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










Publication year range
1.
Oncogene ; 36(10): 1364-1373, 2017 03.
Article in English | MEDLINE | ID: mdl-27869166

ABSTRACT

EZH2 is crucial for the progression of prostate cancer (PCa) and castration-resistant prostate cancer (CRPC) through upregulation and activation of progenitor genes, as well as androgen receptor (AR)-target genes. However, the mechanisms by which EZH2 is regulated in PCa and CRPC remain elusive. Here we report that EZH2 is post-transcriptionally regulated by SKP2 in vitro in cultured cells and in vivo in mouse models. We observed aberrant upregulation of Skp2, Ezh2 and histone H3 lysine 27 trimethylation (H3K27me3) in both Pten null mouse embryonic fibroblasts (MEFs) and Pten null mouse prostate tissues. Loss of Skp2 resulted in a striking decrease of Ezh2 levels in Pten/Trp53 double-null MEFs and in prostate tumors of Pten/Trp53 double-null mutant mice. SKP2 knockdown decreased EZH2 levels in human PCa cells through upregulation of TRAF6-mediated and lysine(K) 63-linked ubiquitination of EZH2 for degradation. Ectopic expression of TRAF6 promoted the K63-linked ubiquitination of EZH2 to decrease EZH2 and H3K27me3 levels in PCa cells. In contrast, TRAF6 knockdown resulted in a reduced EZH2 ubiquitination with an increase of EZH2 and H3K27me3 levels in PCa cells. Furthermore, the catalytically dead mutant TRAF6 C70A abolished the TRAF6-mediated polyubiquitination of recombinant human EZH2 in vitro. Most importantly, a concurrent elevation of Skp2 and Ezh2 was found in CRPC tumors of Pten/Trp53 mutant mice, and expression levels of SKP2 and EZH2 were positively correlated in human PCa specimens. Taken together, our findings revealed a novel mechanism on EZH2 ubiquitination and an important signaling network of SKP2-TRAF6-EZH2/H3K27me3, and targeting SKP2-EZH2 pathway may be a promising therapeutic strategy for CRPC treatment.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , S-Phase Kinase-Associated Proteins/genetics , TNF Receptor-Associated Factor 6/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Knockout Techniques , Histones/metabolism , Humans , Lysine/metabolism , Male , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostate/metabolism , Prostatic Neoplasms/pathology , Protein Stability , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitination
2.
Oncogene ; 34(28): 3700-10, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25220414

ABSTRACT

Androgen receptor splicing variants (ARVs) that lack the ligand-binding domain (LBD) are associated with the development of castration-resistant prostate cancer (CRPC), including resistance to the new generation of high-affinity anti-androgens. However, the mechanism by which ARV expression is regulated is not fully understood. In this study, we show that the activation of classical nuclear factor-kappa B (NF-κB) signaling increases the expression of ARVs in prostate cancer (PCa) cells and converts androgen-sensitive PCa cells to become androgen-insensitive, whereas downregulation of NF-κB signaling inhibits ARV expression and restores responsiveness of CRPC to anti-androgen therapy. In addition, we demonstrated that combination of anti-androgen with NF-κB-targeted therapy inhibits efficiently tumor growth of human CRPC xenografts. These results indicate that induction of ARVs by activated NF-κB signaling in PCa cells is a critical mechanism by which the PCa progresses to CRPC. This has important implications as it can prolong the survival of CRPC patients by restoring the tumors to once again respond to conventional androgen-deprivation therapy (ADT).


Subject(s)
Androgen Antagonists/administration & dosage , Antineoplastic Agents/administration & dosage , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Androgen Antagonists/pharmacology , Anilides/administration & dosage , Anilides/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/administration & dosage , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Humans , Male , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitriles/administration & dosage , Nitriles/pharmacology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Pyrazines/administration & dosage , Pyrazines/pharmacology , Receptors, Androgen/genetics , Tosyl Compounds/administration & dosage , Tosyl Compounds/pharmacology , Xenograft Model Antitumor Assays
3.
Prostate Cancer Prostatic Dis ; 17(4): 301-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25091041

ABSTRACT

BACKGROUND: SOX2 is a member of SOX (SRY-related high mobility group box) family of transcription factors. METHODS: In this study, we examined the expression of SOX2 in murine and human prostatic specimens by immunohistochemistry. RESULTS: We found that SOX2 was expressed in murine prostates during budding morphogenesis and in neuroendocrine (NE) prostate cancer (PCa) murine models. Expression of SOX2 was also examined in human prostatic tissue. We found that SOX2 was expressed in 26 of the 30 BPH specimens. In these BPH samples, expression of SOX2 was limited to basal epithelial cells. In contrast, 24 of the 25 primary PCa specimens were negative for SOX2. The only positive primary PCa was the prostatic NE tumor, which also showed co-expression of synaptophysin. Additionally, the expression of SOX2 was detected in all prostatic NE tumor xenograft lines. Furthermore, we have examined the expression of SOX2 on a set of tissue microarrays consisting of metastatic PCa tissues. Expression of SOX2 was detected in at least one metastatic site in 15 of the 24 patients with metastatic castration-resistant PCa; and the expression of SOX2 was correlated with synaptophysin. CONCLUSIONS: SOX2 was expressed in developing prostates, basal cells of BPH, as well as prostatic NE tumors.


