1
|
Liu H, Li Z, Xu X, Xu B, Li Z. Network pharmacology and in vitro analyses reveal EGCG inhibits breast cancer progression via suppression of the EGFR/Src pathway. Biochem Biophys Res Commun 2025; 769:151942. [PMID: 40359763 DOI: 10.1016/j.bbrc.2025.151942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/15/2025]
Abstract
Breast cancer (BRCA) has emerged as a significant threat to women's health. Epigallocatechin gallate (EGCG) has shown promising therapeutic potential. However, its mechanisms of action are not yet fully understood. This study employed a network pharmacology (NP) approach to investigate the key targets and signaling pathways regulated by EGCG in BRCA, and validated the findings through cell experiments. Our comprehensive analysis identified 10 key targets of EGCG, suggesting that EGCG may inhibit the EGFR/Src pathway. Consistently, our cell experiments revealed that EGCG could significantly inhibit the migration, invasion, and proliferation of BRCA cells. Furthermore, EGCG downregulated the protein levels of EGFR, Src, PI3K, Akt, STAT3, and Bcl2. These experimental findings support the results of the NP analysis. In conclusion, our NP and in vitro studies indicate that EGCG inhibits BRCA progression by suppressing the EGFR/Src pathway and its downstream signals transduction including the PI3K/Akt and STAT3/Bcl2 pathways.
Collapse
Affiliation(s)
- Huiying Liu
- The Institute of Translational Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; The Jiangxi Province Key Laboratory of Precision Cell Therapy, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; Department of Rehabilitation Medicine, The Department of Rehabilitation Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Zhiqiang Li
- The Institute of Translational Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; The Jiangxi Province Key Laboratory of Precision Cell Therapy, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; Department of Rehabilitation Medicine, The Department of Rehabilitation Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaohui Xu
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Shandong, 266000, China
| | - Binwu Xu
- The Institute of Translational Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; The Jiangxi Province Key Laboratory of Precision Cell Therapy, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; Department of Rehabilitation Medicine, The Department of Rehabilitation Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Zhipeng Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China.
| |
Collapse
|
2
|
Kotwal A, Goldner WS, Bennett RG. Role of Relaxin Signaling in Cancer: A Review. Biochem Pharmacol 2024; 230:116634. [PMID: 39547477 DOI: 10.1016/j.bcp.2024.116634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/29/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
The investigation into relaxin (RLN), additional RLN-like proteins, and RLN family peptide receptors (RXFP) has demonstrated their role in modulating the extracellular matrix (ECM), immune cells, specifically macrophages, and angiogenesis, with recent evidence showing an effect on signaling pathways in tumor cells. These findings serve as the basis for our narrative review to collate pertinent studies in this field and provide our perspective on their clinical and investigational significance. In the article, we discuss findings from pertinent studies focusing on evaluating the expression or effect of RLN1, RLN2, or RXFP1 in various cancers. We also briefly discuss the potential role that other RLN family peptides and their receptors play in cancer. Specifically, we delve into questions regarding RLN signaling in terms of parity/pregnancy-associated protection from mammary tumors, expression in tumors, detection in serum in the setting of cancers, effect on tumor-adjacent cells, effect on tumorigenesis depending on endogenous expression or delivery, and last, but not the least, impact on the effectiveness of anti-cancer therapies. We expect that summarizing the available literature to answer these questions will allow readers to understand the role of RLN-receptor interaction in cancer as well as identify areas of uncertainty and avenues for future investigation.
