1
|
Zhao S, Ali AS, Kong X, Zhang Y, Liu X, Skidmore MA, Forsyth CM, Savage GP, Wu D, Xu Y, Francis CL. 1-Benzyloxy-5-phenyltetrazole derivatives highly active against androgen receptor-dependent prostate cancer cells. Eur J Med Chem 2023; 246:114982. [PMID: 36495632 DOI: 10.1016/j.ejmech.2022.114982] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/04/2022]
Abstract
A series of 1-benzyloxy-5-phenyltetrazole derivatives and similar compounds were synthesized and evaluated for their in vitro inhibitory activity against androgen-receptor-dependent (22Rv1) and androgen-receptor independent (PC3) prostate cancer cells. The most active compounds had in vitro IC50 values against 22Rv1 cells of <50 nM and showed apparent selectivity for this cell type over PC3 cells; however, these active compounds had short half-lives when incubated with mouse liver microsomes and/or when plasma concentration was monitored during in vivo pharmacokinetic studies in mice or rats. Importantly, lead compound 1 exhibited promising inhibitory effects on cell proliferation, expression of AR and its splicing variant AR-v7 as well as AR regulated target genes in 22Rv1 cells, which are so called castration-resistant prostate cancer (CRPC) cells, and a 22Rv1 CRPC xenograft tumour model in mice. Structural changes which omitted the N-O-benzyl moiety led to dramatic or total loss of activity and S-benzylation of a cysteine derivative, as a surrogate for in vivo S-nucleophiles, by representative highly active compounds, suggested a possible chemical reactivity basis for this "activity cliff" and poor pharmacokinetic profile. However, representative highly active compounds did not inhibit a cysteine protease, indicating that the mode of activity is unlikely to be protein modification by S-benzylation. Despite our efforts to elucidate the mode of action, the mechanism remains unclear.
Collapse
Affiliation(s)
- Shiting Zhao
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; Guangzhou Medical University, Guangzhou, 511436, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Abdelsalam S Ali
- Drug Discovery Chemistry Team, CSIRO, Clayton, VIC, 3168, Australia
| | - Xinyu Kong
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan Zhang
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaomin Liu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | | | - Craig M Forsyth
- School of Chemistry, Monash University, Clayton, VIC, 3800, Australia
| | - G Paul Savage
- Drug Discovery Chemistry Team, CSIRO, Clayton, VIC, 3168, Australia
| | - Donghai Wu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; Guangzhou Medical University, Guangzhou, 511436, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yong Xu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; Guangzhou Medical University, Guangzhou, 511436, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Craig L Francis
- Drug Discovery Chemistry Team, CSIRO, Clayton, VIC, 3168, Australia.
| |
Collapse
|
2
|
Yang Y, Sun X, Cui W, Liu N, Wang K, Qu L, Pan C. The detection of mutation within goat cell division cycle 25 A and its effect on kidding number. Anim Biotechnol 2022; 33:1504-1509. [PMID: 33879023 DOI: 10.1080/10495398.2021.1910519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cell division cycle 25 A (CDC25A) accounts for an essential function on early folliculogenesis of female mammals, especially regulating the function of intra-ovarian, thus this gene is pinpointed as a candidate gene that influences the kidding number of goat. On this ground, the purpose of this study was to investigate whether the reported 20-nt nucleotide variants locus (rs639467625) of the CDC25A gene influences kidding number in Shaanbei white cashmere goat (SBWC). The χ2-test showed that there were more ID genotypes in mothers of multiple lambs than in mothers of single lambs. Interestingly, this indel locus was related to the first-born kidding number in the group of SBWC goats (p < 0.05). Similarly, the result of the t-test was consistent with the result of the χ2-test, showed the kidding number of ID genotype individuals was large than that of II individuals (p < 0.05). These findings proved that the different genotypes of CDC25A have impacts on goat kidding numbers. Thus, the results led us to speculate that the ID genotype of CDC25A was one of the main indel influencing goat kidding numbers. Simultaneously, this study was expected to provide useful DNA markers for superior individuals selection by marker-assisted selection (MAS) and make a contribution to goats breeding.
