73 results on '"Burnstein KL"'
Search Results
2. Challenges and opportunities in training cancer researchers.
- Author
-
Keith B, Welch DR, Agarwal N, Blair C, Chukwudozie IB, Shevde LA, Boise LH, Burnstein KL, Antalis TM, O'Connor KL, Dalal SN, Ellis S, and Nakshatri H
- Abstract
Competing Interests: Competing interests: The authors declare no competing interests.
- Published
- 2025
- Full Text
- View/download PDF
3. Pan-Cancer Drug Sensitivity Prediction from Gene Expression using Deep Learning.
- Author
-
Ocasio BA, Hu J, Stathias V, Martinez MJ, Burnstein KL, and Schürer SC
- Abstract
Cancer is a group of complex diseases, with tumor heterogeneity, durable drug efficacy, emerging resistance, and host toxicity presenting major challenges to the development of effective cancer therapeutics. While traditionally used methods have remained limited in their capacity to overcome these challenges in cancer drug development, efforts have been made in recent years toward applying "big data" to cancer research and precision oncology. By curating, standardizing, and integrating data from various databases, we developed deep learning architectures that use perturbation and baseline transcriptional signatures to predict efficacious small molecule compounds and genetic dependencies in cancer. A series of internal validations followed by prospective validation in prostate cancer cell lines were performed to ensure consistent performance and model applicability. We report SensitivitySeq , a novel bioinformatics tool for prioritizing small molecule compounds and gene dependencies in silico to drive the development of targeted therapies for cancer. To the best of our knowledge, this is the first supervised deep learning approach, validated in vitro , to predict drug sensitivity using baseline cancer cell line gene expression alongside cell line-independent perturbation-response consensus signatures., Competing Interests: Declaration of interests The authors declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
4. International Union of Basic and Clinical Pharmacology CXIII: Nuclear Receptor Superfamily-Update 2023.
- Author
-
Burris TP, de Vera IMS, Cote I, Flaveny CA, Wanninayake US, Chatterjee A, Walker JK, Steinauer N, Zhang J, Coons LA, Korach KS, Cain DW, Hollenberg AN, Webb P, Forrest D, Jetten AM, Edwards DP, Grimm SL, Hartig S, Lange CA, Richer JK, Sartorius CA, Tetel M, Billon C, Elgendy B, Hegazy L, Griffett K, Peinetti N, Burnstein KL, Hughes TS, Sitaula S, Stayrook KR, Culver A, Murray MH, Finck BN, and Cidlowski JA
- Subjects
- Humans, Receptors, Cytoplasmic and Nuclear metabolism, Transcription Factors metabolism, Carrier Proteins, Ligands, Pharmacology, Clinical
- Abstract
The NR superfamily comprises 48 transcription factors in humans that control a plethora of gene network programs involved in a wide range of physiologic processes. This review will summarize and discuss recent progress in NR biology and drug development derived from integrating various approaches, including biophysical techniques, structural studies, and translational investigation. We also highlight how defective NR signaling results in various diseases and disorders and how NRs can be targeted for therapeutic intervention via modulation via binding to synthetic lipophilic ligands. Furthermore, we also review recent studies that improved our understanding of NR structure and signaling. SIGNIFICANCE STATEMENT: Nuclear receptors (NRs) are ligand-regulated transcription factors that are critical regulators of myriad physiological processes. NRs serve as receptors for an array of drugs, and in this review, we provide an update on recent research into the roles of these drug targets., (U.S. Government work not protected by U.S. copyright.)
- Published
- 2023
- Full Text
- View/download PDF
5. Automation, live-cell imaging, and endpoint cell viability for prostate cancer drug screens.
- Author
-
Lyles RDZ, Martinez MJ, Sherman B, Schürer S, and Burnstein KL
- Subjects
- Male, Humans, Androgens metabolism, Reproducibility of Results, Androgen Antagonists therapeutic use, Cell Survival, Cell Line, Tumor, Receptors, Androgen metabolism, Automation, Prostatic Neoplasms, Castration-Resistant metabolism
- Abstract
Androgen deprivation therapy (ADT) is the standard of care for high risk and advanced prostate cancer; however, disease progression from androgen-dependent prostate cancer (ADPC) to lethal and incurable castration-resistant prostate cancer (CRPC) and (in a substantial minority of cases) neuroendocrine prostate cancer (NEPC) is common. Identifying effective targeted therapies is challenging because of acquired resistance to established treatments and the vast heterogeneity of advanced prostate cancer (PC). To streamline the identification of potentially active prostate cancer therapeutics, we have developed an adaptable semi-automated protocol which optimizes cell growth and leverages automation to enhance robustness, reproducibility, and throughput while integrating live-cell imaging and endpoint viability assays to assess drug efficacy in vitro. In this study, culture conditions for 72-hr drug screens in 96-well plates were established for a large, representative panel of human prostate cell lines including: BPH-1 and RWPE-1 (non-tumorigenic), LNCaP and VCaP (ADPC), C4-2B and 22Rv1 (CRPC), DU 145 and PC3 (androgen receptor-null CRPC), and NCI-H660 (NEPC). The cell growth and 72-hr confluence for each cell line was optimized for real-time imaging and endpoint viability assays prior to screening for novel or repurposed drugs as proof of protocol validity. We demonstrated effectiveness and reliability of this pipeline through validation of the established finding that the first-in-class BET and CBP/p300 dual inhibitor EP-31670 is an effective compound in reducing ADPC and CRPC cell growth. In addition, we found that insulin-like growth factor-1 receptor (IGF-1R) inhibitor linsitinib is a potential pharmacological agent against highly lethal and drug-resistant NEPC NCI-H660 cells. This protocol can be employed across other cancer types and represents an adaptable strategy to optimize assay-specific cell growth conditions and simultaneously assess drug efficacy across multiple cell lines., Competing Interests: The authors have declared that no competing interests exist., (Copyright: This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.)
- Published
- 2023
- Full Text
- View/download PDF
6. Inhibition of the serine/threonine kinase BUB1 reverses taxane resistance in prostate cancer.
- Author
-
Martinez MJ, Lyles RDZ, Peinetti N, Grunfeld AM, and Burnstein KL
- Abstract
Men with incurable castration resistant prostate cancer (CRPC) are typically treated with taxanes; however, drug resistance rapidly develops. We previously identified a clinically relevant seven gene network in aggressive CRPC, which includes the spindle assembly checkpoint (SAC) kinase BUB1. Since SAC is deregulated in taxane resistant PC, we evaluated BUB1 and found that it was over-expressed in advanced PC patient datasets and taxane resistant PC cells. Treatment with a specific BUB1 kinase inhibitor re-sensitized resistant CRPC cells, including cells expressing constitutively active androgen receptor (AR) variants, to clinically used taxanes. Consistent with a role of AR variants in taxane resistance, ectopically expressed AR-V7 increased BUB1 levels and reduced sensitivity to taxanes. This work shows that disruption of BUB1 kinase activity reverted resistance to taxanes, which is essential to advancing BUB1 as a potential therapeutic target for intractable chemotherapy resistant CRPC including AR variant driven CRPC, which lacks durable treatment options., Competing Interests: The authors declare no competing interests., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
7. Accurate and Early Metastases Diagnosis in Live Animals With Multimodal X-ray and Optical Imaging.
- Author
-
Chen J, Zhao N, Copello V, Ru Y, Burnstein KL, and Yang Y
- Subjects
- Animals, Male, Mice, Prostate, Tomography, X-Ray Computed, X-Rays, Prostatic Neoplasms diagnostic imaging, Tomography, Optical methods
- Abstract
Purpose: In vivo optical imaging systems are essential to track disease progression and evaluate therapeutic efficacy in animal studies. However, current approaches are limited by their inability to accurately capture 3-dimensional (3-D) image information. To overcome this hindrance, we adopted x-ray computed tomography (CT) as a prior for 3-D optical image reconstruction and further challenged the multimodal imaging performance with a metastasis model., Methods and Materials: The iSMAART system, an integrated small animal research platform, features coregistered high-quality quantitative optical tomography and CT. In the synergistic dual-modality imaging, CT provides both 3-D anatomy information and animal structure mesh for optical tomography reconstruction, which is performed using bioluminescence projections acquired from 4 orthogonal angles. The multimodal imaging system was challenged with a prostate cancer metastasis model, and a double-blind histopathology diagnosis was obtained to validate the imaging results., Results: The iSMAART located, visualized, and quantified early tumor metastases at the millimeter scale, and can accurately track deep tumors as small as 1.5 mm in live animals. Tumors metastasized into the liver, diaphragm, and tibia in 4 mice were all successfully diagnosed by the integrated tomographic imaging., Conclusions: Instead of roughly comparing surface-light intensities, as traditionally performed in 2-dimensional optical imaging, iSMAART provides accurate tumor imaging and quantitative assessment capabilities with integrated CT and optical tomography for cancer metastasis research. With the powerful 3-D optical/CT imaging capability, iSMAART has the potential to tackle more complex research needs with higher targeting accuracy., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
8. Is androgen receptor activity in metastatic prostate cancer a good biomarker for bipolar androgen therapy?
- Author
-
Peinetti N, Bilusic M, and Burnstein KL
- Subjects
- Male, Humans, Androgens, Receptors, Androgen genetics, Androgen Antagonists pharmacology, Androgen Antagonists therapeutic use, Biomarkers, Prostatic Neoplasms drug therapy, Prostatic Neoplasms genetics
- Abstract
Androgen deprivation therapy (ADT) is the longstanding treatment for advanced prostate cancer (PC) because androgen receptor (AR) is the key therapeutic vulnerability for this disease. Bipolar androgen therapy (BAT) - the rapid cycling of supraphysiologic androgen (SPA) and low serum testosterone levels - is an alternative concept, but not all patients respond and acquired resistance can occur. In this issue of the JCI, Sena et al. developed a gene signature indicative of high AR activity to predict patient response to BAT, including a decline in both serum prostate-specific antigen (PSA) and tumor volume. Preclinical models showed that AR-mediated suppression of MYC, known to drive PC, was associated with decreased cell growth following SPA treatment. Because BAT eventually leads to resistance, the authors tested cycling between SPA and AR antagonism in a patient-derived xenograft and observed a delay in tumor growth. These findings represent a major step toward the informed use of BAT for advanced PC.
- Published
- 2022
- Full Text
- View/download PDF
9. Exploiting Dependence of Castration-Resistant Prostate Cancer on the Arginine Vasopressin Signaling Axis by Repurposing Vaptans.
- Author
-
Heidman LM, Peinetti N, Copello VA, and Burnstein KL
- Subjects
- Androgen Antagonists, Arginine Vasopressin therapeutic use, Cell Line, Tumor, Humans, Male, Neoplasm Recurrence, Local, Receptors, Androgen metabolism, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant metabolism, Receptors, Vasopressin genetics, Receptors, Vasopressin metabolism
- Abstract
Men with advanced prostate cancer are treated by androgen deprivation therapy but the disease recurs as incurable castration-resistant prostate cancer (CRPC), requiring new treatment options. We previously demonstrated that the G protein-coupled receptor (GPCR) arginine vasopressin receptor type1A (AVPR1A) is expressed in CRPC and promotes castration-resistant growth in vitro and in vivo. AVPR1A is part of a family of GPCR's including arginine vasopressin receptor type 2 (AVPR2). Interrogation of prostate cancer patient sample data revealed that coexpression of AVPR1A and AVPR2 is highly correlated with disease progression. Stimulation of AVPR2 with a selective agonist desmopressin promoted CRPC cell proliferation through cAMP/protein kinase A signaling, consistent with AVPR2 coupling to the G protein subunit alpha s. In contrast, blocking AVPR2 with a selective FDA-approved antagonist, tolvaptan, reduced cell growth. In CRPC xenografts, antagonizing AVPR2, AVPR1A, or both significantly reduced CRPC tumor growth as well as decreased on-target markers of tumor burden. Combinatorial use of AVPR1A and AVPR2 antagonists promoted apoptosis synergistically in CRPC cells. Furthermore, we found that castration-resistant cells produced AVP, the endogenous ligand for arginine vasopressin receptors, and knockout of AVP in CRPC cells significantly reduced proliferation suggesting possible AVP autocrine signaling. These data indicate that the AVP/arginine vasopressin receptor signaling axis represents a promising and clinically actionable target for CRPC., Implications: The arginine vasopressin signaling axis in CRPC provides a therapeutic window that is targetable through repurposing safe and effective AVPR1A and AVPR2 antagonists., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
10. The kinesin KIF20A promotes progression to castration-resistant prostate cancer through autocrine activation of the androgen receptor.
