230 results on '"Ralph Buttyan"'
Search Results
2. FOXC1 Activates Smoothened-Independent Hedgehog Signaling in Basal-like Breast Cancer
- Author
-
Bingchen Han, Ying Qu, Yanli Jin, Yi Yu, Nan Deng, Kolja Wawrowsky, Xiao Zhang, Na Li, Shikha Bose, Qiang Wang, Sugunadevi Sakkiah, Ravinder Abrol, Tor W. Jensen, Benjamin P. Berman, Hisashi Tanaka, Jeffrey Johnson, Bowen Gao, Jijun Hao, Zhenqiu Liu, Ralph Buttyan, Partha S. Ray, Mien-Chie Hung, Armando E. Giuliano, and Xiaojiang Cui
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The mesoderm- and epithelial-mesenchymal transition-associated transcription factor FOXC1 is specifically overexpressed in basal-like breast cancer (BLBC), but its biochemical function is not understood. Here, we demonstrate that FOXC1 controls cancer stem cell (CSC) properties enriched in BLBC cells via activation of Smoothened (SMO)-independent Hedgehog (Hh) signaling. This non-canonical activation of Hh is specifically mediated by Gli2. Furthermore, we show that the N-terminal domain of FOXC1 (aa 1–68) binds directly to an internal region (aa 898–1168) of Gli2, enhancing the DNA-binding and transcription-activating capacity of Gli2. FOXC1 expression correlates with that of Gli2 and its targets in human breast cancers. Moreover, FOXC1 overexpression reduces sensitivity to anti-Hedgehog (Hh) inhibitors in BLBC cells and xenograft tumors. Together, these findings reveal FOXC1-mediated non-canonical Hh signaling that determines the BLBC stem-like phenotype and anti-Hh sensitivity, supporting inhibition of FOXC1 pathways as potential approaches for improving BLBC treatment.
- Published
- 2015
- Full Text
- View/download PDF
3. Effects of androgen receptor and androgen on gene expression in prostate stromal fibroblasts and paracrine signaling to prostate cancer cells.
- Author
-
Matthew J Tanner, R Charles Welliver, Mengqian Chen, Michael Shtutman, Alejandro Godoy, Gary Smith, Badar M Mian, and Ralph Buttyan
- Subjects
Medicine ,Science - Abstract
The androgen receptor (AR) is expressed in a subset of prostate stromal cells and functional stromal cell AR is required for normal prostate developmental and influences the growth of prostate tumors. Although we are broadly aware of the specifics of the genomic actions of AR in prostate cancer cells, relatively little is known regarding the gene targets of functional AR in prostate stromal cells. Here, we describe a novel human prostate stromal cell model that enabled us to study the effects of AR on gene expression in these cells. The model involves a genetically manipulated variant of immortalized human WPMY-1 prostate stromal cells that overexpresses wildtype AR (WPMY-AR) at a level comparable to LNCaP cells and is responsive to dihydrotestosterone (DHT) stimulation. Use of WPMY-AR cells for gene expression profiling showed that the presence of AR, even in the absence of DHT, significantly altered the gene expression pattern of the cells compared to control (WPMY-Vec) cells. Treatment of WPMY-AR cells, but not WPMY-Vec control cells, with DHT resulted in further changes that affected the expression of 141 genes by 2-fold or greater compared to vehicle treated WPMY-AR cells. Remarkably, DHT significantly downregulated more genes than were upregulated but many of these changes reversed the initial effects of AR overexpression alone on individual genes. The genes most highly effected by DHT treatment were categorized based upon their role in cancer pathways or in cell signaling pathways (transforming growth factor-β, Wnt, Hedgehog and MAP Kinase) thought to be involved in stromal-epithelial crosstalk during prostate or prostate cancer development. DHT treatment of WPMY-AR cells was also sufficient to alter their paracrine potential for prostate cancer cells as conditioned medium from DHT-treated WPMY-AR significantly increased growth of LNCaP cells compared to DHT-treated WPMY-Vec cell conditioned medium.
- Published
- 2011
- Full Text
- View/download PDF
4. Supplementary Figures S1-S4 from Transient Sox9 Expression Facilitates Resistance to Androgen-Targeted Therapy in Prostate Cancer
- Author
-
Ralph Buttyan, Xuesen Dong, Colin Collins, Colleen Nelson, Brett G. Hollier, Martin E. Gleave, Jane Foo, Mike Wang, Jackson Moore, Jessica M. Lovnicki, Varune R. Ramnarine, Ladan Fazli, Ahn R. Lee, Sarah Truong, Na Li, Amy A. Lubik, Josselin Caradec, Shabnam Massah, and Mannan Nouri
- Abstract
Supplementary Figures S1-S4
- Published
- 2023
5. Supplementary Figure 4 from Not all NOTCH Is Created Equal: The Oncogenic Role of NOTCH2 in Bladder Cancer and Its Implications for Targeted Therapy
- Author
-
Peter C. Black, Akio Matsubara, Ladan Fazli, Ralph Buttyan, Manuel Altamirano-Dimas, Htoo Zarni Oo, Na Li, Shannon Awrey, Wolfgang Jäger, Akihiro Goriki, Alexander W. Wyatt, Kilian M. Gust, and Tetsutaro Hayashi
- Abstract
NOTCH2-inactivating antibody inhibited tumor progression in vitro and in vivo.
- Published
- 2023
6. Supplementary Figure 3 from Not all NOTCH Is Created Equal: The Oncogenic Role of NOTCH2 in Bladder Cancer and Its Implications for Targeted Therapy
- Author
-
Peter C. Black, Akio Matsubara, Ladan Fazli, Ralph Buttyan, Manuel Altamirano-Dimas, Htoo Zarni Oo, Na Li, Shannon Awrey, Wolfgang Jäger, Akihiro Goriki, Alexander W. Wyatt, Kilian M. Gust, and Tetsutaro Hayashi
- Abstract
NOTCH2 silencing inhibited tumor progression through decreased cell proliferation and invasion.
- Published
- 2023
7. Supplementary Figure 2 from Not all NOTCH Is Created Equal: The Oncogenic Role of NOTCH2 in Bladder Cancer and Its Implications for Targeted Therapy
- Author
-
Peter C. Black, Akio Matsubara, Ladan Fazli, Ralph Buttyan, Manuel Altamirano-Dimas, Htoo Zarni Oo, Na Li, Shannon Awrey, Wolfgang Jäger, Akihiro Goriki, Alexander W. Wyatt, Kilian M. Gust, and Tetsutaro Hayashi
- Abstract
NOTCH2-induced cell invasion was blocked by downstream inhibition of canonical NOTCH2 signaling.
- Published
- 2023
8. Supplementary Materials and Methods from Transient Sox9 Expression Facilitates Resistance to Androgen-Targeted Therapy in Prostate Cancer
- Author
-
Ralph Buttyan, Xuesen Dong, Colin Collins, Colleen Nelson, Brett G. Hollier, Martin E. Gleave, Jane Foo, Mike Wang, Jackson Moore, Jessica M. Lovnicki, Varune R. Ramnarine, Ladan Fazli, Ahn R. Lee, Sarah Truong, Na Li, Amy A. Lubik, Josselin Caradec, Shabnam Massah, and Mannan Nouri
- Abstract
Supplementary Materials and Methods with detailed methods and primer and antibody lists.
- Published
- 2023
9. Data from Not all NOTCH Is Created Equal: The Oncogenic Role of NOTCH2 in Bladder Cancer and Its Implications for Targeted Therapy
- Author
-
Peter C. Black, Akio Matsubara, Ladan Fazli, Ralph Buttyan, Manuel Altamirano-Dimas, Htoo Zarni Oo, Na Li, Shannon Awrey, Wolfgang Jäger, Akihiro Goriki, Alexander W. Wyatt, Kilian M. Gust, and Tetsutaro Hayashi
- Abstract
Purpose: Recent molecular analyses of bladder cancer open the door to significant advances in targeted therapies. NOTCH has been identified as a tumor suppressor in bladder cancer, but prior reports have focused on NOTCH1. Here we hypothesized that NOTCH2 is an oncogene suitable for therapeutic targeting in bladder cancer.Experimental design: We studied genomic aberrations of NOTCH, compared survival and tumor progression according to NOTCH2 expression levels, and studied NOTCH2 function in vitro and vivo.Results: We report a high rate of NOTCH2 copy number gain in bladder cancer. High NOTCH2 expression was identified especially in the basal subtype and in mesenchymal tumors. NOTCH2 activation correlated with adverse disease parameters and worse prognosis by immunohistochemistry. Forced overexpression of the intracellular domain of NOTCH2 (N2ICD) induced cell growth and invasion by cell-cycle progression, maintenance of stemness and epithelial-to-mesenchymal transition (EMT). These effects were abrogated by silencing of CSL, indicating that the effects were mediated through the canonical NOTCH signaling pathway. In an orthotopic xenograft model, forced overexpression of N2ICD increased growth, invasion, and metastasis. To explore the potential for therapeutic targeting of NOTCH2, we first silenced the receptor with shRNA and subsequently treated with a specific inhibitory antibody. Both interventions decreased cell growth, invasion, and metastasis in vitro and in the orthotopic xenograft model.Conclusions: We have demonstrated that NOTCH2 acts as an oncogene that promotes bladder cancer growth and metastasis through EMT, cell-cycle progression, and maintenance of stemness. Inhibition of NOTCH2 is a rational novel treatment strategy for invasive bladder cancer. Clin Cancer Res; 22(12); 2981–92. ©2016 AACR.
- Published
- 2023
10. Data from Transient Sox9 Expression Facilitates Resistance to Androgen-Targeted Therapy in Prostate Cancer
- Author
-
Ralph Buttyan, Xuesen Dong, Colin Collins, Colleen Nelson, Brett G. Hollier, Martin E. Gleave, Jane Foo, Mike Wang, Jackson Moore, Jessica M. Lovnicki, Varune R. Ramnarine, Ladan Fazli, Ahn R. Lee, Sarah Truong, Na Li, Amy A. Lubik, Josselin Caradec, Shabnam Massah, and Mannan Nouri
- Abstract
Purpose:Patients with metastatic prostate cancer are increasingly presenting with treatment-resistant, androgen receptor–negative/low (AR−/Low) tumors, with or without neuroendocrine characteristics, in processes attributed to tumor cell plasticity. This plasticity has been modeled by Rb1/p53 knockdown/knockout and is accompanied by overexpression of the pluripotency factor, Sox2. Here, we explore the role of the developmental transcription factor Sox9 in the process of prostate cancer therapy response and tumor progression.Experimental Design:Unique prostate cancer cell models that capture AR−/Low stem cell–like intermediates were analyzed for features of plasticity and the functional role of Sox9. Human prostate cancer xenografts and tissue microarrays were evaluated for temporal alterations in Sox9 expression. The role of NF-κB pathway activity in Sox9 overexpression was explored.Results:Prostate cancer stem cell–like intermediates have reduced Rb1 and p53 protein expression and overexpress Sox2 as well as Sox9. Sox9 was required for spheroid growth, and overexpression increased invasiveness and neural features of prostate cancer cells. Sox9 was transiently upregulated in castration-induced progression of prostate cancer xenografts and was specifically overexpressed in neoadjuvant hormone therapy (NHT)–treated patient tumors. High Sox9 expression in NHT-treated patients predicts biochemical recurrence. Finally, we link Sox9 induction to NF-κB dimer activation in prostate cancer cells.Conclusions:Developmentally reprogrammed prostate cancer cell models recapitulate features of clinically advanced prostate tumors, including downregulated Rb1/p53 and overexpression of Sox2 with Sox9. Sox9 is a marker of a transitional state that identifies prostate cancer cells under the stress of therapeutic assault and facilitates progression to therapy resistance. Its expression may index the relative activity of the NF-κB pathway.