Collapse
Affiliation(s)
- Anupam Kotwal
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Whitney S Goldner
- Department of Internal Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, USA
| | - Robert G Bennett
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
3
|
Deng X, Qiu Z, Chen X, Liu J, Wang X, Li J, Zhang J, Cui X, Fu Y, Jiang M. Exploring the potential mechanism of ginsenoside Rg1 to regulate ferroptosis in Alzheimer's disease based on network pharmacology. Eur J Pharmacol 2024; 979:176859. [PMID: 39067563 DOI: 10.1016/j.ejphar.2024.176859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVES To explore the pathogenesis of Alzheimer's disease (AD), the potential targets and signaling pathways of ginsenoside Rg1 against AD were investigated by network pharmacology. METHODS Ginsenoside Rg1 targets were identified through PubChem, PharmMapper, and Uniprot databases, while the GeneCards database was used to examine the respective targets of amyloid precursor protein (APP) and AD. Then, the common targets between ginsenoside Rg1 and APP were explored by the Venny tool, the interaction network diagram between the active components and the targets was built via Cytoscape software, as well as GO enrichment and KEGG pathway annotation analysis were performed. Furthermore, genes associated with ferroptosis were found by the GeneCards and FerrDb databases. Besides, the connection among ginsenoside Rg1, APP, ferroptosis, and AD was predicted and analyzed. Finally, the effects of ginsenosides Rg1 and liproxstain-1 on the proliferation and differentiation of APP/PS1 mice were evaluated by immunohistochemistry. RESULTS Ginsenoside Rg1, APP, ferroptosis, and AD had 12 hub genes. GO enrichment and KEGG pathway annotation analysis showed that EGFR, SRC, protein hydrolysis, protein phosphorylation, the Relaxin pathway, and the FoxO signaling pathway play an important role in the potential mechanism of ginsenoside Rg1's under regulation of ferroptosis anti-AD through the modulation of APP-related signaling pathways. The APP/PS1 mice experiment verified that ginsenosides Rg1 and liproxstain-1 can promote the proliferation and differentiation. CONCLUSION Ginsenoside Rg1, APP and ferroptosis may act on EGFR, SRC, the Relaxin and FoxO signaling pathways to regulate protein metabolism, protein phosphorylation and other pathways to improve AD symptoms.
Collapse
Affiliation(s)
- Xu Deng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Zixiong Qiu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaoshuai Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiangxiu Liu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaowei Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jie Li
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiankai Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaojun Cui
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Yuan Fu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Mei Jiang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
4
|
Jung J, Han H. The diverse influences of relaxin-like peptide family on tumor progression: Potential opportunities and emerging challenges. Heliyon 2024; 10:e24463. [PMID: 38298643 PMCID: PMC10828710 DOI: 10.1016/j.heliyon.2024.e24463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
Relaxin-like peptide family exhibit differential expression patterns in various types of cancers and play a role in cancer development. This family participates in tumorigenic processes encompassing proliferation, migration, invasion, tumor microenvironment, immune microenvironment, and anti-cancer resistance, ultimately influencing patient prognosis. In this review, we explore the mechanisms underlying the interaction between the RLN-like peptide family and tumors and provide an overview of therapeutic approaches utilizing this interaction.
Collapse
Affiliation(s)
| | - Hyunho Han
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
5
|
Hossain MA, Praveen P, Noorzi NA, Wu H, Harrison IP, Handley T, Selemidis S, Samuel CS, Bathgate RAD. Development of Novel High-Affinity Antagonists for the Relaxin Family Peptide Receptor 1. ACS Pharmacol Transl Sci 2023; 6:842-853. [PMID: 37200817 PMCID: PMC10186362 DOI: 10.1021/acsptsci.3c00053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Indexed: 05/20/2023]
Abstract
H2 relaxin is a peptide hormone that exerts its biological actions through the G protein-coupled receptor, RXFP1. The numerous important biological functions of H2 relaxin, including potent renal, vasodilatory, cardioprotective, and anti-fibrotic actions, have resulted in considerable interest in its use as a therapeutic for various cardiovascular diseases and other fibrotic indications. Interestingly though, H2 relaxin and RXFP1 have been shown to be overexpressed in prostate cancer, allowing for the downregulation or blocking of relaxin/RXFP1 to decrease prostate tumor growth. These findings suggest the application of an RXFP1 antagonist for the treatment of prostate cancer. However, these therapeutically relevant actions are still poorly understood and have been hindered by the lack of a high-affinity antagonist. In this study, we chemically synthesized three novel H2 relaxin analogues that have complex insulin-like structures with two chains (A and B) and three disulfide bridges. We report here the structure-activity relationship studies on H2 relaxin that resulted in the development of a novel high-affinity RXFP1 antagonist, H2 B-R13HR (∼40 nM), that has only one extra methylene group in the side chain of arginine 13 in the B-chain (ArgB13) of H2 relaxin. Most notably, the synthetic peptide was shown to be active in a mouse model of prostate tumor growth in vivo where it inhibited relaxin-mediated tumor growth. Our compound H2 B-R13HR will be an important research tool to understand relaxin actions through RXFP1 and may be a potential lead compound for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Mohammed Akhter Hossain