- Author
-
Copello VA and Burnstein KL
- Subjects
- Androgen Antagonists pharmacology, Androgens metabolism, Cell Line, Tumor, Humans, Male, Neoplasm Recurrence, Local, Kinesins genetics, Prostatic Neoplasms, Castration-Resistant metabolism, Receptors, Androgen genetics, Receptors, Androgen metabolism
- Abstract
Prostate cancer that recurs following androgen-deprivation therapy is termed castration-resistant, which is incurable and is marked by reactivation of androgen receptor (AR) signaling. KIF20A, a kinesin with unique structural features, is overexpressed in human castration-resistant prostate cancer (CRPC) compared to androgen-dependent PC and benign tissue. KIF20A has well-described roles in mitotic processes, but it has a less characterized function in vesicle fission and trafficking within Golgi-driven secretory pathways. Stable expression of KIF20A in androgen-dependent PC cells promoted progression to CRPC through the activation of AR signaling in vitro and in vivo. KIF20A expression resulted in the secretion of autocrine factors in the conditioned media that activated AR and caused castration-resistant proliferation of naïve androgen-dependent cells. Pharmacologic disruption of vesicle biogenesis blocked KIF20A-driven castration-resistant proliferation of androgen-dependent PC. KIF20A depletion or treatment with the KIF20A-specific inhibitor, paprotrain, reduced CRPC. These data are the first to establish KIF20A as a driver of CRPC progression through AR activation and as a promising therapeutic target against CRPC., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
11. Arginine vasopressin receptor 1a is a therapeutic target for castration-resistant prostate cancer.
- Author
-
Zhao N, Peacock SO, Lo CH, Heidman LM, Rice MA, Fahrenholtz CD, Greene AM, Magani F, Copello VA, Martinez MJ, Zhang Y, Daaka Y, Lynch CC, and Burnstein KL
- Subjects
- Animals, Calcium metabolism, Cell Line, Tumor, Cell Proliferation genetics, Cyclic AMP Response Element-Binding Protein metabolism, Drug Resistance, Neoplasm genetics, Gene Expression Regulation, Neoplastic, Humans, Indoles pharmacology, Indoles therapeutic use, MAP Kinase Signaling System drug effects, Male, Mice, Nude, Osteogenesis drug effects, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Proto-Oncogene Proteins c-vav metabolism, Pyrrolidines pharmacology, Pyrrolidines therapeutic use, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Androgen metabolism, Receptors, Vasopressin genetics, Molecular Targeted Therapy, Prostatic Neoplasms, Castration-Resistant drug therapy, Receptors, Vasopressin metabolism
- Abstract
Castration-resistant prostate cancer (CRPC) recurs after androgen deprivation therapy (ADT) and is incurable. Reactivation of androgen receptor (AR) signaling in the low androgen environment of ADT drives CRPC. This AR activity occurs through a variety of mechanisms, including up-regulation of AR coactivators such as VAV3 and expression of constitutively active AR variants such as the clinically relevant AR-V7. AR-V7 lacks a ligand-binding domain and is linked to poor prognosis. We previously showed that VAV3 enhances AR-V7 activity to drive CRPC progression. Gene expression profiling after depletion of either VAV3 or AR-V7 in CRPC cells revealed arginine vasopressin receptor 1a ( AVPR1A ) as the most commonly down-regulated gene, indicating that this G protein-coupled receptor may be critical for CRPC. Analysis of publicly available human PC datasets showed that AVPR1A has a higher copy number and increased amounts of mRNA in advanced PC. Depletion of AVPR1A in CRPC cells resulted in decreased cell proliferation and reduced cyclin A. In contrast, androgen-dependent PC, AR-negative PC, or nontumorigenic prostate epithelial cells, which have undetectable AVPR1A mRNA, were minimally affected by AVPR1A depletion. Ectopic expression of AVPR1A in androgen-dependent PC cells conferred castration resistance in vitro and in vivo. Furthermore, treatment of CRPC cells with the AVPR1A ligand, arginine vasopressin (AVP), activated ERK and CREB, known promoters of PC progression. A clinically safe and selective AVPR1A antagonist, relcovaptan, prevented CRPC emergence and decreased CRPC orthotopic and bone metastatic growth in mouse models. Based on these preclinical findings, repurposing AVPR1A antagonists is a promising therapeutic approach for CRPC., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
12. Reduced Arginyltransferase 1 is a driver and a potential prognostic indicator of prostate cancer metastasis.
- Author
-
Birnbaum MD, Zhao N, Moorthy BT, Patel DM, Kryvenko ON, Heidman L, Kumar A, Morgan WM, Ban Y, Reis IM, Chen X, Gonzalgo ML, Jorda M, Burnstein KL, and Zhang F
- Subjects
- Aminoacyltransferases genetics, Animals, HEK293 Cells, Humans, Male, Mice, Mice, Knockout, Neoplasm Metastasis, Neoplasm Proteins genetics, Prognosis, Prostatic Neoplasms diagnosis, Prostatic Neoplasms pathology, Aminoacyltransferases metabolism, Cell Movement, Neoplasm Proteins metabolism, Prostatic Neoplasms enzymology, Prostatic Neoplasms metabolism
- Abstract
Most prostate cancer cases remain indolent for long periods of time, but metastatic progression quickly worsens the prognosis and leads to mortality. However, little is known about what promotes the metastasis of prostate cancer and there is a lack of effective prognostic indicators, making it immensely difficult to manage options for treatment or surveillance. Arginyltransferase 1 (Ate1) is the enzyme mediating post-translational protein arginylation, which has recently been identified as a master regulator affecting many cancer-relevant pathways including stress response, cell cycle checkpoints, and cell migration/adhesion. However, the precise role of Ate1 in cancer remains unknown. In this study, we found the occurrence of metastasis of prostate cancer is inversely correlated with the levels of Ate1 protein and mRNA in the primary tumor. We also found that metastatic prostate cancer cell lines have a reduced level of Ate1 protein compared to non-metastatic cell lines, and that a depletion of Ate1 drives prostate cancer cells towards more aggressive pro-metastatic phenotypes without affecting proliferation rates. Furthermore, we demonstrated that a reduction of Ate1 can result from chronic stress, and that shRNA-reduced Ate1 increases cellular resistance to stress, and drives spontaneous and stress-induced genomic mutations. Finally, by using a prostate orthotropic xenograft mouse model, we found that a reduction of Ate1 was sufficient to enhance the metastatic phenotypes of prostate cancer cell line PC-3 in vivo. Our study revealed a novel role of Ate1 in suppressing prostate cancer metastasis, which has a profound significance for establishing metastatic indicators for prostate cancer, and for finding potential treatments to prevent its metastasis.
- Published
- 2019
- Full Text
- View/download PDF
13. Alterations of tumor microenvironment by nitric oxide impedes castration-resistant prostate cancer growth.
- Author
-
Arora H, Panara K, Kuchakulla M, Kulandavelu S, Burnstein KL, Schally AV, Hare JM, and Ramasamy R
- Subjects
- Animals, Cell Differentiation physiology, Cell Line, Tumor, Gene Expression Regulation, Neoplastic physiology, Inflammation metabolism, Inflammation pathology, MAP Kinase Signaling System physiology, Macrophages metabolism, Macrophages pathology, Male, Mice, Mice, Inbred C57BL, Nitric Oxide Donors metabolism, Tumor Burden physiology, Xenograft Model Antitumor Assays methods, Cell Proliferation physiology, Nitric Oxide metabolism, Prostatic Neoplasms, Castration-Resistant metabolism, Prostatic Neoplasms, Castration-Resistant pathology, Tumor Microenvironment physiology
- Abstract
Immune targeted therapy of nitric oxide (NO) synthases are being considered as a potential frontline therapeutic to treat patients diagnosed with locally advanced and metastatic prostate cancer. However, the role of NO in castration-resistant prostate cancer (CRPC) is controversial because NO can increase in nitrosative stress while simultaneously possessing antiinflammatory properties. Accordingly, we tested the hypothesis that increased NO will lead to tumor suppression of CRPC through tumor microenvironment. S-nitrosoglutathione (GSNO), an NO donor, decreased the tumor burden in murine model of CRPC by targeting tumors in a cell nonautonomous manner. GSNO inhibited both the abundance of antiinflammatory (M2) macrophages and expression of pERK, indicating that tumor-associated macrophages activity is influenced by NO. Additionally, GSNO decreased IL-34, indicating suppression of tumor-associated macrophage differentiation. Cytokine profiling of CRPC tumor grafts exposed to GSNO revealed a significant decrease in expression of G-CSF and M-CSF compared with grafts not exposed to GSNO. We verified the durability of NO on CRPC tumor suppression by using secondary xenograft murine models. This study validates the significance of NO on inhibition of CRPC tumors through tumor microenvironment (TME). These findings may facilitate the development of previously unidentified NO-based therapy for CRPC., Competing Interests: Conflict of interest: J.M.H. discloses a relationship with Vestion Inc. that includes equity, board membership, and consulting. J.M.H. is the Chief Scientific Officer, a compensated consultant, and advisory board member for Longeveron and holds equity in Longeveron. J.M.H. is also the coinventor of intellectual property licensed to Longeveron.
- Published
- 2018
- Full Text
- View/download PDF
14. Identification of an oncogenic network with prognostic and therapeutic value in prostate cancer.
- Author
-
Magani F, Bray ER, Martinez MJ, Zhao N, Copello VA, Heidman L, Peacock SO, Wiley DJ, D'Urso G, and Burnstein KL
- Subjects
- Cell Line, Tumor, Disease-Free Survival, Drug Resistance, Neoplasm genetics, Gene Expression Profiling methods, Gene Expression Regulation, Neoplastic, Humans, Male, Prognosis, Prostatic Neoplasms pathology, RNA Splicing genetics, Receptors, Androgen chemistry, Schizosaccharomyces genetics, Signal Transduction genetics, Carcinogenesis genetics, Cell Proliferation genetics, Prostatic Neoplasms genetics, Receptors, Androgen genetics
- Abstract
Identifying critical pathways governing disease progression is essential for accurate prognosis and effective therapy. We developed a broadly applicable and novel systems-level gene discovery strategy. This approach focused on constitutively active androgen receptor (AR) splice variant-driven pathways as representative of an intractable mechanism of prostate cancer (PC) therapeutic resistance. We performed a meta-analysis of human prostate samples using weighted gene co-expression network analysis combined with experimental AR variant transcriptome analyses. An AR variant-driven gene module that is upregulated during human PC progression was identified. We filtered this module by identifying genes that functionally interacted with AR variants using a high-throughput synthetic genetic array screen in Schizosaccharomyces pombe This strategy identified seven AR variant-regulated genes that also enhance AR activity and drive cancer progression. Expression of the seven genes predicted poor disease-free survival in large independent PC patient cohorts. Pharmacologic inhibition of interacting members of the gene set potently and synergistically decreased PC cell proliferation. This unbiased and novel gene discovery strategy identified a clinically relevant, oncogenic, interacting gene hub with strong prognostic and therapeutic potential in PC., (© 2018 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2018
- Full Text
- View/download PDF
15. Targeting AR Variant-Coactivator Interactions to Exploit Prostate Cancer Vulnerabilities.
- Author
-
Magani F, Peacock SO, Rice MA, Martinez MJ, Greene AM, Magani PS, Lyles R, Weitz JR, and Burnstein KL
- Subjects
- Alternative Splicing, Binding Sites, Cell Line, Tumor, Cell Movement, Cell Proliferation, Cell Survival, Gene Expression Regulation, Neoplastic, Humans, Male, Mutation, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant therapy, Protein Binding, Proto-Oncogene Proteins c-vav chemistry, Receptors, Androgen chemistry, Signal Transduction, Up-Regulation, Prostatic Neoplasms, Castration-Resistant metabolism, Proto-Oncogene Proteins c-vav genetics, Proto-Oncogene Proteins c-vav metabolism, Receptors, Androgen genetics, Receptors, Androgen metabolism
- Abstract
Castration-resistant prostate cancer (CRPC) progresses rapidly and is incurable. Constitutively active androgen receptor splice variants (AR-Vs) represent a well-established mechanism of therapeutic resistance and disease progression. These variants lack the AR ligand-binding domain and, as such, are not inhibited by androgen deprivation therapy (ADT), which is the standard systemic approach for advanced prostate cancer. Signaling by AR-Vs, including the clinically relevant AR-V7, is augmented by Vav3, an established AR coactivator in CRPC. Using mutational and biochemical studies, we demonstrated that the Vav3 Diffuse B-cell lymphoma homology (DH) domain interacted with the N-terminal region of AR-V7 (and full length AR). Expression of the Vav3 DH domain disrupted Vav3 interaction with and enhancement of AR-V7 activity. The Vav3 DH domain also disrupted AR-V7 interaction with other AR coactivators: Src1 and Vav2, which are overexpressed in PC. This Vav3 domain was used in proof-of-concept studies to evaluate the effects of disrupting the interaction between AR-V7 and its coactivators on CRPC cells. This disruption decreased CRPC cell proliferation and anchorage-independent growth, caused increased apoptosis, decreased migration, and resulted in the acquisition of morphological changes associated with a less aggressive phenotype. While disrupting the interaction between FL-AR and its coactivators decreased N-C terminal interaction, disrupting the interaction of AR-V7 with its coactivators decreased AR-V7 nuclear levels. Implications: This study demonstrates the potential therapeutic utility of inhibiting constitutively active AR-V signaling by disrupting coactivator binding. Such an approach is significant, as AR-Vs are emerging as important drivers of CRPC that are particularly recalcitrant to current therapies. Mol Cancer Res; 15(11); 1469-80. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
16. Thioredoxin-1 protects against androgen receptor-induced redox vulnerability in castration-resistant prostate cancer.
- Author
-
Samaranayake GJ, Troccoli CI, Huynh M, Lyles RDZ, Kage K, Win A, Lakshmanan V, Kwon D, Ban Y, Chen SX, Zarco ER, Jorda M, Burnstein KL, and Rai P
- Subjects
- Cell Death drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Disease Progression, Disulfides pharmacology, Humans, Imidazoles pharmacology, Male, Reactive Oxygen Species metabolism, Prostatic Neoplasms, Castration-Resistant metabolism, Prostatic Neoplasms, Castration-Resistant pathology, Receptors, Androgen metabolism, Thioredoxins metabolism
- Abstract
Androgen deprivation (AD) therapy failure leads to terminal and incurable castration-resistant prostate cancer (CRPC). We show that the redox-protective protein thioredoxin-1 (TRX1) increases with prostate cancer progression and in androgen-deprived CRPC cells, suggesting that CRPC possesses an enhanced dependency on TRX1. TRX1 inhibition via shRNA or a phase I-approved inhibitor, PX-12 (untested in prostate cancer), impedes the growth of CRPC cells to a greater extent than their androgen-dependent counterparts. TRX1 inhibition elevates reactive oxygen species (ROS), p53 levels and cell death in androgen-deprived CRPC cells. Unexpectedly, TRX1 inhibition also elevates androgen receptor (AR) levels under AD, and AR depletion mitigates both TRX1 inhibition-mediated ROS production and cell death, suggesting that AD-resistant AR expression in CRPC induces redox vulnerability. In vivo TRX1 inhibition via shRNA or PX-12 reverses the castration-resistant phenotype of CRPC cells, significantly inhibiting tumor formation under systemic AD. Thus, TRX1 is an actionable CRPC therapeutic target through its protection against AR-induced redox stress.