Collapse
Affiliation(s)
- Yuta Yang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China.,College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaomei Sun
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Wenbo Cui
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Nuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Ke Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Lei Qu
- College of Life Sciences, Yulin University, Yulin, Shaanxi, PR China.,Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Life Science Research Center, Yulin University, Yulin, Shaanxi, China
| | - Chuanying Pan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
3
|
Venkadakrishnan VB, Ben-Salem S, Heemers HV. AR-dependent phosphorylation and phospho-proteome targets in prostate cancer. Endocr Relat Cancer 2020; 27:R193-R210. [PMID: 32276264 PMCID: PMC7583603 DOI: 10.1530/erc-20-0048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
Prostate cancer (CaP) is the second leading cause of cancer-related deaths in Western men. Because androgens drive CaP by activating the androgen receptor (AR), blocking AR's ligand activation, known as androgen deprivation therapy (ADT), is the default treatment for metastatic CaP. Despite an initial remission, CaP eventually develops resistance to ADT and progresses to castration-recurrent CaP (CRPC). CRPC continues to rely on aberrantly activated AR that is no longer inhibited effectively by available therapeutics. Interference with signaling pathways downstream of activated AR that mediate aggressive CRPC behavior may lead to alternative CaP treatments. Developing such therapeutic strategies requires a thorough mechanistic understanding of the most clinically relevant and druggable AR-dependent signaling events. Recent proteomics analyses of CRPC clinical specimens indicate a shift in the phosphoproteome during CaP progression. Kinases and phosphatases represent druggable entities, for which clinically tested inhibitors are available, some of which are incorporated already in treatment plans for other human malignancies. Here, we reviewed the AR-associated transcriptome and translational regulon, and AR interactome involved in CaP phosphorylation events. Novel and for the most part mutually exclusive AR-dependent transcriptional and post-transcriptional control over kinase and phosphatase expression was found, with yet other phospho-regulators interacting with AR. The multiple mechanisms by which AR can shape and fine-tune the CaP phosphoproteome were reflected in diverse aspects of CaP biology such as cell cycle progression and cell migration. Furthermore, we examined the potential, limitations and challenges of interfering with AR-mediated phosphorylation events as alternative strategy to block AR function during CaP progression.
Collapse
Affiliation(s)
- Varadha Balaji Venkadakrishnan
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio, USA
| | - Salma Ben-Salem
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
| | | |
Collapse
|
4
|
Holmes TR, Dindu S, Hansen LA. Aberrant localization of signaling proteins in skin cancer: Implications for treatment. Mol Carcinog 2019; 58:1631-1639. [PMID: 31062427 DOI: 10.1002/mc.23036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 01/01/2023]
Abstract
Aberrant subcellular localization of signaling proteins can provide cancer cells with advantages such as resistance to apoptotic cell death, increased invasiveness and more rapid proliferation. Nuclear to cytoplasmic shifts in tumor-promoting proteins can lead to worse patient outcomes, providing opportunities to target cancer-specific processes. Herein, we review the significance of dysregulated protein localization with a focus on skin cancer. Altered localization of signaling proteins controlling cell cycle progression or cell death is a common feature of cancer. In some instances, aberrant subcellular localization results in an acquired prosurvival function. Taking advantage of this knowledge reveals novel targets useful in the development of cancer therapeutics.
Collapse
Affiliation(s)
- Thomas R Holmes
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Shravya Dindu
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Laura A Hansen
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| |
Collapse
|
5
|
Nickkholgh B, Sittadjody S, Ordonez K, Rothberg MB, Balaji KC. Protein kinase D1 induces G1-phase cell-cycle arrest independent of Checkpoint kinases by phosphorylating Cell Division Cycle Phosphatase 25. Prostate 2019; 79:1053-1058. [PMID: 30958903 PMCID: PMC8514115 DOI: 10.1002/pros.23807] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 01/12/2023]
Abstract
Protein Kinase D1 (PrKD1) functions as a tumor and metastasis suppressor in several human cancers by influencing cell-cycle progression. However, the exact mechanism of cell-cycle regulation by PrKD1 is unclear. Overexpression and ectopic expression of PrKD1 induces G1 arrest in cancer cell lines. Because checkpoint kinases (CHEKs) are known to play a role in progression through the G1 phase, we downregulated CHEK1, which did not overcome the G1 arrest induced by PrKD1. Using in vitro phosphorylation and Western blot assays, we showed that PrKD1 phosphorylates all CDC25 isoforms (known substrates of CHEK kinases), independent from CHEK kinases, suggesting that direct phosphorylation of CDC25 by PrKD1 may be an alternate mechanism of G1 arrest. The study has identified a molecular mechanism for the influence of PrKD1 in cell-cycle progression.