- Published
- 2023
11. Supplementary Figure Legends and Data Tables from Transient Sox9 Expression Facilitates Resistance to Androgen-Targeted Therapy in Prostate Cancer
- Author
-
Ralph Buttyan, Xuesen Dong, Colin Collins, Colleen Nelson, Brett G. Hollier, Martin E. Gleave, Jane Foo, Mike Wang, Jackson Moore, Jessica M. Lovnicki, Varune R. Ramnarine, Ladan Fazli, Ahn R. Lee, Sarah Truong, Na Li, Amy A. Lubik, Josselin Caradec, Shabnam Massah, and Mannan Nouri
- Abstract
Supplementary Figure Legends and Data Table containing developmental reprogramming 59-Gene commonly regulated gene list.
- Published
- 2023
12. Supplementary Figure 1 from Not all NOTCH Is Created Equal: The Oncogenic Role of NOTCH2 in Bladder Cancer and Its Implications for Targeted Therapy
- Author
-
Peter C. Black, Akio Matsubara, Ladan Fazli, Ralph Buttyan, Manuel Altamirano-Dimas, Htoo Zarni Oo, Na Li, Shannon Awrey, Wolfgang Jäger, Akihiro Goriki, Alexander W. Wyatt, Kilian M. Gust, and Tetsutaro Hayashi
- Abstract
NOTCH2 overexpression promoted bladder cancer progression through increased proliferation and invasion.
- Published
- 2023
13. Supplementary Materials and Methods from Not all NOTCH Is Created Equal: The Oncogenic Role of NOTCH2 in Bladder Cancer and Its Implications for Targeted Therapy
- Author
-
Peter C. Black, Akio Matsubara, Ladan Fazli, Ralph Buttyan, Manuel Altamirano-Dimas, Htoo Zarni Oo, Na Li, Shannon Awrey, Wolfgang Jäger, Akihiro Goriki, Alexander W. Wyatt, Kilian M. Gust, and Tetsutaro Hayashi
- Abstract
The lists of antibodies, primers and patients characteristics and detailed information of methods were described in Supplementary Material and Methods.
- Published
- 2023
14. Supplementary Figure 1 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Author
-
Alexandre de la Taille, Francis Vacherot, Ralph Buttyan, Stéphane Culine, Laurent Salomon, Bernard Paule, Estelle Coppolani, Nathalie Nicolaiew, Pascale Soyeux, Laurence Kheuang, Nanor Sirab, Yves Allory, Pascale Maillé, Stéphane Terry, and Guillaume Ploussard
- Abstract
Supplementary Figure 1 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Published
- 2023
15. Supplementary Table 1 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Author
-
Alexandre de la Taille, Francis Vacherot, Ralph Buttyan, Stéphane Culine, Laurent Salomon, Bernard Paule, Estelle Coppolani, Nathalie Nicolaiew, Pascale Soyeux, Laurence Kheuang, Nanor Sirab, Yves Allory, Pascale Maillé, Stéphane Terry, and Guillaume Ploussard
- Abstract
Supplementary Table 1 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Published
- 2023
16. Supplementary Figure 5 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Author
-
Alexandre de la Taille, Francis Vacherot, Ralph Buttyan, Stéphane Culine, Laurent Salomon, Bernard Paule, Estelle Coppolani, Nathalie Nicolaiew, Pascale Soyeux, Laurence Kheuang, Nanor Sirab, Yves Allory, Pascale Maillé, Stéphane Terry, and Guillaume Ploussard
- Abstract
Supplementary Figure 5 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Published
- 2023
17. Supplementary Figure 3 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Author
-
Alexandre de la Taille, Francis Vacherot, Ralph Buttyan, Stéphane Culine, Laurent Salomon, Bernard Paule, Estelle Coppolani, Nathalie Nicolaiew, Pascale Soyeux, Laurence Kheuang, Nanor Sirab, Yves Allory, Pascale Maillé, Stéphane Terry, and Guillaume Ploussard
- Abstract
Supplementary Figure 3 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Published
- 2023
18. Supplementary Methods from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Author
-
Alexandre de la Taille, Francis Vacherot, Ralph Buttyan, Stéphane Culine, Laurent Salomon, Bernard Paule, Estelle Coppolani, Nathalie Nicolaiew, Pascale Soyeux, Laurence Kheuang, Nanor Sirab, Yves Allory, Pascale Maillé, Stéphane Terry, and Guillaume Ploussard
- Abstract
Supplementary Methods from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Published
- 2023
19. Supplementary Figure Legends 1-5 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Author
-
Alexandre de la Taille, Francis Vacherot, Ralph Buttyan, Stéphane Culine, Laurent Salomon, Bernard Paule, Estelle Coppolani, Nathalie Nicolaiew, Pascale Soyeux, Laurence Kheuang, Nanor Sirab, Yves Allory, Pascale Maillé, Stéphane Terry, and Guillaume Ploussard
- Abstract
Supplementary Figure Legends 1-5 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Published
- 2023
20. Supplementary Figure 2 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Author
-
Alexandre de la Taille, Francis Vacherot, Ralph Buttyan, Stéphane Culine, Laurent Salomon, Bernard Paule, Estelle Coppolani, Nathalie Nicolaiew, Pascale Soyeux, Laurence Kheuang, Nanor Sirab, Yves Allory, Pascale Maillé, Stéphane Terry, and Guillaume Ploussard
- Abstract
Supplementary Figure 2 from Class III β-Tubulin Expression Predicts Prostate Tumor Aggressiveness and Patient Response to Docetaxel-Based Chemotherapy
- Published
- 2023
21. Abstract 3080: Characterization of steroid receptor mediated activation of GLI as driving force of breast cancer growth
- Author
-
Shabnam Massah, Maria Guo, Jane Foo, Ralph Buttyan, Artem Cherkasov, and Nada Lallous
- Subjects
Cancer Research ,Oncology - Abstract
Background: Hedgehog (Hh) signaling pathway plays a fundamental role in the early stages of development by regulating morphogenesis. GLI transcription factors are drivers of this signaling pathway by regulating the expression of growth related genes. Hyperactivation of GLI proteins have been associated with several cancers including medulloblastoma, glioblastoma, ovarian, prostate and breast cancers. Our study in breast cancer (BCa) suggests transcriptional activation of GLI3 in estrogen receptor alpha (ERα) positive cells upon estradiol stimulation. Loss of ERα greatly decreases GLI3 protein stability and stimulation with estradiol significantly increases GLI3 stability in ER-positive BCa cells. We have discovered that GLI3 forms nuclear complexes with ERα upon estradiol stimulation and that the loss of GLI3 reduces BCa cells growth. Therefore, we hypothesize that ERα-GLI3 complex orchestrates BCa transcriptome required for cell growth and development. Our study focuses on characterizing ERα mediated activation of GLI3 in BCa cells and the possible compensation by other steroid receptors such as the androgen receptor (AR) and the glucocorticoid receptor (GR) in ER-negative BCa cells. Method: We examined ERα- GLI transcriptional activity by RNA sequencing upon stimulation with estradiol and inhibition of GLI activity by GANT61 in ER-positive MCF7 cells. We also identified ERα domains that are essential for GLI3 binding by immunoprecipitation and proximity ligation assay (PLA). To identify ERα-GLI3 interactome, we have optimized Rapid Immunoprecipitation Mass Spectrometry of Endogenous protein (RIME). We also examined the role of GLI3 in ER-negative BCa cell growth and characterized the role of AR and GR in activating GLI3 by luciferase reporter assay.Results: Inhibition of GLI DNA binding by GANT61 reduced the expression of GLI regulated (CDC20, CDK1, UBE2C) and modified expression of ERα regulated (FOXM1, SPC24, KIF20A, BIRC5) genes, suggesting cooperative role of ERα-GLI3 in mediating growth and metastasis. The optimized RIME assay suggests immunoprecipitation of ERα with GLI3 in BCa cells upon estradiol induction. Immunoprecipitation and PLA studies suggest that ERα-N terminal, DNA binding and C-terminal domains interact with GLI3. We also showed that knockdown of GLI3 reduces growth in ER negative BCa cells and that AR or GR stimulation by dihydrotestosterone and dexamethasone respectively, are important for GLI3 transcriptional activity. Conclusion: Collectively, our results suggest a new role of steroid receptors in regulating GLI oncogenic transcriptional activity in BCa, leading to new therapeutic possibilities for patients. Citation Format: Shabnam Massah, Maria Guo, Jane Foo, Ralph Buttyan, Artem Cherkasov, Nada Lallous. Characterization of steroid receptor mediated activation of GLI as driving force of breast cancer growth [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3080.
- Published
- 2023
22. The evolution of long noncoding RNA acceptance in prostate cancer initiation, progression, and its clinical utility in disease management
- Author
-
Maxim Kobelev, Amina Zoubeidi, Ewan A. Gibb, Mannan Nouri, Yuzhuo Wang, Colin Collins, Dong Lin, Ralph Buttyan, Elai Davicioni, and Varune Rohan Ramnarine
- Subjects
Male ,Urology ,030232 urology & nephrology ,Context (language use) ,Computational biology ,medicine.disease_cause ,Genome ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Drug Discovery ,Biomarkers, Tumor ,medicine ,Humans ,business.industry ,Prostatic Neoplasms ,Cancer ,Prognosis ,medicine.disease ,Long non-coding RNA ,Clinical trial ,Pharmacogenetics ,030220 oncology & carcinogenesis ,RNA, Long Noncoding ,Identification (biology) ,business ,Carcinogenesis ,Procedures and Techniques Utilization - Abstract
Context It is increasingly evident that non–protein-coding regions of the genome can give rise to transcripts that form functional layers of the cancer genome. One of most abundant classes in these regions is long noncoding RNAs (lncRNAs). They have gained increasing attention in prostate cancer (PCa) and paved the way for a greater understanding of these cryptic regulators in cancer. Objective To review current research exploring the functional biology of lncRNAs in PCa over the past three decades. Evidence acquisition A systematic review was performed using PubMed to search for reports with terms “long noncoding RNA”, “prostate”, and “cancer” over the past 30 yr (1988–2018). Evidence synthesis We comprehensively surveyed the literature collected and summarise experiments leading to the characterisation of lncRNAs in PCa. A historical timeline of lncRNA identification is described, where each lncRNA is categorised mechanistically and within the primary areas of carcinogenesis: tumour risk and initiation, tumour promotion, tumour suppression, and tumour treatment resistance. We describe select lncRNAs that exemplify these areas. We also review whether these lncRNAs have a clinical utility in PCa diagnosis, prognosis, and prediction, and as therapeutic targets. Conclusions The biology of lncRNA is multifaceted, demonstrating a complex array of molecular and cellular functions. These studies reveal that lncRNAs are involved in every stage of PCa. Their clinical utility for diagnosis, prognosis, and prediction of PCa is well supported, but further evaluation for their therapeutic candidacy is needed. We provide a detailed resource and view inside the lncRNA landscape for other cancer biologists, oncologists, and clinicians. Patient summary In this study, we review current knowledge of the non–protein-coding genome in prostate cancer (PCa). We conclude that many of these regions are functional and a source of accurate biomarkers in PCa. With a strong research foundation, they hold promise as future therapeutic targets, yet clinical trials are necessary to determine their intrinsic value to PCa disease management.