- Published
- 2017
- Full Text
- View/download PDF
17. The microRNA-23b/-27b cluster suppresses prostate cancer metastasis via Huntingtin-interacting protein 1-related.
- Author
-
Rice MA, Ishteiwy RA, Magani F, Udayakumar T, Reiner T, Yates TJ, Miller P, Perez-Stable C, Rai P, Verdun R, Dykxhoorn DM, and Burnstein KL
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Cell Line, Tumor, Cell Movement genetics, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, Microfilament Proteins, Neoplasm Invasiveness genetics, Neoplasm Metastasis, Prostate pathology, Prostatic Neoplasms pathology, Xenograft Model Antitumor Assays, MicroRNAs genetics, Prostatic Neoplasms genetics, Vesicular Transport Proteins genetics
- Abstract
Deregulation of microRNAs (miRs) contributes to progression and metastasis of prostate and other cancers. miR-23b and -27b, encoded in the same miR cluster (miR-23b/-27b), are downregulated in human metastatic prostate cancer compared with primary tumors and benign tissue. Expression of miR-23b/-27b decreases prostate cancer cell migration, invasion and results in anoikis resistance. Conversely, antagomiR-mediated miR-23b and -27b silencing produces the opposite result in a more indolent prostate cancer cell line. However, neither miR-23b/-27b expression or inhibition impacts prostate cancer cell proliferation suggesting that miR-23b/-27b selectively suppresses metastasis. To examine the effects of miR-23b/-27b on prostate cancer metastasis in vivo, orthotopic prostate xenografts were established using aggressive prostate cancer cells transduced with miR-23b/-27b or non-targeting control miRNA. Although primary tumor formation was similar between miR-23b/-27b-transduced cells and controls, miR-23b/-27b expression in prostate cancer cells decreased seminal vesicle invasion and distant metastases. Gene-expression profiling identified the endocytic adaptor, Huntingtin-interacting protein 1-related (HIP1R) as being downregulated by miR-23b/-27b. Increased HIP1R expression in prostate cancer cells inversely phenocopied the effects of miR-23b/-27b overexpression on migration, invasion and anchorage-independent growth. HIP1R rescued miR-23b/-27b-mediated repression of migration in prostate cancer cells. HIP1R mRNA levels were decreased in seminal vesicle tissue from mice bearing miR-23b/-27b-transduced prostate cancer cell xenografts compared with scrambled controls, suggesting HIP1R is a key functional target of miR-23b/-27b. In addition, depletion of HIP1R led to a more rounded, less mesenchymal-like cell morphology, consistent with decreased metastatic properties. Together, these data demonstrate that the miR-23b/-27b cluster functions as a metastasis-suppressor by decreasing HIP1R levels in pre-clinical models of prostate cancer.
- Published
- 2016
- Full Text
- View/download PDF
18. Edelfosine Promotes Apoptosis in Androgen-Deprived Prostate Tumors by Increasing ATF3 and Inhibiting Androgen Receptor Activity.
- Author
-
Udayakumar TS, Stoyanova R, Shareef MM, Mu Z, Philip S, Burnstein KL, and Pollack A
- Subjects
- Androgen Antagonists pharmacology, Animals, Apoptosis, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Drug Synergism, Gene Expression Regulation, Neoplastic drug effects, Humans, Male, Mice, Neoplasm Transplantation, Phospholipid Ethers pharmacology, Promoter Regions, Genetic drug effects, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Proto-Oncogene Proteins c-akt metabolism, Receptors, Androgen metabolism, Signal Transduction drug effects, Up-Regulation, Xenograft Model Antitumor Assays, Activating Transcription Factor 3 metabolism, Androgen Antagonists administration & dosage, Phospholipid Ethers administration & dosage, Prostatic Neoplasms drug therapy, Receptors, Androgen genetics
- Abstract
Edelfosine is a synthetic alkyl-lysophospholipid that possesses significant antitumor activity in several human tumor models. Here, we investigated the effects of edelfosine combined with androgen deprivation (AD) in LNCaP and VCaP human prostate cancer cells. This treatment regimen greatly decreased cell proliferation compared with single agent or AD alone, resulting in higher levels of apoptosis in LNCaP compared with VCaP cells. Edelfosine caused a dose-dependent decrease in AKT activity, but did not affect the expression of total AKT in either cell line. Furthermore, edelfosine treatment inhibited the expression of androgen receptor (AR) and was associated with an increase in activating transcription factor 3 (ATF3) expression levels, a stress response gene and a negative regulator of AR transactivation. ATF3 binds to AR after edelfosine + AD and represses the transcriptional activation of AR as demonstrated by PSA promoter studies. Knockdown of ATF3 using siRNA-ATF3 reversed the inhibition of PSA promoter activity, suggesting that the growth inhibition effect of edelfosine was ATF3 dependent. Moreover, expression of AR variant 7 (ARv7) and TMPRSS2-ERG fusion gene were greatly inhibited after combined treatment with AD and edelfosine in VCaP cells. In vivo experiments using an orthotopic LNCaP model confirmed the antitumor effects of edelfosine + AD over the individual treatments. A significant decrease in tumor volume and PSA levels was observed when edelfosine and AD were combined, compared with edelfosine alone. Edelfosine shows promise in combination with AD for the treatment of prostate cancer patients. Mol Cancer Ther; 15(6); 1353-63. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
19. A novel calcium-dependent mechanism of acquired resistance to IGF-1 receptor inhibition in prostate cancer cells.
- Author
-
Fahrenholtz CD, Greene AM, Beltran PJ, and Burnstein KL
- Subjects
- Antibodies, Monoclonal, Humanized, Apoptosis drug effects, Calcium metabolism, Calcium Signaling genetics, Cell Cycle Checkpoints drug effects, Cell Line, Tumor, Cell Proliferation, Drug Resistance, Neoplasm, Focal Adhesion Kinase 2 antagonists & inhibitors, Focal Adhesion Kinase 2 metabolism, Humans, Male, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local prevention & control, Phospholipase C gamma antagonists & inhibitors, Phospholipase C gamma metabolism, Phosphorylation, Proto-Oncogene Proteins c-akt metabolism, Receptor, IGF Type 1 biosynthesis, Receptor, Insulin biosynthesis, TOR Serine-Threonine Kinases metabolism, Xenograft Model Antitumor Assays, Antibodies, Monoclonal pharmacology, Calcium Chelating Agents pharmacology, Calcium Signaling drug effects, Prostatic Neoplasms drug therapy, Receptor, IGF Type 1 antagonists & inhibitors
- Abstract
Inhibition of the mitogenic insulin-like growth factor receptor 1 (IGF-1R) signaling axis is a compelling treatment strategy for prostate cancer. Combining the IGF-1R inhibitor ganitumab (formerly AMG 479) with standard of care androgen-deprivation therapy greatly delays prostate cancer recurrence in xenograft models; however, a significant proportion of these tumors ultimately acquire resistance to ganitumab. Here we describe the development of a stable and reproducible ganitumab-resistant VCaP human prostate cancer cell derivative termed VCaP/GanR to investigate the mechanism of acquired resistance to IGF-1R inhibition. Unlike parental VCaP, VCaP/GanR did not undergo apoptosis following ganitumab treatment. VCaP/GanR did not express increased levels of IGF-1R, insulin receptor, or phospho-AKT compared to parental VCaP. VCaP/GanR exhibited increased levels of phospho-S6 indicative of increased mTOR activity. However, acquired resistance to ganitumab was not dependent on increased mTOR activity in VCaP/GanR. Phospho-proteomic arrays revealed alterations in several calcium-regulated signaling components in VCaP/GanR compared to VCaP. Reduction of intracellular calcium using cell-permeable calcium-specific chelators restored ganitumab sensitivity to VCaP/GanR through inhibition of cell-cycle progression. These data suggest a new mechanism of resistance to IGF-1R inhibition involving calcium-mediated proliferation effects. Such pathways should be considered in future clinical studies of IGF-1R inhibitors in prostate cancer.
- Published
- 2014
- Full Text
- View/download PDF
20. Coactivator MYST1 regulates nuclear factor-κB and androgen receptor functions during proliferation of prostate cancer cells.
- Author
-
Jaganathan A, Chaurasia P, Xiao GQ, Philizaire M, Lv X, Yao S, Burnstein KL, Liu DP, Levine AC, and Mujtaba S
- Subjects
- Acetylation, Apoptosis, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Tumor, G2 Phase Cell Cycle Checkpoints, Gene Expression, Gene Expression Regulation, Neoplastic, Humans, Male, Prostatic Neoplasms, Protein Processing, Post-Translational, Response Elements, Sirtuin 1 metabolism, Transcriptional Activation, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Cell Proliferation, Histone Acetyltransferases physiology, NF-kappa B physiology, Receptors, Androgen physiology
- Abstract
In prostate cancer (PCa), the functional synergy between androgen receptor (AR) and nuclear factor-κ B (NF-κB) escalates the resistance to therapeutic regimens and promotes aggressive tumor growth. Although the underlying mechanisms are less clear, gene regulatory abilities of coactivators can bridge the transcription functions of AR and NF-κB. The present study shows that MYST1 (MOZ, YBF2 and SAS2, and TIP60 protein 1) costimulates AR and NF-κB functions in PCa cells. We demonstrate that activation of NF-κB promotes deacetylation of MYST1 by sirtuin 1. Further, the mutually exclusive interactions of MYST1 with sirtuin 1 vs AR regulate the acetylation of lysine 16 on histone H4. Notably, in AR-lacking PC3 cells and in AR-depleted LNCaP cells, diminution of MYST1 activates the cleavage of poly(ADP-ribose) polymerase and caspase 3 that leads to apoptosis. In contrast, in AR-transformed PC3 cells (PC3-AR), depletion of MYST1 induces cyclin-dependent kinase (CDK) N1A/p21, which results in G2M arrest. Concomitantly, the levels of phospho-retinoblastoma, E2F1, CDK4, and CDK6 are reduced. Finally, the expression of tumor protein D52 (TPD52) was unequivocally affected in PC3, PC3-AR, and LNCaP cells. Taken together, the results of this study reveal that the functional interactions of MYST1 with AR and NF-κB are critical for PCa progression.
- Published
- 2014
- Full Text
- View/download PDF
21. VAV3 mediates resistance to breast cancer endocrine therapy.
- Author
-
Aguilar H, Urruticoechea A, Halonen P, Kiyotani K, Mushiroda T, Barril X, Serra-Musach J, Islam A, Caizzi L, Di Croce L, Nevedomskaya E, Zwart W, Bostner J, Karlsson E, Pérez Tenorio G, Fornander T, Sgroi DC, Garcia-Mata R, Jansen MP, García N, Bonifaci N, Climent F, Soler MT, Rodríguez-Vida A, Gil M, Brunet J, Martrat G, Gómez-Baldó L, Extremera AI, Figueras A, Balart J, Clarke R, Burnstein KL, Carlson KE, Katzenellenbogen JA, Vizoso M, Esteller M, Villanueva A, Rodríguez-Peña AB, Bustelo XR, Nakamura Y, Zembutsu H, Stål O, Beijersbergen RL, and Pujana MA
- Subjects
- Androstadienes therapeutic use, Antineoplastic Agents, Hormonal pharmacology, Aromatase Inhibitors therapeutic use, Biomarkers, Tumor genetics, Breast pathology, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Enzyme Activators pharmacology, ErbB Receptors antagonists & inhibitors, Erlotinib Hydrochloride, Estrogen Receptor alpha antagonists & inhibitors, Estrogen Receptor alpha genetics, Female, Gene Expression Regulation, Neoplastic, Genetic Association Studies, Genetic Variation, Humans, Letrozole, MCF-7 Cells, Nitriles therapeutic use, Protein Kinase Inhibitors pharmacology, Quinazolines pharmacology, RNA Interference, RNA, Small Interfering, Tamoxifen pharmacology, Tamoxifen therapeutic use, Toremifene pharmacology, Toremifene therapeutic use, Triazoles therapeutic use, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm genetics, Estrogen Receptor alpha metabolism, Indazoles pharmacology, Proto-Oncogene Proteins c-vav genetics
- Abstract
Introduction: Endocrine therapies targeting cell proliferation and survival mediated by estrogen receptor α (ERα) are among the most effective systemic treatments for ERα-positive breast cancer. However, most tumors initially responsive to these therapies acquire resistance through mechanisms that involve ERα transcriptional regulatory plasticity. Herein we identify VAV3 as a critical component in this process., Methods: A cell-based chemical compound screen was carried out to identify therapeutic strategies against resistance to endocrine therapy. Binding to ERα was evaluated by molecular docking analyses, an agonist fluoligand assay and short hairpin (sh)RNA-mediated protein depletion. Microarray analyses were performed to identify altered gene expression. Western blot analysis of signaling and proliferation markers, and shRNA-mediated protein depletion in viability and clonogenic assays, were performed to delineate the role of VAV3. Genetic variation in VAV3 was assessed for association with the response to tamoxifen. Immunohistochemical analyses of VAV3 were carried out to determine its association with therapeutic response and different tumor markers. An analysis of gene expression association with drug sensitivity was carried out to identify a potential therapeutic approach based on differential VAV3 expression., Results: The compound YC-1 was found to comparatively reduce the viability of cell models of acquired resistance. This effect was probably not due to activation of its canonical target (soluble guanylyl cyclase), but instead was likely a result of binding to ERα. VAV3 was selectively reduced upon exposure to YC-1 or ERα depletion, and, accordingly, VAV3 depletion comparatively reduced the viability of cell models of acquired resistance. In the clinical scenario, germline variation in VAV3 was associated with the response to tamoxifen in Japanese breast cancer patients (rs10494071 combined P value = 8.4 × 10-4). The allele association combined with gene expression analyses indicated that low VAV3 expression predicts better clinical outcome. Conversely, high nuclear VAV3 expression in tumor cells was associated with poorer endocrine therapy response. Based on VAV3 expression levels and the response to erlotinib in cancer cell lines, targeting EGFR signaling may be a promising therapeutic strategy., Conclusions: This study proposes VAV3 as a biomarker and a rationale for its use as a signaling target to prevent and/or overcome resistance to endocrine therapy in breast cancer.