Collapse
Affiliation(s)
- Bita Nickkholgh
- Wake Forest Institute for Regenerative medicine (WFRM), Wake Forest University School of Medicine, Winston-Salem, North Carolina
- Department of Physiology-Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Sivanandane Sittadjody
- Wake Forest Institute for Regenerative medicine (WFRM), Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Karina Ordonez
- Department of Biological Sciences, Winston-Salem State University, Winston-Salem, North Carolina
| | | | - K C Balaji
- Wake Forest Institute for Regenerative medicine (WFRM), Wake Forest University School of Medicine, Winston-Salem, North Carolina
- Department of Urology, University of Florida, Jacksonville, Florida
| |
Collapse
|
6
|
Accumulation of cytoplasmic CDC25A in cutaneous squamous cell carcinoma leads to a dependency on CDC25A for cancer cell survival and tumor growth. Cancer Lett 2017; 410:41-49. [DOI: 10.1016/j.canlet.2017.09.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 01/29/2023]
|
7
|
Lee E, Decker AM, Cackowski FC, Kana LA, Yumoto K, Jung Y, Wang J, Buttitta L, Morgan TM, Taichman RS. Growth Arrest-Specific 6 (GAS6) Promotes Prostate Cancer Survival by G 1 Arrest/S Phase Delay and Inhibition of Apoptosis During Chemotherapy in Bone Marrow. J Cell Biochem 2016; 117:2815-2824. [PMID: 27153245 PMCID: PMC5223280 DOI: 10.1002/jcb.25582] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 05/02/2016] [Indexed: 12/11/2022]
Abstract
Prostate cancer (PCa) is known to develop resistance to chemotherapy. Growth arrest-specific 6 (GAS6), plays a role in tumor progression by regulating growth in many cancers. Here, we explored how GAS6 regulates the cell cycle and apoptosis of PCa cells in response to chemotherapy. We found that GAS6 is sufficient to significantly increase the fraction of cells in G1 and the duration of phase in PCa cells. Importantly, the effect of GAS6 on G1 is potentiated during docetaxel chemotherapy. GAS6 altered the levels of several key cell cycle regulators, including the downregulation of Cyclin B1 (G2 /M phase), CDC25A, Cyclin E1, and CDK2 (S phase entry), while the upregulation of cell cycle inhibitors p27 and p21, Cyclin D1, and CDK4. Importantly, these changes became further accentuated during docetaxel treatment in the presence of GAS6. Moreover, GAS6 alters the apoptotic response of PCa cells during docetaxel chemotherapy. Docetaxel induced PCa cell apoptosis is efficiently suppressed in PCa cell culture in the presence of GAS6 or GAS6 secreted from co-cultured osteoblasts. Similarly, the GAS6-expressing bone environment protects PCa cells from apoptosis within primary tumors in vivo studies. Docetaxel induced significant levels of Caspase-3 and PARP cleavage in PCa cells, while GAS6 protected PCa cells from docetaxel-induced apoptotic signaling. Together, these data suggest that GAS6, expressed by osteoblasts in the bone marrow, plays a significant role in the regulation of PCa cell survival during chemotherapy, which will have important implications for targeting metastatic disease. J. Cell. Biochem. 117: 2815-2824, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eunsohl Lee
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Ann M Decker
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Frank C Cackowski
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, 48109, Michigan
| | - Lulia A Kana
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Kenji Yumoto
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Younghun Jung
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Jingcheng Wang
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan
| | - Laura Buttitta
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, 48109, Michigan
| | - Todd M Morgan
- Department of Urology, University of Michigan School of Medicine, Ann Arbor, 48109, Michigan
| | - Russell S Taichman
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, 48109, Michigan.
| |
Collapse
|
8
|
Novío S, Cartea ME, Soengas P, Freire-Garabal M, Núñez-Iglesias MJ. Effects of Brassicaceae Isothiocyanates on Prostate Cancer. Molecules 2016; 21:E626. [PMID: 27187332 PMCID: PMC6272898 DOI: 10.3390/molecules21050626] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/13/2016] [Accepted: 05/03/2016] [Indexed: 12/21/2022] Open
Abstract
Despite the major progress made in the field of cancer biology, cancer is still one of the leading causes of mortality, and prostate cancer (PCa) is one of the most encountered malignancies among men. The effective management of this disease requires developing better anticancer agents with greater efficacy and fewer side effects. Nature is a large source for the development of chemotherapeutic agents, with more than 50% of current anticancer drugs being of natural origin. Isothiocyanates (ITCs) are degradation products from glucosinolates that are present in members of the family Brassicaceae. Although they are known for a variety of therapeutic effects, including antioxidant, immunostimulatory, anti-inflammatory, antiviral and antibacterial properties, nowadays, cell line and animal studies have additionally indicated the chemopreventive action without causing toxic side effects of ITCs. In this way, they can induce cell cycle arrest, activate apoptosis pathways, increase the sensitivity of resistant PCa to available chemodrugs, modulate epigenetic changes and downregulate activated signaling pathways, resulting in the inhibition of cell proliferation, progression and invasion-metastasis. The present review summarizes the chemopreventive role of ITCs with a particular emphasis on specific molecular targets and epigenetic alterations in in vitro and in vivo cancer animal models.
Collapse
Affiliation(s)
- Silvia Novío
- Lennart Levi Stress and Neuroimmunology Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, 15782 Santiago de Compostela, A Coruña, Spain.
| | - María Elena Cartea
- Group of Genetics, Breeding and Biochemistry of Brassicas, Misión Biológica de Galicia (CSIC) Aptdo. 28, 36080 Pontevedra, Spain.
| | - Pilar Soengas
- Group of Genetics, Breeding and Biochemistry of Brassicas, Misión Biológica de Galicia (CSIC) Aptdo. 28, 36080 Pontevedra, Spain.
| | - Manuel Freire-Garabal
- Lennart Levi Stress and Neuroimmunology Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, 15782 Santiago de Compostela, A Coruña, Spain.
| | - María Jesús Núñez-Iglesias
- Lennart Levi Stress and Neuroimmunology Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, 15782 Santiago de Compostela, A Coruña, Spain.
