , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 2 , Nuclear Receptor Coactivator 2 , Phosphorylation , Protein Processing, Post-Translational , Repressor Proteins/metabolism , Reproduction , Serine/metabolism , Steroidogenic Factor 1 , Stress, Physiological , Transcription Factors/genetics , Transcriptional Activation , Transfection , Tumor Cells, Cultured
18.
Hum Mol Genet ; 8(5): 731-41, 1999 May.
Article in English | MEDLINE | ID: mdl-10196362

ABSTRACT

Spinal bulbar muscular atrophy is a neurodegenerative disorder caused by a polyglutamine expansion in the androgen receptor (AR). We show in transiently transfected HeLa cells that an AR containing 48 glutamines (ARQ48) accumulates in a hormone-dependent manner in both cytoplasmic and nuclear aggregates. Electron microscopy reveals both types of aggregates to have a similar ultrastructure. ARQ48 aggregates sequester mitochondria and steroid receptor coactivator 1 and stain positively for NEDD8, Hsp70, Hsp90 and HDJ-2/HSDJ. Co-expression of HDJ-2/HSDJ significantly represses aggregate formation. ARQ48 aggregates also label with antibodies recognizing the PA700 proteasome caps but not 20S core particles. These results suggest that ARQ48 accumulates due to protein misfolding and a breakdown in proteolytic processing. Furthermore, the homeostatic disturbances associated with aggregate formation may affect normal cell function.


Subject(s)
Cysteine Endopeptidases/metabolism , Heat-Shock Proteins/metabolism , Multienzyme Complexes/metabolism , Peptides/metabolism , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Green Fluorescent Proteins , HSP40 Heat-Shock Proteins , HeLa Cells/drug effects , HeLa Cells/metabolism , Heat-Shock Proteins/genetics , Histone Acetyltransferases , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mitosis , NEDD8 Protein , Nuclear Receptor Coactivator 1 , Peptides/genetics , Proteasome Endopeptidase Complex , Receptors, Androgen/genetics , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism
19.
J Mol Med (Berl) ; 76(7): 469-79, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9660165

ABSTRACT

In addition to the conventional hormone-dependent regulation of the activity of steroid/thyroid receptor family members, many studies have shown that there is substantial cross-talk between signal transduction pathways and steroid receptors. In a number of cases the modulation of kinase/phosphatase activity in cells leads to activation of steroid receptors in the absence of hormone. This novel mechanism may not be ubiquitous as the glucocorticoid receptor appears to be refractory to activation in the absence of hormone. However, estrogen receptors, progesterone receptors, androgen receptors, retinoic acid receptors, retinoid X receptors, and vitamin D receptors all exhibit ligand-independent activation under appropriate conditions. Whether a steroid receptor responds to a signal by inducing transcription of a target gene in the absence of hormone depends upon the cell type, promoter, and activator. The mechanism(s) by which ligand-independent activation is induced is currently a subject of great interest. Because the signals that activate receptors induce protein phosphorylation, altered phosphorylation of the receptors, and/or proteins that associate with the receptors are likely to be key to ligand-independent activation. In the case of the estrogen receptor there is good evidence that altered receptor phosphorylation plays a role in ligand-independent activation. Other likely targets are proteins in the heat shock protein complexes, corepressors, and/or coactivators of steroid receptors.


Subject(s)
Receptors, Steroid/metabolism , Signal Transduction , Steroids/metabolism , Animals , Humans , Ligands
20.
Mol Cell Biol ; 18(3): 1369-78, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9488452

ABSTRACT

Previously, we defined a novel class of ligands for the human progesterone receptor (PR) which function as mixed agonists. These compounds induce a conformational change upon binding the receptor that is different from those induced by agonists and antagonists. This establishes a correlation between the structure of a ligand-receptor complex and its transcriptional activity. In an attempt to define the cellular components which distinguish between different ligand-induced PR conformations, we have determined, by using a mammalian two-hybrid assay, that the nuclear receptor corepressor (NCoR) and the silencing mediator for retinoid and thyroid hormone receptor (SMRT) differentially associate with PR depending upon the class of ligand bound to the receptor. Specifically, we observed that the corepressors preferentially associate with antagonist-occupied PR and that overexpression of these corepressors suppresses the partial agonist activity of antagonist-occupied PR. Binding studies performed in vitro, however, reveal that recombinant SMRT can interact with PR in a manner which is not influenced by the nature of the bound ligand. Thus, the inability of SMRT or NCoR to interact with agonist-activated PR when assayed in vivo may relate more to the increased affinity of PR for coactivators, with a subsequent displacement of corepressors, than to an inherent low affinity for the corepressor proteins. Previous work from other groups has shown that 8-bromo-cyclic AMP (8-bromo-cAMP) can convert the PR antagonist RU486 into an agonist and, additionally, can potentiate the transcriptional activity of agonist-bound PR. In this study, we show that exogenous expression of NCoR or SMRT suppresses all 8-bromo-cAMP-mediated potentiation of PR transcriptional activity. Further analysis revealed that 8-bromo-cAMP addition decreases the association of NCoR and SMRT with PR. Thus, we propose that 8-bromo-cAMP-mediated potentiation of PR transcriptional activity is due, at least in part, to a disruption of the interaction between PR and the corepressors NCoR and SMRT. Cumulatively, these results suggest that NCoR and SMRT expression may play a pivotal role in PR pharmacology.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/metabolism , DNA-Binding Proteins/metabolism , Ligands , Nuclear Proteins/metabolism , Receptors, Progesterone/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Mifepristone/metabolism , Mifepristone/pharmacology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Receptors, Progesterone/agonists , Receptors, Progesterone/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Sequence Deletion , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...