Subject(s)
Neuroendocrine Tumors/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , SOXB1 Transcription Factors/biosynthesis , Animals , Blotting, Western , Heterografts , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Prostate/embryology , Prostatic Hyperplasia/metabolism , Tissue Array Analysis
4.
Oncogene ; 30(16): 1868-79, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21151173

ABSTRACT

Our previous studies have found that activation of Wnt/ß-catenin signaling resulted in mouse prostatic intraepithelial neoplasia (mPIN). In the large probasin promoter directed SV40-large T-antigen (LPB-Tag) expressing mouse prostate, mPIN forms with rare areas of adenocarcinoma. Combining expression of both Wnt-signaling and Tag expression in the mouse prostate, we have studied the role of Wnt/ß-catenin signaling in the progression from mPIN to adenocarcinoma. Our results show that the prostates of mice expressing Tag alone or nuclear ß-catenin alone developed mPIN, whereas the activation of both Tag and the Wnt/ß-catenin pathway resulted in invasive prostate adenocarcinoma. Furthermore, Foxa2, a forkhead transcription factor, was induced by active Wnt/ß-catenin signaling, and the expression of Foxa2 was associated with the invasive phenotype in the primary prostate cancer. In the LPB-Tag/dominant active (DA) ß-catenin prostates, MMP7, a Wnt/ß-catenin target gene, was upregulated. Furthermore, we also assessed AR and AR signaling pathway in these LPB-Tag/DA ß-catenin mice. Although ß-catenin is a well-known AR co-activator in vitro, our study provides strong in vivo evidences indicating that both AR protein and the AR pathway were downregulated in the prostate of LPB-Tag/DA ß-catenin mice. Histological analysis shows that prostate sections derived from the LPB-Tag/DA ß-catenin mice display neuroendocrine differentiation (NED), but NE cancer does not develop. Together, our findings indicate that Wnt/ß-catenin signaling has an important role in the progression of mPIN to prostate adenocarcinoma.


Subject(s)
Adenocarcinoma/pathology , Prostatic Neoplasms/pathology , Wnt Proteins/metabolism , beta Catenin/metabolism , Adenocarcinoma/metabolism , Animals , Disease Progression , Male , Mice , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction
5.
Cell Death Differ ; 17(3): 469-81, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19834493

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPARgamma) regulates the interface between cellular lipid metabolism, redox status and organelle differentiation. Conditional prostatic epithelial knockout of PPARgamma in mice resulted in focal hyperplasia which developed into mouse prostatic intraepithelial neoplasia (mPIN). The grade of PIN became more severe with time. Electron microscopy (EM) showed accumulated secondary lysosomes containing cellular organelles and debris suggestive of autophagy. Consistent with this analysis the autophagy marker LC-3 was found to be upregulated in areas of PIN in PPARgamma KO tissues. We selectively knocked down PPARgamma2 isoform in wild-type mouse prostatic epithelial cells and examined the consequences of this in a tissue recombination model. Histopathologically grafted tissues resembled the conditional PPARgamma KO mouse prostates. EM studies of PPARgamma- and PPARgamma2-deficient epithelial cells in vitro were suggestive of autophagy, consistent with the prostatic tissue analysis. This was confirmed by examining expression of beclin-1 and LC-3. Gene expression profiling in PPARgamma-/gamma2-deficient cells indicated a major dysregulation of cell cycle control and metabolic signaling networks related to peroxisomal and lysosomal maturation, lipid oxidation and degradation. The putative autophagic phenotypes of PPARgamma-deficient cells could be rescued by re-expression of either gamma1 or gamma2 isoform. We conclude that disruption of PPARgamma signaling results in autophagy and oxidative stress during mPIN pathogenesis.