| |
Collapse
|
9
|
Gabrielli B, Burgess A. Cdc25 Family Phosphatases in Cancer. PROTEIN TYROSINE PHOSPHATASES IN CANCER 2016:283-306. [DOI: 10.1007/978-1-4939-3649-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
Lu X, Sun W, Tang Y, Zhu L, Li Y, Ou C, Yang C, Su J, Luo C, Hu Y, Cao J. Identification of key genes in hepatocellular carcinoma and validation of the candidate gene, cdc25a, using gene set enrichment analysis, meta-analysis and cross-species comparison. Mol Med Rep 2015; 13:1172-8. [PMID: 26647881 PMCID: PMC4732839 DOI: 10.3892/mmr.2015.4646] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 10/26/2015] [Indexed: 12/31/2022] Open
Abstract
The aim of the present study was to determine key pathways and genes involved in the pathogenesis of hepatocellular carcinoma (HCC) through bioinformatic analyses of HCC microarray data based on cross-species comparison. Microarray data of gene expression in HCC in different species were analyzed using gene set enrichment analysis (GSEA) and meta-analysis. Reverse transcription-quantitative polymerase chain reaction and western blotting were performed to determine the mRNA and protein expression levels of cdc25a, one of the identified candidate genes, in human, rat and tree shrew samples. The cell cycle pathway had the largest overlap between the GSEA and meta-analysis. Meta-analyses showed that 25 genes, including cdc25a, in the cell cycle pathway were differentially expressed. Cdc25a mRNA levels in HCC tissues were higher than those in normal liver tissues in humans, rats and tree shrews, and the expression level of cdc25a in HCC tissues was higher than in corresponding paraneoplastic tissues in humans and rats. In human HCC tissues, the cdc25a mRNA level was significantly correlated with clinical stage, portal vein tumor thrombosis and extrahepatic metastasis. Western blotting showed that, cdc25a protein levels were significantly upregulated in HCC tissues in humans, rats and tree shrews. In conclusion, GSEA and meta-analysis can be combined to identify key molecules and pathways involved in HCC. This study demonstrated that the cell cycle pathway and the cdc25a gene may be crucial in the pathogenesis and progression of HCC.
Collapse
Affiliation(s)
- Xiaoxu Lu
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wen Sun
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yanping Tang
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Lingqun Zhu
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yuan Li
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chao Ou
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chun Yang
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jianjia Su
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chengpiao Luo
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yanling Hu
- The Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Ji Cao
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
11
|
Ta HQ, Ivey ML, Frierson HF, Conaway MR, Dziegielewski J, Larner JM, Gioeli D. Checkpoint Kinase 2 Negatively Regulates Androgen Sensitivity and Prostate Cancer Cell Growth. Cancer Res 2015; 75:5093-105. [PMID: 26573794 DOI: 10.1158/0008-5472.can-15-0224] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 09/12/2015] [Indexed: 12/18/2022]
Abstract
Prostate cancer is the second leading cause of cancer death in American men, and curing metastatic disease remains a significant challenge. Nearly all patients with disseminated prostate cancer initially respond to androgen deprivation therapy (ADT), but virtually all patients will relapse and develop incurable castration-resistant prostate cancer (CRPC). A high-throughput RNAi screen to identify signaling pathways regulating prostate cancer cell growth led to our discovery that checkpoint kinase 2 (CHK2) knockdown dramatically increased prostate cancer growth and hypersensitized cells to low androgen levels. Mechanistic investigations revealed that the effects of CHK2 were dependent on the downstream signaling proteins CDC25C and CDK1. Moreover, CHK2 depletion increased androgen receptor (AR) transcriptional activity on androgen-regulated genes, substantiating the finding that CHK2 affects prostate cancer proliferation, partly, through the AR. Remarkably, we further show that CHK2 is a novel AR-repressed gene, suggestive of a negative feedback loop between CHK2 and AR. In addition, we provide evidence that CHK2 physically associates with the AR and that cell-cycle inhibition increased this association. Finally, IHC analysis of CHK2 in prostate cancer patient samples demonstrated a decrease in CHK2 expression in high-grade tumors. In conclusion, we propose that CHK2 is a negative regulator of androgen sensitivity and prostate cancer growth, and that CHK2 signaling is lost during prostate cancer progression to castration resistance. Thus, perturbing CHK2 signaling may offer a new therapeutic approach for sensitizing CRPC to ADT and radiation.