- Published
- 2019
23. Abstract 5283: Characterization of Gli activation by the estrogen receptor in breast cancer cells
- Author
-
Shabnam Massah, Jane Foo, Na Li, Sarah Truong, Mannan Nouri, Lishi Xie, Gail Prins, Ralph Buttyan, Nada Lallous, and Artem Cherkasov
- Subjects
Cancer Research ,Oncology - Abstract
Background: The nuclear steroid receptor superfamily encompasses a group of proteins best known fortheir functions as primary transcription factors that are conditionally active when bound to a ligand.Here, we show that a prominent member of this family, the estrogen receptor [ER-α] have a secondaryfunction of activating the Gli family of transcription factors. Gli is recognized as the mediator of activeHedgehog (Hh) signaling and plays an important role in cell development and growth. Gli activity isregulated by a post-translational proteolytic process that is suppressed by Hedgehog signaling.Previously we found that in prostate cancer, the ligand activated androgen receptor [AR] recognizes andbinds to Gli proteins at their Protein Processing Domains. This binding stabilizes Gli proteins in their un-proteolyzed active form and bypasses the Hedgehog signaling, thus promoting cancer progressionthrough non-canonical activation of Gli proteins. Due to the high similarity between AR and ER, wehypothesized that a similar Gli regulation could play a role in Breast Cancer (BrCA) progression. We thustested the ability of human ER-α to bind and activate Gli in BrCa and evaluated the role of this pathwayin tumor cell growth. Methods: we measured Gli activity in 293T and BrCa cells (MCF7, T47D, MDA-MB-453) in presence andabsence of steroid ligand using Gli-luciferase reporter assay. We evaluated the interaction between Gli3and ER-α by co-immunoprecipitation and proximity ligation assay and assessed the stability of Gli3stability in BrCa cell extracts by western blots. We also studied the effect of ER-α knockdown ordestabilization (by fulvestrant treatment) on Gli3 stability, formation of intranuclear ER-α-Gli3complexes and Gli reporter activity. We also measured the expression level of Gli target genes in thepresence and absence of estradiol by qPCR in BrCa cells. Lastly, we evaluated the importance of Gli3expression on BrCa growth by Gli3 knockdown and Cyquant assay. Results: We found that ER co-immunoprecipitates with Gli3. Transfection with ER-α increased Glireporter activity which was further increased by estradiol treatment. Acute (2hr) estradiol treatmentincreased intranuclear ER-α-Gli3 complex formation in BrCa cells. Chronic (48hr) estradiol treatmentincreased Gli3 stability and endogenous activity in BrCa cells. Destabilization or knockdown of ER-αdecreased estradiol-induced formation of ER-α-Gli3 complexes as well as Gli activity and stability in BrCacells. In addition, siRNA knockdown of Gli3 reduced growth in BrCa cells. Conclusion: Collectively our results uncovered a new role of the steroid receptors ER and AR inregulating Gli oncogenic transcriptional activity in BrCa and PCa, respectively. Citation Format: Shabnam Massah, Jane Foo, Na Li, Sarah Truong, Mannan Nouri, Lishi Xie, Gail Prins, Ralph Buttyan, Nada Lallous, Artem Cherkasov. Characterization of Gli activation by the estrogen receptor in breast cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5283.
- Published
- 2022
24. Gli activation by the estrogen receptor in breast cancer cells: Regulation of cancer cell growth by Gli3
- Author
-
Gail S. Prins, Jane Foo, Na Li, Shabnam Massah, Mannan Nouri, Ralph Buttyan, Lishi Xie, and Sarah Truong
- Subjects
musculoskeletal diseases ,0301 basic medicine ,animal structures ,Estrogen receptor ,030209 endocrinology & metabolism ,Breast Neoplasms ,Proximity ligation assay ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Zinc Finger Protein Gli3 ,Cell Line, Tumor ,medicine ,Humans ,skin and connective tissue diseases ,Molecular Biology ,Cell Proliferation ,Cell Nucleus ,Gene knockdown ,Fulvestrant ,Estradiol ,Cell growth ,Chemistry ,Protein Stability ,fungi ,Transfection ,Androgen receptor ,030104 developmental biology ,HEK293 Cells ,Receptors, Estrogen ,embryonic structures ,Cancer cell ,Cancer research ,Female ,medicine.drug - Abstract
Background Gli is an oncogenic transcription factor family thought to be involved in breast cancer (BrCa) cell growth. Gli activity is regulated by a post-translational proteolytic process that is suppressed by Hedgehog signaling. In prostate cancer cells, however, Gli activation is mediated by an interaction of active androgen receptor proteins with Gli3 that stabilizes Gli3 in its un-proteolyzed form. Here we show that the estrogen receptor (ER), ERα, also binds Gli3 and activates Gli in BrCa cells. Moreover, we show that ER + BrCa cells are dependent on Gli3 for cancer cell growth. Methods Transfection with Gli-luciferase reporter was used to report Gli activity in 293FT or BrCa cells (MCF7, T47D, MDA-MB-453) with or without steroid ligands. Co-immunoprecipitation and proximity ligation were used to show association of Gli3 with ERα. Gli3 stability was determined by western blots of BrCa cell extracts. ERα knockdown or destabilization (by fulvestrant) was used to assess how loss of ERα affects estradiol-induced Gli reporter activity, formation of intranuclear ERα-Gli3 complexes and Gli3 stability. Expression of Gli1 and/or other endogenous Gli-target genes in BrCa cells were measured by qPCR in the presence or absence of estradiol. Gli3 knockdown was assessed for effects on BrCa cell growth using the Cyquant assay. Results ERα co-transfection increased Gli reporter activity in 293FT cells that was further increased by estradiol. Gli3 co-precipitated in ERα immunoprecipitates. Acute (2 h) estradiol increased Gli reporter activity and the formation of intranuclear ERα-Gli3 complexes in ER + BrCa cells but more chronic estradiol (48 h) reduced ERα-Gli complexes commensurate with reduced ERα levels. Gli3 stability and endogenous activity was only increased by more chronic estradiol treatment. Fulvestrant or ERα knockdown suppressed E2-induction of Gli activity, intranuclear ERα-Gli3 complexes and stabilization of Gli3. Gli3 knockdown significantly reduced the growth of BrCa cells. Conclusions ERα interacts with Gli3 in BrCa cells and estradiol treatment leads to Gli3 stabilization and increased expression of Gli-target genes. Furthermore, we found tthat Gli3 is necessary for BrCa cell growth. These results support the idea that the ERα-Gli interaction and Gli3 may be novel targets for effective control of BrCa growth.
- Published
- 2020
25. Decision letter: MEIS-mediated suppression of human prostate cancer growth and metastasis through HOXB13-dependent regulation of proteoglycans
- Author
-
Ralph Buttyan and Wilbert Zwart
- Subjects
business.industry ,Cancer research ,medicine ,Cancer ,medicine.disease ,business ,Human prostate ,Metastasis - Published
- 2020
26. A molecular portrait of epithelial–mesenchymal plasticity in prostate cancer associated with clinical outcome
- Author
-
Ralph Buttyan, Brett G. Hollier, Jennifer H. Gunter, Martin C. Sadowski, Chenwei Wang, Abhishek S. Kashyap, Martin E. Gleave, Colleen C. Nelson, Elizabeth D. Williams, Nataly Stylianou, Anja Rockstroh, Atefeh Taherian Fard, Micheal S. Ward, Melanie Lehman, Lidija Jovanovic, Thomas F. Westbrook, and Ladan Fazli
- Subjects
0301 basic medicine ,Cancer Research ,Mesenchymal stem cell ,Cancer ,Biology ,Gene signature ,medicine.disease ,Phenotype ,3. Good health ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Cancer cell ,Genetics ,Cancer research ,medicine ,Carcinoma ,Epithelial–mesenchymal transition ,Molecular Biology - Abstract
The propensity of cancer cells to transition between epithelial and mesenchymal phenotypic states via the epithelial–mesenchymal transition (EMT) program can regulate metastatic processes, cancer progression, and treatment resistance. Transcriptional investigations using reversible models of EMT, revealed the mesenchymal-to-epithelial reverting transition (MErT) to be enriched in clinical samples of metastatic castrate resistant prostate cancer (mCRPC). From this enrichment, a metastasis-derived gene signature was identified that predicted more rapid cancer relapse and reduced survival across multiple human carcinoma types. Additionally, the transcriptional profile of MErT is not a simple mirror image of EMT as tumour cells retain a transcriptional “memory” following a reversible EMT. This memory was also enriched in mCRPC samples. Cumulatively, our studies reveal the transcriptional profile of epithelial–mesenchymal plasticity and highlight the unique transcriptional properties of MErT. Furthermore, our findings provide evidence to support the association of epithelial plasticity with poor clinical outcomes in multiple human carcinoma types.
