101
|
Xu FP, Liu YH, Luo XL, Zhang F, Zhou HY, Ge Y, Liu C, Chen J, Luo DL, Yan LX, Mei P, Xu J, Zhuang HG. Overexpression of SRC-3 promotes esophageal squamous cell carcinoma aggressiveness by enhancing cell growth and invasiveness. Cancer Med 2016; 5:3500-3511. [PMID: 27781415 PMCID: PMC5224859 DOI: 10.1002/cam4.884] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 02/06/2023] Open
Abstract
Steroid receptor coactivator‐3 (SRC‐3), a transcriptional coactivator for nuclear receptors and other transcription factors, plays an important role in the genesis and progression of several cancers. However, studies investigated the role of SRC‐3 in esophageal squamous cell carcinomas (ESCCs) are limited, and the role of SRC‐3 in tumor progression remains unclear. We examined the expression of SRC‐3 in 8 ESCC cell lines and 302 human ESCC tissues by qPCR, Western blot, and immunohistochemistry. In addition, ESCC cell lines were subjected to proliferation and invasion assays, tumorigenicity assay, flow cytometry assay, qPCR, Western blot, and Chromatin Immunoprecipitation assay to investigate the role of SRC‐3 in cancer progression. SRC‐3 was overexpressed in 48% of cases and correlated with poor overall (P = 0.0076) and progression‐free (P = 0.0069) survival of surgically resected ESCC patient. Cox regression analysis revealed that SRC‐3 is an independent prognostic marker. Furthermore, we found that activation of insulin‐like growth factor (IGF)/AKT) was involved in the SRC‐3 on the cell growth and invasiveness in two ESCC cell lines, Eca109 and EC18 cells. SRC‐3 overexpression is clinically and functionally relevant to the progression of human ESCC, and might be a useful molecular target for ESCC prognosis and treatment.
Collapse
Affiliation(s)
- Fang-Ping Xu
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yan-Hui Liu
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xin-Lan Luo
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fen Zhang
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hai-Yu Zhou
- Department of Thoracic Surgery, Cancer Center, Guangdong General Hospital, Guangzhou, China
| | - Yan Ge
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chao Liu
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jie Chen
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dong-Lan Luo
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Li-Xu Yan
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Mei
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jie Xu
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Heng-Guo Zhuang
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
102
|
Lonard DM, O'Malley BW. Molecular Pathways: Targeting Steroid Receptor Coactivators in Cancer. Clin Cancer Res 2016; 22:5403-5407. [PMID: 27654711 DOI: 10.1158/1078-0432.ccr-15-1958] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 08/24/2016] [Accepted: 09/01/2016] [Indexed: 11/16/2022]
Abstract
Coactivators represent a large class of proteins that partner with nuclear receptors and other transcription factors to regulate gene expression. Given their pleiotropic roles in the control of transcription, coactivators have been implicated in a broad range of human disease states, including cancer. This is best typified by the three members of the steroid receptor coactivator (SRC) family, each of which integrates steroid hormone signaling and growth factor pathways to drive oncogenic gene expression programs in breast, endometrial, ovarian, prostate, and other cancers. Because of this, coactivators represent emerging targets for cancer therapeutics, and efforts are now being made to develop SRC-targeting agents, such as the SI-2 inhibitor and the novel SRC stimulator, MCB-613, that are able to block cancer growth in cell culture and animal model systems. Here, we will discuss the mechanisms through which coactivators drive cancer progression and how targeting coactivators represent a novel conceptual approach to combat tumor growth that is distinct from the use of other targeted therapeutic agents. We also will describe efforts to develop next-generation SRC inhibitors and stimulators that can be taken into the clinic for the treatment of recurrent, drug-resistant cancers. Clin Cancer Res; 22(22); 5403-7. ©2016 AACR.
Collapse
Affiliation(s)
- David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
103
|
Groner AC, Brown M, Theurillat JP. Targeting transcriptional co-activators in advanced prostate cancer. Cell Cycle 2016; 15:3333-3334. [PMID: 27610751 DOI: 10.1080/15384101.2016.1229019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Anna C Groner
- a Department of Medical Oncology , Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA.,b Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA
| | - Myles Brown
- a Department of Medical Oncology , Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA.,b Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA
| | - Jean-Philippe Theurillat
- c Institute of Oncology Research, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland.,d Faculty of Biology and Medicine, Center Hospitalier, Universitaire Vaudois, University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
104
|
Tabarestani S, Motallebi M, Akbari ME. Are Estrogen Receptor Genomic Aberrations Predictive of Hormone Therapy Response in Breast Cancer? IRANIAN JOURNAL OF CANCER PREVENTION 2016; 9:e6565. [PMID: 27761212 PMCID: PMC5056018 DOI: 10.17795/ijcp-6565] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/21/2016] [Accepted: 08/06/2016] [Indexed: 11/25/2022]
Abstract
CONTEXT Breast cancer is the most common cancer in women worldwide. Estrogen receptor (ER) positive breast cancer constitutes the majority of these cancers. Hormone therapy has significantly improved clinical outcomes for early- and late-stage hormone receptor positive breast cancer. Although most patients with early stage breast cancer are treated with curative intent, approximately 20% - 30% of patients eventually experience a recurrence. During the last two decades, there have been tremendous efforts to understand the biological mechanisms of hormone therapy resistance, with the ultimate goal of implementing new therapeutic strategies to improve the current treatments for ER positive breast cancer. Several mechanisms of hormone therapy resistance have been proposed, including genetic alterations that lead to altered ER expression or ERs with changed protein sequence. EVIDENCE ACQUISITION A Pubmed search was performed utilizing various related terms. Articles over the past 20 years were analyzed and selected for review. RESULTS On the basis of published studies, the frequencies of ESR1 (the gene encoding ER) mutations in ER positive metastatic breast cancer range from 11% to 55%. Future larger prospective studies with standardized mutation detection methods may be necessary to determine the true incidence of ESR1 mutations. ESR1 amplification in breast cancer remains a controversial issue, with numerous studies either confirmed or challenged the reports of ESR1 amplification. The combination of intra-tumor heterogeneity regarding ESR1 copy number alterations and low level ESR1 copy number increase may account for these discrepancies. CONCLUSIONS While numerous unknown issues on the role of ESR1 mutations in advanced breast cancer remain, these new findings will certainly deepen current knowledge on molecular evolution of breast cancer and acquired resistance to hormone therapy.
Collapse
Affiliation(s)
- Sanaz Tabarestani
- Cancer Research Center, Shahid Beheshti Univeristy of Medical Sciences, Tehran, IR Iran
| | - Marzieh Motallebi
- Cancer Research Center, Shahid Beheshti Univeristy of Medical Sciences, Tehran, IR Iran
| | | |
Collapse
|
105
|
Qin L, Xu Y, Xu Y, Ma G, Liao L, Wu Y, Li Y, Wang X, Wang X, Jiang J, Wang J, Xu J. NCOA1 promotes angiogenesis in breast tumors by simultaneously enhancing both HIF1α- and AP-1-mediated VEGFa transcription. Oncotarget 2016; 6:23890-904. [PMID: 26287601 PMCID: PMC4695159 DOI: 10.18632/oncotarget.4341] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/09/2015] [Indexed: 01/03/2023] Open
Abstract
Nuclear receptor coactivator 1 (NCOA1) is overexpressed in a subset of breast cancer and its increased expression positively correlates with disease recurrence and metastasis. Although NCOA1 is known to promote breast cancer metastasis through working with multiple transcription factors to upregulate the expression of Twist1, ITGA5, CSF-1, SDF1 and CXCR4, the role of NCOA1 in breast tumor angiogenesis has not been investigated. In this study, we found that the microvascular density (MVD) was significantly decreased and increased in Ncoa1-knockout and NCOA1-overexpressing mammary tumors, respectively, in several breast cancer mouse models. Knockout or knockdown of NCOA1 in breast cancer cell lines also markedly compromised their capability to induce angiogenesis in Matrigel plugs embedded subcutaneously in mice, while this compromised capability could be rescued by VEGFa treatment. At the molecular level, NCOA1 upregulates VEGFa expression in both mouse mammary tumors and cultured breast cancer cells, and it does so by associating with both c-Fos, which is recruited to the AP-1 site at bp −938 of the VEGFa promoter, and HIF1α, which is recruited to the HIF1α-binding element at bp −979 of the VEGFa promoter, to enhance VEGFa transcription. In 140 human breast tumors, high NCOA1 protein correlates with high MVD and patients with both high NCOA1 and high MVD showed significantly shorter survival time. In summary, this study revealed a novel mechanism that NCOA1 potentiates breast cancer angiogenesis through upregulating HIF1α and AP-1-mediated VEGFa expression, which reinforces the rational of targeting NCOA1 in controlling breast cancer progression and metastasis.