Subject(s)
Autophagy/physiology , PPAR gamma/metabolism , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/metabolism , Protein Isoforms/metabolism , Signal Transduction/physiology , Animals , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Gene Expression Profiling , Hypoglycemic Agents/metabolism , Male , Mice , Mice, SCID , Mice, Transgenic , Molecular Sequence Data , PPAR gamma/genetics , Phenotype , Prostate/cytology , Prostate/metabolism , Prostate/pathology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Protein Isoforms/genetics , Rosiglitazone , Thiazolidinediones/metabolism , Transplantation, Heterologous
6.
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
7.
Cancer Res ; 61(18): 6795-804, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559553

ABSTRACT

Clinical experience with suicide gene therapy for prostate cancer using first-generation approaches has provided a basis for developing improved strategies. Given the low proliferation rate exhibited by prostate cancer, one improvement would be to develop suicide genes that effectively kill both dividing and nondividing cells. A second improvement would be to restrict cytotoxicity to prostate cancer cells, limiting injury of nondiseased tissue. Here we describe a novel approach to achieving both goals based on: (a) the use of a small, but potent, prostate-specific composite promoter, ARR(2)PB, based on the rat probasin gene; and (b) the use of a powerful artificial death switch, called inducible caspase-9 (iCaspase-9). ARR(2)PB includes two copies of the androgen response region (ARR), each containing two androgen receptor (AR)-binding sites, placed upstream of the probasin promoter elements necessary for basal transcription. Because iCaspase-9 contains two binding sites for the dimeric ligand, AP20187, administration of chemical inducers of dimerization leads to aggregation and caspase activation, followed by rapid apoptosis in both dividing and nondividing cells. Using both reagents, we constructed two novel adenoviruses (ADVs), ADV.ARR(2)PB-iCasp9 expressing iCaspase-9 and control ADV.ARR(2)PB-EGFP expressing enhanced green fluorescent protein (EGFP). We demonstrate that tissue specificity is not sacrificed in an ADV backbone because the marker protein, EGFP, is expressed in R1881-stimulated ADV.ARR(2)PB-EGFP-transduced LNCaP cells but not in AR(-) PC-3, 293, HuH-7, U-87, and MCF-7 cells. Similarly, Pro-iCaspase-9 is expressed in ADV.ARR(2)PB-iCasp9-infected LNCaP cells after R1881 administration and is activated after AP20187 administration. In vitro experiments revealed rapid and efficient iCaspase-9-induced apoptosis of LNCaP cells in both an R1881- and AP20187-dependent manner. Only 28, 8, and 0.5% survival of LNCaP cells was seen at multiplicities of infection of 2, 10, and 25, respectively. Furthermore, at a multiplicity of infection of 10, extraordinary sensitivity to AP20187 was seen (IC(50), approximately 3 pM). In vivo experiments showed that ADV.ARR(2)PB-iCasp9 induced apoptosis in LNCaP but not in HuH-7 xenograft tumors in an AP20187-dependent manner. Furthermore, a simple i.p. injection of AP20187 dramatically suppressed LNCaP tumor growth in nude mice and led to a significantly increased host survival. This study demonstrates the feasibility of using tissue-specific expression of cell cycle-independent iCaspases as a nonmutagenic alternative modality for prostate cancer suicide gene therapy.


Subject(s)
Adenocarcinoma/therapy , Apoptosis/genetics , Caspases/genetics , Genetic Therapy/methods , Prostatic Neoplasms/therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoviridae/genetics , Androgen-Binding Protein/genetics , Androgens/biosynthesis , Androgens/physiology , Binding Sites , Caspase 9 , Caspases/biosynthesis , Caspases/metabolism , Enzyme Induction , Genetic Vectors/genetics , HeLa Cells , Humans , Male , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/therapy , Organ Specificity , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Response Elements/genetics , Tumor Cells, Cultured
8.
Cancer Res ; 61(5): 2239-49, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280793

ABSTRACT

Neuroendocrine (NE) cells may be involved not only in growth and differentiation of the normal prostate but also in carcinogenesis and progression of prostate adenocarcinoma (Pca), including development of androgen resistance. However, the exact pathophysiology of NE cells in Pca remains poorly understood. Here we describe a transgenic model of Pca with progressive NE differentiation. Seven lines of transgenic mice with the rat prostate-specific large probasin promoter linked to the SV40-large T antigen (Tag) that develop prostatic neoplasia have been established. In this study, one of the seven lines (12T-10) was characterized by examination of 52 mice aged from 2-12 months. With advancing age, low-grade prostatic intraepithelial neoplasia, high-grade prostatic intraepithelial neoplasia, microinvasion, invasive carcinoma, and poorly or undifferentiated carcinoma with NE differentiation appeared in the prostates in sequential order. Whereas Tag is expressed uniformly in prostate epithelium, only an increasing subset of cells in prostatic intraepithelial neoplasia showed NE differentiation by chromogranin immunostaining. Frankly invasive carcinoma developing subsequently showed occasional definitive glandular differentiation (adenocarcinoma) and particularly undifferentiated carcinoma with NE histological features similar to those observed in NE carcinomas in humans. The NE carcinomas occurred in the dorsolateral and ventral lobes and were generally androgen receptor negative. Twenty-one of 32 (66%) mice aged > or = 6 months and 15 of 17 (88%) mice aged > or = 9 months developed metastatic tumors, as confirmed by histology and/or Tag immunohistochemistry. Metastases occurred at the later time points, with metastasis to regional lymph nodes, liver, and lung being particularly common. Metastases showed histological features of NE differentiation, as confirmed by chromogranin immunostaining and electron microscopy. An athymic nude mouse that received a s.c. implant of a primary NE tumor developed Tag-positive metastatic tumors with similar NE differentiation. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry identified identical protein profiles between the primary NE tumor and lesions in the extraprostatic organs. Hence, in the 12T-10 large probasin promoter-Tag mouse, high-grade prostatic intraepithelial neoplasia develops progressively greater NE differentiation and progresses to invasive adenocarcinoma and NE carcinoma, with a high percentage of metastases. The predictable progression through these stages will allow testing of therapeutic interventions as well as possible further delineation of the role of NE cells in Pca progression.


