Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 22(10): 1166-1181, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37486978

ABSTRACT

Prostate cancers adapt to androgen receptor (AR) pathway inhibitors and progress to castration resistance due to ongoing AR expression and function. To counter this, we developed a new approach to modulate the AR and inhibit castration-resistant prostate cancer (CRPC) using multivalent peptoid conjugates (MPC) that contain multiple copies of the AR-targeting ligand ethisterone attached to a peptidomimetic scaffold. Here, we investigated the antitumor effects of compound MPC309, a trivalent display of ethisterone conjugated to a peptoid oligomer backbone that binds to the AR with nanomolar affinity. MPC309 exhibited potent antiproliferative effects on various enzalutamide-resistant prostate cancer models, including those with AR splice variants, ligand-binding mutations, and noncanonical AR gene expression programs, as well as mouse prostate organoids harboring defined genetic alterations that mimic lethal human prostate cancer subtypes. MPC309 is taken up by cells through macropinocytosis, an endocytic process more prevalent in cancer cells than in normal ones, thus providing an opportunity to target tumors selectively. MPC309 triggers a distinct AR transcriptome compared with DHT and enzalutamide, a clinically used antiandrogen. Specifically, MPC309 enhances the expression of differentiation genes while reducing the expression of genes needed for cell division and metabolism. Mechanistically, MPC309 increases AR chromatin occupancy and alters AR interactions with coregulatory proteins in a pattern distinct from DHT. In xenograft studies, MPC309 produced significantly greater tumor suppression than enzalutamide. Altogether, MPC309 represents a promising new AR modulator that can combat resistant disease by promoting an AR antiproliferative gene expression program.


Subject(s)
Peptoids , Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Animals , Mice , Humans , Receptors, Androgen/metabolism , Peptoids/pharmacology , Ligands , Ethisterone/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Prostatic Neoplasms/pathology , Nitriles/pharmacology , Androgen Receptor Antagonists/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism
2.
PLoS One ; 17(9): e0272986, 2022.
Article in English | MEDLINE | ID: mdl-36054206

ABSTRACT

Acyl-CoA synthetase 1 (ACSL1) is an enzyme that converts fatty acids to acyl-CoA-derivatives for lipid catabolism and lipid synthesis in general and can provide substrates for the production of mediators of inflammation in monocytes and macrophages. Acsl1 expression is increased by hyperglycemia and inflammatory stimuli in monocytes and macrophages, and promotes the pro-atherosclerotic effects of diabetes in mice. Yet, surprisingly little is known about the mechanisms underlying Acsl1 transcriptional regulation. Here we demonstrate that the glucose-sensing transcription factor, Carbohydrate Response Element Binding Protein (CHREBP), is a regulator of the expression of Acsl1 mRNA by high glucose in mouse bone marrow-derived macrophages (BMDMs). In addition, we show that inflammatory stimulation of BMDMs with lipopolysaccharide (LPS) increases Acsl1 mRNA via the transcription factor, NF-kappa B. LPS treatment also increases ACSL1 protein abundance and localization to membranes where it can exert its activity. Using an Acsl1 reporter gene containing the promoter and an upstream regulatory region, which has multiple conserved CHREBP and NF-kappa B (p65/RELA) binding sites, we found increased Acsl1 promoter activity upon CHREBP and p65/RELA expression. We also show that CHREBP and p65/RELA occupy the Acsl1 promoter in BMDMs. In primary human monocytes cultured in high glucose versus normal glucose, ACSL1 mRNA expression was elevated by high glucose and further enhanced by LPS treatment. Our findings demonstrate that CHREBP and NF-kappa B control Acsl1 expression under hyperglycemic and inflammatory conditions.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Coenzyme A Ligases/genetics , Hyperglycemia , Inflammation/metabolism , NF-kappa B p50 Subunit/metabolism , NF-kappa B , Animals , Coenzyme A/metabolism , Glucose/metabolism , Glucose/pharmacology , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Inflammation/genetics , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , NF-kappa B/metabolism , RNA, Messenger/genetics
3.
Mob DNA ; 12(1): 21, 2021 Aug 23.
Article in English | MEDLINE | ID: mdl-34425899