Collapse
Affiliation(s)
- Li Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yan Xu
- Department of Breast and Thyroid Surgery, Daping Hospital, Third Military Medical University, Chongqing, China.,Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yixiang Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Gang Ma
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yelin Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Xian Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Xiaosong Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Jun Jiang
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jin Wang
- Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Institute for Cancer Medicine and College of Basic Medical Sciences, Sichuan Medical University, Luzhou, Sichuan, China
| |
Collapse
|
106
|
Zou Z, Luo X, Nie P, Wu B, Zhang T, Wei Y, Wang W, Geng G, Jiang J, Mi Y. Inhibition of SRC-3 enhances sensitivity of human cancer cells to histone deacetylase inhibitors. Biochem Biophys Res Commun 2016; 478:227-233. [PMID: 27425252 DOI: 10.1016/j.bbrc.2016.07.063] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/13/2016] [Indexed: 12/31/2022]
Abstract
SRC-3 is widely expressed in multiple tumor types and involved in cancer cell proliferation and apoptosis. Histone deacetylase (HDAC) inhibitors are promising antitumor drugs. However, the poor efficacy of HDAC inhibitors in solid tumors has restricted its further clinical application. Here, we reported the novel finding that depletion of SRC-3 enhanced sensitivity of breast and lung cancer cells to HDAC inhibitors (SAHA and romidepsin). In contrast, overexpression of SRC-3 decreased SAHA-induced cancer cell apoptosis. Furthermore, we found that SRC-3 inhibitor bufalin increased cancer cell apoptosis induced by HDAC inhibitors. The combination of bufalin and SAHA was particular efficient in attenuating AKT activation and reducing Bcl-2 levels. Taken together, these accumulating data might guide development of new breast and lung cancer therapies.
Collapse
Affiliation(s)
- Zhengzhi Zou
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510000, China.
| | - Xiaoyong Luo
- Department of Oncology, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang 471000, China
| | - Peipei Nie
- KingMed Diagnostics and KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 510000, China
| | - Baoyan Wu
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510000, China
| | - Tao Zhang
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510000, China
| | - Yanchun Wei
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510000, China
| | - Wenyi Wang
- Xiamen Cancer Center, Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Guojun Geng
- Xiamen Cancer Center, Department of Thoracic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Jie Jiang
- Xiamen Cancer Center, Department of Thoracic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Yanjun Mi
- Xiamen Cancer Center, Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China.
| |
Collapse
|
107
|
Sun P, Feng LX, Zhang DM, Liu M, Liu W, Mi T, Wu WY, Jiang BH, Yang M, Hu LH, Guo DA, Liu X. Bufalin derivative BF211 inhibits proteasome activity in human lung cancer cells in vitro by inhibiting β1 subunit expression and disrupting proteasome assembly. Acta Pharmacol Sin 2016; 37:908-18. [PMID: 27238210 DOI: 10.1038/aps.2016.30] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
AIM Bufalin is one of the active components in the traditional Chinese medicine ChanSu that is used to treat arrhythmia, inflammation and cancer. BF211 is a bufalin derivative with stronger cytotoxic activity in cancer cells. The aim of this study was to identify the putative target proteins of BF211 and the signaling pathways in cancer cells. METHODS A549 human lung cancer cells were treated with BF211. A SILAC-based proteomic analysis was used to detect the protein expression profiles of BF211-treated A549 cells. Cellular proteasome activities were examined using fluorogenic peptide substrates, and the binding affinities of BF211 to recombinant proteasome subunit proteins were evaluated using the Biacore assay. The expression levels of proteasome subunits were determined using RT-PCR and Western blotting, and the levels of the integral 26S proteasome were evaluated using native PAGE analysis. RESULTS The proteomic analysis revealed that 1282 proteins were differentially expressed in BF211-treated A549 cells, and the putative target proteins of BF211 were associated with various cellular functions, including transcription, translation, mRNA splicing, ribosomal protein synthesis and proteasome function. In A549 cells, BF211 (5, 10, and 20 nmol/L) dose-dependently inhibited the enzymatic activities of proteasome. But BF211 displayed a moderate affinity in binding to proteasome β1 subunit and no binding affinity to the β2 and β5 subunits. Moreover, BF211 (0.1, 1, and 10 nmol/L) did not inhibit the proteasome activities in the cell lysates. BF211 (5, 10, and 20 nmol/L) significantly decreased the expression level of proteasome β1 subunit and the levels of integral 26S proteasome in A549 cells. Similarly, knockdown of the β1 subunit with siRNA in A549 cells significantly decreased integral 26S proteasome and proteasome activity. CONCLUSION BF211 inhibits proteasome activity in A549 cells by decreasing β1 subunit expression and disrupting proteasome assembly.
Collapse
|
108
|
Shi J, Liu W, Sui F, Lu R, He Q, Yang Q, Lv H, Shi B, Hou P. Frequent amplification of AIB1, a critical oncogene modulating major signaling pathways, is associated with poor survival in gastric cancer. Oncotarget 2016; 6:14344-59. [PMID: 25970779 PMCID: PMC4546471 DOI: 10.18632/oncotarget.3852] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 04/17/2015] [Indexed: 11/26/2022] Open
Abstract
Amplified in breast cancer 1 (AIB1) is a member of p160 steroid receptor coactivator (SRC) family that mediates the transcriptional activities of nuclear receptors and other transcription factors. It acts as a major oncogene in diverse cancers, whereas biological function of AIB1 in gastric cancer remains largely unclear. This study was designed to explore the role of AIB1 in gastric tumorigenesis and its potential as a useful prognostic marker and therapeutic target in this cancer. Our data demonstrated that AIB1 was significantly up-regulated in gastric cancer tissues as compared with control subjects. Moreover, AIB1 amplification was found in 47 of 133 (35.3%) gastric cancer cases, but not in control subjects. AIB1 amplification was positively associated with its protein expression, and was significantly correlated with poor patient survival. AIB1 knockdown in gastric cancer cells dramatically inhibited cell proliferation, invasiveness and tumorigenic potential in nude mice, and induced cell cycle arrest and apoptosis. Mechanically, AIB1 promotes gastric cancer cell proliferation, survival and invasiveness through modulating major signaling pathways such as ErbB and Wnt/β-catenin pathways. Collectively, these findings suggest that AIB1 plays an important role in the pathogenesis of gastric cancer and represents a potential prognostic marker and therapeutic target for this cancer.