Collapse
Affiliation(s)
- Huy Q Ta
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Melissa L Ivey
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Henry F Frierson
- Department of Pathology, University of Virginia Health System, Charlottesville, Virginia. Cancer Center Member, University of Virginia, Charlottesville, Virginia
| | - Mark R Conaway
- Cancer Center Member, University of Virginia, Charlottesville, Virginia. Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia
| | - Jaroslaw Dziegielewski
- Cancer Center Member, University of Virginia, Charlottesville, Virginia. Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia
| | - James M Larner
- Cancer Center Member, University of Virginia, Charlottesville, Virginia. Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia
| | - Daniel Gioeli
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia. Cancer Center Member, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
12
|
Wyatt AW, Mo F, Wang K, McConeghy B, Brahmbhatt S, Jong L, Mitchell DM, Johnston RL, Haegert A, Li E, Liew J, Yeung J, Shrestha R, Lapuk AV, McPherson A, Shukin R, Bell RH, Anderson S, Bishop J, Hurtado-Coll A, Xiao H, Chinnaiyan AM, Mehra R, Lin D, Wang Y, Fazli L, Gleave ME, Volik SV, Collins CC. Heterogeneity in the inter-tumor transcriptome of high risk prostate cancer. Genome Biol 2014; 15:426. [PMID: 25155515 PMCID: PMC4169643 DOI: 10.1186/s13059-014-0426-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 07/28/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Genomic analyses of hundreds of prostate tumors have defined a diverse landscape of mutations and genome rearrangements, but the transcriptomic effect of this complexity is less well understood, particularly at the individual tumor level. We selected a cohort of 25 high-risk prostate tumors, representing the lethal phenotype, and applied deep RNA-sequencing and matched whole genome sequencing, followed by detailed molecular characterization. RESULTS Ten tumors were exposed to neo-adjuvant hormone therapy and expressed marked evidence of therapy response in all except one extreme case, which demonstrated early resistance via apparent neuroendocrine transdifferentiation. We observe high inter-tumor heterogeneity, including unique sets of outlier transcripts in each tumor. Interestingly, outlier expression converged on druggable cellular pathways associated with cell cycle progression, translational control or immune regulation, suggesting distinct contemporary pathway affinity and a mechanism of tumor stratification. We characterize hundreds of novel fusion transcripts, including a high frequency of ETS fusions associated with complex genome rearrangements and the disruption of tumor suppressors. Remarkably, several tumors express unique but potentially-oncogenic non-ETS fusions, which may contribute to the phenotype of individual tumors, and have significance for disease progression. Finally, one ETS-negative tumor has a striking tandem duplication genotype which appears to be highly aggressive and present at low recurrence in ETS-negative prostate cancer, suggestive of a novel molecular subtype. CONCLUSIONS The multitude of rare genomic and transcriptomic events detected in a high-risk tumor cohort offer novel opportunities for personalized oncology and their convergence on key pathways and functions has broad implications for precision medicine.
Collapse
Affiliation(s)
- Alexander W Wyatt
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Fan Mo
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Kendric Wang
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Brian McConeghy
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Sonal Brahmbhatt
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Lina Jong
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Devon M Mitchell
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Rebecca L Johnston
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Anne Haegert
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Estelle Li
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Janet Liew
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Jake Yeung
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Raunak Shrestha
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Anna V Lapuk
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Andrew McPherson
- />Bioinformatics Training Program, University of British Columbia, Vancouver, BC Canada
| | - Robert Shukin
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Robert H Bell
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Shawn Anderson
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Jennifer Bishop
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Antonio Hurtado-Coll
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Hong Xiao
- />Michigan Center for Translational Pathology, Ann Arbor, Michigan USA
| | - Arul M Chinnaiyan
- />Michigan Center for Translational Pathology, Ann Arbor, Michigan USA
| | - Rohit Mehra
- />Michigan Center for Translational Pathology, Ann Arbor, Michigan USA
| | - Dong Lin
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
- />Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC Canada
| | - Yuzhuo Wang
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
- />Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC Canada
| | - Ladan Fazli
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Martin E Gleave
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Stanislav V Volik
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Colin C Collins
- />Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| |
Collapse
|
13
|
Stebbing J, Lit LC, Zhang H, Darrington RS, Melaiu O, Rudraraju B, Giamas G. The regulatory roles of phosphatases in cancer. Oncogene 2014; 33:939-53. [PMID: 23503460 DOI: 10.1038/onc.2013.80] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 02/01/2013] [Indexed: 02/06/2023]
Abstract
The relevance of potentially reversible post-translational modifications required for controlling cellular processes in cancer is one of the most thriving arenas of cellular and molecular biology. Any alteration in the balanced equilibrium between kinases and phosphatases may result in development and progression of various diseases, including different types of cancer, though phosphatases are relatively under-studied. Loss of phosphatases such as PTEN (phosphatase and tensin homologue deleted on chromosome 10), a known tumour suppressor, across tumour types lends credence to the development of phosphatidylinositol 3-kinase inhibitors alongside the use of phosphatase expression as a biomarker, though phase 3 trial data are lacking. In this review, we give an updated report on phosphatase dysregulation linked to organ-specific malignancies.