ABSTRACT

BACKGROUND: The autonomous retroelement Long Interspersed Element-1 (LINE-1) mobilizes though a copy and paste mechanism using an RNA intermediate (retrotransposition). Throughout human evolution, around 500,000 LINE-1 sequences have accumulated in the genome. Most of these sequences belong to ancestral LINE-1 subfamilies, including L1PA2-L1PA7, and can no longer mobilize. Only a small fraction of LINE-1 sequences, approximately 80 to 100 copies belonging to the L1Hs subfamily, are complete and still capable of retrotransposition. While silenced in most cells, many questions remain regarding LINE-1 dysregulation in cancer cells. RESULTS: Here, we optimized CRISPR Cas9 gRNAs to specifically target the regulatory sequence of the L1Hs 5'UTR promoter. We identified three gRNAs that were more specific to L1Hs, with limited binding to older LINE-1 sequences (L1PA2-L1PA7). We also adapted the C-BERST method (dCas9-APEX2 Biotinylation at genomic Elements by Restricted Spatial Tagging) to identify LINE-1 transcriptional regulators in cancer cells. Our LINE-1 C-BERST screen revealed both known and novel LINE-1 transcriptional regulators, including CTCF, YY1 and DUSP1. CONCLUSION: Our optimization and evaluation of gRNA specificity and application of the C-BERST method creates a tool for studying the regulatory mechanisms of LINE-1 in cancer. Further, we identified the dual specificity protein phosphatase, DUSP1, as a novel regulator of LINE-1 transcription.

4.
Biol Reprod ; 103(4): 880-891, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32678429

ABSTRACT

Spermatogenesis is a complex process that establishes male fertility and involves proper communication between the germline (spermatozoa) and the somatic tissue (Sertoli cells). Many factors that are important for spermatozoa production are also required for Sertoli cell function. Recently, we showed that the transcriptional cofactor ubiquitously expressed transcript (UXT) encodes a protein that is essential in germ cells for spermatogenesis and fertility. However, the role of UXT within Sertoli cells and how it affects Sertoli cell function was still unclear. Here we describe a novel role for UXT in the Sertoli cell's ability to support spermatogenesis. We find that the conditional deletion of Uxt in Sertoli cells results in smaller testis size and weight, which coincided with a loss of germ cells in a subset of seminiferous tubules. In addition, the deletion of Uxt has no impact on Sertoli cell abundance or maturity, as they express markers of mature Sertoli cells. Gene expression analysis reveals that the deletion of Uxt in Sertoli cells reduces the transcription of genes involved in the tight junctions of the blood-testis barrier (BTB). Furthermore, tracer experiments and electron microscopy reveal that the BTB is permeable in UXT KO animals. These findings broaden our understanding of UXT's role in Sertoli cells and its contribution to the structural integrity of the BTB.


Subject(s)
Blood-Testis Barrier/physiology , Cell Cycle Proteins/metabolism , Molecular Chaperones/metabolism , Sertoli Cells/metabolism , Animals , Cell Adhesion , Cell Cycle Proteins/genetics , Down-Regulation , Gene Deletion , Gene Expression Regulation , Germ Cells/physiology , Male , Mice , Molecular Chaperones/genetics , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
5.
Oncotarget ; 10(7): 707-716, 2019 Jan 22.
Article in English | MEDLINE | ID: mdl-30774773

ABSTRACT

Ubiquitously-expressed, prefoldin-like chaperone (UXT) also called Androgen Receptor Trapped clone-27 (ART-27) is widely expressed in human tissues. Our previous studies showed that UXT regulates transcription repression including androgen receptor (AR) signaling in prostate cancer. Here we analyzed a tissue microarray consisting of normal prostate, benign prostatic hyperplasia, high grade prostatic intraepithelial neoplasia (HGPIN) and primary prostate cancer cases for UXT protein expression. We found that HGPIN and malignant tumors have significantly decreased UXT expression compared to the normal prostate. Loss of UXT expression in primary prostate cancer is positively associated with high Gleason grade and poor relapse-free survival. We engineered prostate-specific Uxt KO mice that developed a hyperplastic phenotype with apparent prostate secretion fluid blockage as well as PIN by 4-6 months. Doubly mutant Uxt KO /Pten KO mice developed a more aggressive PIN phenotype. UXT depletion in prostate cancer cells also increased retroelements expression, including LINE-1 and Alu. Consistent with this finding Uxt KO mice have increased LINE-1 protein levels in the prostate compared to control mice. In addition, cancer cells with UXT depletion have increased retrotransposition activity and accumulated DNA damage. Our findings demonstrate that loss of UXT is an early event during prostate cancer progression, which may contribute to genome instability.