Subject(s)
Adenocarcinoma/pathology , Androgen-Binding Protein/genetics , Antigens, Polyomavirus Transforming/genetics , Carcinoma, Neuroendocrine/pathology , Prostatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Animals , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/secondary , Cell Differentiation/physiology , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Nude , Mice, Transgenic , Promoter Regions, Genetic , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Mech Dev ; 101(1-2): 61-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231059

ABSTRACT

To facilitate the elucidation of the genetic events that may play an important role in the development or tumorigenesis of the prostate gland, we have generated a transgenic mouse line with prostate-specific expression of Cre recombinase. This line, named PB-Cre4, carries the Cre gene under the control of a composite promoter, ARR2PB which is a derivative of the rat prostate-specific probasin (PB) promoter. Based on RT-PCR detection of Cre mRNA in PB-Cre4 mice or Cre-mediated activation of LacZ activity in PB-Cre4/R26R double transgenic mice, it is conclusively demonstrated that Cre expression is post-natal and prostatic epithelium-specific. Although the Cre recombination is detected in all lobes of the mouse prostate, there is a significant difference in expression levels between the lobes, being highest in the lateral lobe, followed by the ventral, and then the dorsal and anterior lobes. Besides the prostate gland, no other tissues of the adult PB-Cre4 mice demonstrate significant Cre expression, except for a few scattered areas in the gonads and the stroma of the seminal vesicle. By crossing the PB-Cre4 animals with floxed RXRalpha allelic mice, we demonstrate that mice, whose conventional knockout of this gene is lethal in embryogenesis, could be propagated with selective inactivation of RXRalpha in the prostate. Taken together, the results show that the PB-Cre4 mice have high levels of Cre expression and a high penetrance in the prostatic epithelium. The PB-Cre4 mice will be a useful resource for genetic-based studies on prostate development and prostatic disease.


Subject(s)
Epithelium/metabolism , Integrases/biosynthesis , Integrases/genetics , Prostate/metabolism , Viral Proteins , Alleles , Animals , Crosses, Genetic , Female , Galactosides/metabolism , Immunohistochemistry , Indoles/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Ovary/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic , Prostate/growth & development , Prostatic Neoplasms/metabolism , Rats , Receptors, Retinoic Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Testis/metabolism , Time Factors , Tissue Distribution , Transgenes
10.
Cancer Res ; 61(2): 497-503, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11212240

ABSTRACT

15-Lipoxygenase (15-LOX)-2 is expressed in benign prostate secretory cells and benign prostate produces 15S-hydroxyeicosatetraenoic acid (15S-HETE) from exogenous arachidonic acid (AA). In contrast, 15S-LOX-2 and 15S-HETE formation are reduced in prostate carcinoma (Pca). The mechanisms whereby reduced 15-LOX-2 may contribute to Pca development or progression are not known. We investigated the expression of peroxisome proliferator-activated receptor (PPAR) gamma in benign and malignant prostate tissues and the ability of 15S-HETE to activate PPARgamma-dependent transcription and modulate proliferation of the Pca cell line PC3. In contrast to benign prostate and similar to most Pca tissues, 15-LOX-2 mRNA was not detected in PC3 cells, and they did not produce detectable 15-HETE from [14C]AA. By reverse transcription-PCR, PPARgamma mRNA was present in 18 of 18 benign and 9 of 9 tumor specimens. The PPARgamma ligand BRL 49653 and 15S-HETE caused a dose-dependent inhibition of PC3 proliferation in a 14-day soft agar colony-forming assay (IC50 of 3 and 30 microM, respectively). 15S-HETE (10 microM) caused greater inhibition than 10 microM 15R-HETE. At 3 days, BRL 49653 and 15S-HETE caused a slight increase in cells in G0-G1 and a corresponding decrease in cells in S phase. In PC3 cells transiently transfected with a luciferase reporter linked to a PPAR response element, 1 microM BRL 49653 and 10 microM 15S-HETE caused approximately threefold and greater than twofold induction of PPAR-dependent transcription, respectively. By quantitative real-time reverse transcription-PCR and Northern analysis, 3-day treatment with BRL 49653 and 15S-HETE caused a reduction of PPARgamma expression but a marked up-regulation of the PPAR response element containing adipocyte type fatty acid binding protein. These results support the hypothesis that 15-LOX-2-derived 15S-HETE may constitute an endogenous ligand for PPARgamma in the prostate and that loss of this pathway by reduced expression of 15-LOX-2 may contribute to increased proliferation and reduced differentiation in prostate carcinoma.