Collapse
Affiliation(s)
- J Stebbing
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - L C Lit
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - H Zhang
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - R S Darrington
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - O Melaiu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - B Rudraraju
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - G Giamas
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
14
|
Imberg-Kazdan K, Ha S, Greenfield A, Poultney CS, Bonneau R, Logan SK, Garabedian MJ. A genome-wide RNA interference screen identifies new regulators of androgen receptor function in prostate cancer cells. Genome Res 2013; 23:581-91. [PMID: 23403032 PMCID: PMC3613576 DOI: 10.1101/gr.144774.112] [Citation(s) in RCA: 33] [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: 06/18/2012] [Accepted: 01/31/2013] [Indexed: 01/22/2023]
Abstract
The androgen receptor (AR) is a mediator of both androgen-dependent and castration-resistant prostate cancers. Identification of cellular factors affecting AR transcriptional activity could in principle yield new targets that reduce AR activity and combat prostate cancer, yet a comprehensive analysis of the genes required for AR-dependent transcriptional activity has not been determined. Using an unbiased genetic approach that takes advantage of the evolutionary conservation of AR signaling, we have conducted a genome-wide RNAi screen in Drosophila cells for genes required for AR transcriptional activity and applied the results to human prostate cancer cells. We identified 45 AR-regulators, which include known pathway components and genes with functions not previously linked to AR regulation, such as HIPK2 (a protein kinase) and MED19 (a subunit of the Mediator complex). Depletion of HIPK2 and MED19 in human prostate cancer cells decreased AR target gene expression and, importantly, reduced the proliferation of androgen-dependent and castration-resistant prostate cancer cells. We also systematically analyzed additional Mediator subunits and uncovered a small subset of Mediator subunits that interpret AR signaling and affect AR-dependent transcription and prostate cancer cell proliferation. Importantly, targeting of HIPK2 by an FDA-approved kinase inhibitor phenocopied the effect of depletion by RNAi and reduced the growth of AR-positive, but not AR-negative, treatment-resistant prostate cancer cells. Thus, our screen has yielded new AR regulators including drugable targets that reduce the proliferation of castration-resistant prostate cancer cells.
Collapse
Affiliation(s)
- Keren Imberg-Kazdan
- Department of Biochemistry and Department of Molecular Pharmacology, New York University School of Medicine, New York, New York 10016, USA
| | - Susan Ha
- Department of Biochemistry and Department of Molecular Pharmacology, New York University School of Medicine, New York, New York 10016, USA
- Department of Urology, New York University School of Medicine, New York, New York 10016, USA
| | - Alex Greenfield
- Center for Genomics and Systems Biology, New York University, New York, New York 10003, USA
| | | | - Richard Bonneau
- Center for Genomics and Systems Biology, New York University, New York, New York 10003, USA
| | - Susan K. Logan
- Department of Biochemistry and Department of Molecular Pharmacology, New York University School of Medicine, New York, New York 10016, USA
- Department of Urology, New York University School of Medicine, New York, New York 10016, USA
- NYU Cancer Institute, New York University School of Medicine, New York, New York 10016, USA
| | - Michael J. Garabedian
- Department of Urology, New York University School of Medicine, New York, New York 10016, USA
- Center for Genomics and Systems Biology, New York University, New York, New York 10003, USA
- NYU Cancer Institute, New York University School of Medicine, New York, New York 10016, USA
- Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA
| |
Collapse
|
15
|
Chiu YT, Liu J, Tang K, Wong YC, Khanna KK, Ling MT. Inactivation of ATM/ATR DNA damage checkpoint promotes androgen induced chromosomal instability in prostate epithelial cells. PLoS One 2012; 7:e51108. [PMID: 23272087 PMCID: PMC3525593 DOI: 10.1371/journal.pone.0051108] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 10/29/2012] [Indexed: 11/18/2022] Open
Abstract
The ATM/ATR DNA damage checkpoint functions in the maintenance of genetic stability and some missense variants of the ATM gene have been shown to confer a moderate increased risk of prostate cancer. However, whether inactivation of this checkpoint contributes directly to prostate specific cancer predisposition is still unknown. Here, we show that exposure of non-malignant prostate epithelial cells (HPr-1AR) to androgen led to activation of the ATM/ATR DNA damage response and induction of cellular senescence. Notably, knockdown of the ATM gene expression in HPr-1AR cells can promote androgen-induced TMPRSS2: ERG rearrangement, a prostate-specific chromosome translocation frequently found in prostate cancer cells. Intriguingly, unlike the non-malignant prostate epithelial cells, the ATM/ATR DNA damage checkpoint appears to be defective in prostate cancer cells, since androgen treatment only induced a partial activation of the DNA damage response. This mechanism appears to preserve androgen induced autophosphorylation of ATM and phosphorylation of H2AX, lesion processing and repair pathway yet restrain ATM/CHK1/CHK2 and p53 signaling pathway. Our findings demonstrate that ATM/ATR inactivation is a crucial step in promoting androgen-induced genomic instability and prostate carcinogenesis.
Collapse
Affiliation(s)
- Yung-Tuen Chiu
- Department of Anatomy, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Ji Liu
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
| | - Kaidun Tang
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
| | - Yong-Chuan Wong
- Department of Pathology, Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Kum Kum Khanna
- Signal Transduction Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Ming-Tat Ling
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
- * E-mail:
| |
Collapse
|
16
|
Schiewer MJ, Augello MA, Knudsen KE. The AR dependent cell cycle: mechanisms and cancer relevance. Mol Cell Endocrinol 2012; 352:34-45. [PMID: 21782001 PMCID: PMC3641823 DOI: 10.1016/j.mce.2011.06.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 06/08/2011] [Accepted: 06/27/2011] [Indexed: 01/04/2023]
Abstract
Prostate cancer cells are exquisitely dependent on androgen receptor (AR) activity for proliferation and survival. As these functions are critical targets of therapeutic intervention for human disease, it is imperative to delineate the mechanisms by which AR engages the cell cycle engine. More than a decade of research has revealed that elegant intercommunication between AR and the cell cycle machinery governs receptor-dependent cellular proliferation, and that perturbations in this process occur frequently in human disease. Here, AR-cell cycle interplay and associated cancer relevance will be reviewed.