6.
Adv Exp Med Biol ; 1106: 85-94, 2018.
Article in English | MEDLINE | ID: mdl-30484154

ABSTRACT

The Unconventional prefoldin RPB5 interacting protein (URI), also known as RPB5-Mediating Protein (RMP) has been shown to play several regulatory roles in different cellular compartments including the mitochondria, as a phosphatase binding protein; in the cytoplasm, as a chaperone-like protein; and in the nucleus, as a transcriptional regulator through binding to RPB5 and RNA polymerase II (polII). This chapter focuses on the role URI plays in transcriptional regulation in the prostate cell. In prostate cells, URI is tightly bound to another prefoldin-like protein called UXT, a known androgen receptor (AR) cofactor. Part of a multiprotein complex, URI and UXT act as transcriptional repressors, and URI regulates KAP1 through PP2A phosphatase activity. The discovery of the interaction of URI and UXT with KAP1, AR, and PP2A, as well as the numerous interactions between URI and components of the R2TP/prefoldin-like complex, RPB5, and nuclear proteins involved in DNA damage response, chromatin remodeling and gene transcription, reveal a pleiotropic effect of the URI/UXT complex on nuclear processes. The mechanisms by which URI/UXT affect transcription, chromatin structure and regulation, and genome stability, remain to be elucidated but will be of fundamental importance considering the many processes affected by alterations of URI/UXT and other prefoldins and prefoldin-like proteins.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/chemistry , Molecular Chaperones/chemistry , Neoplasm Proteins/chemistry , Cell Cycle Proteins , Humans , Male , Prostate , Repressor Proteins
7.
PLoS One ; 13(4): e0195747, 2018.
Article in English | MEDLINE | ID: mdl-29649254

ABSTRACT

Male mammals must simultaneously produce prodigious numbers of sperm and maintain an adequate reserve of stem cells to ensure continuous production of gametes throughout life. Failures in the mechanisms responsible for balancing germ cell differentiation and spermatogonial stem cell (SSC) self-renewal can result in infertility. We discovered a novel requirement for Ubiquitous Expressed Transcript (UXT) in spermatogenesis by developing the first knockout mouse model for this gene. Constitutive deletion of Uxt is embryonic lethal, while conditional knockout in the male germline results in a Sertoli cell-only phenotype during the first wave of spermatogenesis that does not recover in the adult. This phenotype begins to manifest between 6 and 7 days post-partum, just before meiotic entry. Gene expression analysis revealed that Uxt deletion downregulates the transcription of genes governing SSC self-renewal, differentiation, and meiosis, consistent with its previously defined role as a transcriptional co-factor. Our study has revealed the first in vivo function for UXT in the mammalian germline as a regulator of distinct transcriptional programs in SSCs and differentiating spermatogonia.


Subject(s)
Molecular Chaperones/genetics , Spermatogenesis/genetics , Animals , Caspase 3/metabolism , Cell Cycle , Cell Cycle Proteins , Cell Differentiation/genetics , Gene Deletion , Genes, Lethal , Immunohistochemistry , Machine Learning , Male , Meiosis/genetics , Mice , Mice, Knockout , Molecular Chaperones/metabolism , Phenotype , Spermatogonia/metabolism , Testis/metabolism , Testis/pathology
8.
Mob DNA ; 9: 1, 2018.
Article in English | MEDLINE | ID: mdl-29308092

ABSTRACT

BACKGROUND: Long Interspersed Nuclear Element-1 (LINE-1) is an autonomous retrotransposon that generates new genomic insertions through the retrotransposition of a RNA intermediate. Expression of LINE-1 is tightly repressed in most somatic tissues to prevent DNA damage and ensure genomic integrity. However, the reactivation of LINE-1 has been documented in cancer and the role of LINE-1 protein expression and retrotransposition has become of interest in the development, progression, and adaptation of many epithelial neoplasms, including prostate cancer. RESULTS: Here, we examined endogenous LINE-1 protein expression and localization in a panel of prostate cancer cells and observed a diverse range of LINE-1 expression patterns between cell lines. Subcellular localization of LINE-1 proteins, ORF1p and ORF2p, revealed distinct expression patterns. ORF1p, a nucleic acid chaperone that binds LINE-1 mRNA, was predominantly expressed in the cytoplasm, with minor localization in the nucleus. ORF2p, containing endonuclease and reverse transcriptase domains, exhibited punctate foci in the nucleus and also displayed co-localization with PCNA and γH2AX. Using a retrotransposition reporter assay, we found variations in LINE-1 retrotransposition between cell lines. CONCLUSIONS: Overall, our findings reveal new insight into the expression and retrotransposition of LINE-1 in prostate cancer. The prostate cancer cells we investigated provide a unique model for investigating endogenous LINE-1 activity and provide a functional model for studying LINE-1 mechanisms in prostate cancer.