Collapse
Affiliation(s)
- Jing Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Wei Liu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Fang Sui
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Rong Lu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Qingyuan He
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Qi Yang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Hongjun Lv
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| |
Collapse
|
109
|
Wang J, Zou JX, Xue X, Cai D, Zhang Y, Duan Z, Xiang Q, Yang JC, Louie MC, Borowsky AD, Gao AC, Evans CP, Lam KS, Xu J, Kung HJ, Evans RM, Xu Y, Chen HW. ROR-γ drives androgen receptor expression and represents a therapeutic target in castration-resistant prostate cancer. Nat Med 2016; 22:488-96. [PMID: 27019329 PMCID: PMC5030109 DOI: 10.1038/nm.4070] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 02/19/2016] [Indexed: 02/07/2023]
Abstract
The androgen receptor (AR) is overexpressed and hyperactivated in human castration-resistant prostate cancer (CRPC). However, the determinants of AR overexpression in CRPC are poorly defined. Here we show that retinoic acid receptor-related orphan receptor γ (ROR-γ) is overexpressed and amplified in metastatic CRPC tumors, and that ROR-γ drives AR expression in the tumors. ROR-γ recruits nuclear receptor coactivator 1 and 3 (NCOA1 and NCOA3, also known as SRC-1 and SRC-3) to an AR-ROR response element (RORE) to stimulate AR gene transcription. ROR-γ antagonists suppress the expression of both AR and its variant AR-V7 in prostate cancer (PCa) cell lines and tumors. ROR-γ antagonists also markedly diminish genome-wide AR binding, H3K27ac abundance and expression of the AR target gene network. Finally, ROR-γ antagonists suppressed tumor growth in multiple AR-expressing, but not AR-negative, xenograft PCa models, and they effectively sensitized CRPC tumors to enzalutamide, without overt toxicity, in mice. Taken together, these results establish ROR-γ as a key player in CRPC by acting upstream of AR and as a potential therapeutic target for advanced PCa.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Benzamides
- Cell Survival/drug effects
- Databases, Factual
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Glucose-6-Phosphate Isomerase
- Humans
- Immunoblotting
- Immunohistochemistry
- Male
- Mice
- Neoplasm Transplantation
- Nitriles
- Nuclear Receptor Coactivator 1/metabolism
- Nuclear Receptor Coactivator 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Phenylthiohydantoin/analogs & derivatives
- Phenylthiohydantoin/pharmacology
- Piperazines/pharmacology
- Propanols/pharmacology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- RNA, Messenger/metabolism
- Real-Time Polymerase Chain Reaction
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Response Elements
- Tumor Stem Cell Assay
Collapse
Affiliation(s)
- Junjian Wang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - June X Zou
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Xiaoqian Xue
- Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Demin Cai
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Yan Zhang
- Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhijian Duan
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Qiuping Xiang
- Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Joy C Yang
- Department of Urology, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Maggie C Louie
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, California, USA
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Allen C Gao
- Department of Urology, School of Medicine, University of California, Davis, Sacramento, California, USA
- Comprehensive Cancer Center, University of California, Davis, Sacramento, California, USA
| | - Christopher P Evans
- Department of Urology, School of Medicine, University of California, Davis, Sacramento, California, USA
- Comprehensive Cancer Center, University of California, Davis, Sacramento, California, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
- Comprehensive Cancer Center, University of California, Davis, Sacramento, California, USA
| | - Jianzhen Xu
- Shantou University Medical College, Shantou, China
| | - Hsing-Jien Kung
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
- Comprehensive Cancer Center, University of California, Davis, Sacramento, California, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute, Howard Hughes Medical Institute, Salk Institute, La Jolla, California, USA
| | - Yong Xu
- Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
- Comprehensive Cancer Center, University of California, Davis, Sacramento, California, USA
- Veterans Affairs Northern California Health Care System-Mather, Mather, California, USA
| |
Collapse
|
110
|
Flach KD, Zwart W. The first decade of estrogen receptor cistromics in breast cancer. J Endocrinol 2016; 229:R43-56. [PMID: 26906743 DOI: 10.1530/joe-16-0003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 02/23/2016] [Indexed: 02/03/2023]
Abstract
The advent of genome-wide transcription factor profiling has revolutionized the field of breast cancer research. Estrogen receptor α (ERα), the major drug target in hormone receptor-positive breast cancer, has been known as a key transcriptional regulator in tumor progression for over 30 years. Even though this function of ERα is heavily exploited and widely accepted as an Achilles heel for hormonal breast cancer, only since the last decade we have been able to understand how this transcription factor is functioning on a genome-wide scale. Initial ChIP-on-chip (chromatin immunoprecipitation coupled with tiling array) analyses have taught us that ERα is an enhancer-associated factor binding to many thousands of sites throughout the human genome and revealed the identity of a number of directly interacting transcription factors that are essential for ERα action. More recently, with the development of massive parallel sequencing technologies and refinements thereof in sample processing, a genome-wide interrogation of ERα has become feasible and affordable with unprecedented data quality and richness. These studies have revealed numerous additional biological insights into ERα behavior in cell lines and especially in clinical specimens. Therefore, what have we actually learned during this first decade of cistromics in breast cancer and where may future developments in the field take us?
Collapse
Affiliation(s)
- Koen D Flach
- Division of Molecular PathologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Molecular PathologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
111
|
The Role of Steroid Receptor Coactivators in Hormone Dependent Cancers and Their Potential as Therapeutic Targets. Discov Oncol 2016; 7:229-35. [PMID: 27125199 DOI: 10.1007/s12672-016-0261-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/08/2016] [Indexed: 10/21/2022] Open
Abstract
Steroid receptor coactivator (SRC) family members (SRC-1, SRC-2, SRC-3) interact with nuclear receptors (NRs) and many transcription factors to enhance target gene transcription. Deregulation of SRCs is widely implicated in NR mediated diseases, especially hormone dependent cancers. By integrating steroid hormone signaling and growth factor pathways, SRC proteins exert multiple modes of oncogenic regulation in cancers and represent emerging targets for cancer therapeutics. Recent work has identified SRC-targeting agents that show promise in blocking tumor growth in vitro and in vivo, and have the potential to function as powerful and broadly encompassing treatments for different cancers.
Collapse
|
112
|
Development of potent small-molecule inhibitors to drug the undruggable steroid receptor coactivator-3. Proc Natl Acad Sci U S A 2016; 113:4970-5. [PMID: 27084884 DOI: 10.1073/pnas.1604274113] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Protein-protein interactions (PPIs) play a central role in most biological processes, and therefore represent an important class of targets for therapeutic development. However, disrupting PPIs using small-molecule inhibitors (SMIs) is challenging and often deemed as "undruggable." We developed a cell-based functional assay for high-throughput screening to identify SMIs for steroid receptor coactivator-3 (SRC-3 or AIB1), a large and mostly unstructured nuclear protein. Without any SRC-3 structural information, we identified SI-2 as a highly promising SMI for SRC-3. SI-2 meets all of the criteria of Lipinski's rule [Lipinski et al. (2001) Adv Drug Deliv Rev 46(1-3):3-26] for a drug-like molecule and has a half-life of 1 h in a pharmacokinetics study and a reasonable oral availability in mice. As a SRC-3 SMI, SI-2 can selectively reduce the transcriptional activities and the protein concentrations of SRC-3 in cells through direct physical interactions with SRC-3, and selectively induce breast cancer cell death with IC50 values in the low nanomolar range (3-20 nM), but not affect normal cell viability. Furthermore, SI-2 can significantly inhibit primary tumor growth and reduce SRC-3 protein levels in a breast cancer mouse model. In a toxicology study, SI-2 caused minimal acute cardiotoxicity based on a hERG channel blocking assay and an unappreciable chronic toxicity to major organs based on histological analyses. We believe that this work could significantly improve breast cancer treatment through the development of "first-in-class" drugs that target oncogenic coactivators.
Collapse
|
113
|
Foley C, Mitsiades N. Moving Beyond the Androgen Receptor (AR): Targeting AR-Interacting Proteins to Treat Prostate Cancer. HORMONES & CANCER 2016; 7:84-103. [PMID: 26728473 PMCID: PMC5380740 DOI: 10.1007/s12672-015-0239-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/23/2015] [Indexed: 02/07/2023]
Abstract
Medical or surgical castration serves as the backbone of systemic therapy for advanced and metastatic prostate cancer, taking advantage of the importance of androgen signaling in this disease. Unfortunately, resistance to castration emerges almost universally. Despite the development and approval of new and more potent androgen synthesis inhibitors and androgen receptor (AR) antagonists, prostate cancers continue to develop resistance to these therapeutics, while often maintaining their dependence on the AR signaling axis. This highlights the need for innovative therapeutic approaches that aim to continue disrupting AR downstream signaling but are orthogonal to directly targeting the AR itself. In this review, we discuss the preclinical research that has been done, as well as clinical trials for prostate cancer, on inhibiting several important families of AR-interacting proteins, including chaperones (such as heat shock protein 90 (HSP90) and FKBP52), pioneer factors (including forkhead box protein A1 (FOXA1) and GATA-2), and AR transcriptional coregulators such as the p160 steroid receptor coactivators (SRCs) SRC-1, SRC-2, SRC-3, as well as lysine deacetylases (KDACs) and lysine acetyltransferases (KATs). Researching the effect of-and developing new therapeutic agents that target-the AR signaling axis is critical to advancing our understanding of prostate cancer biology, to continue to improve treatments for prostate cancer and for overcoming castration resistance.