- Published
- 2018
27. Non-canonical activation of hedgehog in prostate cancer cells mediated by the interaction of transcriptionally active androgen receptor proteins with Gli3
- Author
-
Na Li, Nader Al Nakouzi, Amy A. Lubik, Mannan Nouri, Jackson Moore, Ralph Buttyan, and Sarah Truong
- Subjects
Male ,Transcriptional Activation ,0301 basic medicine ,Cancer Research ,animal structures ,Repressor ,Nerve Tissue Proteins ,Article ,03 medical and health sciences ,0302 clinical medicine ,Zinc Finger Protein Gli3 ,Cell Line, Tumor ,GLI2 ,Genetics ,Humans ,Hedgehog Proteins ,Promoter Regions, Genetic ,Receptor ,Molecular Biology ,Transcription factor ,biology ,HEK 293 cells ,Prostatic Neoplasms ,Cell biology ,Ubiquitin ligase ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,HEK293 Cells ,030104 developmental biology ,Receptors, Androgen ,030220 oncology & carcinogenesis ,biology.protein ,Smoothened ,Protein Binding ,Signal Transduction ,Transcription Factors - Abstract
Hedgehog (Hh) is an oncogenic signaling pathway that regulates the activity of Gli transcription factors. Canonical Hh is a Smoothened- (Smo-) driven process that alters the post-translational processing of Gli2/Gli3 proteins. Though evidence supports a role for Gli action in prostate cancer (PCa) cell growth and progression, there is little indication that Smo is involved. Here we describe a non-canonical means for activation of Gli transcription in PCa cells mediated by the binding of transcriptionally-active androgen receptors (ARs) to Gli3. Androgens stimulated reporter expression from a Gli-dependent promoter in a variety of AR + PCa cells and this activity was suppressed by an anti-androgen, Enz, or by AR knockdown. Androgens also upregulated expression of endogenous Gli-dependent genes. This activity was associated with increased intranuclear binding of Gli3 to AR that was antagonized by Enz. Fine mapping of the AR binding domain on Gli2 showed that AR recognizes the Gli protein processing domain (PPD) in the C-terminus. Mutations in the arginine-/serine repeat elements of the Gli2 PPD involved in phosphorylation and ubiquitinylation blocked the binding to AR. β-TrCP, a ubiquitin ligase that recognizes the Gli PPD, competed with AR for binding to this site. AR binding to Gli3 suppressed its proteolytic processing to the Gli3 repressor form (Gli3R) whereas AR knockdown increased Gli3R. Both full-length and truncated ARs were able to activate Gli transcription. Finally, we found that an ARbinding decoy polypeptide derived from the Gli2 C-terminus can compete with Gli3 for binding to AR. Exogenous overexpression of this decoy suppressed Gli transcriptional activity in PCa cells. Collectively, this work identifies a novel pathway for non-canonical activation of Hh signaling in PCa cells and identifies a means for interference that may have clinical relevance for PCa patients.
- Published
- 2018
28. Implications of PI3K/AKT inhibition on REST protein stability and neuroendocrine phenotype acquisition in prostate cancer cells
- Author
-
Ralph Buttyan, Xuesen Dong, Ruiqui Chen, and Yinan Li
- Subjects
0301 basic medicine ,medicine.medical_specialty ,PI3K/AKT inhibition ,neuroendocrine prostate cancer ,Gene signature ,Biology ,Protein degradation ,medicine.disease ,neuroendocrine differentiation ,Neuroendocrine differentiation ,3. Good health ,Androgen receptor ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,Endocrinology ,Oncology ,Internal medicine ,LNCaP ,Cancer research ,medicine ,REST degradation ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Research Paper - Abstract
// Ruiqui Chen 1 , Yinan Li 1 , Ralph Buttyan 1 and Xuesen Dong 1 1 Vancouver Prostate Center, Department of Urologic Sciences, The University of British Columbia, Vancouver, Canada Correspondence to: Xuesen Dong, email: // Keywords : PI3K/AKT inhibition, REST degradation, neuroendocrine differentiation, neuroendocrine prostate cancer Received : July 02, 2017 Accepted : July 06, 2017 Published : July 19, 2017 Abstract Treatment-induced neuroendocrine prostate cancer (t-NEPC) is an aggressive subtype of prostate cancer (PCa) that arises as a consequence of rigorous androgen receptor (AR) pathway inhibition (ARPI) therapies. While the PI3K/AKT pathway has been investigated as a co-therapeutic target with ARPI for advanced PCa, whether this strategy can prevent tumor progression to t-NEPC remains unknown. Here, we report that PI3K/AKT inhibition alone reduces RE-1 silencing transcription factor (REST) protein expression and induces multiple NE markers in PCa cells. The loss of REST by PI3K/AKT inhibition is through protein degradation mediated by the E3-ubiquitin ligase β-TRCP and REST phosphorylations at the S1024, S1027, and S1030 sites. Since AR inhibition can also deplete REST, the combinational inhibition of PI3K/AKT and AR further aggravated REST protein reduction. We profiled the transcriptomes of AKT and AR inhibitions in the LNCaP cells. The Gene Set Enrichment Analysis (GSEA) showed that these transcriptomes are highly correlated with the REST-regulated gene signature. Co-targeting AKT and AR resulted in a higher correlation comparing to those of single treatment. Comparing these transcriptomes to the t-NEPC gene signature in patients by GSEA, we observed that adding AKT inhibition to AR blockade enhanced the expression of neurogenesis-related genes and resulted in a stronger and broader upregulation of REST-regulated genes specific to t-NEPC. These results indicate that AKT pathway inhibition can induce neuroendocrine differentiation of PCa cells via REST protein degradation. It delineates a potential risk for the AR and PI3K/AKT co-targeting strategy as it may further facilitate t-NEPC development.
- Published
- 2017
29. Neuropilin-1 is upregulated in the adaptive response of prostate tumors to androgen-targeted therapies and is prognostic of metastatic progression and patient mortality
- Author
-
Elizabeth D. Williams, K McGowan, Miriam S. Butler, Jennifer H. Gunter, Ellca Ratther, Pamela J. Russell, N. Erho, Mohammed Alshalafa, Melanie Lehman, Mannan Nouri, Edward M. Schaeffer, Ladan Fazli, Robert Jeffrey Karnes, P S Rennie, Ashley E. Ross, Brett G. Hollier, Stephen McPherson, Nataly Stylianou, Rajdeep Das, Ralph Buttyan, Philip A. Gregory, Jacqui A. McGovern, Josselin Caradec, Luke A. Selth, E. Davicioni, Brian W.C. Tse, Marianna Volpert, Robert B. Jenkins, Robert B. Den, Martin E. Gleave, Colleen C. Nelson, Mani Roshan-Moniri, M. Takhar, Cheryl Y. Gregory-Evans, Tse, BWC, Volpert, M, Ratther, E, Stylianou, N, Gregory, PA, and Hollier, B. G.
- Subjects
Male ,0301 basic medicine ,Biochemical recurrence ,PCA3 ,Cancer Research ,Epithelial-Mesenchymal Transition ,medicine.medical_treatment ,Biology ,androgen-targeted ,Metastasis ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Neoplasm Metastasis ,Molecular Biology ,Tissue microarray ,Prostatic Neoplasms ,metastatic progression ,Cancer ,Androgen Antagonists ,medicine.disease ,Survival Analysis ,prostate tumors ,Neuropilin-1 ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Radiation therapy ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Immunology ,Disease Progression ,Cancer research ,Original Article ,Neoplasm Grading ,patient mortality - Abstract
Recent evidence has implicated the transmembrane co-receptor neuropilin-1 (NRP1) in cancer progression. Primarily known as a regulator of neuronal guidance and angiogenesis, NRP1 is also expressed in multiple human malignancies, where it promotes tumor angiogenesis. However, non-angiogenic roles of NRP1 in tumor progression remain poorly characterized. In this study, we define NRP1 as an androgen-repressed gene whose expression is elevated during the adaptation of prostate tumors to androgen-targeted therapies (ATTs), and subsequent progression to metastatic castration-resistant prostate cancer (mCRPC). Using short hairpin RNA (shRNA)-mediated suppression of NRP1, we demonstrate that NRP1 regulates the mesenchymal phenotype of mCRPC cell models and the invasive and metastatic dissemination of tumor cells in vivo. In patients, immunohistochemical staining of tissue microarrays and mRNA expression analyses revealed a positive association between NRP1 expression and increasing Gleason grade, pathological T score, positive lymph node status and primary therapy failure. Furthermore, multivariate analysis of several large clinical prostate cancer (PCa) cohorts identified NRP1 expression at radical prostatectomy as an independent prognostic biomarker of biochemical recurrence after radiation therapy, metastasis and cancer-specific mortality. This study identifies NRP1 for the first time as a novel androgen-suppressed gene upregulated during the adaptive response of prostate tumors to ATTs and a prognostic biomarker of clinical metastasis and lethal PCa. Refereed/Peer-reviewed
- Published
- 2017
30. Transient Sox9 Expression Facilitates Resistance to Androgen-Targeted Therapy in Prostate Cancer
- Author
-
Colin Collins, Josselin Caradec, Ladan Fazli, Jackson Moore, Mike Wang, Varune Rohan Ramnarine, Brett G. Hollier, Na Li, Sarah Truong, Amy A. Lubik, Jessica M. Lovnicki, Jane Foo, Ahn R. Lee, Mannan Nouri, Ralph Buttyan, Martin E. Gleave, Colleen C. Nelson, Shabnam Massah, and Xuesen Dong
- Subjects
0301 basic medicine ,Biochemical recurrence ,Male ,Cancer Research ,medicine.medical_treatment ,Retinoblastoma Protein ,Targeted therapy ,03 medical and health sciences ,Prostate cancer ,Mice ,0302 clinical medicine ,stomatognathic system ,SOX2 ,Neuroendocrine Cells ,Cell Line, Tumor ,medicine ,Androgen Receptor Antagonists ,Animals ,Humans ,Tissue microarray ,business.industry ,SOXB1 Transcription Factors ,Stem Cells ,NF-kappa B ,Cancer ,SOX9 Transcription Factor ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,Androgen receptor ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,Oncology ,Tumor progression ,Drug Resistance, Neoplasm ,Receptors, Androgen ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer research ,Tumor Suppressor Protein p53 ,business ,Signal Transduction - Abstract
Purpose: Patients with metastatic prostate cancer are increasingly presenting with treatment-resistant, androgen receptor–negative/low (AR−/Low) tumors, with or without neuroendocrine characteristics, in processes attributed to tumor cell plasticity. This plasticity has been modeled by Rb1/p53 knockdown/knockout and is accompanied by overexpression of the pluripotency factor, Sox2. Here, we explore the role of the developmental transcription factor Sox9 in the process of prostate cancer therapy response and tumor progression. Experimental Design: Unique prostate cancer cell models that capture AR−/Low stem cell–like intermediates were analyzed for features of plasticity and the functional role of Sox9. Human prostate cancer xenografts and tissue microarrays were evaluated for temporal alterations in Sox9 expression. The role of NF-κB pathway activity in Sox9 overexpression was explored. Results: Prostate cancer stem cell–like intermediates have reduced Rb1 and p53 protein expression and overexpress Sox2 as well as Sox9. Sox9 was required for spheroid growth, and overexpression increased invasiveness and neural features of prostate cancer cells. Sox9 was transiently upregulated in castration-induced progression of prostate cancer xenografts and was specifically overexpressed in neoadjuvant hormone therapy (NHT)–treated patient tumors. High Sox9 expression in NHT-treated patients predicts biochemical recurrence. Finally, we link Sox9 induction to NF-κB dimer activation in prostate cancer cells. Conclusions: Developmentally reprogrammed prostate cancer cell models recapitulate features of clinically advanced prostate tumors, including downregulated Rb1/p53 and overexpression of Sox2 with Sox9. Sox9 is a marker of a transitional state that identifies prostate cancer cells under the stress of therapeutic assault and facilitates progression to therapy resistance. Its expression may index the relative activity of the NF-κB pathway.