9.
J Biol Chem ; 291(49): 25516-25528, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27780869

ABSTRACT

URI (unconventional prefoldin RPB5 interactor protein) is an unconventional prefoldin, RNA polymerase II interactor that functions as a transcriptional repressor and is part of a larger nuclear protein complex. The components of this complex and the mechanism of transcriptional repression have not been characterized. Here we show that KAP1 (KRAB-associated protein 1) and the protein phosphatase PP2A interact with URI. Mechanistically, we show that KAP1 phosphorylation is decreased following recruitment of PP2A by URI. We functionally characterize the novel URI-KAP1-PP2A complex, demonstrating a role of URI in retrotransposon repression, a key function previously demonstrated for the KAP1-SETDB1 complex. Microarray analysis of annotated transposons revealed a selective increase in the transcription of LINE-1 and L1PA2 retroelements upon knockdown of URI. These data unveil a new nuclear function of URI and identify a novel post-transcriptional regulation of KAP1 protein that may have important implications in reactivation of transposable elements in prostate cancer cells.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Protein Phosphatase 2/metabolism , Repressor Proteins/metabolism , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Multiprotein Complexes/genetics , Neoplasm Proteins/genetics , Phosphorylation/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Phosphatase 2/genetics , Repressor Proteins/genetics , Retroelements , Tripartite Motif-Containing Protein 28
10.
PLoS One ; 10(10): e0141589, 2015.
Article in English | MEDLINE | ID: mdl-26509262

ABSTRACT

Despite decades of effort to develop effective therapy and to identify promising new drugs, prostate cancer is lethal once it progresses to castration-resistant disease. Studies show mis-regulation of multiple pathways in castration-resistant prostate cancer (CRPC), reflecting the heterogeneity of the tumors and also hinting that targeting androgen receptor (AR) pathway alone might not be sufficient to treat CRPC. In this study, we present evidence that the Wnt/ß-catenin pathway might be activated in prostate cancer cells after androgen-deprivation to promote androgen-independent growth, partly through enhanced interaction of ß-catenin with TCF4. Androgen-independent prostate cancer cells were more prone to activate a Wnt-reporter, and inhibition of the Wnt/ß-catenin pathway increased sensitivity of these cells to the second-generation antiandrogen, enzalutamide. Combined treatment of enzalutamide and Wnt/ß-catenin inhibitor showed increased growth repression in both androgen-dependent and -independent prostate cancer cells, suggesting therapeutic potential for this approach.


Subject(s)
Receptors, Androgen/metabolism , beta Catenin/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Humans , Immunoprecipitation , Male , Prostatic Neoplasms , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , beta Catenin/genetics
11.
PLoS One ; 8(5): e63879, 2013.
Article in English | MEDLINE | ID: mdl-23667685

ABSTRACT

Unconventional prefoldin RPB5 Interactor (URI) was identified as a transcriptional repressor that binds RNA polymerase II (pol II) through interaction with the RPB5/POLR2E subunit. Despite the fact that many other proteins involved in transcription regulation have been shown to interact with URI, its nuclear function still remains elusive. Previous mass spectrometry analyses reported that URI is part of a novel protein complex called R2TP/prefoldin-like complex responsible for the cytoplasmic assembly of RNA polymerase II. We performed a mass spectrometry (MS)-based proteomic analysis to identify nuclear proteins interacting with URI in prostate cells. We identified all the components of the R2TP/prefoldin-like complex as nuclear URI interactors and we showed that URI binds and regulates RPB5 protein stability and transcription. Moreover, we validated the interaction of URI to the P53 and DNA damage-Regulated Gene 1 (PDRG1) and show that PDRG1 protein is also stabilized by URI binding. We present data demonstrating that URI nuclear/cytoplasmic shuttling is affected by compounds that stall pol II on the DNA (α-amanitin and actinomycin-D) and by leptomycin B, an inhibitor of the CRM1 exportin that mediates the nuclear export of pol II subunits. These data suggest that URI, and probably the entire R2TP/prefoldin-like complex is exported from the nucleus through CRM1. Finally we identified putative URI sites of phosphorylation and acetylation and confirmed URI sites of post-transcriptional modification identified in previous large-scale analyses the importance of which is largely unknown. However URI post-transcriptional modification was shown to be essential for URI function and therefore characterization of novel sites of URI modification will be important to the understanding of URI function.