Collapse
Affiliation(s)
- Matthew J. Schiewer
- Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Cancer Biology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
| | - Michael A. Augello
- Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Cancer Biology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
| | - Karen E. Knudsen
- Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Cancer Biology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Urology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Radiation Oncology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Corresponding author at: Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th St., BLSB 1008, Philadelphia, PA 19107, USA. Tel.: +1 215 503 8574 (office)/+1 215 503 8573 (lab). (K.E. Knudsen)
| |
Collapse
|
17
|
Cdc25A regulates matrix metalloprotease 1 through Foxo1 and mediates metastasis of breast cancer cells. Mol Cell Biol 2011; 31:3457-71. [PMID: 21670150 DOI: 10.1128/mcb.05523-11] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cdc25A is a cell cycle-activating phosphatase, and its overexpression in breast cancers has been shown to correlate with poor prognosis. Most recent studies related to Cdc25A and tumor progression have focused on its role in regulating cell cycle progression. However, less is known about how Cdc25A modulates the metastasis of breast cancer cells. In this study, we revealed that Cdc25A enhances Foxo1 stability by dephosphorylating Cdk2, and Foxo1 was shown to directly regulate transcription of the metastatic factor MMP1. Further studies have shown that overexpression of Cdc25A in breast cancer cells enhances metastasis, whereas its downmodulation inhibits metastasis in mouse models, and the effects of Cdc25A on breast cancer cell metastasis are independent of cell proliferation and apoptosis. Furthermore, we have demonstrated that aberrant Cdc25A in breast cancer patient samples directly correlates with the metastatic phenotype. Further insights into this critical role of Cdc25A in the metastasis of breast cancer cells and the trial of an anti-Cdc25A strategy in mouse models may reveal its therapeutic potential in prevention and treatment of breast cancer cell dissemination.
Collapse
|
18
|
Cui J, Yang Y, Zhang C, Hu P, Kan W, Bai X, Liu X, Song H. FBI-1 functions as a novel AR co-repressor in prostate cancer cells. Cell Mol Life Sci 2011; 68:1091-103. [PMID: 20812024 PMCID: PMC11114496 DOI: 10.1007/s00018-010-0511-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2010] [Revised: 08/12/2010] [Accepted: 08/16/2010] [Indexed: 01/25/2023]
Abstract
The pro-oncogene FBI-1, encoded by Zbtb7a, is a transcriptional repressor that belongs to the POK (POZ/BTB and Krüppel) protein family. In this study, we investigated a potential interaction between androgen receptor (AR) signaling and FBI-1 and demonstrated that overexpression of FBI-1 inhibited ligand-dependent AR activation. A protein-protein interaction was identified between FBI-1 and AR in a ligand-dependent manner. Furthermore, FBI-1, AR and SMRT formed a ternary complex and FBI-1 enhanced the recruitment of NCoR and SMRT to endogenous PSA upstream sequences. Our data also indicated that the FBI-1-mediated inhibition of AR transcriptional activity is partially dependent on HDAC. Interestingly, FBI-1 plays distinct roles in regulating LNCaP (androgen-dependent) and PC-3 cell (androgen-independent) proliferation.
Collapse
Affiliation(s)
- Jiajun Cui
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, 100071 Beijing, People’s Republic of China
- Biotech Pharmaceuticals Co., Ltd, 100176 Beijing, People’s Republic of China
| | - Yutao Yang
- Beijing Institute for Neuroscience, Capital Medical University, 100069 Beijing, People’s Republic of China
| | - Chuanfu Zhang
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, 100071 Beijing, People’s Republic of China
| | - Pinliang Hu
- Biotech Pharmaceuticals Co., Ltd, 100176 Beijing, People’s Republic of China
| | - Wei Kan
- Biotech Pharmaceuticals Co., Ltd, 100176 Beijing, People’s Republic of China
| | - Xianhong Bai
- Biotech Pharmaceuticals Co., Ltd, 100176 Beijing, People’s Republic of China
| | - Xuelin Liu
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, 100071 Beijing, People’s Republic of China
| | - Hongbin Song
- Institute of Disease Control and Prevention, Chinese Academy of Military Medical Sciences, 100071 Beijing, People’s Republic of China
| |
Collapse
|
19
|
Heemers HV, Schmidt LJ, Sun Z, Regan KM, Anderson SK, Duncan K, Wang D, Liu S, Ballman KV, Tindall DJ. Identification of a clinically relevant androgen-dependent gene signature in prostate cancer. Cancer Res 2011; 71:1978-88. [PMID: 21324924 PMCID: PMC3077061 DOI: 10.1158/0008-5472.can-10-2512] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The androgen receptor (AR) is the principal target for treatment of non-organ-confined prostate cancer (PCa). Androgen deprivation therapies (ADT) directed against the AR ligand-binding domain do not fully inhibit androgen-dependent signaling critical for PCa progression. Thus, information that could direct the development of more effective ADTs is desired. Systems and bioinformatics approaches suggest that considerable variation exists in the mechanisms by which AR regulates expression of effector genes, pointing to a role for secondary transcription factors. A combination of microarray and in silico analyses led us to identify a 158-gene signature that relies on AR along with the transcription factor SRF (serum response factor), representing less than 6% of androgen-dependent genes. This AR-SRF signature is sufficient to distinguish microdissected benign and malignant prostate samples, and it correlates with the presence of aggressive disease and poor outcome. The AR-SRF signature described here associates more strongly with biochemical failure than other AR target gene signatures of similar size. Furthermore, it is enriched in malignant versus benign prostate tissues, compared with other signatures. To our knowledge, this profile represents the first demonstration of a distinct mechanism of androgen action with clinical relevance in PCa, offering a possible rationale to develop novel and more effective forms of ADT.