- Published
- 2019
31. Multimodal biomarkers overcome sampling bias to predict presence of aggressive localized prostate cancer
- Author
-
David M. Berman, Jacques Lapointe, Anna Yw Lee, Tamara Jamaspishvili, Dan Dion, Laura A. Lee, Simone Chevalier, Nadia Boufaied, Vasundara Venkateswaran, Paul C. Boutros, Palak G. Patel, Paul C. Park, Karl-Philippe Guérard, Axel A. Thomson, Walead Ebrahimizadeh, John M. S. Bartlett, Jane Bayani, Robert Lesurf, Fadi Brimo, and Ralph Buttyan
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Disease ,medicine.disease ,Tumor grade ,Prostate cancer ,medicine.anatomical_structure ,Prostate ,Internal medicine ,medicine ,business ,Sampling bias - Abstract
209 Background: Histopathologic investigation of diagnostic prostate biopsies both confirms the presence of disease and estimates its potential for distal spread via tumour grade. The accuracy of biopsy grading is limited by intra-tumoral heterogeneity, inter-observer variability, and other factors. To improve risk stratification at the time of diagnosis, we sought to create objective molecular biomarkers of radical prostatectomy grade that are resistant to sampling error and should be useful when applied to biopsy tissue. Methods: We developed and validated a robust objective biomarker of prostate cancer grade using pathologic grading of prostatectomy tissues as the gold standard. We created training (333 patients) and validation (202 patients) cohorts of Cancer of the Prostate Risk Assessment (CAPRA) low- and intermediate-risk prostate cancer patients. To address intra-tumoral heterogeneity, each tumor was sampled at two locations. We profiled the abundance of 342 mRNAs complemented by 100 canonical DNA copy number aberration loci (CNAs) and 14 hypermethylation events. Using the training cohort with cross-validation, we evaluated models for training classifiers of pathologic Grade Group ≥2, Restricting to strategies resulting in true negative rates ≥0.5, true positive (TP) rates ≥0.8, we selected two strategies to train classifiers, PRONTO-e and PRONTO-m. Results: The PRONTO-e classifier comprises 353 mRNA and CNA features, while the PRONTO-m classifier comprises 94 mRNA, CNA, methylation and clinical features. Both classifiers (PRONTO-e, PRONTO-m) validated in the independent cohort, with respective TP rates of 0.809 and 0.760, false positive rates of 0.429 and 0.262, F1 scores of 0.709 and 0.724, and AUCs of 0.792 and 0.818. Conclusions: Two classifiers were developed and validated in separate cohorts, each achieved excellent performance by integrating different types of molecular data. Implementation of classifiers with these performance characteristics could markedly improve current active surveillance approaches without increasing patient morbidity and may help better inform patients on their individual need for definitive therapy versus active surveillance.
- Published
- 2021
32. Paracrine sonic hedgehog signaling contributes significantly to acquired steroidogenesis in the prostate tumor microenvironment
- Author
-
Mazyar Ghaffari, Mannan Nouri, Emma S. Tomlinson Guns, Steven Pham, Parvin Yenki, Ralph Buttyan, Na Li, Sarah Truong, Eva Corey, Hans Adomat, Amy A. Lubik, and Michael E. Cox
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Tumor microenvironment ,Stromal cell ,medicine.drug_class ,Biology ,urologic and male genital diseases ,Androgen ,medicine.disease ,Hedgehog signaling pathway ,3. Good health ,Androgen deprivation therapy ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Oncology ,030220 oncology & carcinogenesis ,Internal medicine ,LNCaP ,medicine ,Smoothened - Abstract
Despite the substantial benefit of androgen deprivation therapy (ADT) for metastatic prostate cancer, patients often progress to castration-resistant disease (CRPC) that is more difficult to treat. CRPC is associated with renewed androgen receptor activity in tumor cells and restoration of tumor androgen levels through acquired intratumoral steroidogenesis (AIS). Although prostate cancer (PCa) cells have been shown to have steroidogenic capability in vitro, we previously found that benign prostate stromal cells (PrSCs) can also synthesize testosterone (T) from an adrenal precursor, DHEA, when stimulated with a hedgehog (Hh) pathway agonist, SAG. Here, we show exposure of PrSCs to a different Smoothened (Smo) agonist, Ag1.5, or to conditioned medium from sonic hedgehog overexpressing LNCaP cells induces steroidogenic enzyme expression in PrSCs and significantly increases production of T and its precursor steroids in a Smo-dependent manner from 22-OH-cholesterol substrate. Hh agonist-/ligand-treated PrSCs produced androgens at a rate similar to or greater than that of PCa cell lines. Likewise, primary bone marrow stromal cells became more steroidogenic and produced T under the influence of Smo agonist. Treatment of mice bearing LNCaP xenografts with a Smo antagonist, TAK-441, delayed the onset of CRPC after castration and substantially reduced androgen levels in residual tumors. These outcomes support the idea that stromal cells in ADT-treated primary or metastatic prostate tumors can contribute to AIS as a consequence of a paracrine Hh signaling microenvironment. As such, Smo antagonists may be useful for targeting prostate tumor stromal cell-derived AIS and delaying the onset of CRPC after ADT.
- Published
- 2016
33. Not all NOTCH Is Created Equal: The Oncogenic Role of NOTCH2 in Bladder Cancer and Its Implications for Targeted Therapy
- Author
-
Na Li, Ladan Fazli, Manuel Altamirano-Dimas, Ralph Buttyan, Akio Matsubara, Akihiro Goriki, Kilian M. Gust, Tetsutaro Hayashi, Alexander W. Wyatt, Wolfgang Jäger, Peter C. Black, Shannon Awrey, and Htoo Zarni Oo
- Subjects
0301 basic medicine ,Oncology ,endocrine system ,Cancer Research ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,endocrine system diseases ,medicine.medical_treatment ,Gene Dosage ,Notch signaling pathway ,Biology ,Metastasis ,Targeted therapy ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Receptor, Notch2 ,Epithelial–mesenchymal transition ,RNA, Small Interfering ,Receptor, Notch1 ,Receptor, Notch3 ,Cell Proliferation ,Bladder cancer ,Oncogene ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Enzyme Activation ,030104 developmental biology ,Urinary Bladder Neoplasms ,Tumor progression ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,RNA Interference ,Signal Transduction - Abstract
Purpose: Recent molecular analyses of bladder cancer open the door to significant advances in targeted therapies. NOTCH has been identified as a tumor suppressor in bladder cancer, but prior reports have focused on NOTCH1. Here we hypothesized that NOTCH2 is an oncogene suitable for therapeutic targeting in bladder cancer. Experimental design: We studied genomic aberrations of NOTCH, compared survival and tumor progression according to NOTCH2 expression levels, and studied NOTCH2 function in vitro and vivo. Results: We report a high rate of NOTCH2 copy number gain in bladder cancer. High NOTCH2 expression was identified especially in the basal subtype and in mesenchymal tumors. NOTCH2 activation correlated with adverse disease parameters and worse prognosis by immunohistochemistry. Forced overexpression of the intracellular domain of NOTCH2 (N2ICD) induced cell growth and invasion by cell-cycle progression, maintenance of stemness and epithelial-to-mesenchymal transition (EMT). These effects were abrogated by silencing of CSL, indicating that the effects were mediated through the canonical NOTCH signaling pathway. In an orthotopic xenograft model, forced overexpression of N2ICD increased growth, invasion, and metastasis. To explore the potential for therapeutic targeting of NOTCH2, we first silenced the receptor with shRNA and subsequently treated with a specific inhibitory antibody. Both interventions decreased cell growth, invasion, and metastasis in vitro and in the orthotopic xenograft model. Conclusions: We have demonstrated that NOTCH2 acts as an oncogene that promotes bladder cancer growth and metastasis through EMT, cell-cycle progression, and maintenance of stemness. Inhibition of NOTCH2 is a rational novel treatment strategy for invasive bladder cancer. Clin Cancer Res; 22(12); 2981–92. ©2016 AACR.
- Published
- 2016
34. Inhibition of GLI2 with antisense-oligonucleotides: A potential therapy for the treatment of bladder cancer
- Author
-
Ralph Buttyan, Peter A. Raven, Summer Lysakowski, Shintaro Narita, Zheng Tan, Ninadh M. D'Costa, Claudia Chavez-Munoz, Werner J. Struss, Alan I. So, Sebastian Frees, Igor Moskalev, and Yoshiyuki Matsui
- Subjects
0301 basic medicine ,animal structures ,Cyclopamine ,Physiology ,Cell Survival ,medicine.medical_treatment ,Clinical Biochemistry ,Antineoplastic Agents ,Zinc Finger Protein Gli2 ,Targeted therapy ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,GLI1 ,GLI2 ,Cell Line, Tumor ,medicine ,Humans ,Sonic hedgehog ,skin and connective tissue diseases ,Transcription factor ,biology ,Chemistry ,Cell Cycle ,Nuclear Proteins ,Cell Biology ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Urinary Bladder Neoplasms ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Signal transduction ,Smoothened - Abstract
The sonic hedgehog (SHH) signaling pathway plays an integral role in the maintenance and progression of bladder cancer (BCa) and SHH inhibition may be an efficacious strategy for BCa treatment. We assessed an in-house human BCa tissue microarray and found that the SHH transcription factors, GLI1 and GLI2, were increased in disease progression. A panel of BCa cell lines show that two invasive lines, UM-UC-3 and 253J-BV, both express these transcription factors but UM-UC-3 produces more SHH ligand and is less responsive in viability to pathway stimulation by recombinant human SHH or smoothened agonist, and less responsive to inhibitors including the smoothened inhibitors cyclopamine and SANT-1. In contrast, 253J-BV was highly responsive to these manipulations. We utilized a GLI1 and GLI2 antisense oligonucleotide (ASO) to bypass pathway mechanics and target the transcription factors directly. UM-UC-3 decreased in viability due to both ASOs but 253J-BV was only affected by GLI2 ASO. We utilized the murine intravesical orthotopic human BCa (mio-hBC) model for the establishment of noninvasive BCa and treated tumors with GLI2 ASO. Tumor size, growth rate, and GLI2 messenger RNA and protein expression were decreased. These results suggest that GLI2 ASO may be a promising new targeted therapy for BCa.