Subject(s)
Cell Nucleus/metabolism , DNA-Directed RNA Polymerases/metabolism , Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Prostate/metabolism , RNA Polymerase II/metabolism , Active Transport, Cell Nucleus/physiology , Blotting, Western , Cell Line, Tumor , DNA Primers/genetics , DNA-Binding Proteins/metabolism , Fatty Acids, Unsaturated , Gene Expression Regulation/genetics , Humans , Immunoprecipitation , Karyopherins/metabolism , Luciferases , Male , Mass Spectrometry , Microscopy, Fluorescence , Polymerase Chain Reaction , Proteomics/methods , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins , Exportin 1 Protein
12.
Cancer ; 119(14): 2532-40, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23605249

ABSTRACT

BACKGROUND: Androgen receptor (AR) expression in breast cancers may serve as a prognostic and predictive marker. We examined the expression pattern of AR and its phosphorylated forms, Ser-213 (AR-Ser[P]-213) and Ser-650 (AR-Ser[P]-650), in breast cancer and evaluated their association with clinicopathological parameters. METHODS: Immunohistochemistry was performed on primary and distant metastatic breast cancers and benign breast tissue using antibodies against AR, AR-Ser(P)-213, and AR-Ser(P)-650. The levels of cytoplasmic and nuclear expression were scored semiquantitatively using a histoscore. RESULTS: Nuclear staining of AR was observed in all benign breast tissue and 67% of cancer cases. Nuclear and cytoplasmic AR-Ser(P)-213 was increased in breast cancers 2-fold (P = .0014) and 1.7-fold (P = .05), respectively, compared with benign controls, whereas nuclear and cytoplasmic AR-Ser(P)-650 expression was decreased in tumors by 1.9-fold and 1.7-fold (both P < .0001), respectively. Increased expression of nuclear or cytoplasmic AR-Ser(P)-213 was observed in metastatic breast cancers (1.3-fold, P = .05), ER-negative (2.6-fold, P = .001), and invasive ductal carcinoma (6.8-fold, P = .04). AR-Ser(P)-650 expression was downregulated in lymph node-positive breast cancers (1.4-fold, P = .02) but was upregulated in invasive ductal carcinomas (3.2-fold, P < .0001) and metastases (1.5-fold, P = .003). Moreover, in ER-negative breast cancers, nuclear AR-Ser(P)-650 was decreased (1.4-fold, P = .005), and cytoplasmic AR-Ser(P)-650 was increased (1.4-fold, P = .003). CONCLUSIONS: AR and its phosphorylation at serines 213 and 650 are differentially expressed in breast cancer tumorigenesis and progression. Phosphorylation of AR at serines 213 and 650 is increased in ER-negative breast cancers, ductal carcinomas, and metastases and may have predictive value in breast cancer prognosis.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Receptors, Androgen/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Cell Nucleus/metabolism , Cytoplasm/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Lymphatic Metastasis , Middle Aged , Neoplasm Staging , Phosphorylation , Predictive Value of Tests , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Up-Regulation
13.
Genome Res ; 23(4): 581-91, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23403032

ABSTRACT

The androgen receptor (AR) is a mediator of both androgen-dependent and castration-resistant prostate cancers. Identification of cellular factors affecting AR transcriptional activity could in principle yield new targets that reduce AR activity and combat prostate cancer, yet a comprehensive analysis of the genes required for AR-dependent transcriptional activity has not been determined. Using an unbiased genetic approach that takes advantage of the evolutionary conservation of AR signaling, we have conducted a genome-wide RNAi screen in Drosophila cells for genes required for AR transcriptional activity and applied the results to human prostate cancer cells. We identified 45 AR-regulators, which include known pathway components and genes with functions not previously linked to AR regulation, such as HIPK2 (a protein kinase) and MED19 (a subunit of the Mediator complex). Depletion of HIPK2 and MED19 in human prostate cancer cells decreased AR target gene expression and, importantly, reduced the proliferation of androgen-dependent and castration-resistant prostate cancer cells. We also systematically analyzed additional Mediator subunits and uncovered a small subset of Mediator subunits that interpret AR signaling and affect AR-dependent transcription and prostate cancer cell proliferation. Importantly, targeting of HIPK2 by an FDA-approved kinase inhibitor phenocopied the effect of depletion by RNAi and reduced the growth of AR-positive, but not AR-negative, treatment-resistant prostate cancer cells. Thus, our screen has yielded new AR regulators including drugable targets that reduce the proliferation of castration-resistant prostate cancer cells.