- Published
- 2014
- Full Text
- View/download PDF
22. Preclinical efficacy of growth hormone-releasing hormone antagonists for androgen-dependent and castration-resistant human prostate cancer.
- Author
-
Fahrenholtz CD, Rick FG, Garcia MI, Zarandi M, Cai RZ, Block NL, Schally AV, and Burnstein KL
- Subjects
- Animals, Body Weight, Cell Line, Tumor, Cell Proliferation, Drug Screening Assays, Antitumor, Humans, Hypothalamus metabolism, Ligands, Male, Mice, Mice, Nude, Neoplasm Transplantation, Prostate-Specific Antigen metabolism, Receptors, G-Protein-Coupled metabolism, Time Factors, Androgens metabolism, Growth Hormone-Releasing Hormone antagonists & inhibitors, Prostatic Neoplasms drug therapy, Prostatic Neoplasms, Castration-Resistant drug therapy
- Abstract
Advanced hormone-sensitive prostate cancer responds to androgen-deprivation therapy (ADT); however, therapeutic options for recurrent castration-resistant disease are limited. Because growth hormone-releasing hormone (GHRH) and GHRH receptor (GHRH-R) are regulated in an autocrine fashion in prostate cancer, inhibition of GHRH-R represents a compelling approach to treatment. We investigated the effects of the latest series of improved, highly potent GHRH antagonists--MIA-602, MIA-606, and MIA-690--on the growth of androgen-dependent as well as castration-resistant prostate cancer (CRPC) cells in vitro and in vivo. GHRH-R and its splice variant, SV1, were present in 22Rv1, LNCaP, and VCaP human prostate cancer cell lines. Androgen-dependent LNCaP and VCaP cells expressed higher levels of GHRH-R protein compared with castration-resistant 22Rv1 cells; however, 22Rv1 expressed higher levels of SV1. In vitro, MIA-602 decreased cell proliferation of 22Rv1, LNCaP, and VCaP prostate cancer cell lines by 70%, 61%, and 20%, respectively (all P < 0.05), indicating direct effects of MIA-602. In vivo, MIA-602 was more effective than MIA-606 and MIA-690 and decreased 22Rv1 xenograft tumor volumes in mice by 63% after 3 wk (P < 0.05). No noticeable untoward effects or changes in body weight occurred. In vitro, the VCaP cell line was minimally inhibited by MIA-602, but in vivo, this line showed a substantial reduction in growth of xenografts in response to MIA-602, indicating both direct and systemic inhibitory effects. MIA-602 also further inhibited VCaP xenografts when combined with ADT. This study demonstrates the preclinical efficacy of the GHRH antagonist MIA-602 for treatment of both androgen-dependent and CRPC.
- Published
- 2014
- Full Text
- View/download PDF
23. Targeting IGF-IR with ganitumab inhibits tumorigenesis and increases durability of response to androgen-deprivation therapy in VCaP prostate cancer xenografts.
- Author
-
Fahrenholtz CD, Beltran PJ, and Burnstein KL
- Subjects
- Androgens metabolism, Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized, Antineoplastic Agents administration & dosage, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Male, Mice, Orchiectomy, Prostatic Neoplasms therapy, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Antibodies, Monoclonal pharmacology, Antineoplastic Agents pharmacology, Cell Transformation, Neoplastic drug effects, Prostatic Neoplasms metabolism, Receptor, IGF Type 1 antagonists & inhibitors
- Abstract
Prostate cancer is the most commonly diagnosed malignancy in men. While tumors initially respond to androgen-deprivation therapy, the standard care for advanced or metastatic disease, tumors eventually recur as castration-resistant prostate cancer (CRPC). Upregulation of the insulin-like growth factor receptor type I (IGF-IR) signaling axis drives growth and progression of prostate cancer by promoting proliferation, survival, and angiogenesis. Ganitumab (formerly AMG 479) is a fully human antibody that inhibits binding of IGF-I and IGF-II to IGF-IR. We evaluated the therapeutic value of ganitumab in several preclinical settings including androgen-dependent prostate cancer, CRPC, and in combination with androgen-deprivation therapy. Ganitumab inhibited IGF-I-induced phosphorylation of the downstream effector AKT and reduced proliferation of multiple androgen-dependent and castration-resistant human prostate cancer cell lines in vitro. Ganitumab inhibited androgen-dependent VCaP xenograft growth and increased tumor-doubling time from 2.3 ± 0.4 weeks to 6.4 ± 0.4 weeks. Ganitumab blocked growth of castration-resistant VCaP xenografts for over 11.5 weeks of treatment. In contrast, ganitumab did not have appreciable effects on the castration-resistant CWR-22Rv1 xenograft model. Ganitumab was most potent against VCaP xenografts when combined with complete androgen-deprivation therapy (castration). Tumor volume was reduced by 72% after 4 weeks of treatment and growth suppression was maintained over 16 weeks of treatment. These data suggest that judicious use of ganitumab particularly in conjunction with androgen-deprivation therapy may be beneficial in the treatment of prostate cancer.
- Published
- 2013
- Full Text
- View/download PDF
24. Novel interaction between the co-chaperone Cdc37 and Rho GTPase exchange factor Vav3 promotes androgen receptor activity and prostate cancer growth.
- Author
-
Wu F, Peacock SO, Rao S, Lemmon SK, and Burnstein KL
- Subjects
- Animals, COS Cells, Cell Line, Tumor, Cell Proliferation, Chlorocebus aethiops, Glutathione Transferase metabolism, Guanine Nucleotide Exchange Factors metabolism, HEK293 Cells, Humans, Male, Molecular Chaperones metabolism, Protein Binding, Proto-Oncogene Proteins c-vav metabolism, Two-Hybrid System Techniques, Cell Cycle Proteins metabolism, Chaperonins metabolism, Gene Expression Regulation, Neoplastic, Prostatic Neoplasms metabolism, Proto-Oncogene Proteins c-vav physiology
- Abstract
Elevated androgen receptor (AR) activity in castration-resistant prostate cancer may occur through increased levels of AR co-activator proteins. Vav3, a guanine nucleotide exchange factor, is up-regulated following progression to castration resistance in preclinical models and is overexpressed in a significant number of human prostate cancers. Vav3 is a novel co-activator of the AR. We sought to identify Vav3 binding partners in an effort to understand the molecular mechanisms underlying Vav3 enhancement of AR activity and to identify new therapeutic targets. The cell division cycle 37 homolog (Cdc37), a protein kinase-specific co-chaperone for Hsp90, was identified as a Vav3 interacting protein by yeast two-hybrid screening. Vav3-Cdc37 interaction was confirmed by GST pulldown and, for native proteins, by co-immunoprecipitation experiments in prostate cancer cells. Cdc37 potentiated Vav3 co-activation of AR transcriptional activity and Vav3 enhancement of AR N-terminal-C-terminal interaction, which is essential for optimal receptor transcriptional activity. Cdc37 increased prostate cancer cell proliferation selectively in Vav3-expressing cells. Cdc37 did not affect Vav3 nucleotide exchange activity, Vav3 protein levels, or subcellular localization. Disruption of Vav3-Cdc37 interaction inhibited Vav3 enhancement of AR transcriptional activity and AR N-C interaction. Diminished Vav3-Cdc37 interaction also caused decreased prostate cancer cell proliferation selectively in Vav3-expressing cells. Taken together, we identified a novel Vav3 interacting protein that enhances Vav3 co-activation of AR and prostate cancer cell proliferation. Vav3-Cdc37 interaction may provide a new therapeutic target in prostate cancer.
- Published
- 2013
- Full Text
- View/download PDF
25. In vivo 17β-estradiol treatment contributes to podocyte actin stabilization in female db/db mice.
- Author
-
Catanuto P, Fornoni A, Pereira-Simon S, Wu F, Burnstein KL, Xia X, Conti F, Lenzi A, and Elliot S
- Subjects
- Animals, Apoptosis, Female, Heat-Shock Proteins metabolism, Mice, Mice, Inbred C57BL, Molecular Chaperones, Neoplasm Proteins metabolism, Neuropeptides metabolism, Phenotype, Placebos, Rhodamines chemistry, Transcriptional Activation, rac GTP-Binding Proteins metabolism, rac1 GTP-Binding Protein, Actins metabolism, Estradiol metabolism, Podocytes cytology
- Abstract
We recently showed that 17β-estradiol (E(2)) treatment ameliorated type 2 diabetic glomerulosclerosis in mice in part by protecting podocyte structure and function. Progressive podocyte damage is characterized by foot process effacement, vacuolization, detachment of podocytes from the glomerular basement membrane, and apoptosis. In addition, podocytes are highly dependent on the preservation of their actin cytoskeleton to ensure proper function and survival. Because E(2) administration prevented podocyte damage in our study on diabetic db/db mice and has been shown to regulate both actin cytoskeleton and apoptosis in other cell types and tissues, we investigated whether actin remodeling and apoptosis were prevented in podocytes isolated from E(2)-treated diabetic db/db mice. We performed G-actin/F-actin assays, Western analysis for Hsp25 expression, Ras-related C(3) botulinum toxin substrate 1 (Rac1) activity, and apoptosis assays on previously characterized podocytes isolated from both in vivo-treated placebo and E(2) female db/db mice. We found that in vivo E(2) protects against a phenotype change in the cultured podocytes characterized by a percent increase of F-actin vs. G-actin, suppression of Hsp25 expression and transcriptional activation, increase of Rac1 activity, and decreased apoptotic intermediates. We conclude from these studies that E(2) treatment protects against podocyte damage and may prevent/reduce diabetes-induced kidney disease.
- Published
- 2012
- Full Text
- View/download PDF
26. Vav3 enhances androgen receptor splice variant activity and is critical for castration-resistant prostate cancer growth and survival.
- Author
-
Peacock SO, Fahrenholtz CD, and Burnstein KL
- Subjects
- Castration, Cell Line, Tumor, Cell Proliferation, Cell Survival, Gene Expression Regulation, Neoplastic, HEK293 Cells, Humans, Male, Neoplasm Transplantation, Neoplasms, Hormone-Dependent genetics, Prostate pathology, Protein Isoforms genetics, Proto-Oncogene Proteins c-vav metabolism, Receptors, Androgen metabolism, Signal Transduction, Transcription, Genetic, Transplantation, Heterologous, Up-Regulation, Prostatic Neoplasms genetics, Protein Isoforms metabolism, Proto-Oncogene Proteins c-vav genetics, RNA Splicing, Receptors, Androgen genetics
- Abstract
Advanced or metastatic prostate cancer is treated by androgen deprivation; however, patients inevitably relapse with castration-resistant prostate cancer (CRPC). CRPC remains dependent on androgen receptor (AR) signaling, which may include constitutive, ligand-independent action of naturally occurring AR splice variants. For example, the AR splice variant AR3 (also termed AR-V7) is expressed in CRPC and is linked to poor prognosis. Vav3, a Rho GTPase guanine nucleotide exchange factor, is an AR coactivator that is up-regulated in human prostate cancer compared with benign tissue and in preclinical models of CRPC. Vav3 confers castration-resistant growth to androgen-dependent human prostate cancer cells. Despite the importance of AR coactivators in promoting CRPC, the potential role of these regulatory proteins in modulating AR splice variant activity is unknown. We examined the contributions of Vav3 to AR activity in two CRPC cell lines that naturally express relatively high levels of Vav3 and AR3. Vav3 or AR3 knockdown greatly attenuated cell proliferation, soft agar growth, and ligand-independent AR activity. Vav3 potently enhanced the transcriptional activity of AR3 and another clinically relevant AR splice variant, ARv567es. Vav3 knockdown resulted in lowered nuclear AR3 levels, whereas total AR3 levels remained similar. Conversely, overexpression of Vav3 resulted in increased nuclear AR3. Coimmunoprecipitation revealed that AR3 and Vav3 interact. These novel data demonstrating physical and functional interactions between Vav3, a unique AR coactivator, and an AR splice variant provide insights into the mechanisms by which Vav3 exploits and enhances AR signaling in the progression to CRPC.