Subject(s)
Cell Division/drug effects , Hydroxyeicosatetraenoic Acids/pharmacology , Prostatic Neoplasms/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Thiazolidinediones , Transcription Factors/genetics , Agar/pharmacology , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Blotting, Northern , Catalysis , Culture Media/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Luciferases/drug effects , Luciferases/genetics , Luciferases/metabolism , Male , Prostatic Neoplasms/pathology , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rosiglitazone , Thiazoles/pharmacology , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Tumor Cells, Cultured
11.
Endocrinology ; 142(3): 1296-308, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181548

ABSTRACT

Using transgenic mice, we have recently shown that 5 kb of the 5'-flanking region of the mouse epididymal retinoic acid-binding protein (mE-RABP) gene contains all of the information required for spatial and temporal gene expression in the epididymis. To identify the important cis-DNA regulatory element(s) involved in the tissue-, region-, and cell-specific expression of the mE-RABP gene, the 5-kb DNA fragment was sequenced. A computer analysis of the nucleotide sequence showed the presence of a new gene located 1.7 kb upstream from the mE-RABP gene transcription initiation site. The analysis of the open reading frame showed that the new gene encoded a putative 17-kDa lipocalin (named mEP17) related to mE-RABP. A 600-bp complementary DNA encoding mEP17 was cloned by rapid amplification of 3'-cDNA ends from epididymal total RNA. Two mEP17 RNA species (1 and 3.1 kb in size) were detected by Northern blot in the epididymis, but not in other tissues tested. In situ hybridization analyses showed that, unlike mE-RABP messenger RNA (mRNA), which is expressed in the distal caput epididymidis, mEP17 mRNA was detected only in the principal cells of the initial segment. The spatial expression and homology with mE-RABP suggest that mEP17 may act as a retinoid carrier protein within the epididymis. mEP17 mRNA expression disappeared 5 days postcastration. Four days after unilateral castration, mEP17 mRNA had nearly disappeared in the epididymis from the castrated side, but not from the intact side. In addition, testosterone replacement to bilaterally castrated mice failed to restore gene expression. We conclude that mEP17 gene expression is dependent on testicular factors circulating in the luminal fluid. Together our results suggest that mE-RABP and mEP17 genes were generated by duplication and that evolution led to a different region-specific gene expression and regulation in the epididymis.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/physiology , Epididymis/metabolism , Gene Duplication , Receptors, Retinoic Acid/genetics , Testis/metabolism , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Conserved Sequence/genetics , Gene Expression Regulation , Genome , Hormones/physiology , Lipocalins , Male , Mice , Mice, Inbred Strains , Molecular Sequence Data , Molecular Weight , Open Reading Frames/genetics , Orchiectomy , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism
12.
Mol Endocrinol ; 15(1): 46-56, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145738

ABSTRACT

The androgen receptor (AR), a member of the steroid receptor superfamily of nuclear transcription factors, mediates androgen signaling in diverse target tissues. Here we report AR gene mutations identified in human prostate cancer and the autochthonous transgenic adenocarcinoma of the mouse prostate model that colocate to residues (668)QPIF(671) at the boundary of the hinge and ligand-binding domain, resulting in receptors that exhibit 2- to 4-fold increased activity compared with wild-type AR in response to dihydrotestosterone, estradiol, progesterone, adrenal androgens, and the AR antagonist, hydroxyflutamide, without an apparent effect on receptor levels, ligand binding kinetics, or DNA binding. The expression of these or similar variants could explain the emergence of hormone refractory disease in a subset of patients. Homology modeling indicates that amino acid residues (668)QPIF(671) form a ridge bordering a potential protein-protein interaction surface. The naturally occurring AR gene mutations reported in this study result in decreased hydrophobicity of this surface, suggesting that altered receptor-protein interaction mediates the precocious activity of the AR variants.