Subject(s)
Genome-Wide Association Study , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA Interference , Receptors, Androgen/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cluster Analysis , Drosophila/genetics , Drosophila/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Mediator Complex/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic
14.
Am J Clin Exp Urol ; 1(1): 53-65, 2013.
Article in English | MEDLINE | ID: mdl-25374900

ABSTRACT

Chronic inflammation has been linked to cancer initiation and progression in a variety of tissues, yet the impact of acute and chronic inflammatory signaling on androgen receptor function has not been widely studied. In this report, we examine the impact of the inflammation-linked cytokine, interleukin-1ß on androgen receptor function in prostate cancer cells. We demonstrate that acute interleukin-1ß treatment inhibits the transcription of the androgen receptor gene itself, resulting in the reduction of androgen receptor protein levels. Interestingly, in cells subjected to chronic interleukin-1ß stimulation, the transcription of the androgen receptor gene is restored within a few cell passages and the cells acquire the ability to grow in the presence of the anti-androgen, bicalutamide. Importantly, the changes that accompany this loss of androgen receptor regulation and gain of anti-androgen resistance are stably heritable since once established, the phenotype is maintained even in the absence of exogenously added interleukin-1ß. Further, bicalutamide resistance correlates with increased transcription of androgen receptor target genes and histone H3K4 dimethylation at M-phase gene enhancers. Overall, our studies demonstrate a novel route to anti-androgen resistance upon exposure to an inflammatory cytokine and provide a new tool to further understand how anti-androgen resistance emerges under chronic inflammation.

15.
Mol Cell Biol ; 31(17): 3639-52, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21730289

ABSTRACT

Androgen receptor (AR)-mediated transcription is modulated by interaction with coregulatory proteins. We demonstrate that the unconventional prefoldin RPB5 interactor (URI) is a new regulator of AR transcription and is critical for antagonist (bicalutamide) action. URI is phosphorylated upon androgen treatment, suggesting communication between the URI and AR signaling pathways. Whereas depletion of URI enhances AR-mediated gene transcription, overexpression of URI suppresses AR transcriptional activation and anchorage-independent prostate cancer cell growth. Repression of AR-mediated transcription is achieved, in part, by URI binding and regulation of androgen receptor trapped clone 27 (Art-27), a previously characterized AR corepressor. Consistent with this idea, genome-wide expression profiling in prostate cancer cells upon depletion of URI or Art-27 reveals substantially overlapping patterns of gene expression. Further, depletion of URI increases the expression of the AR target gene NKX-3.1, decreases the recruitment of Art-27, and increases AR occupancy at the NKX-3.1 promoter. While Art-27 can bind AR directly, URI is bound to chromatin prior to hormone-dependent recruitment of AR, suggesting a role for URI in modulating AR recruitment to target genes.


Subject(s)
Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Receptors, Androgen/genetics , Transcription, Genetic/genetics , Androgen Antagonists/pharmacology , Anilides/pharmacology , Blotting, Western , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/metabolism , Chromatin Immunoprecipitation , Gene Expression Profiling , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Metribolone/pharmacology , Molecular Chaperones , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nitriles/pharmacology , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Prostatic Neoplasms/embryology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Binding , RNA Interference , Receptors, Androgen/metabolism , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Tosyl Compounds/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects
16.
Endocr Relat Cancer ; 18(2): 245-55, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21317204