Collapse
Affiliation(s)
- Hannelore V Heemers
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Nemoto K. G2/M accumulation in prostate cancer cell line PC-3 is induced by Cdc25 inhibitor 7-chloro-6-(2-morpholin-4-ylethylamino) quinoline-5, 8-dione (DA 3003-2). Exp Ther Med 2010; 1:647-650. [PMID: 22993588 DOI: 10.3892/etm_00000101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 05/13/2010] [Indexed: 12/28/2022] Open
Abstract
Cdc25 phosphatases are dual-specific phosphatases that play a role in cell cycle progression. In many human cancers, Cdc25 phosphatases are overexpressed as compared with normal tissues. In addition, overexpression of Cdc25 phosphatases in prostate cancer is correlated with disease progression. The antiproliferative efficacy of Cdc25 phosphatase inhibitor 7-chloro-6-(2-morpholin-4-ylethylamino) quinoline-5, 8-dione (DA 3003-2) was investigated in the PC-3 asynchronous human prostate cancer cell line using a cell-based assay. The time course changes in cell cycle distribution and the modulation of cell cycle regulators after DA 3003-2 administration were analyzed using the MTT assay. We found that the relative IC(50) of DA 3003-2 was 2-fold lower as compared with its congener (2-mercaptoethanol)-3-methyl-1, 4-naphthoquinone (NSC 672121). Asynchronous PC-3 cells accumulated in the G2/M phase at 24 h after treatment with 10 μM DA 3003-2 or 20 μM NSC 672121, which represent IC(70) concentrations. Treatment of cells with DA 3003-2 caused hyperphosphorylation of Cdc2 tyr(15) in cyclin B(1) and cyclin A complexes. DA 3003-2 did not downregulate the protein expression levels of Cdc25s, cyclins and cyclin-dependent kinases (Cdks). To conclude, after DA 3003-2 administration asynchronous PC-3 cells accumulated in the G2/M phase, with hyperphosphorylation of the G2/M cyclin-Cdk complex.
Collapse
Affiliation(s)
- Kaoru Nemoto
- Department of Urology, Nippon Medical School, Chiba Hokusoh Hospital, 1715 Kamagari, Inzai, Chiba 270-1694, Japan
| |
Collapse
|
21
|
Wu Y, Feng X, Jin Y, Wu Z, Hankey W, Paisie C, Li L, Liu F, Barsky SH, Zhang W, Ganju R, Zou X. A novel mechanism of indole-3-carbinol effects on breast carcinogenesis involves induction of Cdc25A degradation. Cancer Prev Res (Phila) 2010; 3:818-28. [PMID: 20587702 DOI: 10.1158/1940-6207.capr-09-0213] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The natural compound indole-3-carbinol (I3C; found in vegetables of the genus Brassica) is a promising cancer prevention or therapy agent. The cell division cycle 25A (Cdc25A) phosphatase is overexpressed in a variety of human cancers and other diseases. In the present study, I3C induced degradation of Cdc25A, arrest of the G(1) cell cycle, and inhibition of the growth of breast cancer cells. We also showed that the Ser124 site of Cdc25A, which is related to cyclin-dependent kinase 2, is required for I3C-induced degradation of Cdc25A in breast cancer cells, and that interruption of the ATM-Chk2 pathway suppressed I3C-induced destruction of Cdc25A. Our in vivo studies of different mutated forms of Cdc25A found that the mutation Cdc25A(S124A) (Ser124 to Ala124), which confers resistance to I3C-induced degradation of Cdc25A, attenuated I3C inhibition of breast tumorigenesis in a mouse xenograft model. The present in vitro and in vivo studies together show that I3C-induced activation of the ATM-Chk2 pathway and degradation of Cdc25A represent a novel molecular mechanism of I3C in arresting the G(1) cell cycle and inhibiting the growth of breast cancer cells. The finding that I3C induces Cdc25A degradation underscores the potential use of this agent for preventing and treating cancers and other human diseases with Cdc25A overexpression.
Collapse
|