Subject(s)
Flutamide/analogs & derivatives , Mutation , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Transcriptional Activation , Adenocarcinoma/genetics , Androgen Antagonists/pharmacology , Androgens/pharmacology , Animals , Binding Sites , COS Cells , Cell Line , DNA/metabolism , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Flutamide/pharmacology , Humans , Male , Mice , Mice, Transgenic , Models, Molecular , Mutagenesis , Progesterone/pharmacology , Prostatic Neoplasms/genetics , Protein Structure, Secondary , Receptors, Androgen/physiology , Structure-Activity Relationship , Transfection
13.
Endocrinology ; 141(12): 4698-710, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108285

ABSTRACT

Transient transfection studies have shown that the probasin (PB) promoter confers androgen selectivity over other steroid hormones, and transgenic animal studies have demonstrated that the PB promoter will target androgen, but not glucocorticoid, regulation in a prostate-specific manner. Previous PB promoters either targeted low levels of transgene expression or became too large to be conveniently used. The goal was to design a PB promoter that would be small, yet target high levels of prostate-specific transgene expression. Thus, a composite probasin promoter (ARR2PB) coupled to the bacterial chloramphenicol acetyltransferase reporter (ARR2PBCAT) was generated and tested in prostatic and nonprostatic cell lines and in a transgenic mouse model. In PC-3, LNCaP, and DU145 prostate cancer cell lines, the ARR2PB promoter gave basal expression and was induced in response to androgen and glucocorticoid treatment after cotransfection with the respective steroid receptor. Basal expression of ARR2PBCAT in the nonprostatic COS-1, MCF-7, ZR-75-1, and PANC-1 cell lines was very low; however, CAT activity could be induced in response to androgens and glucocorticoids when cells were cotransfected with either the AR or GR. In contrast to the transfection studies, ARR2PBCAT transgene expression remained highly specific for prostatic epithelium in transgenic mice. CAT activity decreased after castration, and could be induced by androgens and, in addition, glucocorticoids. This demonstrates that the necessary sequences required to target prostate-specific epithelial expression are contained within the composite ARR2PB minimal promoter, and that high transgene expression can now be regulated by both androgens and glucocorticoids. The ARR2PB promoter represents a novel glucocorticoid inducible promoter that can be used for the generation of transgenic mouse models and in viral gene therapy vectors for the treatment of prostate cancer in humans.


Subject(s)
Androgen-Binding Protein/genetics , Androgens/pharmacology , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Promoter Regions, Genetic , Prostate/metabolism , Receptors, Androgen/metabolism , Animals , Binding Sites , Breast Neoplasms , COS Cells , Chloramphenicol O-Acetyltransferase/genetics , Epithelium/metabolism , Female , Haplorhini , Humans , Kidney , Male , Mice , Mice, Transgenic , Pancreatic Neoplasms , Prostatic Neoplasms , Tumor Cells, Cultured
14.
Biol Reprod ; 63(6): 1881-92, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090461

ABSTRACT

The epididymis provides the optimal milieu for sperm maturation and storage. Epididymal secretory proteins are believed to be involved in that process. Androgens are the major endocrine and paracrine regulatory signals that regulate gene expression in the epididymis. We have previously identified an androgen-dependent retinoic acid-binding protein (mE-RABP) that is secreted into the luminal fluid from the mouse mid/distal caput epididymidis. The mE-RABP protein belongs to the lipocalin superfamily and may be involved in the trafficking of retinoic acid within the epididymis. We have recently demonstrated that 5 kilobases of the 5' flanking region of the mE-RABP gene contained all the information for the hormonal regulation and the tissue-, region-, and cell-specific expression of the mE-RABP gene. In this study, we have identified a complex androgen-specific response region (ARR) within the first 600 base pairs of the mE-RABP gene promoter. Androgen (DHT) but not glucocorticoid (DEX) activates the ARR in HeLa and PC-3 cells. Two androgen receptor binding sites have been located at positions -445/-459 and -102/-88 and were named ARBS-1 and ARBS-0, respectively. Point mutations of ARBS-0 resulted in a slight decrease of the androgen response. However, mutations of ARBS-1 led to a total loss of the androgen responsiveness, suggesting that it was a major cis-acting element. When ARBS-1 is isolated from its promoter context, it serves as a weak androgen-responsive element that was activated by both androgens and glucocorticoids. Also, the -543/-88 DNA promoter fragment behaved as a poor androgen-responsive region, suggesting that regulatory elements located within the proximal mE-RABP promoter were required for a full androgen response. In conclusion, the mE-RABP ARR is a good model for the study of molecular mechanisms that lead to an androgen-specific responsiveness in vivo.