ABSTRACT

Multiple lines of evidence suggest a functional link between the androgen receptor (AR) and the serine/threonine kinase Akt in the development and progression of prostate cancer. To investigate the impact of Akt activity on AR homeostasis, we treated androgen-dependent LNCaP and LAPC-4 prostate cancer cells with Akt inhibitor. Akt inhibition decreased AR expression, suggesting that Akt activity was required for regulation of AR protein levels. However, while androgen-independent LNCaP-abl cells also showed diminished AR protein levels in response to Akt inhibition, treatment of androgen-independent LNCaP-AI cells failed to alter AR protein levels upon similar treatment, suggesting that AR protein levels in these androgen-independent prostate cells were regulated by mechanisms independent of Akt activation. Regulation of AR, downstream of activated Akt, also was observed in vivo when examining transgenic mice that overexpress constitutively active mutant myristoylated (myr)-Akt1 in the prostate. Transgenic mice expressing activated myr-Akt1 exhibited higher levels of AR mRNA and protein. Expression of activated myr-Akt1 did not alter prostate cell growth and no significant size differences between prostate tissues derived from transgenic animals were observed when comparing transgenic mice with wild-type mice. Still, transgenic mice overexpressing Akt exhibited higher levels of γH2AX and phosphorylated Chk2 in prostate tissue. These changes in markers associated with oncogene-induced senescence confirmed significant altered signaling in the transgenic mouse model. Overall, results presented here suggest that AR levels are regulated by the Akt pathway.


Subject(s)
Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/biosynthesis , Androgens/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Checkpoint Kinase 2 , Chromones/pharmacology , Histones/biosynthesis , Humans , Male , Metribolone/pharmacology , Mice , Mice, Transgenic , Morpholines/pharmacology , Prostate/anatomy & histology , Prostate/drug effects , Prostate/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
17.
Cancer Res ; 69(7): 3140-7, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19318562

ABSTRACT

The androgen receptor (AR) directs diverse biological processes through interaction with coregulators such as AR trapped clone-27 (ART-27). Our results show that ART-27 is recruited to AR-binding sites by chromatin immunoprecipitation analysis. In addition, the effect of ART-27 on genome-wide transcription was examined. The studies indicate that loss of ART-27 enhances expression of many androgen-regulated genes, suggesting that ART-27 inhibits gene expression. Surprisingly, classes of genes that are up-regulated upon ART-27 depletion include regulators of DNA damage checkpoint and cell cycle progression, suggesting that ART-27 functions to keep expression levels of these genes low. Consistent with this idea, stable reduction of ART-27 by short-hairpin RNA enhances LNCaP cell proliferation compared with control cells. The effect of ART-27 loss was also examined in response to the antiandrogen bicalutamide. Unexpectedly, cells treated with ART-27 siRNA no longer exhibited gene repression in response to bicalutamide. To examine ART-27 loss in prostate cancer progression, immunohistochemistry was conducted on a tissue array containing samples from primary tumors of individuals who were clinically followed and later shown to have either recurrent or nonrecurrent disease. Comparison of ART-27 and AR staining indicated that nuclear ART-27 expression was lost in the majority of AR-positive recurrent prostate cancers. Our studies show that reduction of ART-27 protein levels in prostate cancer may facilitate antiandrogen-resistant disease.


Subject(s)
Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Androgen Antagonists/pharmacology , Androgens/genetics , Anilides/pharmacology , Cell Cycle Proteins , Cell Growth Processes/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Molecular Chaperones , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Nitriles/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/biosynthesis , Tosyl Compounds/pharmacology
18.
Methods Mol Biol ; 505: 221-35, 2009.
Article in English | MEDLINE | ID: mdl-19117148

ABSTRACT

Protein phosphorylation is a versatile posttranslational modification that can regulate nuclear receptor function. Although the precise role of receptor phosphorylation is not fully understood, it appears that it functions to direct or refine receptor activity in response to particular physiological requirements. Identifying and characterizing specific nuclear receptor phosphorylation sites is an important step in elucidating the role(s) receptor phosphorylation plays in function. Although traditional methods of metabolic labeling and in vitro protein phosphorylation have been informative, receptor phosphorylation site-specific antibodies are simple and reliable tools to study receptor phosphorylation. This chapter will discuss how to develop nuclear receptor phosphorylation site-specific antibodies to elucidate function.