- Published
- 2018
35. Hedgehog in prostate cancer explained
- Author
-
Na Li, Shabnam Massah, and Ralph Buttyan
- Subjects
Cancer Research ,business.industry ,medicine.disease ,prostate cancer ,Steroid Receptors ,Androgen receptor ,Prostate cancer ,Editorial ,Oncology ,Cancer research ,Medicine ,business ,Hedgehog ,Androgen Receptors ,Gli - Published
- 2018
36. Semaphorin 3 C drives epithelial-to-mesenchymal transition, invasiveness, and stem-like characteristics in prostate cells
- Author
-
Kevin J. Tam, Ralph Buttyan, James W. Peacock, Daniel H. F. Hui, Shahram Khosravi, Ario Takeuchi, Mingshu Dong, Ivy Z. F. Jiao, Michael E. Cox, Wilson W. Lee, Christopher J. Ong, Tabitha Tombe, Martin E. Gleave, Larissa Ivanova, and Igor Moskalev
- Subjects
0301 basic medicine ,Male ,Epithelial-Mesenchymal Transition ,Cell ,lcsh:Medicine ,Gene Expression ,Context (language use) ,Semaphorins ,Biology ,Bioinformatics ,Article ,Immunophenotyping ,03 medical and health sciences ,Prostate cancer ,Mice ,Semaphorin ,Cell Movement ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,lcsh:Science ,Cell Proliferation ,Multidisciplinary ,Cell growth ,lcsh:R ,Prostatic Neoplasms ,medicine.disease ,3. Good health ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,Neoplastic Stem Cells ,Heterografts ,Ectopic expression ,lcsh:Q ,Biomarkers - Abstract
Prostate cancer (PCa) is among the most commonly-occurring cancers worldwide and a leader in cancer-related deaths. Local non-invasive PCa is highly treatable but limited treatment options exist for those with locally-advanced and metastatic forms of the disease underscoring the need to identify mechanisms mediating PCa progression. The semaphorins are a large grouping of membrane-associated or secreted signalling proteins whose normal roles reside in embryogenesis and neuronal development. In this context, semaphorins help establish chemotactic gradients and direct cell movement. Various semaphorin family members have been found to be up- and down-regulated in a number of cancers. One family member, Semaphorin 3 C (SEMA3C), has been implicated in prostate, breast, ovarian, gastric, lung, and pancreatic cancer as well as glioblastoma. Given SEMA3C’s roles in development and its augmented expression in PCa, we hypothesized that SEMA3C promotes epithelial-to-mesenchymal transition (EMT) and stem-like phenotypes in prostate cells. In the present study we show that ectopic expression of SEMA3C in RWPE-1 promotes the upregulation of EMT and stem markers, heightened sphere-formation, and cell plasticity. In addition, we show that SEMA3C promotes migration and invasion in vitro and cell dissemination in vivo.
- Published
- 2017
37. Directed Developmental Reprogramming of Prostate Cancer Cells to Stem-Like Cells
- Author
-
Mannan Nouri, Josselin Caradec, Sarah Truong, Bronwen Dekker, Na Li, Ralph Buttyan, and Amy A. Lubik
- Subjects
Oncology ,medicine.medical_specialty ,Prostate cancer ,business.industry ,Cancer stem cell ,Internal medicine ,General Earth and Planetary Sciences ,Medicine ,business ,medicine.disease ,Reprogramming ,General Environmental Science - Published
- 2017
38. PD33-01 NON-CANONICAL ACTIVATION OF HEDGEHOG SIGNALING IN PROSTATE CANCER CELLS MEDIATED BY THE BINDING OF TRANSCRIPTIONALLY ACTIVE ANDROGEN RECEPTORS TO GLI TRANSCRIPTION FACTORS
- Author
-
Sarah Truong, Mannan Nouri, Ralph Buttyan, Amy A. Lubik, and Na Li
- Subjects
Androgen receptor ,Prostate cancer ,Non canonical ,business.industry ,Urology ,Cancer research ,Medicine ,business ,medicine.disease ,Transcription factor ,Hedgehog signaling pathway - Published
- 2017
39. Determinants of Gli2 co-activation of wildtype and naturally truncated androgen receptors
- Author
-
Chunhong Yan, Ralph Buttyan, Mengqian Chen, Sarah Truong, and Na Li
- Subjects
Androgen receptor ,Transactivation ,animal structures ,Oncology ,Transcription (biology) ,Urology ,LNCaP ,Biology ,Chromatin immunoprecipitation ,Molecular biology ,Transcription factor ,Hedgehog signaling pathway ,Chromatin - Abstract
BACKGROUND Gli2, a transcription factor in the Hedgehog pathway, is overexpressed in castrate-resistant prostate cancer (PCa). Previously we showed that Gli2 overexpression increased transcriptional activity of androgen receptor (AR) and conferred androgen growth-independence to normally growth-dependent PCa cells. Here we localized the regions of AR-Gli2 protein interaction and determined the domains within Gli2 needed for AR co-activation. METHODS Co-immunoprecipitation and GST-pulldown assays were used to define AR-Gli binding domains. Co-activation assays using androgen-responsive promoter reporters were used to define Gli2 regions needed for AR co-activation. Chromatin immunoprecipitation (ChIP) assays were used to confirm nuclear interactions of Gli2 with AR in PCa cells. RESULTS The Gli2 C-terminal domain (CTD) is sufficient for AR co-activation. Two elements within the CTD were required: (1) an AR binding domain within aa628-897; and (2) at least part of the Gli2 transactivation domain within aa1252-1586. In turn, Gli2 binds the tau5/AF5 ligand-independent activation domain in the AR N-terminus. Mutations in the WxxLF motif in tau5/AF5 greatly diminished binding to Gli2-CTD. Gli2 interaction with AR tau5/AF5 was further substantiated by the ability of Gli2/Gli2-CTD to co-activate truncated AR splice variants (AR-V7/ARV567es). ChIP assays confirmed that Gli2 associates with chromatin at androgen response elements found near androgen-responsive genes in LNCaP cells. These assays also showed that AR associates with chromatin containing a Gli-response element near a Gli-responsive gene. CONCLUSION Our findings indicate that Gli2 overexpression in PCa cells might support development of castration resistant PCa through AR co-activation and suggests that AR might modulate transcription from Gli2. Prostate 74:1400–1410, 2014. © 2014 Wiley Periodicals, Inc.
- Published
- 2014
40. FOXA1 deletion in luminal epithelium causes prostatic hyperplasia and alteration of differentiated phenotype
- Author
-
Ralph Buttyan, William J. Hayward, Michael A. Walter, Klaus H. Kaestner, David J. DeGraff, Simon W. Hayward, Nan Gao, Douglas W. Strand, Magdalena M. Grabowska, Mary K. Herrick, Robert J. Matusik, Tom Case, Justin M M Cates, Xiuping Yu, and Yajun Yi
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Male ,Cellular differentiation ,Prostatic Hyperplasia ,Morphogenesis ,Mice, Transgenic ,Biology ,FOX proteins ,Cell morphology ,Article ,Epithelium ,Pathology and Forensic Medicine ,Mice ,Prostate cancer ,medicine ,Animals ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Integrases ,Reverse Transcriptase Polymerase Chain Reaction ,Prostate ,Seminal Vesicles ,Cell Differentiation ,Cell Biology ,Hyperplasia ,medicine.disease ,Immunohistochemistry ,Molecular biology ,medicine.anatomical_structure ,Microscopy, Fluorescence ,FOXA2 ,Transcriptome - Abstract
The forkhead box (Fox) superfamily of transcription factors has essential roles in organogenesis and tissue differentiation. Foxa1 and Foxa2 are expressed during prostate budding and ductal morphogenesis, whereas Foxa1 expression is retained in adult prostate epithelium. Previous characterization of prostatic tissue rescued from embryonic Foxa1 knockout mice revealed Foxa1 to be essential for ductal morphogenesis and epithelial maturation. However, it is unknown whether Foxa1 is required to maintain the differentiated status in adult prostate epithelium. Here, we employed the PBCre4 transgenic system and determined the impact of prostate-specific Foxa1 deletion in adult murine epithelium. PBCre4/Foxa1(loxp/loxp) mouse prostates showed progressive florid hyperplasia with extensive cribriform patterning, with the anterior prostate being most affected. Immunohistochemistry studies show mosaic Foxa1 KO consistent with PBCre4 activity, with Foxa1 KO epithelial cells specifically exhibiting altered cell morphology, increased proliferation, and elevated expression of basal cell markers. Castration studies showed that, while PBCre4/Foxa1(loxp/loxp) prostates did not exhibit altered sensitivity in response to hormone ablation compared with control prostates, the number of Foxa1-positive cells in mosaic Foxa1 KO prostates was significantly reduced compared with Foxa1-negative cells following castration. Unexpectedly, gene expression profile analyses revealed that Foxa1 deletion caused abnormal expression of seminal vesicle-associated genes in KO prostates. In summary, these results indicate Foxa1 expression is required for the maintenance of prostatic cellular differentiation.
- Published
- 2014
41. Abstract 4396: Gli, not Androgen Receptor, is the primary driver of prostate cell growth
- Author
-
Jane Foo, Na Li, Shabnam Massah, Mannan Nouri, Mike Wang, and Ralph Buttyan
- Subjects
Cancer Research ,Oncology - Abstract
Introduction Hedgehog (Hh) signaling regulates the activity of Gli transcription factors. Gli controls genes needed for development, cell growth and cell motility. Canonical Hh signaling through Smoothened [Smo] suppresses the proteolysis of Gli2 and Gli3, maintaining them in high molecular weight active forms. Previously, we showed that active androgen receptors ([AR] liganded AR full length and AR-V7) recognize and binds Gli3 its Protein Processing Domain. This binding overrides the need for Smo action and provides a means for stabilization and activation of Gli3 in prostate cancer (PCa) cells without Hh. Studies on the effects of AR binding to Gli2 are more challenging because it is expressed at much lower levels than Gli3. Here, we report that AR also affects Gli2 protein stability and show that Gli2, like Gli3, is a driver of PCa cell growth that is overexpressed in castration resistant disease. Method Immunohistochemistry (IHC) using Gli2- and Gli3-specific antibodies was used to assess expression of Gli2 and Gli3 in human PCa tumor microarrays. Western blot were used to identify and quantify levels of active Gli2 and Gli3 in androgen-treated PCa cells (LNCaP, LNCaP-AI, LAPC4). Proximity ligation assays (PLA) was used to visualize in situ and quantify AR-Gli2 complexes in PCa cells. Gli2- and Gli3-specific siRNAs were used to knock down Gli expression in AR+ (LNCaP-AI, LAPC4, 22Rv1) and AR- (PC3) PCa, cells followed by the Gli reporter assay and the cell growth assay. Results IHC outcomes showed that nuclear Gli2 and Gli3 protein expression was significantly higher in castration resistant tumors compared to primary disease. Androgens increased expression of active Gli2 and Gli3 protein expression in PCa cells and stimulated Gli reporter activity. PLA showed the presence of intranuclear complexes of Gli2/Gli3-AR-Full-length in androgen-treated LNCaP and Gli2-AR-V7 complexes in CWR22rv1 cells. Knockdown of Gli2 or Gli3 present significant reduction in Gli transcriptional activity as well as inhibition on growth of both AR+ and AR- PCa cells that were tested. Conclusion AR activity affects Gli2 processing, as it does for Gli3. We can visualize and quantify AR-Gli2/Gli3 complexes in situ in PCa cells. Gli2 suppression, as well as Gli3 suppression, has a profound effect on PCa cell growth but combined suppression has the strongest effects in certain PCa cells. Finally, Gli2 and Gli3 protein expression is elevated in castration resistant disease. Citation Format: Jane Foo, Na Li, Shabnam Massah, Mannan Nouri, Mike Wang, Ralph Buttyan. Gli, not Androgen Receptor, is the primary driver of prostate cell growth [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4396.