- Published
- 2012
- Full Text
- View/download PDF
27. A novel nuclear role for the Vav3 nucleotide exchange factor in androgen receptor coactivation in prostate cancer.
- Author
-
Rao S, Lyons LS, Fahrenholtz CD, Wu F, Farooq A, Balkan W, and Burnstein KL
- Subjects
- Animals, Binding Sites genetics, Blotting, Western, Cell Line, Tumor, Chromatin Immunoprecipitation, Cytoplasm metabolism, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, Male, Metribolone pharmacology, Mice, Mice, SCID, Microscopy, Fluorescence, Mutation, Orchiectomy, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Protein Binding drug effects, Proto-Oncogene Proteins c-vav genetics, Receptors, Androgen genetics, Trans-Activators genetics, Trans-Activators metabolism, Transplantation, Heterologous, Tumor Burden, Cell Nucleus metabolism, Prostatic Neoplasms metabolism, Proto-Oncogene Proteins c-vav metabolism, Receptors, Androgen metabolism
- Abstract
Increased androgen receptor (AR) transcriptional activity mediated by coactivator proteins may drive castration-resistant prostate cancer (CRPC) growth. Vav3, a Rho GTPase guanine nucleotide exchange factor (GEF), is overexpressed in human prostate cancers, particularly in models of CRPC progression. Vav3 coactivates AR in a Vav3 pleckstrin homology (PH) domain-dependent but GEF-independent manner. Ectopic expression of Vav3 in androgen-dependent human prostate cancer cells conferred robust castration-resistant xenograft tumor growth. Vav3 but not a Vav3 PH mutant greatly stimulated interaction between the AR amino and carboxyl termini (N-C interaction), which is required for maximal receptor transcriptional activity. Vav3 was distributed between the cytoplasm and nucleus with nuclear localization-dependent on the Vav3 PH domain. Membrane targeting of Vav3 abolished Vav3 potentiation of AR activity, whereas nuclear targeting of a Vav3 PH mutant rescued AR coactivation, suggesting that nuclear localization is an important function of the Vav3 PH domain. A nuclear role for Vav3 was further demonstrated by sequential chromatin immunoprecipitation assays, which revealed that Vav3 and AR were recruited to the same transcriptional complexes of an AR target gene enhancer. These data demonstrate the importance of Vav3 in CRPC and define a novel nuclear function of Vav3 in regulating AR activity.
- Published
- 2012
- Full Text
- View/download PDF
28. The microRNA -23b/-27b cluster suppresses the metastatic phenotype of castration-resistant prostate cancer cells.
- Author
-
Ishteiwy RA, Ward TM, Dykxhoorn DM, and Burnstein KL
- Subjects
- Apoptosis, Blotting, Western, Cadherins genetics, Cell Adhesion, Cell Proliferation, Flow Cytometry, Humans, Male, Neoplasms, Hormone-Dependent secondary, Prostatic Neoplasms pathology, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, rac1 GTP-Binding Protein genetics, Cadherins metabolism, Cell Movement, MicroRNAs genetics, Neoplasms, Hormone-Dependent genetics, Orchiectomy, Prostatic Neoplasms genetics, rac1 GTP-Binding Protein metabolism
- Abstract
MicroRNAs (miRs) are small, endogenous, non-coding RNAs that regulate the stability and/or translation of complementary mRNA targets. MiRs have emerged not only as critical modulators of normal physiologic processes, but their deregulation may significantly impact prostate and other cancers. The expression of miR-23b and miR-27b, which are encoded by the same miR cluster (miR-23b/-27b), are downregulated in metastatic, castration-resistant tumors compared to primary prostate cancer and benign tissue; however, their possible role in prostate cancer progression is unknown. We found that ectopic expression of miR-23b/-27b in two independent castration-resistant prostate cancer cell lines resulted in suppression of invasion and migration, as well as reduced survival in soft agar (a measure of anoikis). However, there was no effect of miR-23b/-27b on cell proliferation suggesting that these miRs function as metastasis (but not growth) suppressors in prostate cancer. Conversely, inhibition of miR-23b/-27b in the less aggressive androgen-dependent LNCaP prostate cancer cell line resulted in enhanced invasion and migration also without affecting proliferation. Mechanistically, we found that introduction of miR-23b/-27b in metastatic, castration-resistant prostate cancer cell lines resulted in a significant attenuation of Rac1 activity without affecting total Rac1 levels and caused increased levels of the tumor suppressor E-cadherin. Inhibition of these miRs had the opposite effect in androgen-dependent LNCaP cells. These results suggest that miR-23b/-27b are metastasis suppressors that might serve as novel biomarkers and therapeutic agents for castration-resistant disease.
- Published
- 2012
- Full Text
- View/download PDF
29. Inhibition of the Rho GTPase, Rac1, decreases estrogen receptor levels and is a novel therapeutic strategy in breast cancer.
- Author
-
Rosenblatt AE, Garcia MI, Lyons L, Xie Y, Maiorino C, Désiré L, Slingerland J, and Burnstein KL
- Subjects
- Blotting, Western, Breast Neoplasms enzymology, Cell Line, Tumor, Cell Proliferation drug effects, Down-Regulation physiology, Enzyme Inhibitors pharmacology, Female, Humans, Proto-Oncogene Proteins c-vav metabolism, Pyrones pharmacology, Quinolines pharmacology, RNA chemistry, Reverse Transcriptase Polymerase Chain Reaction, Transcription, Genetic, rac1 GTP-Binding Protein genetics, rac1 GTP-Binding Protein metabolism, rho GTP-Binding Proteins metabolism, Breast Neoplasms metabolism, Estrogen Receptor alpha metabolism, p21-Activated Kinases metabolism, rac1 GTP-Binding Protein antagonists & inhibitors, rho GTP-Binding Proteins antagonists & inhibitors
- Abstract
Rac1, a Rho GTPase, modulates diverse cellular processes and is hyperactive in some cancers. Estrogen receptor-alpha (ERα) in concert with intracellular signaling pathways regulates genes associated with cell proliferation, tumor development, and breast cancer cell survival. Therefore, we examined the possibility of Rac1 and ERα crosstalk in breast cancer cells. We found that Rac1 enhanced ERα transcriptional activity in breast cancer cells. Vav3, a Rho guanine nucleotide exchange factor that activates Rac1, was an upstream mediator, and P21/Cdc42/Rac1 activating kinase-1 (Pak-1) was a downstream effector of Rac1 enhancement of ERα activity. These results suggest that Rac1 may prove to be a therapeutic target. To test this hypothesis, we used a small molecule Rac inhibitor, EHT 1864, and found that EHT 1864 inhibited ERα transcriptional activity. Furthermore, EHT 1864 inhibited estrogen-induced cell proliferation in breast cancer cells and decreased tamoxifen-resistant breast cancer cell growth. EHT 1864 decreased activity of the promoter of the ERα gene resulting in down-regulation of ERα mRNA and protein levels. Therefore, ERα down-regulation by EHT 1864 is the likely mechanism of EHT 1864-mediated inhibition of ERα activity and estrogen-stimulated breast cancer cell proliferation. Since ERα plays a critical role in the pathogenesis of breast cancer and the Rac inhibitor EHT 1864 down-regulates ERα expression and breast cancer cell proliferation, further investigation of the therapeutic potential of Rac1 targeting in the treatment of breast cancer is warranted.
- Published
- 2011
- Full Text
- View/download PDF
30. GADD45gamma: a new vitamin D-regulated gene that is antiproliferative in prostate cancer cells.
- Author
-
Flores O and Burnstein KL
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Antineoplastic Agents, Hormonal pharmacology, Antineoplastic Agents, Hormonal therapeutic use, Biomarkers, Pharmacological analysis, Biomarkers, Pharmacological metabolism, Carcinoma drug therapy, Carcinoma pathology, Cell Line, Tumor, Cluster Analysis, Drug Resistance, Neoplasm genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Humans, Intracellular Signaling Peptides and Proteins metabolism, Intracellular Signaling Peptides and Proteins physiology, Male, Oligonucleotide Array Sequence Analysis, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Vitamin D therapeutic use, Carcinoma genetics, Cell Proliferation drug effects, Intracellular Signaling Peptides and Proteins genetics, Prostatic Neoplasms genetics, Vitamin D pharmacology
- Abstract
1,25-dihydroxyvitamin D3 [1,25-(OH)2D3] inhibits proliferation of normal and malignant prostate epithelial cells at least in part through inhibition of G1 to S phase cell cycle progression. The mechanisms of the antiproliferative effects of 1,25-(OH)2D3 have yet to be fully elucidated but are known to require the vitamin D receptor. We previously developed a 1,25-(OH)2D3-resistant derivative of the human prostate cancer cell line, LNCaP, which retains active vitamin D receptors but is not growth inhibited by 1,25-(OH)2D3. Gene expression profiling revealed two novel 1,25-(OH)2D3-inducible genes, growth arrest and DNA damage-inducible gene gamma (GADD45γ) and mitogen induced gene 6 (MIG6), in LNCaP but not in 1,25-(OH)2D3-resistant cells. GADD45γ up-regulation was associated with growth inhibition by 1,25-(OH)2D3 in human prostate cancer cells. Ectopic expression of GADD45γ in either LNCaP or ALVA31 cells resulted in G1 accumulation and inhibition of proliferation equal to or greater than that caused by 1,25-(OH)2D3 treatment. In contrast, ectopic expression of MIG6 had only minimal effects on cell cycle distribution and proliferation. Whereas GADD45γ has been shown to be induced by androgens in prostate cancer cells, up-regulation of GADD45γ by 1,25-(OH)2D3 was not dependent on androgen receptor signaling, further refuting a requirement for androgens/androgen receptor in vitamin D-mediated growth inhibition. These data introduce two novel 1,25-(OH)2D3-regulated genes and establish GADD45γ as a growth-inhibitory protein in prostate cancer. Furthermore, the induction of GADD45γ gene expression by 1,25-(OH)2D3 may mark therapeutic response in prostate cancer.
- Published
- 2010
- Full Text
- View/download PDF
31. Nuclear targeting of cyclin-dependent kinase 2 reveals essential roles of cyclin-dependent kinase 2 localization and cyclin E in vitamin D-mediated growth inhibition.
- Author
-
Flores O, Wang Z, Knudsen KE, and Burnstein KL
- Subjects
- Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Cytoplasm metabolism, Enzyme Activation, G1 Phase, Humans, Male, Phosphorylation, Protein Transport, Retinoblastoma Protein metabolism, Up-Regulation, Adenocarcinoma metabolism, Calcitriol metabolism, Cell Proliferation, Cyclin E metabolism, Cyclin-Dependent Kinase 2 metabolism, Prostatic Neoplasms metabolism
- Abstract
1,25-Dihydroxyvitamin D(3) (1,25-(OH)(2)D(3)), inhibits proliferation of a variety of cell types including adenocarcinoma of the prostate. We have previously shown that 1,25-(OH)(2)D(3) increases the stability of the cyclin-dependent kinase inhibitor p27(KIP1), decreases cyclin-dependent kinase 2 (CDK2) activity, and promotes G(1) phase accumulation in human prostate cancer cells. These effects correlate with cytoplasmic relocalization of CDK2. In this study, we investigated the role of CDK2 cytoplasmic relocalization in the antiproliferative effects of 1,25-(OH)(2)D(3). CDK2 was found to be necessary for prostate cancer cell proliferation. Although induced by 1,25-(OH)(2)D(3), the cyclin-dependent kinase inhibitor p27(KIP1) was dispensable for 1,25-(OH)(2)D(3)-mediated growth inhibition. Reduction in CDK2 activity by 1,25-(OH)(2)D(3) was associated with decreased T160 phosphorylation, a residue whose phosphorylation in the nucleus is essential for CDK2 activity. Ectopic expression of cyclin E was sufficient to overcome 1,25-(OH)(2)D(3)-mediated cytoplasmic mislocalization of CDK2 and all antiproliferative effects of 1,25-(OH)(2)D(3), yet endogenous levels of cyclin E or binding to CDK2 were not affected by 1,25-(OH)(2)D(3). Similarly, knockdown of the CDK2 substrate retinoblastoma, which causes cyclin E up-regulation, resulted in resistance to 1,25-(OH)(2)D(3)-mediated growth inhibition. Human prostate cancer cells resistant to growth inhibition by 1,25-(OH)(2)D(3) but retaining fully functional vitamin D receptors were developed. These cells did not exhibit 1,25-(OH)(2)D(3)-mediated cytoplasmic relocalization of CDK2. Targeting CDK2 to the nucleus of 1,25-(OH)(2)D(3)-sensitive cancer cells blocked G(1) accumulation and growth inhibition by 1,25-(OH)(2)D(3). These data establish central roles for CDK2 nuclear-cytoplasmic trafficking and cyclin E in the mechanism of 1,25-(OH)(2)D(3)-mediated growth inhibition in prostate cancer cells.
- Published
- 2010
- Full Text
- View/download PDF
32. Inhibition of androgen receptor transcriptional activity as a novel mechanism of action of arsenic.
- Author
-
Rosenblatt AE and Burnstein KL
- Subjects
- Androgen Antagonists pharmacology, Androgens metabolism, Androgens pharmacology, Anilides administration & dosage, Anilides pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Arsenic Trioxide, Arsenicals administration & dosage, Cell Nucleus drug effects, Cell Nucleus metabolism, Cell Proliferation drug effects, Drug Evaluation, Preclinical, Drug Synergism, Humans, Male, Nitriles administration & dosage, Nitriles pharmacology, Nuclear Receptor Coactivator 2 metabolism, Oxides administration & dosage, Prostatic Neoplasms pathology, Protein Binding drug effects, Protein Structure, Tertiary, Protein Transport drug effects, Receptors, Androgen chemistry, Receptors, Androgen metabolism, Receptors, Androgen physiology, Response Elements physiology, Tosyl Compounds administration & dosage, Tosyl Compounds pharmacology, Tumor Cells, Cultured, Androgen Receptor Antagonists, Arsenicals pharmacology, Oxides pharmacology, Transcriptional Activation drug effects
- Abstract
Environmental sodium arsenite is a toxin that is associated with male infertility due to decreased and abnormal sperm production. Arsenic trioxide (ATO), another inorganic trivalent semimetal, is an effective therapy for acute promyelocytic leukemia, and there is investigation of its possible efficacy in prostate cancer. However, the mechanism of arsenic action in male urogenital tract tissues is not clear. Because the androgen receptor (AR) plays an important role in spermatogenesis and prostate cancer, we explored the possibility that trivalent arsenic regulates AR function. We found that arsenic inhibited AR transcriptional activity in prostate cancer and Sertoli cells using reporter gene assays testing several androgen response element-containing regions and by assessing native target gene expression. Arsenic inhibition of AR activity was not due to down-regulation of AR protein levels, decreased hormone binding to AR, disruption of AR nuclear translocation, or interference with AR-DNA binding in vitro. However, chromatin immunoprecipitation studies revealed that arsenic inhibited AR recruitment to an AR target gene enhancer in vivo. Consistent with a deficiency in AR-chromatin binding, arsenic disrupted AR amino and carboxyl termini interaction. Furthermore, ATO caused a significant decrease in prostate cancer cell proliferation that was more pronounced in cells expressing AR compared with cells depleted of AR. In addition, inhibition of AR activity by ATO and by the AR antagonist, bicalutamide, was additive. Thus, arsenic-induced male infertility may be due to inhibition of AR activity. Further, because AR is an important target in prostate cancer therapy, arsenic may serve as an effective therapeutic option.