Subject(s)
Antibodies, Phospho-Specific , Protein Engineering/methods , Receptors, Cytoplasmic and Nuclear/chemistry , Amino Acid Sequence , Animals , Antibodies, Phospho-Specific/chemistry , Antibodies, Phospho-Specific/metabolism , Antibody Specificity , Biological Assay/methods , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Liver X Receptors , Molecular Sequence Data , Orphan Nuclear Receptors , Phosphorylation , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Reproducibility of Results
19.
J Biol Chem ; 280(49): 40916-24, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16210317

ABSTRACT

The biological ramifications of phosphorylation of the androgen receptor (AR) are largely unknown. To examine the phosphorylation of AR at serine 213, a putative substrate for Akt, a phosphorylation site-specific antibody was generated. The use of this antibody indicated that AR Ser-213 is phosphorylated in vivo and that phosphorylation is tightly regulated in a cell type-specific manner. Furthermore, Ser-213 phosphorylation took place with rapid kinetics and was inhibited by the phosphatidylinositol 3-kinase inhibitor LY294002. Phosphorylation occurred in response to R1881 and dihydrotestosterone but weakly if at all in response to testosterone. It did not occur in response to AR antagonists or growth factor stimulation in the absence of an AR agonist. Transcription assays using an AR-responsive reporter gene construct showed that activated phosphatidylinositol 3-kinase inhibited transcription mediated by wild type AR but not that of a mutant AR variant (S213A), which could not be phosphorylated at Ser-213. By immunohistochemistry, the AR Ser(P)-213 antigen was detected in prostate epithelial but not stromal cells despite the fact that an antibody recognizing both phosphorylated and non-phosphorylated forms of AR demonstrates that AR is present in both cell types as expected. In fetal tissue the AR-Ser(P)-213 antigen was present in epithelial cells of the urogenital sinus when endogenous androgen levels were high and activated Akt was prevalent, but absent at a later stage of development when endogenous androgen levels were low and Akt activation was minimal. Immunoreactivity was evident in differentiated cells lining the lumen of the urogenital sinus but not in rapidly dividing, Ki67 positive cells within the developing prostate or stromal tissue, suggesting that site-specific phosphorylation of AR Ser-213 by cellular kinases occurs in a non-proliferating cellular milieu.


Subject(s)
Homeostasis , Prostate/cytology , Protein Kinases/metabolism , Receptors, Androgen/metabolism , Amino Acid Sequence , Antibody Specificity , Cell Division , Cell Line , Chromones/pharmacology , Dihydrotestosterone/pharmacology , Embryo, Mammalian , Epidermal Growth Factor/pharmacology , Epithelial Cells/chemistry , Fetus/chemistry , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunohistochemistry , Insulin-Like Growth Factor I/pharmacology , Ki-67 Antigen/analysis , Kidney , Male , Metribolone/pharmacology , Molecular Sequence Data , Morpholines/pharmacology , Mutation , Peptide Fragments/chemistry , Peptide Fragments/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Prostate/chemistry , Prostate/embryology , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Serine/metabolism , Stromal Cells/chemistry , Structure-Activity Relationship , Transcription, Genetic , Transfection
20.
Mol Endocrinol ; 19(9): 2273-82, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15919721

ABSTRACT

The transcriptional activity of the androgen receptor (AR) is modulated by interactions with coregulatory molecules. It has been proposed that aberrant interactions between AR and its coregulators may contribute to diseases related to AR activity, such as prostate cancer and androgen insensitivity syndrome (AIS); however, evidence linking abnormal receptor-cofactor interactions to disease is scant. ART-27 is a recently identified AR N-terminal coactivator that is associated with AR-mediated growth inhibition. Here we analyze a number of naturally occurring AR mutations identified in prostate cancer and AIS for their ability to affect AR response to ART-27. Although the vast majority of AR mutations appeared capable of increased activation in response to ART-27, an AR mutation identified in prostate cancer (AR P340L) and AIS (AR E2K) show reduced transcriptional responses to ART-27, whereas their response to the p160 class of coactivators was not diminished. Relative to the wild-type receptor, less ART-27 protein associated with the AR E2K substitution, consistent with reduced transcriptional response. Surprisingly, more ART-27 associated with AR P340L, despite the fact that the mutation decreased transcriptional activation in response to ART-27. Our findings suggest that aberrant AR-coactivator association interferes with normal ART-27 coactivator function, resulting in suppression of AR activity, and may contribute to the pathogenesis of diseases related to alterations in AR activity, such as prostate cancer and AIS.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Mutation , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Trans-Activators/metabolism , Amino Acid Substitution , Androgen-Insensitivity Syndrome/metabolism , Cell Cycle Proteins , Humans , Male , Molecular Chaperones , Neoplasm Proteins , Prostatic Neoplasms/metabolism , Protein Structure, Tertiary , Receptors, Androgen/chemistry , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...