- Published
- 2019
42. Correction: A molecular portrait of epithelial–mesenchymal plasticity in prostate cancer associated with clinical outcome
- Author
-
Micheal S. Ward, Chenwei Wang, Atefeh Taherian Fard, Thomas F. Westbrook, Anja Rockstroh, Martin E. Gleave, Nataly Stylianou, Colleen C. Nelson, Brett G. Hollier, Lidija Jovanovic, Ralph Buttyan, Abhishek S. Kashyap, Jennifer H. Gunter, Ladan Fazli, Elizabeth D. Williams, Martin C. Sadowski, and Melanie Lehman
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Epithelial mesenchymal plasticity ,Biology ,Outcome (game theory) ,Article ,Disease-Free Survival ,Metastasis ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Portrait ,Cell Line, Tumor ,Internal medicine ,Genetics ,medicine ,Humans ,Neoplasm Metastasis ,Molecular Biology ,Correction ,medicine.disease ,Survival Rate ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,030220 oncology & carcinogenesis - Abstract
The propensity of cancer cells to transition between epithelial and mesenchymal phenotypic states via the epithelial–mesenchymal transition (EMT) program can regulate metastatic processes, cancer progression, and treatment resistance. Transcriptional investigations using reversible models of EMT, revealed the mesenchymal-to-epithelial reverting transition (MErT) to be enriched in clinical samples of metastatic castrate resistant prostate cancer (mCRPC). From this enrichment, a metastasis-derived gene signature was identified that predicted more rapid cancer relapse and reduced survival across multiple human carcinoma types. Additionally, the transcriptional profile of MErT is not a simple mirror image of EMT as tumour cells retain a transcriptional “memory” following a reversible EMT. This memory was also enriched in mCRPC samples. Cumulatively, our studies reveal the transcriptional profile of epithelial–mesenchymal plasticity and highlight the unique transcriptional properties of MErT. Furthermore, our findings provide evidence to support the association of epithelial plasticity with poor clinical outcomes in multiple human carcinoma types.
- Published
- 2018
43. Expression and Function of the Progesterone Receptor in Human Prostate Stroma Provide Novel Insights to Cell Proliferation Control
- Author
-
Ning Xie, Liangliang Liu, Yue Yu, Yuzhuo Wang, Xuesen Dong, Martin E. Gleave, Ladan Fazli, Ralph Buttyan, and Hui Xue
- Subjects
Male ,medicine.medical_specialty ,Cell type ,Stromal cell ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Estrogen receptor ,Cell Cycle Proteins ,Cyclin A ,Cyclin B ,Biology ,Biochemistry ,Endocrinology ,Prostate ,Internal medicine ,Progesterone receptor ,medicine ,Endocrine Research ,Humans ,Protein Isoforms ,cdc25 Phosphatases ,Receptor ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Cell growth ,Biochemistry (medical) ,Prostatic Neoplasms ,Epithelial Cells ,Muscle, Smooth ,Hyperplasia ,medicine.disease ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Gene Expression Regulation ,Stromal Cells ,Receptors, Progesterone ,Cell Division - Abstract
Like other tissues, the prostate is an admixture of many different cell types that can be segregated into components of the epithelium or stroma. Reciprocal interactions between these 2 types of cells are critical for maintaining prostate homeostasis, whereas aberrant stromal cell proliferation can disrupt this balance and result in diseases such as benign prostatic hyperplasia. Although the androgen and estrogen receptors are relatively well studied for their functions in controlling stromal cell proliferation and differentiation, the role of the progesterone receptor (PR) remains unclear.The aim of the study was to investigate the expression and function of the PR in the prostate.Human prostate biopsies, renal capsule xenografts, and prostate stromal cells were used. Immunohistochemistry, Western blotting, real-time quantitative PCR, cell proliferation, flow cytometry, and gene microarray analyses were performed.Two PR isoforms, PRA and PRB, are expressed in prostate stromal fibroblasts and smooth muscle cells, but not in epithelial cells. Both PR isoforms suppress prostate stromal cell proliferation through inhibition of the expression of cyclinA, cyclinB, and cdc25c, thus delaying cell cycling through S and M phases. Gene microarray analyses further demonstrated that PRA and PRB regulated different transcriptomes. However, one of the major gene groups commonly regulated by both PR isoforms was the one associated with regulation of cell proliferation.PR plays an inhibitory role in prostate stromal cell proliferation.
- Published
- 2013
44. Therapy-induced developmental reprogramming of prostate cancer cells and acquired therapy resistance
- Author
-
Mani Roshan-Moniri, Miriam S. Butler, Mandeep Takhar, Mannan Nouri, Dawn R. Cochrane, Josselin Caradec, Colin Collins, Brett G. Hollier, Elai Davicioni, Ralph Buttyan, Michael E. Cox, Sheryl Gregory-Evans, Martin E. Gleave, Mengqian Chen, Colleen C. Nelson, Amy A. Lubik, Nicholas Erho, Jennifer L. Bishop, R. Jeffrey Karnes, Eric A. Klein, Melanie Lehman, Na Li, Mohamed Alshalafa, Manuel Altimirano-Dimas, Sarah Truong, Shih Chieh Huang, and Robert B. Jenkins
- Subjects
0301 basic medicine ,Gerontology ,Male ,cancer stem cell ,Blotting, Western ,Fluorescent Antibody Technique ,Biology ,urologic and male genital diseases ,Real-Time Polymerase Chain Reaction ,Metastasis ,03 medical and health sciences ,Prostate cancer ,Mice ,Cancer stem cell ,medicine ,Animals ,Humans ,Zebrafish ,Oligonucleotide Array Sequence Analysis ,neuroendocrine transdifferentiation ,Transdifferentiation ,Neural crest ,Prostatic Neoplasms ,Gene signature ,medicine.disease ,Cellular Reprogramming ,Flow Cytometry ,prostate cancer ,3. Good health ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Cell Transdifferentiation ,Cancer research ,Disease Progression ,Neoplastic Stem Cells ,Heterografts ,Stem cell ,hormone resistance ,Reprogramming ,neural crest ,Research Paper - Abstract
Treatment-induced neuroendocrine transdifferentiation (NEtD) complicates therapies for metastatic prostate cancer (PCa). Based on evidence that PCa cells can transdifferentiate to other neuroectodermally-derived cell lineages in vitro, we proposed that NEtD requires first an intermediary reprogramming to metastable cancer stem-like cells (CSCs) of a neural class and we demonstrate that several different AR+/PSA+ PCa cell lines were efficiently reprogrammed to, maintained and propagated as CSCs by growth in androgen-free neural/neural crest (N/NC) stem medium. Such reprogrammed cells lost features of prostate differentiation; gained features of N/NC stem cells and tumor-initiating potential; were resistant to androgen signaling inhibition; and acquired an invasive phenotype in vitro and in vivo. When placed back into serum-containing mediums, reprogrammed cells could be re-differentiated to N-/NC-derived cell lineages or return back to an AR+ prostate-like state. Once returned, the AR+ cells were resistant to androgen signaling inhibition. Acute androgen deprivation or anti-androgen treatment in serum-containing medium led to the transient appearance of a sub-population of cells with similar characteristics. Finally, a 132 gene signature derived from reprogrammed PCa cell lines distinguished tumors from PCa patients with adverse outcomes. This model may explain neural manifestations of PCa associated with lethal disease. The metastable nature of the reprogrammed stem-like PCa cells suggests that cycles of PCa cell reprogramming followed by re-differentiation may support disease progression and therapeutic resistance. The ability of a gene signature from reprogrammed PCa cells to identify tumors from patients with metastasis or PCa-specific mortality implies that developmental reprogramming is linked to aggressive tumor behaviors.
- Published
- 2016
45. MP84-09 INTERFERENCE WITH GLI BINDING TO ANDROGEN RECEPTORS ALTERS THE AR TRANSCRIPTOME AND EFFECTIVELY INHIBITS ANDROGEN GROWTH-INDEPENDENCE OF PROSTATE CANCER CELLS
- Author
-
Josselin Caradec, Amy A. Lubik, Mannan Nouri, Na Li, Sarah Truong, and Ralph Buttyan
- Subjects
medicine.medical_specialty ,medicine.drug_class ,business.industry ,Urology ,media_common.quotation_subject ,medicine.disease ,Interference (genetic) ,Androgen ,Independence ,Transcriptome ,Androgen receptor ,Prostate cancer ,Endocrinology ,Internal medicine ,medicine ,Cancer research ,business ,media_common - Published
- 2016
46. Paracrine sonic hedgehog signaling contributes significantly to acquired steroidogenesis in the prostate tumor microenvironment
- Author
-
Amy A, Lubik, Mannan, Nouri, Sarah, Truong, Mazyar, Ghaffari, Hans H, Adomat, Eva, Corey, Michael E, Cox, Na, Li, Emma S, Guns, Parvin, Yenki, Steven, Pham, and Ralph, Buttyan
- Subjects
Male ,Mice, Nude ,Mice ,Prostatic Neoplasms, Castration-Resistant ,Bone Marrow ,Receptors, Androgen ,Cell Line, Tumor ,Paracrine Communication ,Androgens ,Tumor Microenvironment ,Animals ,Humans ,Hedgehog Proteins ,Testosterone ,Castration ,Stromal Cells ,Signal Transduction - Abstract
Despite the substantial benefit of androgen deprivation therapy (ADT) for metastatic prostate cancer, patients often progress to castration-resistant disease (CRPC) that is more difficult to treat. CRPC is associated with renewed androgen receptor activity in tumor cells and restoration of tumor androgen levels through acquired intratumoral steroidogenesis (AIS). Although prostate cancer (PCa) cells have been shown to have steroidogenic capability in vitro, we previously found that benign prostate stromal cells (PrSCs) can also synthesize testosterone (T) from an adrenal precursor, DHEA, when stimulated with a hedgehog (Hh) pathway agonist, SAG. Here, we show exposure of PrSCs to a different Smoothened (Smo) agonist, Ag1.5, or to conditioned medium from sonic hedgehog overexpressing LNCaP cells induces steroidogenic enzyme expression in PrSCs and significantly increases production of T and its precursor steroids in a Smo-dependent manner from 22-OH-cholesterol substrate. Hh agonist-/ligand-treated PrSCs produced androgens at a rate similar to or greater than that of PCa cell lines. Likewise, primary bone marrow stromal cells became more steroidogenic and produced T under the influence of Smo agonist. Treatment of mice bearing LNCaP xenografts with a Smo antagonist, TAK-441, delayed the onset of CRPC after castration and substantially reduced androgen levels in residual tumors. These outcomes support the idea that stromal cells in ADT-treated primary or metastatic prostate tumors can contribute to AIS as a consequence of a paracrine Hh signaling microenvironment. As such, Smo antagonists may be useful for targeting prostate tumor stromal cell-derived AIS and delaying the onset of CRPC after ADT.