- Published
- 2009
- Full Text
- View/download PDF
33. Endothelin-1 enhances the expression of the androgen receptor via activation of the c-myc pathway in prostate cancer cells.
- Author
-
Lee JG, Zheng R, McCafferty-Cepero JM, Burnstein KL, Nanus DM, and Shen R
- Subjects
- Base Sequence, Blotting, Western, Cell Line, Tumor, Chromatin Immunoprecipitation, DNA Primers, Humans, Male, Phosphatidylinositol 3-Kinases metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Receptors, Androgen genetics, Reverse Transcriptase Polymerase Chain Reaction, Endothelin-1 physiology, Genes, myc, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism
- Abstract
Increasing evidence suggests that androgen independent prostate cancer (PC) maintains a functional androgen receptor (AR) pathway despite the low levels of circulating androgen following androgen withdrawal, the molecular mechanisms of which are not well defined yet. To address this question, we investigated the effects of endothelin-1 (ET-1) on AR expression. Western analysis and RT-PCR revealed that in the presence of ET-1, levels of AR significantly increased in a time- and dose-dependent manner in LNCaP cells. Pretreatments with inhibitors of Src and phosphoinositide kinase 3 (PI-3K) suppressed ET-1-induced AR expression. As ET-1 was reported to cause a transient increase in c-myc mRNA levels, we examined the involvement of c-myc in ET-1-mediated AR expression. Transient transfection of c-myc siRNA neutralized ET-1-induced AR expression, suggesting that AR induction by ET-1 is c-myc dependent. AR can regulate the transcription of its own gene via a mechanism in which c-myc plays a crucial role. Therefore, we assessed if ET-1-induced-c-myc leads to the enhancement of AR transcription. Reporter gene assays using the previously identified AR gene enhancer containing a c-myc binding site were conducted in LNCaP cells. We found that ET-1 induced reporter gene activity from the construct containing the wild-type but not mutant c-myc binding site. Chromatin immunoprecipitation assays confirmed that ET-1 increased interaction between c-myc and c-myc binding sites in AR enhancer, suggesting that ET-1-induced AR transcription occurs via c-myc-mediated AR transcription. Together, these data support the notion that ET-1, via Src/PI-3K signaling, augments c-myc expression leading to enhanced AR expression in PC., (Copyright 2008 Wiley-Liss, Inc.)
- Published
- 2009
- Full Text
- View/download PDF
34. Ligand-independent activation of androgen receptors by Rho GTPase signaling in prostate cancer.
- Author
-
Lyons LS, Rao S, Balkan W, Faysal J, Maiorino CA, and Burnstein KL
- Subjects
- Animals, Cell Line, Tumor, Guanine Nucleotide Exchange Factors biosynthesis, Guanine Nucleotide Exchange Factors genetics, Humans, MAP Kinase Signaling System, Male, Mice, Mice, Nude, Mitogen-Activated Protein Kinases metabolism, Neoplasm Transplantation, Neoplasms, Hormone-Dependent enzymology, Prostatic Neoplasms enzymology, Proto-Oncogene Proteins c-vav biosynthesis, Proto-Oncogene Proteins c-vav genetics, RNA, Messenger biosynthesis, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Transcription, Genetic, Transplantation, Heterologous, Up-Regulation, rac1 GTP-Binding Protein biosynthesis, rac1 GTP-Binding Protein genetics, Guanine Nucleotide Exchange Factors metabolism, Neoplasms, Hormone-Dependent metabolism, Prostatic Neoplasms metabolism, Proto-Oncogene Proteins c-vav metabolism, Receptors, Androgen metabolism, rac1 GTP-Binding Protein metabolism
- Abstract
Prostate cancer invariably recurs after androgen deprivation therapy. Growth of this recurrent/androgen-independent form of prostate cancer may be due to increased androgen receptor (AR) transcriptional activity in the absence of androgen. This ligand-independent AR activation is promoted by some growth factors but the mechanism is not well understood. Vav3, a Rho guanosine triphosphatase guanine nucleotide exchange factor, which is activated by growth factors, is up-regulated in human prostate cancer. We show here that Vav3 levels increase during in vivo progression of prostate cancer to androgen independence. Vav3 strikingly enhanced growth factor activation of AR in the absence of androgen. Because Vav3 may be chronically activated in prostate cancer by growth factor receptors, we examined the effects of a constitutively active (Ca) form of Vav3 on AR transcriptional activity. Ca Vav3 caused nuclear localization and ligand-independent activation of AR via the Rho guanosine triphosphatase, Rac1. Ca Rac1 activation of AR occurred, in part, through MAPK/ERK signaling. Expression of active Rac1 conferred androgen-independent growth of prostate cancer cells in culture, soft agar, and mice. These findings suggest that Vav3/Rac 1 signaling is an important modulator of ligand-independent AR transcriptional activity in prostate cancer progression.
- Published
- 2008
- Full Text
- View/download PDF
35. International Union of Pharmacology. LXV. The pharmacology and classification of the nuclear receptor superfamily: glucocorticoid, mineralocorticoid, progesterone, and androgen receptors.
- Author
-
Lu NZ, Wardell SE, Burnstein KL, Defranco D, Fuller PJ, Giguere V, Hochberg RB, McKay L, Renoir JM, Weigel NL, Wilson EM, McDonnell DP, and Cidlowski JA
- Subjects
- Animals, Hormone Antagonists adverse effects, Hormone Antagonists therapeutic use, Humans, Ligands, Mutation, Receptors, Androgen genetics, Receptors, Cytoplasmic and Nuclear classification, Receptors, Cytoplasmic and Nuclear genetics, Receptors, Cytoplasmic and Nuclear physiology, Receptors, Glucocorticoid genetics, Receptors, Mineralocorticoid genetics, Receptors, Progesterone genetics, Receptors, Androgen physiology, Receptors, Glucocorticoid physiology, Receptors, Mineralocorticoid physiology, Receptors, Progesterone physiology
- Published
- 2006
- Full Text
- View/download PDF
36. Vav3, a Rho GTPase guanine nucleotide exchange factor, increases during progression to androgen independence in prostate cancer cells and potentiates androgen receptor transcriptional activity.
- Author
-
Lyons LS and Burnstein KL
- Subjects
- Androgens pharmacology, Cell Line, Tumor, Disease Progression, Guanine Nucleotide Exchange Factors genetics, Humans, Male, Mutation, Phosphatidylinositol 3-Kinases metabolism, Prostate-Specific Antigen genetics, Prostatic Neoplasms genetics, Protein Structure, Tertiary genetics, Proto-Oncogene Proteins c-vav genetics, RNA, Messenger analysis, RNA, Messenger metabolism, Receptors, Androgen genetics, Transcription, Genetic, Up-Regulation, Androgens metabolism, Gene Expression Regulation, Neoplastic, Guanine Nucleotide Exchange Factors metabolism, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Proto-Oncogene Proteins c-vav metabolism, Receptors, Androgen metabolism
- Abstract
The progression of prostate cancer from androgen dependence to androgen independence is often accompanied by enhanced androgen receptor (AR) transcriptional activity. We observed a marked increase in the expression of Vav3, a Rho GTPase guanine nucleotide exchange factor (GEF), during the progression of human prostate cancer LNCaP cells to the androgen-independent derivative, LNCaP-R1. GEFs activate Rho family GTPases by promoting the exchange of GDP for GTP. Reporter gene assays showed that Vav3 potentiated AR transcriptional activity, and knock down of Vav3 resulted in decreased AR transactivation. Vav3 also increased androgen-induced levels of prostate-specific antigen mRNA. Furthermore, Vav3 enhanced AR activity at subnanomolar concentrations of androgen. This finding is particularly relevant because low androgen levels may be present in prostate tissue of patients undergoing androgen deprivation therapy. Enhancement of AR activity by Vav3 required amino terminal activation function 1 (AF1) of AR; however, Vav3 did not interact with AR or increase AR levels. Neither GEF function nor the C-terminal domains of Vav3 were required for Vav3-mediated enhancement of AR activity; however, the pleckstrin homology domain was obligatory. These data show that Vav3 levels rise during progression to androgen independence and support continued AR signaling (even under conditions of low androgen) by a novel GEF-independent cross-talk mechanism.
- Published
- 2006
- Full Text
- View/download PDF
37. Regulation of androgen receptor levels: implications for prostate cancer progression and therapy.
- Author
-
Burnstein KL
- Subjects
- Animals, Disease Progression, Humans, Male, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Signal Transduction, Gene Expression Regulation, Neoplastic, Prostatic Neoplasms pathology, Prostatic Neoplasms therapy, Receptors, Androgen metabolism
- Abstract
Androgen deprivation has been the standard therapy for advanced and metastatic prostate cancer for over half a century, as prostate tumors are initially dependent on androgens for growth and survival. Unfortunately, in most patients undergoing androgen ablation, relapse (recurrent tumor growth) eventually occurs. The actions of the principal androgens, testosterone and dihydrotestosterone (DHT), are mediated via androgen receptors (ARs), ligand-activated transcription factors that belong to the nuclear receptor superfamily. Because of the presence of transcriptionally active ARs in tumors from recurrent or androgen-independent disease, there is a heightened interest in new therapeutic paradigms that target the AR and its regulatory pathways. The regulation of AR levels is highly complex with control exerted by several pathways and in a cell-, tissue-, and developmental-stage specific manner. Androgens are important regulators of AR mRNA and protein through transcriptional and post-transcriptional mechanisms. This article reviews the evidence implicating the AR in recurrent prostate cancer and discusses the multiple mechanisms that regulate AR levels in normal and neoplastic cells. The complexity of AR regulation suggests that there will be an ample array of potential new drug targets for modulating levels of this receptor, a key signaling molecule in prostate cancer.
- Published
- 2005
- Full Text
- View/download PDF
38. Androgen-induced mineralization by MC3T3-E1 osteoblastic cells reveals a critical window of hormone responsiveness.
- Author
-
Balkan W, Burnstein KL, Schiller PC, Perez-Stable C, D'Ippolito G, Howard GA, and Roos BA
- Subjects
- 3T3 Cells, Adaptation, Physiological drug effects, Adaptation, Physiological physiology, Animals, Calcification, Physiologic drug effects, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Line, Dose-Response Relationship, Drug, Mice, Nandrolone pharmacology, Osteoblasts drug effects, Androgen Receptor Antagonists, Androgens pharmacology, Calcification, Physiologic physiology, Nandrolone analogs & derivatives, Osteoblasts cytology, Osteoblasts physiology, Receptors, Androgen metabolism, Testosterone pharmacology
- Abstract
Despite their clinical importance for skeletal growth and homeostasis, the actions of androgens on osteoblastic cells are not well understood. MC3T3-E1 cells, a nontransformed murine preosteoblastic cell line, that traverse the stages of osteoblastic differentiation within 30 days in vitro, were exposed to mibolerone (an androgen receptor (AR) agonist) or 5alpha-dihydroxytestosterone (DHT) from days 3 to 30 post-plating. Cells exposed to this hormonal regimen exhibited a significant increase in mineralization (calcium deposition) compared to vehicle-treated cells. Delaying treatment for 4-11 days (treatment still completed on day 30 post-plating) enhanced mineralization further. Within 2 days post-plating, AR protein increased 7.2-fold in androgen-treated cells and 2.5-fold in vehicle-treated cells. MC3T3-E1 cells transfected with an androgen- and glucocorticoid-responsive reporter construct on day 1 post-plating followed by a 2 day exposure to DHT, mibolerone, or dexamethasone (dex; a glucocorticoid receptor agonist) exhibited reporter gene activation only with dex treatment. In contrast, delaying transfection and treatment for at least 1 day resulted in comparable androgen- and dex-mediated reporter gene transactivation. Therefore, the ability of MC3T3-E1 cells to respond to androgens is dependent on the timing of androgen administration.
- Published
- 2005
- Full Text
- View/download PDF
39. Deregulation of the Rho GTPase, Rac1, suppresses cyclin-dependent kinase inhibitor p21(CIP1) levels in androgen-independent human prostate cancer cells.
- Author
-
Knight-Krajewski S, Welsh CF, Liu Y, Lyons LS, Faysal JM, Yang ES, and Burnstein KL
- Subjects
- Androgens metabolism, CDC2-CDC28 Kinases antagonists & inhibitors, Cell Division physiology, Cyclin-Dependent Kinase 2, Cyclin-Dependent Kinase Inhibitor p21, Cyclins genetics, Humans, Male, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclins metabolism, Prostatic Neoplasms metabolism, rac1 GTP-Binding Protein metabolism
- Abstract
Abnormally suppressed levels of cyclin-dependent kinase inhibitors (CKIs) are associated with aggressive androgen-independent prostate cancer and contribute to uncontrolled proliferation. The androgen-independent human prostate cancer cell lines, LNCaP-104R1, ALVA31 and PC-3, express low levels of the CKI, p21(CIP1), compared to the less-malignant, androgen-dependent LNCaP cells. We investigated the mechanism underlying this suppression by examining the role of Rho GTPases, signaling proteins that play important roles in cell cycle progression, at least in part through regulation of CKIs. Inhibition of Rac1 induced p21 expression in androgen-independent lines but had no effect on the higher p21 levels characteristic of LNCaP cells. This induction of p21 was functionally significant as evidenced by inhibition of cyclin-dependent kinase 2 activity and decreased cell proliferation. Conversely, overexpression of constitutively active Rac1 suppressed the higher p21 levels seen in LNCaP cells. Thus, Rac1 activity is both necessary and sufficient for suppression of p21 in prostate cancer cells. Furthermore, Rac1 activity was significantly higher in all three androgen-independent cell lines compared to LNCaP cells. Thus in three models of aggressive human prostate cancer, hyperactivity of Rac1 corresponds to suppressed levels of p21. These results are unique in describing a role for Rac1 in p21 regulation and may implicate the Rac1 signaling pathway as a potential therapeutic target for controlling prostate cancer cell growth following progression to androgen independence.