Collapse
Affiliation(s)
- Christopher Foley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Suite R407, MS: BCM187, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Suite R407, MS: BCM187, Houston, TX, 77030, USA
| | - Nicholas Mitsiades
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Suite R407, MS: BCM187, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Suite R407, MS: BCM187, Houston, TX, 77030, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
114
|
DePriest AD, Fiandalo MV, Schlanger S, Heemers F, Mohler JL, Liu S, Heemers HV. Regulators of Androgen Action Resource: a one-stop shop for the comprehensive study of androgen receptor action. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2016; 2016:bav125. [PMID: 26876983 PMCID: PMC4752970 DOI: 10.1093/database/bav125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 12/14/2015] [Indexed: 12/20/2022]
Abstract
Androgen receptor (AR) is a ligand-activated transcription factor that is the main target for treatment of non-organ-confined prostate cancer (CaP). Failure of life-prolonging AR-targeting androgen deprivation therapy is due to flexibility in steroidogenic pathways that control intracrine androgen levels and variability in the AR transcriptional output. Androgen biosynthesis enzymes, androgen transporters and AR-associated coregulators are attractive novel CaP treatment targets. These proteins, however, are characterized by multiple transcript variants and isoforms, are subject to genomic alterations, and are differentially expressed among CaPs. Determining their therapeutic potential requires evaluation of extensive, diverse datasets that are dispersed over multiple databases, websites and literature reports. Mining and integrating these datasets are cumbersome, time-consuming tasks and provide only snapshots of relevant information. To overcome this impediment to effective, efficient study of AR and potential drug targets, we developed the Regulators of Androgen Action Resource (RAAR), a non-redundant, curated and user-friendly searchable web interface. RAAR centralizes information on gene function, clinical relevance, and resources for 55 genes that encode proteins involved in biosynthesis, metabolism and transport of androgens and for 274 AR-associated coregulator genes. Data in RAAR are organized in two levels: (i) Information pertaining to production of androgens is contained in a ‘pre-receptor level’ database, and coregulator gene information is provided in a ‘post-receptor level’ database, and (ii) an ‘other resources’ database contains links to additional databases that are complementary to and useful to pursue further the information provided in RAAR. For each of its 329 entries, RAAR provides access to more than 20 well-curated publicly available databases, and thus, access to thousands of data points. Hyperlinks provide direct access to gene-specific entries in the respective database(s). RAAR is a novel, freely available resource that provides fast, reliable and easy access to integrated information that is needed to develop alternative CaP therapies. Database URL: http://www.lerner.ccf.org/cancerbio/heemers/RAAR/search/
Collapse
Affiliation(s)
| | | | - Simon Schlanger
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, USA
| | | | - James L Mohler
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Hannelore V Heemers
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, USA Department of Urology Department of Hematology/Medical Oncology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
115
|
Durlacher CT, Chow K, Chen XW, He ZX, Zhang X, Yang T, Zhou SF. Targeting Na⁺/K⁺ -translocating adenosine triphosphatase in cancer treatment. Clin Exp Pharmacol Physiol 2016; 42:427-43. [PMID: 25739707 DOI: 10.1111/1440-1681.12385] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/09/2015] [Accepted: 02/21/2015] [Indexed: 12/24/2022]
Abstract
The Na(+) /K(+) -translocating adenosine triphosphatase (ATPase) transports sodium and potassium across the plasma membrane and represents a potential target in cancer chemotherapy. Na(+) /K(+) -ATPase belongs to the P-type ATPase family (also known as E1-E2 ATPase), which is involved in transporting certain ions, metals, and lipids across the plasma membrane of mammalian cells. In humans, the Na(+) /K(+) -ATPase is a binary complex of an α-subunit that has four isoforms (α1 -α4 ) and a β-subunit that has three isoforms (β1 -β3 ). This review aims to update our knowledge on the role of Na(+) /K(+) -ATPase in cancer development and metastasis, as well as on how Na(+) /K(+) -ATPase inhibitors kill tumour cells. The Na(+) /K(+) -ATPase has been found to be associated with cancer initiation, growth, development, and metastasis. Cardiac glycosides have exhibited anticancer effects in cell-based and mouse studies via inhibition of the Na(+) /K(+) -ATPase and other mechanisms. Na(+) /K(+) -ATPase inhibitors may kill cancer cells via induction of apoptosis and autophagy, radical oxygen species production, and cell cycle arrest. They also modulate multiple signalling pathways that regulate cancer cell survival and death, which contributes to their antiproliferative activities in cancer cells. The clinical evidence supporting the use of Na(+) /K(+) -ATPase inhibitors as anticancer drugs is weak. Several phase I and phase II clinical trials with digoxin, Anvirzel, and huachansu (an intravenous formulated extract of the venom of the wild toad), either alone or more often in combination with other anticancer agents, have shown acceptable safety profiles but limited efficacy in cancer patients. Well-designed randomized clinical trials with reasonable sample sizes are certainly warranted to confirm the efficacy and safety of cardiac glycosides for the treatment of cancer.
Collapse
Affiliation(s)
- Cameron T Durlacher
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | | | | | | | | | | | | |
Collapse
|
116
|
Liu X, Li H, Rajurkar M, Li Q, Cotton JL, Ou J, Zhu LJ, Goel HL, Mercurio AM, Park JS, Davis RJ, Mao J. Tead and AP1 Coordinate Transcription and Motility. Cell Rep 2016; 14:1169-1180. [PMID: 26832411 DOI: 10.1016/j.celrep.2015.12.104] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 11/11/2015] [Accepted: 12/23/2015] [Indexed: 11/25/2022] Open
Abstract
The Tead family transcription factors are the major intracellular mediators of the Hippo-Yap pathway. Despite the importance of Hippo signaling in tumorigenesis, Tead-dependent downstream oncogenic programs and target genes in cancer cells remain poorly understood. Here, we characterize Tead4-mediated transcriptional networks in a diverse range of cancer cells, including neuroblastoma, colorectal, lung, and endometrial carcinomas. By intersecting genome-wide chromatin occupancy analyses of Tead4, JunD, and Fra1/2, we find that Tead4 cooperates with AP1 transcription factors to coordinate target gene transcription. We find that Tead-AP1 interaction is JNK independent but engages the SRC1-3 co-activators to promote downstream transcription. Furthermore, we show that Tead-AP1 cooperation regulates the activity of the Dock-Rac/CDC42 module and drives the expression of a unique core set of target genes, thereby directing cell migration and invasion. Together, our data unveil a critical regulatory mechanism underlying Tead- and AP1-controlled transcriptional and functional outputs in cancer cells.