Subject(s)
Androgens/genetics , Epididymis/metabolism , Response Elements/genetics , Retinol-Binding Proteins/genetics , Animals , Electrophoresis , Histidine/metabolism , Male , Mice , Mice, Transgenic , Nuclease Protection Assays , Promoter Regions, Genetic/genetics , Retinol-Binding Proteins/biosynthesis , Transfection
15.
Biochim Biophys Acta ; 1482(1-2): 249-58, 2000 Oct 18.
Article in English | MEDLINE | ID: mdl-11058766

ABSTRACT

Probasin (PB) occurs both as a secreted and a nuclear protein that is abundantly expressed in the epithelial cells of the rat prostate. A genomic clone of 17.5 kb gene was isolated from a rat liver genomic library, determining that the probasin gene was comprised of seven exons where the splice donor/acceptor sites conformed to the GT/AG consensus sequence. The exon number and size are remarkably similar to those of aphrodisin, rat alpha(2)-urinary globulin and major urinary protein, outlier members of the lipocalin superfamily. In addition, alignment of the deduced amino acids determined that the probasin gene also contains the glycine-X-tryptophan (G-X-W) motif similar to that of human retinol serum binding protein which binds retinol, and the C-X-X-X-C motif also found in insect lipocalins that bind pheromones. The cysteine residues in exons 3 and 6 are conserved, predicting a secondary structure of eight beta-sheets and the alpha-helix commonly seen in the lipocalin superfamily. Unique PB characteristics include a large genomic fragment (17.5 kb compared to the 3-5 kb seen in other lipocalin genes) and an isoelectric point (pI) of 11.5 which is very basic compared to that of the other more acidic lipocalins. Functionally, PB gene expression is regulated by androgens and zinc in the epithelial cells of the rodent prostate. The 5'-flanking region of probasin contains two androgen receptor binding sites that allow androgen-specific gene expression as well as prostate-specific elements that target and maintain high levels of transgene expression in several PB transgenic mouse models.


Subject(s)
Androgen-Binding Protein/chemistry , Prostate/metabolism , Amino Acid Sequence , Androgen-Binding Protein/genetics , Androgen-Binding Protein/physiology , Animals , Gene Expression Regulation , Male , Molecular Sequence Data , Rats , Transcription, Genetic
16.
J Mol Endocrinol ; 22(3): 313-25, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10343290

ABSTRACT

Glucocorticoid and androgen receptors have been shown to function through the same palindromic glucocorticoid response element (GRE) and yet have differential effects on gene transcription. In this study, we examined the functional and structural relationship of the androgen and glucocorticoid receptors with the androgen responsive region (ARR) of the probasin (PB) gene containing two androgen receptor binding sites, ARBS-1 and ARBS-2. Transfection studies indicated that one copy of each cis-acting DNA element was essential for maximal androgen-induced chloramphenicol acetyltransferase (CAT) activity and that androgen selectivity was maintained when multiple copies of the minimal wild type (wt) androgen responsive region containing both ARBS-1 and ARBS-2 (-244 to -96) were subcloned in front of the thymidine kinase promoter. Furthermore, replacing the androgen response region with 1, 2 or 3 copies of either ARBS-1 or ARBS-2 restored less than 4% of the biological activity seen with the wt PB ARR. Multiple copies of either ARBS-1 or ARBS-2 did not result in glucocorticoid-induced CAT gene activity. By comparison, 1 or 2 copies of the tyrosine aminotransferase (TAT) GRE, as well as the mouse mammary tumour virus GRE, were strong inducers of CAT activity in response to both androgen and glucocorticoid treatment. In addition, band shift assays demonstrated that although the synthetic glucocorticoid receptor, GR-DNA binding domain (GR-DBD), and the synthetic androgen receptor, AR2, could interact with the TAT GRE (dissociation constants Kd of 63.9 and 14.1 respectively), only AR2 but not GR-DBD binding could be detected on ARBS-1 and ARBS-2. Our findings provide further evidence that androgen-induced regulation of gene transcription can occur through androgen-specific DNA binding sites that are distinct from the common GRE.


Subject(s)
Androgen-Binding Protein/metabolism , Androgens/pharmacology , Glucocorticoids/pharmacology , Receptors, Androgen/metabolism , Androgen-Binding Protein/genetics , Androgens/metabolism , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , DNA Footprinting , Gene Expression Regulation/drug effects , Humans , Molecular Sequence Data , Receptors, Glucocorticoid/metabolism , Tumor Cells, Cultured
17.
J Biol Chem ; 274(12): 8282-90, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075734

ABSTRACT

The murine epididymis synthesizes and secretes a retinoic acid-binding protein (mE-RABP) that belongs to the lipocalin superfamily. The gene encoding mE-RABP is specifically expressed in the mouse mid/distal caput epididymidis under androgen control. In transgenic mice, a 5-kilobase pair (kb) promoter fragment, but not a 0.6-kb fragment, of the mE-RABP gene driving the chloramphenicol acetyltransferase (CAT) reporter gene restricted high level of transgene expression to the caput epididymidis. No transgene expression was detected in any other male or female tissues. Immunolocalization of the CAT protein and in situ hybridization of the corresponding CAT mRNA indicated that transgene expression occurred in the principal cells of the mid/distal caput epididymidis, thereby mimicking the spatial endogenous mE-RABP gene expression. Transgene and mE-RABP gene expression was detected from 30 days and progressively increased until 60 days of age. Castration, efferent duct ligation, and hormone replacement studies demonstrated that transgene expression was specifically regulated by androgen but not by any other testicular factors. Altogether, our results demonstrate that the 5-kb promoter fragment of the mE-RABP gene contains all of the information required for the hormonal regulation and the spatial and temporal expression of the mE-RABP gene in the epididymis.