- Published
- 2004
- Full Text
- View/download PDF
40. Pancreatic cancer cells express 25-hydroxyvitamin D-1 alpha-hydroxylase and their proliferation is inhibited by the prohormone 25-hydroxyvitamin D3.
- Author
-
Schwartz GG, Eads D, Rao A, Cramer SD, Willingham MC, Chen TC, Jamieson DP, Wang L, Burnstein KL, Holick MF, and Koumenis C
- Subjects
- Base Sequence, Cell Cycle drug effects, Cell Line, Tumor, Cytochrome P-450 Enzyme System genetics, DNA Primers, Humans, Pancreatic Neoplasms pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Steroid Hydroxylases genetics, Vitamin D3 24-Hydroxylase, Calcifediol pharmacology, Cell Division drug effects, Cytochrome P-450 Enzyme System metabolism, Pancreatic Neoplasms enzymology, Steroid Hydroxylases metabolism
- Abstract
The steroid hormone 1,25-dihydroxyvitamin D(3), [1,25(OH)(2)D(3), calcitriol], the active metabolite of vitamin D, exerts pleiotropic antitumor effects against several malignancies. However, the clinical use of this hormone is limited by hypercalcemia. 25-Hydroxyvitamin D(3), the prohormone of 1,25(OH)(2)D(3), is hydroxylated to the active hormone by the enzyme 25-hydroxyvitamin-1-alpha-hydroxylase [1 alpha(OH)ase]. 1 alpha(OH)ase is found primarily in the kidney, but also is expressed in the prostate, colon and other tissues. Using immunohistochemistry, we report that 1 alpha(OH)ase is highly expressed in both normal and malignant pancreatic tissue. Expression of this enzyme and enzymatic activity was also detected in four pancreatic tumor cell lines. 25(OH)D(3) inhibited the growth of three of four pancreatic cell lines in a manner that correlated with the level of induction of the cyclin-dependent kinase inhibitors p21 and p27 and with the induction of cell cycle arrest at the G(1)/S checkpoint. The growth of a cell line stably transfected with a mutant Ki-ras allele and of a second cell line with an endogenous Ki-ras activating mutation was also inhibited by 25(OH)D(3), indicating that activating Ki-Ras mutations, which occur in almost 90% of pancreatic adenocarcinomas, do not interfere with the growth-inhibitory effects of 25(OH)D(3). The expression of 1 alpha(OH)ase in normal and malignant pancreatic tissue and the antiproliferative effects of the prohormone in these cells, suggest that 25(OH)D(3) may offer possible therapeutic and chemopreventive options for pancreatic cancer.
- Published
- 2004
- Full Text
- View/download PDF
41. Hormone resistance: it's SMRT to fight repression.
- Author
-
Burnstein KL and Luetje CW
- Subjects
- Drug Resistance, Humans, Nuclear Receptor Co-Repressor 2, DNA-Binding Proteins physiology, Hormones physiology, Repressor Proteins physiology
- Published
- 2004
- Full Text
- View/download PDF
42. Antiandrogenic activity of extracts of diesel exhaust particles emitted from diesel-engine truck under different engine loads and speeds.
- Author
-
Okamura K, Kizu R, Toriba A, Murahashi T, Mizokami A, Burnstein KL, Klinge CM, and Hayakawa K
- Subjects
- Adenocarcinoma enzymology, Adenocarcinoma genetics, Cell Line, Tumor drug effects, Genes, Reporter, Humans, Luciferases genetics, Luciferases metabolism, Male, Prostate-Specific Antigen genetics, Prostate-Specific Antigen metabolism, Prostatic Neoplasms enzymology, Prostatic Neoplasms genetics, Receptors, Aryl Hydrocarbon agonists, Receptors, Aryl Hydrocarbon antagonists & inhibitors, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae genetics, Androgen Antagonists toxicity, Androgen Receptor Antagonists, Androgens, Vehicle Emissions toxicity
- Abstract
To clarify the alteration of androgenic and antiandrogenic activities by diesel engine conditions, we collected diesel exhaust particles (DEP) samples emitted from a diesel-engine truck under different conditions of engine loads and vehicle speeds, and DEP extract (DEPE) samples were prepared from each. The androgenic and antiandrogenic activities of the DEPE samples were examined using a prostate specific antigen (PSA) promoter-luciferase reporter gene assay in PC3/AR human prostate cancer cells. While all DEPE samples did not exhibit androgenic effects, the antiandrogenic effects were enhanced by higher engine load but not by higher vehicle speed. In this study, significant correlations between antiandrogenic and aryl hydrocarbon receptor (AhR) agonistic activities were demonstrated in PC3/AR cells by 16 polycyclic aromatic compounds and beta-naphthoflavone. Yeast two-hybrid assay and cytochrome P450 (CYP) 1A1 promoter-luciferase reporter gene assay showed that the antiandrogenic constituents acting as androgen receptor (AR) antagonists and AhR agonists were increased by only the higher engine load. In conclusion, the antiandrogenic effects of DEPE samples were enhanced by a higher engine load which resulted in DEPC samples with elevated AhR agonistic and AR antagonistic activities.
- Published
- 2004
- Full Text
- View/download PDF
43. A new luciferase reporter gene assay for the detection of androgenic and antiandrogenic effects based on a human prostate specific antigen promoter and PC3/AR human prostate cancer cells.
- Author
-
Kizu R, Otsuki N, Kishida Y, Toriba A, Mizokami A, Burnstein KL, Klinge CM, and Hayakawai K
- Subjects
- Cell Line, Tumor, Humans, Male, Androgen Antagonists pharmacology, Androgens pharmacology, Genes, Reporter, Luciferases genetics, Promoter Regions, Genetic, Prostate-Specific Antigen genetics
- Abstract
We developed a new mammalian cell-based luciferase reporter gene assay for androgenic and antiandrogenic activities of chemicals and environmental samples. Environmental samples usually have a complex matrix that may contain the constituents acting as androgen receptor (AR) agonists, AR antagonists or aryl hydrocarbon receptor (AhR) agonists. AhR agonists are known to elicit the antiandrogenic effect through cross-talk between AR and AhR signal transduction pathways. In this study, PC3/AR human prostate carcinoma cells were transiently transfected with a prostate-specific antigen (PSA) promoter-driven luciferase expression plasmid. The cells were treated with a test compound or an environmental sample for 24 h at 37 degrees C and then measured for luciferase activity. The luciferase activity was induced by dihydrotestosterone (DHT) in a concentration-dependent manner in a concentration range from 10 fM to 1 nM. R1881, a synthetic androgen receptor agonist, induced luciferase activity and its inductive effects was additive to that of DHT. The luciferase activity was not induced by cortisol, a glucocorticoid, progesterone, a progestin, and 17beta-estradiol, an estrogen in a concentration range of up to 1 microM. DHT-induced luciferase activity was reduced by bicalutamide and cyproterone acetate, AR antagonists, and also by benzo[a]pyrene, an aryl hydrocarbon receptor agonist, through AhR-mediated pathways. All of these findings indicate that the present assay system correctly responds to AR agonists, AR antagonists and AhR agonist and, therefore, it is a powerful tool for the sensitive and selective screening of chemicals and environmental samples for their androgenic and antiandrogenic activities. We developed the first assay system, in which the expression of luciferase was driven by the promoter of a prostate-specific antigen gene, a typical human androgen-regulated gene.
- Published
- 2004
- Full Text
- View/download PDF
44. Antiandrogenic activities of diesel exhaust particle extracts in PC3/AR human prostate carcinoma cells.
- Author
-
Kizu R, Okamura K, Toriba A, Mizokami A, Burnstein KL, Klinge CM, and Hayakawa K
- Subjects
- Adenocarcinoma enzymology, Adenocarcinoma genetics, Anilides pharmacology, Benzoflavones pharmacology, Cell Line, Tumor drug effects, Cytochrome P-450 CYP1A1 antagonists & inhibitors, Cytochrome P-450 CYP1A1 biosynthesis, Dihydrotestosterone pharmacology, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Humans, Luciferases antagonists & inhibitors, Luciferases genetics, Luciferases metabolism, Male, Nitriles, Polycyclic Aromatic Hydrocarbons analysis, Polycyclic Aromatic Hydrocarbons toxicity, Proadifen pharmacology, Prostate-Specific Antigen genetics, Prostate-Specific Antigen metabolism, Prostatic Neoplasms enzymology, Prostatic Neoplasms genetics, Receptors, Androgen metabolism, Receptors, Aryl Hydrocarbon agonists, Receptors, Aryl Hydrocarbon antagonists & inhibitors, Receptors, Aryl Hydrocarbon metabolism, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae genetics, Tosyl Compounds, Vehicle Emissions analysis, beta-Galactosidase genetics, beta-Galactosidase metabolism, Androgen Antagonists toxicity, Androgen Receptor Antagonists, Androgens, Vehicle Emissions toxicity
- Abstract
We collected diesel exhaust particles (DEPs) emitted from three diesel-engine vehicles--a car, a bus, and a truck--in daily use, and prepared DEP extracts (DEPEs), designated as EC, EB, or ET, respectively. The androgenic and antiandrogenic effects of the DEPE samples were examined by a luciferase reporter assay in human prostate carcinoma PC3/AR cells transiently transfected with a prostate specific antigen gene promoter-driven luciferase expression vector pGLPSA5.8. PC3/AR is a subline of human prostate carcinoma PC3 transformed to stably express wild-type human androgen receptor (AR). While DEPE samples did not exhibit any androgenic effect, they exerted antiandrogenic effect, inhibiting dihydrotestosterone (10 pM) -induced luciferase activity by 24 to 52% at an extract concentration of 10 microg/ml. The antiandrogenic effect was greater in the following order: ET > EB > EC. Co-treatment of PC3/AR cells with SKF-525A, a nonselective inhibitor of cytochrome P450 (CYP) enzymes, enhanced the antiandrogenic effect, indicating that the antiandrogenic effect is caused by intact species of DEPE constituents. The antiandrogenic effect of DEPE samples was reversed by alpha-naphthoflavone, an aryl hydrocarbon receptor (AhR) antagonist. The antiandrogenic activity of a DEPE sample correlated with its AhR agonist activity assayed in PC3/AR cells transiently transfected with CYP1A1 gene promoter-driven luciferase expression vector pLUC1A1. Equimolar mixtures of ten polycyclic aromatic hydrocarbons (PAHs) having four or more rings, structures found in the DEPEs, showed significant antiandrogenic effects and AhR agonist activity at concentrations equivalent to those found in DEPE samples. Further, DEPE samples elicited only antiandrogenic effects in recombinant yeast cells, which express beta-galactosidase in response to androgen. A competitive AR binding assay showed that AR-binding constituents exist in DEPE samples, indicating that greater part of AR-binding constituents in DEPEs are AR antagonists. All these findings show that DEPE samples exhibit significant antiandrogenic effect in cell-based transcription assay and that this effect is due in part to the constituents with AhR agonist activity including PAHs and to the constituents with AR antagonist activity.
- Published
- 2003
- Full Text
- View/download PDF
45. Vitamin D inhibits G1 to S progression in LNCaP prostate cancer cells through p27Kip1 stabilization and Cdk2 mislocalization to the cytoplasm.
- Author
-
Yang ES and Burnstein KL
- Subjects
- CDC2-CDC28 Kinases antagonists & inhibitors, Cell Cycle Proteins biosynthesis, Cell Cycle Proteins genetics, Cell Division drug effects, Cell Line, Tumor, Cyclin-Dependent Kinase 2, Cyclin-Dependent Kinase Inhibitor p27, G1 Phase physiology, Half-Life, Humans, Male, Phosphorylation, Protein Transport, S Phase, S-Phase Kinase-Associated Proteins metabolism, Tumor Suppressor Proteins biosynthesis, Tumor Suppressor Proteins genetics, CDC2-CDC28 Kinases metabolism, Calcitriol pharmacology, Cell Cycle Proteins metabolism, Cytoplasm metabolism, G1 Phase drug effects, Prostatic Neoplasms pathology, Tumor Suppressor Proteins metabolism
- Abstract
1,25-(OH)2 vitamin D3 (1,25-(OH)2D3) exerts antiproliferative effects via cell cycle regulation in a variety of tumor cells, including prostate. We have previously shown that in the human prostate cancer cell line LN-CaP, 1,25-(OH)2D3 mediates an increase in cyclin-dependent kinase inhibitor p27Kip1 levels, inhibition of cyclin-dependent kinase 2 (Cdk2) activity, hypophosphorylation of retinoblastoma protein, and accumulation of cells in G1. In this study, we investigated the mechanism whereby 1,25-(OH)2D3 increases p27 levels. 1,25-(OH)2D3 had no effect on p27 mRNA levels or on the regulation of a 3.5-kb fragment of the p27 promoter. The rate of p27 protein synthesis was not affected by 1,25-(OH)2D3 as measured by luciferase activity driven by the 5'- and 3'-untranslated regions of p27 that regulate p27 protein synthesis. Pulse-chase analysis of 35S-labeled p27 revealed an increased p27 protein half-life with 1,25-(OH)2D3 treatment. Because Cdk2-mediated phosphorylation of p27 at Thr187 targets p27 for Skp2-mediated degradation, we examined the phosphorylation status of p27 in 1,25-(OH)2D3-treated cells. 1,25-(OH)2D3 decreased levels of Thr187 phosphorylated p27, consistent with inhibition of Thr187 phosphorylation-dependent p27 degradation. In addition, 1,25-(OH)2D3 reduced Skp2 protein levels in LNCaP cells. Cdk2 is activated in the nucleus by Cdk-activating kinase through Thr160 phosphorylation and by cdc25A phosphatase via Thr14 and Tyr15 dephosphorylation. Interestingly, 1,25-(OH)2D3 decreased nuclear Cdk2 levels as assessed by subcellular fractionation and confocal microscopy. Inhibition of Cdk2 by 1,25-(OH)2D3 may thus involve two mechanisms: 1) reduced nuclear Cdk2 available for cyclin binding and activation and 2) impairment of cyclin E-Cdk2-dependent p27 degradation through cytoplasmic mislocalization of Cdk2. These data suggest that Cdk2 mislocalization is central to the antiproliferative effects of 1,25-(OH)2D3.