Collapse
Affiliation(s)
- Xiangfan Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Huapeng Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Mihir Rajurkar
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Qi Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jennifer L Cotton
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jianhong Ou
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lihua J Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Hira L Goel
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Arthur M Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Joo-Seop Park
- Divisions of Pediatric Urology and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, Worcester, MA 01605, USA
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
117
|
Wang T, Mu L, Jin H, Zhang P, Wang Y, Ma X, Pan J, Miao J, Yuan Y. The effects of bufadienolides on HER2 overexpressing breast cancer cells. Tumour Biol 2015; 37:7155-63. [DOI: 10.1007/s13277-015-4381-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/04/2015] [Indexed: 10/22/2022] Open
|
118
|
Szwarc MM, Lydon JP, O'Malley BW. Steroid receptor coactivators as therapeutic targets in the female reproductive system. J Steroid Biochem Mol Biol 2015; 154:32-8. [PMID: 26151740 PMCID: PMC5201167 DOI: 10.1016/j.jsbmb.2015.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 11/25/2022]
Abstract
The steroid receptor coactivators (SRCs/p160/NCOA) are a family of three transcriptional coregulators initially discovered to transactivate the transcriptional potency of steroid hormone receptors. Even though SRCs were also found to modulate the activity of multiple other transcription factors, their function is still strongly associated with regulation of steroid hormone action and many studies have found that they are critical for the regulation of reproductive biology. In the case of the female reproductive tract, SRCs have been found to play crucial roles in its physiology, ranging from ovulation, implantation, to parturition. Not surprisingly, SRCs' action has been linked to numerous abnormalities and debilitating disorders of female reproductive tissues, including infertility, cancer, and endometriosis. Many of these pathologies are still in critical need of therapeutic intervention and "proof-of-principle" studies have found that SRCs are excellent targets in pathological states. Therefore, small molecule modulators of SRCs' activity could be applied in the future in the treatment of many diseases of the female reproductive system.
Collapse
Affiliation(s)
- Maria M Szwarc
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA.
| |
Collapse
|
119
|
Steroid Receptor Coactivator-3 (SRC-3/AIB1) as a Novel Therapeutic Target in Triple Negative Breast Cancer and Its Inhibition with a Phospho-Bufalin Prodrug. PLoS One 2015; 10:e0140011. [PMID: 26431029 PMCID: PMC4592245 DOI: 10.1371/journal.pone.0140011] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 09/08/2015] [Indexed: 11/19/2022] Open
Abstract
Triple negative breast cancer (TNBC) has the poorest prognosis of all types of breast cancer and currently lacks efficient targeted therapy. Chemotherapy is the traditional standard-of-care for TNBC, but is frequently accompanied by severe side effects. Despite the fact that high expression of steroid receptor coactivator 3 (SRC-3) is correlated with poor survival in estrogen receptor positive breast cancer patients, its role in TNBC has not been extensively investigated. Here, we show that high expression of SRC-3 correlates with both poor overall survival and post progression survival in TNBC patients, suggesting that SRC-3 can serve as a prognostic marker for TNBC. Furthermore, we demonstrated that bufalin, a SRC-3 small molecule inhibitor, when introduced even at nM concentrations, can significantly reduce TNBC cell viability and motility. However, because bufalin has minimal water solubility, its in vivo application is limited. Therefore, we developed a water soluble prodrug, 3-phospho-bufalin, to facilitate its in vivo administration. In addition, we demonstrated that 3-phospho-bufalin can effectively inhibit tumor growth in an orthotopic TNBC mouse model, suggesting its potential application as a targeted therapy for TNBC treatment.
Collapse
|
120
|
Bufadienolides with cytotoxic activity from the skins of Bufo bufo gargarizans. Fitoterapia 2015; 105:7-15. [DOI: 10.1016/j.fitote.2015.05.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 05/17/2015] [Indexed: 11/18/2022]
|
121
|
Szwarc MM, Lydon JP, O'Malley BW. Reprint of "Steroid receptor coactivators as therapeutic targets in the female reproductive system". J Steroid Biochem Mol Biol 2015; 153:144-50. [PMID: 26291832 DOI: 10.1016/j.jsbmb.2015.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 11/22/2022]
Abstract
The steroid receptor coactivators (SRCs/p160/NCOA) are a family of three transcriptional coregulators initially discovered to transactivate the transcriptional potency of steroid hormone receptors. Even though SRCs were also found to modulate the activity of multiple other transcription factors, their function is still strongly associated with regulation of steroid hormone action and many studies have found that they are critical for the regulation of reproductive biology. In the case of the female reproductive tract, SRCs have been found to play crucial roles in its physiology, ranging from ovulation, implantation, to parturition. Not surprisingly, SRCs' action has been linked to numerous abnormalities and debilitating disorders of female reproductive tissues, including infertility, cancer, and endometriosis. Many of these pathologies are still in critical need of therapeutic intervention and "proof-of-principle" studies have found that SRCs are excellent targets in pathological states. Therefore, small molecule modulators of SRCs' activity could be applied in the future in the treatment of many diseases of the female reproductive system.
Collapse
Affiliation(s)
- Maria M Szwarc
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA.
| |
Collapse
|
122
|
Wang L, Yu Y, Chow DC, Yan F, Hsu CC, Stossi F, Mancini MA, Palzkill T, Liao L, Zhou S, Xu J, Lonard DM, O'Malley BW. Characterization of a Steroid Receptor Coactivator Small Molecule Stimulator that Overstimulates Cancer Cells and Leads to Cell Stress and Death. Cancer Cell 2015; 28:240-52. [PMID: 26267537 PMCID: PMC4536575 DOI: 10.1016/j.ccell.2015.07.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 03/12/2015] [Accepted: 07/10/2015] [Indexed: 12/18/2022]
Abstract
By integrating growth pathways on which cancer cells rely, steroid receptor coactivators (SRC-1, SRC-2, and SRC-3) represent emerging targets in cancer therapeutics. High-throughput screening for SRC small molecule inhibitors (SMIs) uncovered MCB-613 as a potent SRC small molecule "stimulator" (SMS). We demonstrate that MCB-613 can super-stimulate SRCs' transcriptional activity. Further investigation revealed that MCB-613 increases SRCs' interactions with other coactivators and markedly induces ER stress coupled to the generation of reactive oxygen species (ROS). Because cancer cells overexpress SRCs and rely on them for growth, we show that we can exploit MCB-613 to selectively induce excessive stress in cancer cells. This suggests that over-stimulating the SRC oncogenic program can be an effective strategy to kill cancer cells.
Collapse
Affiliation(s)
- Lei Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yang Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dar-Chone Chow
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fei Yan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chih-Chao Hsu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Timothy Palzkill
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Suoling Zhou
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
123
|
Nardone A, De Angelis C, Trivedi MV, Osborne CK, Schiff R. The changing role of ER in endocrine resistance. Breast 2015; 24 Suppl 2:S60-6. [PMID: 26271713 DOI: 10.1016/j.breast.2015.07.015] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Estrogen receptor (ER) is expressed in approximately 70% of newly diagnosed breast tumors. Although endocrine therapy targeting ER is highly effective, intrinsic or acquired resistance is common, significantly jeopardizing treatment outcomes and minimizing overall survival. Even in the presence of endocrine resistance, a continued role of ER signaling is suggested by several lines of clinical and preclinical evidence. Indeed, inhibition or down-regulation of ER reduces tumor growth in preclinical models of acquired endocrine resistance, and many patients with recurrent ER+ breast tumors progressing on one type of ER-targeted treatment still benefit from sequential endocrine treatments that target ER by a different mechanism. New insights into the nature and biology of ER have revealed several mechanisms sustaining altered ER signaling in endocrine-resistant tumors, including deregulated growth factor receptor signaling that results in ligand-independent ER activation, unbalanced ER co-regulator activity, and genomic alterations involving the ER gene ESR1. Therefore, biopsies of recurrent lesions are needed to assess the changes in epi/genomics and signaling landscape of ER and associated pathways in order to tailor therapies to effectively overcome endocrine resistance. In addition, more completely abolishing the levels and activity of ER and its co-activators, in combination with selected signal transduction inhibitors or agents blocking the upstream or downstream targets of the ER pathway, may provide a better therapeutic strategy in combating endocrine resistance.