- Published
- 2016
47. The Stress Response Mediator ATF3 Represses Androgen Signaling by Binding the Androgen Receptor
- Author
-
Hongbo Wang, Zhengxin Wang, Mengqian Chen, Hongmei Cui, Chunhong Yan, Simon W. Hayward, Ming Jiang, Ralph Buttyan, and Tsonwin Hai
- Subjects
Male ,medicine.drug_class ,Activating transcription factor ,Repressor ,Biology ,Models, Biological ,Epithelium ,Mice ,Prostate cancer ,Cell Line, Tumor ,medicine ,Animals ,Homeostasis ,Humans ,Molecular Biology ,Cell Proliferation ,Mice, Knockout ,Regulation of gene expression ,ATF3 ,Activating Transcription Factor 3 ,Prostate ,Prostatic Neoplasms ,Articles ,Cell Biology ,medicine.disease ,Androgen ,Protein Structure, Tertiary ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,Gene Expression Regulation ,Receptors, Androgen ,Cancer research ,Signal transduction ,Signal Transduction - Abstract
Activating transcription factor 3 (ATF3) is a common mediator of cellular stress response signaling and is often aberrantly expressed in prostate cancer. We report here that ATF3 can directly bind the androgen receptor (AR) and consequently repress AR-mediated gene expression. The ATF3-AR interaction requires the leucine zipper domain of ATF3 that independently binds the DNA-binding and ligand-binding domains of AR, and the interaction prevents AR from binding to cis-acting elements required for expression of androgen-dependent genes while inhibiting the AR N- and C-terminal interaction. The functional consequences of the loss of ATF3 expression include increased transcription of androgen-dependent genes in prostate cancer cells that correlates with increased ability to grow in low-androgen-containing medium and increased proliferative activity of the prostate epithelium in ATF3 knockout mice that is associated with prostatic hyperplasia. Our results thus demonstrate that ATF3 is a novel repressor of androgen signaling that can inhibit AR functions, allowing prostate cells to restore homeostasis and maintain integrity in the face of a broad spectrum of intrinsic and environmental insults.
- Published
- 2012
48. Paracrine hedgehog increases the steroidogenic potential of prostate stromal cells in a Gli-dependent manner
- Author
-
Michael Shtutman, Ralph Buttyan, Elina Levina, Richard D Carkner, and Mengqian Chen
- Subjects
medicine.medical_specialty ,Stromal cell ,Urology ,Steroid biosynthesis ,Biology ,medicine.disease ,Paracrine signalling ,Prostate cancer ,Endocrinology ,Oncology ,GLI1 ,CYP17A1 ,Internal medicine ,GLI2 ,medicine ,biology.protein ,Cancer research ,Hedgehog - Abstract
Acquired intratumoral steroidogenesis is involved in progression of prostate cancer to castration resistant disease (CRPC) and a target for improved therapeutics. Recent work has shown that prostate cancer cells can acquire steroidogenic activity as they progress to a therapeutic-resistant state. However, benign prostate stromal cells (PrSCs) also have steroidogenic potential though they are often overlooked as a source of intratumoral androgens. Here, we present preliminary studies showing that the steroidogenic activity of primary human PrSCs is significantly increased by exposure to a Hedgehog agonist (SAG) or by transduction of PrSCs with lentiviruses that expresses active Gli2 (Gli2ΔN), a transcription factor that is triggered by Hh signaling. Comparative gene expression profiling on Chips, that was confirmed by quantitative real-time PCR, revealed that hedgehog agonist treatment induced in these cells expressions of hedgehog target genes (Gli1, Ptch1, and SCUBE1) plus a specific cadre of genes involved in cholesterol/steroid biosynthesis, metabolism, and transport. Genes involved downstream in steroid hormone generation, including CYP17A1 and CYP19A1 were also induced. Both the hedgehog agonist and the Gli2-expressing lentivirus significantly increased the output of testosterone (T) from PrSCs that were supplemented with dihydroepiandrosterone (DHEA), an adrenal precursor of T. Finally, knockdown of Gli2 by siRNA suppressed the ability of SAG to induce this response. Collectively, our data indicate that hedgehog/Gli signaling may be a factor in acquired intratumoral steroidogenesis of a prostate tumor through its actions on stromal cells in the tumor microenvironment and an influence for the development of CRPC. Prostate 72:817–824, 2012. © 2011 Wiley Periodicals, Inc.
- Published
- 2011
49. Networks of intergenic long-range enhancers and snpRNAs drive castration-resistant phenotype of prostate cancer and contribute to pathogenesis of multiple common human disorders
- Author
-
Ian Guest, Anna B. Glinskii, Ralph Buttyan, Gennadi V. Glinsky, Chang-uk Lim, Denise Grant, Shuang Ma, Stewart Sell, and Jun Ma
- Subjects
Male ,Bioinformatics ,Single-nucleotide polymorphism ,NLR Proteins ,Regulatory Sequences, Ribonucleic Acid ,Biology ,Genetics & Genomics ,Polymorphism, Single Nucleotide ,Mice ,Intergenic region ,Transcription (biology) ,Report ,Cell Line, Tumor ,Molecular Cell Biology ,Gene expression ,microRNA ,Animals ,Humans ,General Materials Science ,Luciferases ,Enhancer ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Cancer ,Genetics ,Reverse Transcriptase Polymerase Chain Reaction ,Microarray analysis techniques ,Gene Expression Profiling ,Computational Biology ,Prostatic Neoplasms ,Cell Differentiation ,Sequence Analysis, DNA ,Cell Biology ,Flow Cytometry ,Microarray Analysis ,Molecular biology ,Phenotype ,Chromatin ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,DNA, Intergenic ,Apoptosis Regulatory Proteins ,Chromosomes, Human, Pair 17 ,Chromosomes, Human, Pair 8 ,Biotechnology ,Developmental Biology - Abstract
Biological and mechanistic relevance of intergenic disease-associated genetic loci (IDAGL) containing highly statistically significant disease-linked SNPs remains unknown. Here we present the experimental and clinical evidence revealing important role of IDAGL in human diseases. Targeted RT-PCR screen coupled with sequencing of purified PCR products detects widespread transcription at multiple intergenic disease-associated genomic loci (IDAGL) and identifies 96 small non-coding trans-regulatory RNAs of ~ 100-300 nt in length containing SNPs associated with 21 common human disorders (snpRNAs). Functionality of snpRNAs is supported by multiple independent lines of experimental evidence demonstrating their cell-type-specific expression and evolutionary conservation of sequences, genomic coordinates, and biological effects. Analysis of chromatin state signatures, expression profiling experiments using microarray and Q-PCR technologies, and luciferase reporter assays indicate that many IDAGL are Polycomb-regulated long-range enhancers. Expression of snpRNAs in human and mouse cells markedly affects cellular behavior and induces allele-specific clinically-relevant phenotypic changes: NLRP1-locus snpRNAs exert regulatory effects on monocyte/macrophage trans-differentiation, induce prostate cancer (PC) susceptibility snpRNAs, and transform low-malignancy hormone-dependent human PC cells into highly malignant androgen-independent PC. Q-PCR analysis and luciferase reporter assays demonstrate that snpRNA sequences represent allele-specific “decoy” targets of microRNAs which function as SNP-allele-specific modifiers of microRNA expression and activity. We demonstrate that trans-acting RNA molecules facilitating androgen depletion-independent growth (ADIG) in vitro and castration-resistant (CR) phenotype in vivo of PC contain intergenic 8q24-locus SNP variants which were recently linked with increased risk of developing PC. Expression level of 8q24-locus PC susceptibility snpRNAs is regulated by NLRP1-locus snpRNAs, which are transcribed from the intergenic long-range enhancer sequence located in 17p13 region at ~ 30 kb distance from the NLRP1 gene. Q-PCR analysis of clinical PC samples reveals markedly increased snpRNA expression levels in tumor tissues compared to the adjacent normal prostate [122-fold and 45-fold in Gleason 7 tumors (p = 0.03); 370-fold and 127-fold in Gleason 8 tumors (p = 0.0001); for NLRP1-locus and 8q24-locus SnpRNAs, respectively]. Highly concordant expression profiles of the NLRP1-locus snpRNAs and 8q24 CR-locus snpRNAs (r = 0.896; p < 0.0001) in clinical PC samples and experimental evidence of trans-regulatory effects of NLRP1-locus snpRNAs on expression of 8q24-locus SnpRNAs indicate that ADIG and CR phenotype of human PC cells can be triggered by RNA molecules transcribed from the NLRP1-locus intergenic enhancer and down-stream activation of the 8q24-locus snpRNAs. Our results define the intergenic NLRP1 and 8q24 regions as regulatory loci of ADIG and CR phenotype of human PC, reveal previously unknown molecular links between the innate immunity/inflammasome system and development of hormone-independent PC, and identify novel diagnostic and therapeutic targets exploration of which should be highly beneficial for clinical management of PC.
- Published
- 2011
50. Tumor-specific silencing of COPZ2 gene encoding coatomer protein complex subunit ζ2 renders tumor cells dependent on its paralogous gene COPZ1
- Author
-
Mikhail A. Nikiforov, Eugenia V. Broude, Elina Levina, Michael Shtutman, Gregory Hurteau, Chang-uk Lim, Felix T. Wieland, Igor B. Roninson, C. Steven Carmack, Timothy T. Harkins, Ye Ding, Ralph Buttyan, and Mirza S. Baig
- Subjects
Male ,COPZ1 ,Golgi Apparatus ,Apoptosis ,Biology ,Coatomer Protein ,Suppression, Genetic ,Cell Line, Tumor ,Neoplasms ,microRNA ,Autophagy ,Gene Knockdown Techniques ,Humans ,Protein Isoforms ,Cytotoxic T cell ,Gene silencing ,Gene Silencing ,RNA, Neoplasm ,RNA, Small Interfering ,Gene knockdown ,Multidisciplinary ,Base Sequence ,DNA, Neoplasm ,COPI ,Biological Sciences ,MicroRNAs ,Cell culture ,Cancer research ,Female - Abstract
Anticancer drugs are effective against tumors that depend on the molecular target of the drug. Known targets of cytotoxic anticancer drugs are involved in cell proliferation; drugs acting on such targets are ineffective against nonproliferating tumor cells, survival of which leads to eventual therapy failure. Function-based genomic screening identified the coatomer protein complex ζ1 ( COPZ1 ) gene as essential for different tumor cell types but not for normal cells. COPZ1 encodes a subunit of coatomer protein complex 1 (COPI) involved in intracellular traffic and autophagy. The knockdown of COPZ1, but not of COPZ2 encoding isoform coatomer protein complex ζ2, caused Golgi apparatus collapse, blocked autophagy, and induced apoptosis in both proliferating and nondividing tumor cells. In contrast, inhibition of normal cell growth required simultaneous knockdown of both COPZ1 and COPZ2. COPZ2 (but not COPZ1) was down-regulated in the majority of tumor cell lines and in clinical samples of different cancer types. Reexpression of COPZ2 protected tumor cells from killing by COPZ1 knockdown, indicating that tumor cell dependence on COPZ1 is the result of COPZ2 silencing. COPZ2 displays no tumor-suppressive activities, but it harbors microRNA 152, which is silenced in tumor cells concurrently with COPZ2 and acts as a tumor suppressor in vitro and in vivo. Silencing of microRNA 152 in different cancers and the ensuing down-regulation of its host gene COPZ2 offer a therapeutic opportunity for proliferation-independent selective killing of tumor cells by COPZ1-targeting agents.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.