Subject(s)
Androgens/physiology , Epididymis/metabolism , Promoter Regions, Genetic , Receptors, Retinoic Acid/genetics , Animals , Chloramphenicol O-Acetyltransferase/genetics , DNA Fragmentation , Female , Genes, Reporter , In Situ Hybridization , Male , Mice , Mice, Transgenic , Receptors, Retinoic Acid/metabolism , Retinol-Binding Proteins, Plasma , Transgenes
19.
Prostate ; 36(4): 256-63, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9719026

ABSTRACT

BACKGROUND: The purpose of this study was to determine the contribution of different transactivating regions of the androgen receptor (AR) to the induction of androgen-regulated promoters in poorly (PC3 cells) and well-differentiated (LNCaP cells) prostate cancer cell lines. METHODS: PC3 and LNCaP cells were co-transfected with plasmids expressing full-length AR or deletion mutants together with luciferase reporters linked to the probasin (PB) and PSA promoters; as well as to ARR3tk, a PB-derived recombinant promoter. RESULTS: Androgen induction of the ARR3tk promoter in the presence of AR was 8- to 10-fold higher than that seen with the PB promoter. Activation of ARR3tk was greatest with an androgen-independent construct in which the first 231 amino acids and the ligand binding domain had been removed, indicating that this promoter is more responsive to activating functions in the N-terminal domain than in the ligand binding domain. By comparison, induction of the PB promoter was greatest with the full-length AR, which suggests that the ligand binding domain also makes a major contribution to the activation of this promoter. In similar analyses with the PSA promoter, AR regions required for promoter induction was dependent on the host cell type. In PC3 cells, the predominant AR transactivation function was androgen-independent and resided in the N-terminal domain, whereas in LNCaP cells, the highest level of induction was androgen dependent and also required participation of the ligand binding domain. CONCLUSIONS: Our results indicate that the relative utilization of transactivating functions in N-terminal and ligand binding domains of the AR is promoter and cell specific.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Androgen-Binding Protein/genetics , Base Sequence , Binding Sites/genetics , Cell Differentiation , DNA Primers/genetics , Genes, Reporter , Humans , Luciferases/genetics , Male , Promoter Regions, Genetic , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Sequence Deletion , Thymidine Kinase/genetics , Transcriptional Activation , Transfection , Tumor Cells, Cultured
20.
Mol Reprod Dev ; 50(4): 387-95, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9669522

ABSTRACT

The murine epididymal retinoic acid-binding protein (mE-RABP) is specifically synthesized in the mouse mid/distal caput epididymidis and secreted in the lumen. In this report, we have demonstrated by Southern blot analysis of genomic DNA that mE-RABP is encoded by a single-copy gene. A mouse 129/SvJ genomic bacterial artificial chromosome (BAC) library was screened using a cDNA encoding the minor form of mE-RABP. One positive BAC clone was characterized and sequenced to determine the nucleotide sequence of the entire mE-RABP gene. The molecular cloning of the mE-RABP gene completes the characterization of the 20.5-kDa-predicted preprotein leading to the minor and major forms of mE-RABP. Comparison of the DNA sequence of the promoter and coding regions with that of the rat epididymal secretory protein I (ESP I) gene showed that the mE-RABP gene is the orthologue of the ESP I gene that encodes a rat epididymal retinoic acid-binding protein. Several regulatory elements, including a putative androgen receptor binding site, "CACCC-boxes," NF-1, Oct-1, and SP-1 recognition sites, are conserved in the proximal promoter. Analysis of the nucleotide sequence of the mE-RABP gene revealed the presence of seven exons and showed that the genomic organization is highly related to other genes encoding lipocalins. The mE-RABP gene was mapped by fluorescent in situ hybridization to the [A3-B] region of the murine chromosome 2. Our data, combined with that of others, suggest that the proximal segment of the mouse chromosome 2 may be a rich region for genes encoding lipocalins with a genomic organization highly related to the mE-RABP gene.


Subject(s)
Chromosome Mapping , Receptors, Retinoic Acid/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Gene Dosage , Male , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Retinol-Binding Proteins, Plasma , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...