Collapse
Affiliation(s)
- Agostina Nardone
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA
| | - Carmine De Angelis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA; Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Italy
| | - Meghana V Trivedi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Pharmacy Practice and Translational Research, University of Houston, College of Pharmacy, Houston, TX 77030, USA
| | - C Kent Osborne
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA.
| |
Collapse
|
124
|
Tong Z, Li M, Wang W, Mo P, Yu L, Liu K, Ren W, Li W, Zhang H, Xu J, Yu C. Steroid Receptor Coactivator 1 Promotes Human Hepatocellular Carcinoma Progression by Enhancing Wnt/β-Catenin Signaling. J Biol Chem 2015; 290:18596-608. [PMID: 26082485 PMCID: PMC4513118 DOI: 10.1074/jbc.m115.640490] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Indexed: 02/05/2023] Open
Abstract
Steroid receptor coactivator 1 (SRC-1) is a transcriptional coactivator not only for steroid receptors, such as androgen receptor and estrogen receptor, but also for other transcription factors. SRC-1 has been shown to play an important role in the progression of breast cancer and prostate cancer. However, its role in liver cancer progression remains unknown. In this study, we report that SRC-1 was overexpressed in 25 (62.5%) of 40 human hepatocellular carcinoma (HCC) specimens. Down-regulation of SRC-1 decreased HCC cell proliferation and impaired tumor maintenance in HCC xenografts. Knockdown of SRC-1 reduced protein levels of the proliferation marker proliferating cell nuclear antigen (PCNA) and the oncogene c-Myc. Knockout of SRC-1 in mice reduced diethylnitrosamine/CCl4-induced tumor formation in the liver and the expression of c-Myc and PCNA in liver tumors. SRC-1 promoted c-Myc expression, at least in part, by directly interacting with β-catenin to enhance Wnt/β-catenin signaling. Consistent with these results, the expression of SRC-1 was positively correlated with PCNA expression in human HCC specimens, and the expression levels of c-Myc in SRC-1-positive HCC specimens were higher than in SRC-1-negative HCC specimens. In addition, SRC-1 and SRC-3 were co-overexpressed in 47.5% of HCC specimens, and they cooperated to promote HCC cell proliferation. Simultaneous down-regulation of SRC-1 and SRC-3 dramatically inhibited HCC cell proliferation. Our results demonstrate that SRC-1 promotes HCC progression by enhancing Wnt/β-catenin signaling and suggest that SRC-1 is a potential therapeutic molecular target for HCC.
Collapse
Affiliation(s)
- Zhangwei Tong
- From the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China, the Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Ming Li
- From the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China, the Department of Hepatobiliary Pancreas and Vessel Surgery, Chenggong Hospital of Xiamen University, Xiamen, Fujian 361003, China
| | - Wei Wang
- From the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Pingli Mo
- From the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Li Yu
- From the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Kun Liu
- From the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China, the Department of Pathology, Chenggong Hospital of Xiamen University, Xiamen, China
| | - Wenjing Ren
- From the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wengang Li
- the Department of Hepatobiliary Pancreas and Vessel Surgery, Chenggong Hospital of Xiamen University, Xiamen, Fujian 361003, China
| | - Hao Zhang
- the Cancer Research Center at Shantou University Medical College, Shantou, Guangdong 515041, China, and
| | - Jianming Xu
- the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Chundong Yu
- From the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China, the Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China,
| |
Collapse
|
125
|
Abstract
Approximately 70% of breast cancers are oestrogen receptor α (ER) positive, and are, therefore, treated with endocrine therapies. However, about 25% of patients with primary disease and almost all patients with metastases will present with or eventually develop endocrine resistance. Despite the magnitude of this clinical challenge, the mechanisms underlying the development of resistance remain largely unknown. In the past 2 years, several studies unveiled gain-of-function mutations in ESR1, the gene encoding the ER, in approximately 20% of patients with metastatic ER-positive disease who received endocrine therapies, such as tamoxifen and aromatase inhibitors. These mutations are clustered in a 'hotspot' within the ligand-binding domain (LBD) of the ER and lead to ligand-independent ER activity that promotes tumour growth, partial resistance to endocrine therapy, and potentially enhanced metastatic capacity; thus, ER LBD mutations might account for a mechanism of acquired endocrine resistance in a substantial fraction of patients with metastatic disease. In general, the absence of detectable ESR1 mutations in patients with treatment-naive disease, and the correlation between the frequency of patients with tumours harbouring these mutations and the number of endocrine treatments received suggest that, under selective treatment pressure, clonal expansion of rare mutant clones occurs, leading to resistance. Preclinical and clinical development of rationale-based novel therapeutic strategies that inhibit these ER mutants has the potential to substantially improve treatment outcomes. We discuss the contribution of ESR1 mutations to the development of acquired resistance to endocrine therapy, and evaluate how mutated ER can be detected and targeted to overcome resistance and improve patient outcomes.
Collapse
|
126
|
Eedunuri VK, Rajapakshe K, Fiskus W, Geng C, Chew SA, Foley C, Shah SS, Shou J, Mohamed JS, Coarfa C, O'Malley BW, Mitsiades N. miR-137 Targets p160 Steroid Receptor Coactivators SRC1, SRC2, and SRC3 and Inhibits Cell Proliferation. Mol Endocrinol 2015; 29:1170-83. [PMID: 26066330 DOI: 10.1210/me.2015-1080] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The p160 family of steroid receptor coactivators (SRCs) are pleiotropic transcription factor coactivators and "master regulators" of gene expression that promote cancer cell proliferation, survival, metabolism, migration, invasion, and metastasis. Cancers with high p160 SRC expression exhibit poor clinical outcomes and resistance to therapy, highlighting the SRCs as critical oncogenic drivers and, thus, therapeutic targets. microRNAs are important epigenetic regulators of protein expression. To examine the regulation of p160 SRCs by microRNAs, we used and combined 4 prediction algorithms to identify microRNAs that could target SRC1, SRC2, and SRC3 expression. For validation of these predictions, we assessed p160 SRC protein expression and cell viability after transfection of corresponding microRNA mimetics in breast cancer, uveal melanoma, and prostate cancer (PC) cell lines. Transfection of selected microRNA mimetics into breast cancer, uveal melanoma, and PC cells depleted SRC protein expression levels and exerted potent antiproliferative activity in these cell types. In particular, microRNA-137 (miR-137) depleted expression of SRC1, SRC2, and very potently, SRC3. The latter effect can be attributed to the presence of 3 miR-137 recognition sequences within the SRC3 3'-untranslated region. Using reverse phase protein array analysis, we identified a network of proteins, in addition to SRC3, that were modulated by miR-137 in PC cells. We also found that miR-137 and its host gene are epigenetically silenced in human cancer specimens and cell lines. These results support the development and testing of microRNA-based therapies (in particular based on restoring miR-137 levels) for targeting the oncogenic family of p160 SRCs in cancer.
Collapse
Affiliation(s)
- Vijay Kumar Eedunuri
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Kimal Rajapakshe
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Warren Fiskus
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Chuandong Geng
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Sue Anne Chew
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Christopher Foley
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Shrijal S Shah
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - John Shou
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Junaith S Mohamed
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Cristian Coarfa
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Bert W O'Malley
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Nicholas Mitsiades
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
127
|
Zeino M, Brenk R, Gruber L, Zehl M, Urban E, Kopp B, Efferth T. Cytotoxicity of cardiotonic steroids in sensitive and multidrug-resistant leukemia cells and the link with Na(+)/K(+)-ATPase. J Steroid Biochem Mol Biol 2015; 150:97-111. [PMID: 25797029 DOI: 10.1016/j.jsbmb.2015.03.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 03/04/2015] [Accepted: 03/17/2015] [Indexed: 12/12/2022]
Abstract
Cardiotonic steroids have long been in clinical use for treatment of heart failure and are now emerging as promising agents in various diseases, especially cancer. Their main target is Na(+)/K(+)-ATPase, a membrane protein involved in cellular ion homeostasis. Na(+)/K(+)-ATPase has been implicated in cancer biology by affecting several cellular events and signaling pathways in both sensitive and drug-resistant cancer cells. Hence, we investigated the cytotoxic activities of 66 cardiotonic steroids and cardiotonic steroid derivatives in sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000 leukemia cells. Data were then subjected to quantitative structure-activity relationship analysis (QSAR) and molecular docking into Na(+)/K(+)-ATPase, which both indicated a possible differential expression of the pump in the mentioned cell lines. This finding was confirmed by western blotting, intracellular potassium labeling and next generation sequencing which showed that Na(+)/K(+)-ATPase was less expressed in multidrug-resistant than in sensitive cells.