- Published
- 2003
- Full Text
- View/download PDF
46. A role of aryl hydrocarbon receptor in the antiandrogenic effects of polycyclic aromatic hydrocarbons in LNCaP human prostate carcinoma cells.
- Author
-
Kizu R, Okamura K, Toriba A, Kakishima H, Mizokami A, Burnstein KL, and Hayakawa K
- Subjects
- Gene Expression Regulation, Neoplastic drug effects, Humans, Male, Prostate-Specific Antigen biosynthesis, Prostate-Specific Antigen genetics, Proto-Oncogene Proteins c-fos, Proto-Oncogene Proteins c-jun, RNA, Messenger, Receptors, Androgen drug effects, Receptors, Androgen physiology, Transcription Factor AP-1 physiology, Androgen Antagonists pharmacology, Polycyclic Aromatic Hydrocarbons pharmacology, Prostatic Neoplasms metabolism, Receptors, Aryl Hydrocarbon physiology, Tumor Cells, Cultured metabolism
- Abstract
The role of aryl hydrocarbon receptor (AhR) on the antiandrogenic effects of polycyclic aromatic hydrocarbons (PAHs) was studied in LNCaP cells. The PAHs used in this study were chrysene (Chr), benzo[ k]fluoranthene (BkF), benzo[ a]pyrene (BaP), anthracene (Ant) and pyrene (Pyr). Chr, BkF and BaP acted as AhR agonists in LNCaP cells, while Ant and Pyr did not. The antiandrogenic effects of the PAHs were evaluated on the basis of regulation of prostate-specific antigen (PSA) mRNA and protein levels by 5alpha-dihydrotestosterone (DHT). Chr, BkF and BaP exhibited an antiandrogenic effect, but Ant and Pyr did not. alpha-Naphthoflavone (alpha-NF), an AhR antagonist, reversed the antiandrogen action of Chr, BkF and BaP, suggesting a requirement for activated AhR. The antiandrogenic PAHs did not significantly decrease androgen receptor (AR) levels or cellular DHT concentrations. Gel mobility shift assays revealed that Chr, BkF and BaP inhibited the binding of AR in nuclear extracts to oligonucleotide probes containing the AR-responsive element (ARE), whereas Ant and Pyr had no effect. The antiandrogenic PAHs elevated mRNA levels of c-fos and c-jun. Since activator protein-1 (AP-1), a heterodimer of c-jun and c-fos proteins, is known to inhibit binding of AR to ARE by protein-protein interaction with AR, the findings in the present study suggest a possible involvement of AP-1 in the antiandrogenic effects of PAHs acting as AhR agonists. These results suggest that AhR can stimulate AP-1 expression resulting in inhibition of the binding of AR to ARE in the transcription regulatory region of target genes such as PSA.
- Published
- 2003
- Full Text
- View/download PDF
47. Evaluation of vitamin D analogs as therapeutic agents for prostate cancer.
- Author
-
Chen TC, Holick MF, Lokeshwar BL, Burnstein KL, and Schwartz GG
- Subjects
- Animals, Calcitriol analogs & derivatives, Cell Division drug effects, Chloramphenicol O-Acetyltransferase genetics, Chloramphenicol O-Acetyltransferase metabolism, Disease Models, Animal, Humans, Lung Neoplasms pathology, Lung Neoplasms prevention & control, Male, Prostatic Neoplasms pathology, Rats, Receptors, Calcitriol metabolism, Transcriptional Activation drug effects, Tumor Cells, Cultured transplantation, Antineoplastic Agents therapeutic use, Calcitriol therapeutic use, Lung Neoplasms secondary, Prostatic Neoplasms drug therapy, Receptors, Calcitriol genetics
- Abstract
Prostate cancer cells contain specific receptors (VDR) for la,25-dihydroxyvitamin D (1alpha,25(OH)2D), which is known to inhibit the proliferation and invasiveness of these cells. These findings support the use of 1alph,25(OH)2D for prostate cancer therapy. However, because 1alpha,25(OH)2D can cause hypercalcemia, analogs of 1alpha,25(OH)2D that are less calcemic but which exhibit potent antiproliferative activity would be attractive as therapeutic agents. We studied four vitamin D compounds: 25-hydroxyvitaminD3 [25(OH)D3], which is converted to 1alpha,25(OH)2D3 in prostate cells, and three analogs of 1alpha,25(OH)2D3: EB1089, 19-nor-1alpha,25(OH)2D2 and hexafluoro-1alpha,25(OH)2D3 (F6-1alpha,25(OH)2D3). 19-nor-1alpha,25(OH)2D2 has been shown to be less calcemic than 1alpha,25(OH)2D3 in clinical trials. F6-1alpha,25(OH)2D3 has been shown to be 100-fold more active than 1alpha,25(OH)2D3 and to be longer-lasting in inhibiting keratinocyte proliferation in vitro. EB1089 has been shown to be less calcemic than 1alpha,25(OH)2D3 in rats implanted with Leydig cell tumors. For 25(OH)D3, 19-nor-1alpha,25(OH)2D2 and F6-1alpha,25(OH)2D3, we studied the in vitro effects and compared their activity to 1alpha,25(OH)2D3 on cellular proliferation by 3H-thymidine incorporation assay. In addition, we studied transactivation of the VDR in the presence of 25(OH)D3 and 19-nor-1alpha,25(OH)2D2 in prostate cells. For EB1089, we compared its inhibition of prostate cancer metastasis to that induced by 1alpha,25(OH)2D3 in vivo in the rat Dunning MAT LyLu prostate cancer model. We found that 1alpha,25(OH)2D3 and 19-nor-1alpha,25(OH)2D2 caused similar dose-dependent inhibition in 3H-thymidine incorporation into DNA in prostate cells and behaved similarly in the CAT reporter gene transactivation assay in PC-3/VDR cells. F6-1alpha,25(OH)2D3 is 10- to 50-fold more active than 1alpha,25(OH)2D3 in 3H-thymidine incorporation into DNA in the primary cultured prostate cells. Likewise, 25(OH)D3 had comparable antiproliferative activity to la,25(OH)2D3. In the rat model, tumor volumes and the number of metastases in the lungs were significantly reduced by both 1alpha,25(OH)2D3 (10.4 +/- 2.81 tumor foci) and EB1089 (7.7+/-1.29 tumor foci) compared to controls (22.7 +/- 1.98 tumor foci). Although serum calcium levels were significantly elevated in both 1alph,25(OH)2D3- and EB1089-treated rats, EB1089 was significantly less calcemic than 1alpha,25(OH)2D3 (12.59+/-0.21 mg/dl versus 14.47+/-.46 mg/dL; 1 microg/kg; p < 0.001). In conclusion, our data indicate that 25(OH)D3 and the three 1alpha,25(OH)2D analogs represent two different solutions to the problem of hypercalcemia associated with vitamin D-based prostate cancer therapies: 25(OH)D3 requires the presence of 25-hydroxyvitaminD-1alpha-hydroxylase, whereas 19-nor-1alpha,25(OH)2D2, F6-1alpha,25(OH)2D3 and EB1089 do not. These compounds may be good candidates for human clinical trials in prostate cancer.
- Published
- 2003
- Full Text
- View/download PDF
48. 25-Hydroxyvitamin D-1alpha-hydroxylase activity is diminished in human prostate cancer cells and is enhanced by gene transfer.
- Author
-
Whitlatch LW, Young MV, Schwartz GG, Flanagan JN, Burnstein KL, Lokeshwar BL, Rich ES, Holick MF, and Chen TC
- Subjects
- 25-Hydroxyvitamin D3 1-alpha-Hydroxylase genetics, Cells, Cultured, Humans, Male, Prostate cytology, Prostate enzymology, Prostatic Hyperplasia enzymology, Tumor Cells, Cultured, 25-Hydroxyvitamin D3 1-alpha-Hydroxylase metabolism, Prostatic Neoplasms enzymology, Transfection
- Abstract
The hormone 1alpha,25-dihydroxyvitamin D (1alpha,25(OH)(2)D) inhibits growth and induces differentiation of prostate cells. The enzyme responsible for 1alpha,25(OH)(2)D synthesis, 25-hydroxyvitamin D (25(OH)D)-1alpha-hydroxylase (1alpha-OHase), has been demonstrated in human prostate cells. We compared the levels of 1alpha-OHase activity in prostate cancer cell lines, LNCaP, DU145 and PC-3 and in primary cultures of normal, cancerous and benign prostatic hyperplasia (BPH) prostate cells. We observed a marked decrease in 1alpha-OHase activity in prostate cancer cells, including an undetectable level of activity in LNCaP cells. Transient or stable transfection of 1alpha-OHase cDNA into LNCaP cells increased 1alpha-OHase activity from undetectable to 4.95pmole/mg+/-0.69pmole/mg and 5.8pmole/mg+/-0.7pmole/mg protein per hour, respectively. In response to 25(OH)D, the prohormone of 1alpha,25(OH)(2)D, the transfected LNCaP cells showed a significant inhibition of 3H-thymidine incorporation (37%+/-6% and 56%+/-4% at 10(-8)M for transiently and stably transfected cells, respectively). These findings support an important autocrine role for 1alpha,25(OH)(2)D in the prostate and suggest that the re-introduction of the 1alpha-OHase gene to prostate cancer cells, in conjunction with the systemic administration of 25(OH)D, constitutes an endocrine form of gene therapy that may be less toxic than the systemic administration of 1alpha,25(OH)(2)D.
- Published
- 2002
- Full Text
- View/download PDF
49. Vitamin D-mediated growth inhibition of an androgen-ablated LNCaP cell line model of human prostate cancer.
- Author
-
Yang ES, Maiorino CA, Roos BA, Knight SR, and Burnstein KL
- Subjects
- Androgens pharmacology, Cell Cycle, Cell Cycle Proteins metabolism, Cell Division drug effects, Cell Survival, Cyclin-Dependent Kinase Inhibitor p27, Humans, Male, Receptors, Androgen biosynthesis, Receptors, Androgen genetics, Transfection, Tumor Cells, Cultured drug effects, Tumor Suppressor Proteins metabolism, Prostatic Neoplasms pathology, Vitamin D pharmacology
- Abstract
1,25-(OH)(2) vitamin D(3) (1,25-(OH)(2) D), the active metabolite of vitamin D, exerts antiproliferative effects on a variety of tumor cells including prostate. This inhibition requires vitamin D receptors (VDRs) as well as downstream effects on the G1 to S phase checkpoint of the cell cycle. Recent data raise the possibility that androgen plays a role in the antiproliferative effects of 1,25-(OH)(2) D in prostate cancer cells; however, this hypothesis has been difficult to test rigorously as the majority of prostate cancer cell lines (unlike human prostate tumors) lack androgen receptors (ARs). We utilized two different models of androgen-independent prostate cancer that express functional ARs and VDRs to evaluate a possible role of androgen in 1,25-(OH)(2) D mediated growth inhibition. We stably introduced the AR cDNA into the human prostate cancer cell line ALVA 31, which expresses functional VDR but is relatively resistant to growth inhibition by 1,25-(OH)(2) D. Neither ALVA-AR nor the control cells, ALVA-NEO, exhibited substantial growth inhibition by 1,25-(OH)(2) D in the presence or absence of androgen. This observation suggests that the basis for the resistance of ALVA 31 to 1,25-(OH)(2) D-mediated growth inhibition is not the lack of AR. The second model was LNCaP-104R1, an AR-expressing androgen independent prostate cancer cell line derived from androgen dependent LNCaP. 1,25-(OH)(2) D inhibited the growth of LNCaP-104R1 cells in the absence of androgen and this effect was not blocked by the antiandrogen Casodex. As was observed in the parental LNCaP cells, this effect was correlated with G1 phase cell cycle accumulation and upregulation of the cyclin dependent kinase inhibitor (CKI) p27, as well as increased association of p27 with cyclin dependent kinase 2. These findings suggest that the antiproliferative effects of 1,25-(OH)(2) D do not require androgen-activated AR but do involve 1,25-(OH)(2) D induction of CKIs required for G1 cell cycle checkpoint control.
- Published
- 2002
- Full Text
- View/download PDF
50. Correspondence re: S. E. Blutt, T. C. Polek, L. V. Stewart, M. W. Kattan, and N. L. Weigel, A calcitriol analogue, EB1089, inhibits the growth of LNCaP tumors in nude mice. Cancer Res., 60: 779-782, 2000.
- Author
-
Schwartz GG, Lokeshwar BL, and Burnstein KL
- Subjects
- Animals, Cell Division drug effects, Humans, Male, Prostatic Neoplasms pathology, Rats, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Calcitriol analogs & derivatives, Calcitriol pharmacology, Prostatic Neoplasms drug therapy
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.