Collapse
Affiliation(s)
- Maen Zeino
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Ruth Brenk
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Lisa Gruber
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Martin Zehl
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, 1090 Vienna, Austria; Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Ernst Urban
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Brigitte Kopp
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
128
|
Calderón-Montaño JM, Burgos-Morón E, Orta ML, García-Domínguez I, Maldonado-Navas D, López-Lázaro M. Bufalin is a steroid receptor coactivator inhibitor-letter. Cancer Res 2015; 75:1156. [PMID: 25736686 DOI: 10.1158/0008-5472.can-14-0398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | | | - Manuel Luis Orta
- Department of Cell Biology, Faculty of Biology, University of Seville, Seville, Spain
| | | | | | - Miguel López-Lázaro
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| |
Collapse
|
129
|
Lonard DM, Xu J, O'Malley BW. Bufalin is a steroid receptor coactivator inhibitor-response. Cancer Res 2015; 75:1157. [PMID: 25736684 DOI: 10.1158/0008-5472.can-14-2648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
130
|
López-Lázaro M. Two preclinical tests to evaluate anticancer activity and to help validate drug candidates for clinical trials. Oncoscience 2015; 2:91-8. [PMID: 25859551 PMCID: PMC4381701 DOI: 10.18632/oncoscience.132] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/18/2015] [Indexed: 11/25/2022] Open
Abstract
Current approaches to assessing preclinical anticancer activity do not reliably predict drug efficacy in cancer patients. Most of the compounds that show remarkable anticancer effects in preclinical models actually fail when tested in clinical trials. We blame these failures on the complexity of the disease and on the limitations of the preclinical tools we require for our research. This manuscript argues that this lack of clinical response may also be caused by poor in vitro and in vivo preclinical designs, in which cancer patients' needs are not fully considered. Then, it proposes two patient-oriented tests to assess in vitro and in vivo anticancer activity and to help validate drug candidates for clinical evaluation.
Collapse
Affiliation(s)
- Miguel López-Lázaro
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Spain
| |
Collapse
|
131
|
NCOA3-mediated upregulation of mucin expression via transcriptional and post-translational changes during the development of pancreatic cancer. Oncogene 2014; 34:4879-89. [PMID: 25531332 DOI: 10.1038/onc.2014.409] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/12/2014] [Accepted: 10/19/2014] [Indexed: 01/28/2023]
Abstract
Pancreatic cancer (PC) is characterized by aberrant overexpression of mucins that contribute to its pathogenesis. Although the inflammatory cytokines contribute to mucin overexpression, the mucin profile of PC is markedly distinct from that of normal or inflamed pancreas. We postulated that de novo expression of various mucins in PC involves chromatin modifications. Analysis of chromatin modifying enzymes by PCR array identified differential expression of NCOA3 in MUC4-expressing PC cell lines. Immunohistochemistry analysis in tumor tissues from patients and spontaneous mouse models, and microarray analysis following the knockdown of NCOA3 were performed to elucidate its role in mucin regulation and overall impact on PC. Silencing of NCOA3 in PC cell lines resulted in significant downregulation of two most differentially expressed mucins in PC, MUC4 and MUC1 (P<0.01). Immunohistochemistry analysis in PC tissues and metastatic lesions established an association between NCOA3 and mucin (MUC1 and MUC4) expression. Spontaneous mouse model of PC (K-ras(G12D); Pdx-1cre) showed early expression of Ncoa3 during pre-neoplastic lesions. Mechanistically, NCOA3 knockdown abrogated retinoic acid-mediated MUC4 upregulation by restricting MUC4 promoter accessibility as demonstrated by micrococcus nuclease digestion (P<0.05) and chromatin immuno-precipitation analysis. NCOA3 also created pro-inflammatory conditions by upregulating chemokines like CXCL1, 2, 5 and CCL20 (P<0.001). AKT, ubiquitin C, ERK1/2 and NF-κB occupied dominant nodes in the networks significantly modulated after NCOA3 silencing. In addition, NCOA3 stabilized mucins post translationally through fucosylation by FUT8, as the knockdown of FUT8 resulted in the downregulation of MUC4 and MUC1 at protein levels.
Collapse
|
132
|
Dasgupta S, O'Malley BW. Transcriptional coregulators: emerging roles of SRC family of coactivators in disease pathology. J Mol Endocrinol 2014; 53:R47-59. [PMID: 25024406 PMCID: PMC4152414 DOI: 10.1530/jme-14-0080] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Transcriptional coactivators have evolved as an important new class of functional proteins that participate with virtually all transcription factors and nuclear receptors (NRs) to intricately regulate gene expression in response to a wide variety of environmental cues. Recent findings have highlighted that coactivators are important for almost all biological functions, and consequently, genetic defects can lead to severe pathologies. Drug discovery efforts targeting coactivators may prove valuable for treatment of a variety of diseases.
Collapse
Affiliation(s)
- Subhamoy Dasgupta
- Department of Molecular and Cellular BiologyBaylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular BiologyBaylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| |
Collapse
|
133
|
Geng C, Rajapakshe K, Shah SS, Shou J, Eedunuri VK, Foley C, Fiskus W, Rajendran M, Chew SA, Zimmermann M, Bond R, He B, Coarfa C, Mitsiades N. Androgen receptor is the key transcriptional mediator of the tumor suppressor SPOP in prostate cancer. Cancer Res 2014; 74:5631-43. [PMID: 25274033 PMCID: PMC4209379 DOI: 10.1158/0008-5472.can-14-0476] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Somatic missense mutations in the substrate-binding pocket of the E3 ubiquitin ligase adaptor SPOP are present in up to 15% of human prostate adenocarcinomas, but are rare in other malignancies, suggesting a prostate-specific mechanism of action. SPOP promotes ubiquitination and degradation of several protein substrates, including the androgen receptor (AR) coactivator SRC-3. However, the relative contributions that SPOP substrates may make to the pathophysiology of SPOP-mutant (mt) prostate adenocarcinomas are unknown. Using an unbiased bioinformatics approach, we determined that the gene expression profile of prostate adenocarcinoma cells engineered to express mt-SPOP overlaps greatly with the gene signature of both SRC-3 and AR transcriptional output, with a stronger similarity to AR than SRC-3. This finding suggests that in addition to its SRC-3-mediated effects, SPOP also exerts SRC-3-independent effects that are AR-mediated. Indeed, we found that wild-type (wt) but not prostate adenocarcinoma-associated mutants of SPOP promoted AR ubiquitination and degradation, acting directly through a SPOP-binding motif in the hinge region of AR. In support of these results, tumor xenografts composed of prostate adenocarcinoma cells expressing mt-SPOP exhibited higher AR protein levels and grew faster than tumors composed of prostate adenocarcinoma cells expressing wt-SPOP. Furthermore, genetic ablation of SPOP was sufficient to increase AR protein levels in mouse prostate. Examination of public human prostate adenocarcinoma datasets confirmed a strong link between transcriptomic profiles of mt-SPOP and AR. Overall, our studies highlight the AR axis as the key transcriptional output of SPOP in prostate adenocarcinoma and provide an explanation for the prostate-specific tumor suppressor role of wt-SPOP.
Collapse
Affiliation(s)
- Chuandong Geng
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Shrijal S Shah
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - John Shou
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | | | - Christopher Foley
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Warren Fiskus
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Mahitha Rajendran
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Sue Anne Chew
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Martin Zimmermann
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Richard Bond
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Bin He
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Nicholas Mitsiades
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas. Center for Drug Discovery, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
134
|
Mo P, Zhou Q, Guan L, Wang Y, Wang W, Miao M, Tong Z, Li M, Majaz S, Liu Y, Su G, Xu J, Yu C. Amplified in breast cancer 1 promotes colorectal cancer progression through enhancing notch signaling. Oncogene 2014; 34:3935-3945. [PMID: 25263446 PMCID: PMC4377317 DOI: 10.1038/onc.2014.324] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 08/01/2014] [Accepted: 08/25/2014] [Indexed: 12/21/2022]
Abstract
Aberrant activation of Notch signaling has an essential role in colorectal cancer (CRC) progression. Amplified in breast cancer 1 (AIB1), also known as steroid receptor coactivator 3 or NCOA3, is a transcriptional coactivator that promotes cancer cell proliferation and invasiveness. However, AIB1 implication in CRC progression through enhancing Notch signaling is unknown. In this study, we found that several CRC cell lines expressed high levels of AIB1, and knockdown of AIB1 decreased cell proliferation, colony formation and tumorigenesis of these CRC cells. Specifically, knockdown of AIB1 inhibited cell cycle progression at G1 phase by decreasing the mRNA levels of cyclin A2, cyclin B1, cyclin E2 and hairy and enhancer of split (Hes) 1. Furthermore, AIB1 interacted with Notch intracellular domain and Mastermind-like 1 and was recruited to the Hes1 promoter to enhance Notch signaling. Downregulation of AIB1 also decreased CRC cell invasiveness in vitro and lung metastasis in vivo. Besides that, knockout of AIB1 in mice inhibited colon carcinogenesis induced by azoxymethane/dextran sodium sulfate treatment. The mRNA levels of cyclin B1 and Hes5 were downregulated, but p27, ATOH1 and MUC2 were upregulated in the colon tumors from AIB1-deficient mice compared with those from wild-type mice. Thus, our results signify the importance of AIB1 in CRC and demonstrate that AIB1 promotes CRC progression at least in part through enhancing Notch signaling, suggesting that AIB1 is a potential molecular target for CRC treatment.
Collapse
Affiliation(s)
- Pingli Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qiling Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Lei Guan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yi Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Wei Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Mengmeng Miao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zhangwei Tong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Ming Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Sidra Majaz
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yonghong Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Guoqiang Su
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
135
|
Stashi E, York B, O'Malley BW. Steroid receptor coactivators: servants and masters for control of systems metabolism. Trends Endocrinol Metab 2014; 25:337-47. [PMID: 24953190 PMCID: PMC4108168 DOI: 10.1016/j.tem.2014.05.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 11/30/2022]
Abstract
Coregulator recruitment to nuclear receptors (NRs) and other transcription factors is essential for proper metabolic gene regulation, with coactivators enhancing and corepressors attenuating gene transcription. The steroid receptor coactivator (SRC) family is composed of three homologous members (SRC-1, SRC-2, and SRC-3), which are uniquely important for mediating steroid hormone and mitogenic actions. An accumulating body of work highlights the diverse array of metabolic functions regulated by the SRCs, including systemic metabolite homeostasis, inflammation, and energy regulation. We discuss here the cooperative and unique functions among the SRCs to provide a comprehensive atlas of systemic SRC metabolic regulation. Deciphering the fractional and synergistic contributions of the SRCs to metabolic homeostasis is crucial to understanding fully the networks underlying metabolic transcriptional regulation.
Collapse
Affiliation(s)
- Erin Stashi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
136
|
Evaluating the cancer therapeutic potential of cardiac glycosides. BIOMED RESEARCH INTERNATIONAL 2014; 2014:794930. [PMID: 24895612 PMCID: PMC4033509 DOI: 10.1155/2014/794930] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 11/23/2022]
Abstract
Cardiac glycosides, also known as cardiotonic steroids, are a group of natural products that share a steroid-like structure with an unsaturated lactone ring and the ability to induce cardiotonic effects mediated by a selective inhibition of the Na+/K+-ATPase. Cardiac glycosides have been used for many years in the treatment of cardiac congestion and some types of cardiac arrhythmias. Recent data suggest that cardiac glycosides may also be useful in the treatment of cancer. These compounds typically inhibit cancer cell proliferation at nanomolar concentrations, and recent high-throughput screenings of drug libraries have therefore identified cardiac glycosides as potent inhibitors of cancer cell growth. Cardiac glycosides can also block tumor growth in rodent models, which further supports the idea that they have potential for cancer therapy. Evidence also suggests, however, that cardiac glycosides may not inhibit cancer cell proliferation selectively and the potent inhibition of tumor growth induced by cardiac glycosides in mice xenografted with human cancer cells is probably an experimental artifact caused by their ability to selectively kill human cells versus rodent cells. This paper reviews such evidence and discusses experimental approaches that could be used to reveal the cancer therapeutic potential of cardiac glycosides in preclinical studies.
Collapse
|
137
|
Yang R, Bai Y, Qin Z, Yu T. EgoNet: identification of human disease ego-network modules. BMC Genomics 2014; 15:314. [PMID: 24773628 PMCID: PMC4234496 DOI: 10.1186/1471-2164-15-314] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/16/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Mining novel biomarkers from gene expression profiles for accurate disease classification is challenging due to small sample size and high noise in gene expression measurements. Several studies have proposed integrated analyses of microarray data and protein-protein interaction (PPI) networks to find diagnostic subnetwork markers. However, the neighborhood relationship among network member genes has not been fully considered by those methods, leaving many potential gene markers unidentified. The main idea of this study is to take full advantage of the biological observation that genes associated with the same or similar diseases commonly reside in the same neighborhood of molecular networks. RESULTS We present EgoNet, a novel method based on egocentric network-analysis techniques, to exhaustively search and prioritize disease subnetworks and gene markers from a large-scale biological network. When applied to a triple-negative breast cancer (TNBC) microarray dataset, the top selected modules contain both known gene markers in TNBC and novel candidates, such as RAD51 and DOK1, which play a central role in their respective ego-networks by connecting many differentially expressed genes. CONCLUSIONS Our results suggest that EgoNet, which is based on the ego network concept, allows the identification of novel biomarkers and provides a deeper understanding of their roles in complex diseases.
Collapse
Affiliation(s)
| | | | | | - Tianwei Yu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, 1518 Clifton Rd, N,E, Atlanta, GA, USA.
| |
Collapse
|
138
|
Yan F, Yu Y, Chow DC, Palzkill T, Madoux F, Hodder P, Chase P, Griffin PR, O'Malley BW, Lonard DM. Identification of verrucarin a as a potent and selective steroid receptor coactivator-3 small molecule inhibitor. PLoS One 2014; 9:e95243. [PMID: 24743578 PMCID: PMC3990629 DOI: 10.1371/journal.pone.0095243] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/24/2014] [Indexed: 12/22/2022] Open
Abstract
Members of the steroid receptor coactivator (SRC) family are overexpressed in numerous types of cancers. In particular, steroid receptor coactivator 3 (SRC-3) has been recognized as a critical coactivator associated with tumor initiation, progression, recurrence, metastasis, and chemoresistance where it interacts with multiple nuclear receptors and other transcription factors to enhance their transcriptional activities and facilitate cross-talk between pathways that stimulate cancer progression. Because of its central role as an integrator of growth signaling pathways, development of small molecule inhibitors (SMIs) against SRCs have the potential to simultaneously disrupt multiple signal transduction networks and transcription factors involved in tumor progression. Here, high-throughput screening was performed to identify compounds able to inhibit the intrinsic transcriptional activities of the three members of the SRC family. Verrucarin A was identified as a SMI that can selectively promote the degradation of the SRC-3 protein, while affecting SRC-1 and SRC-2 to a lesser extent and having no impact on CARM-1 and p300 protein levels. Verrucarin A was cytotoxic toward multiple types of cancer cells at low nanomolar concentrations, but not toward normal liver cells. Moreover, verrucarin A was able to inhibit expression of the SRC-3 target genes MMP2 and MMP13 and attenuated cancer cell migration. We found that verrucarin A effectively sensitized cancer cells to treatment with other anti-cancer drugs. Binding studies revealed that verrucarin A does not bind directly to SRC-3, suggesting that it inhibits SRC-3 through its interaction with an upstream effector. In conclusion, unlike other SRC SMIs characterized by our laboratory that directly bind to SRCs, verrucarin A is a potent and selective SMI that blocks SRC-3 function through an indirect mechanism.
Collapse
Affiliation(s)
- Fei Yan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Yang Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Dar-Chone Chow
- Department of Pharmacology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Timothy Palzkill
- Department of Pharmacology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Franck Madoux
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Peter Hodder
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Peter Chase
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Patrick R. Griffin
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Bert W. O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - David M. Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|