- Florey
Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Victoria, Australia
- School
of Chemistry, University of Melbourne, Parkville 3010, Victoria, Australia
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Parkville 3010, Victoria, Australia
| | - Praveen Praveen
- Florey
Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Victoria, Australia
| | - Nurhayati Ahmad Noorzi
- Cardiovascular
Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
- Department
of Pharmacology, Monash University, Clayton 3800, Victoria, Australia
| | - Hongkang Wu
- Florey
Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Victoria, Australia
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Parkville 3010, Victoria, Australia
| | - Ian P. Harrison
- Cardiovascular
Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
- Department
of Pharmacology, Monash University, Clayton 3800, Victoria, Australia
| | - Thomas Handley
- Florey
Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Victoria, Australia
| | - Stavros Selemidis
- School
of
Health and Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia
| | - Chrishan S. Samuel
- Cardiovascular
Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
- Department
of Pharmacology, Monash University, Clayton 3800, Victoria, Australia
| | - Ross A. D. Bathgate
- Florey
Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Victoria, Australia
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Parkville 3010, Victoria, Australia
| |
Collapse
|
6
|
Chunduri P, Patel SA, Levick SP. Relaxin/serelaxin for cardiac dysfunction and heart failure in hypertension. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:183-211. [PMID: 35659372 DOI: 10.1016/bs.apha.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The pregnancy related hormone relaxin is produced throughout the reproductive system. However, relaxin also has important cardiovascular effects as part of the adaptation that the cardiovascular system undergoes in response to the extra demands of pregnancy. These effects are primarily mediated by the relaxin family peptide receptor 1, which is one of four known relaxin receptors. The effects of relaxin on the cardiovascular system during pregnancy, as well as its anti-fibrotic and anti-inflammatory properties, have led to extensive studies into the potential of relaxin therapy as an approach to treat heart failure. Cardiomyocytes, cardiac fibroblasts, and endothelial cells all possess relaxin family peptide receptor 1, allowing for direct effects of therapeutic relaxin on the heart. Many pre-clinical animal studies have demonstrated a beneficial effect of exogenous relaxin on adverse cardiac remodeling including inflammation, fibrosis, cardiomyocyte hypertrophy and apoptosis, as well as effects on cardiac contractile function. Despite this, clinical studies have yielded disappointing results for the synthetic seralaxin, even though seralaxin was well tolerated. This article will provide background on relaxin in the context of normal physiology, as well as the role of relaxin in pregnancy-related adaptations of the cardiovascular system. We will also present evidence from pre-clinical animal studies that demonstrate the potential benefits of relaxin therapy, as well as discussing the results from clinical trials. Finally, we will discuss possible reasons for the failure of these clinical trials as well as steps being taken to potentially improve relaxin therapy for heart failure.
Collapse
Affiliation(s)
- Prasad Chunduri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Shrey A Patel
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Scott P Levick
- Physiology and Pharmacology, West Virginia University, Morgantown, WV, United States.
| |
Collapse
|
7
|
Use of RNA-Seq and a Transgenic Mouse Model to Identify Genes Which May Contribute to Mutant p53-Driven Prostate Cancer Initiation. BIOLOGY 2022; 11:biology11020218. [PMID: 35205085 PMCID: PMC8869245 DOI: 10.3390/biology11020218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/03/2022]
Abstract
Simple Summary We use RNA-seq analysis to identify genes that may contribute to mutant p53-mediated prostate cancer initiation in a genetically engineered mouse model (B6.129S4-Trp53tm3.1Tyj/J). A total of 1378 differentially expressed genes, including wildtype p53 target genes (e.g. Cdkn1a, Bax, Bcl2, Kras, Mdm2), p53 gain-of-function-related genes (Mgmt, Id4), and prostate cancer-related genes (Cav-1, Raf1, Kras), were identified. Mice that were homozygous or heterozygous for the Trp53 R270H mutation developed grade one PIN lesions at 3 months and 5 months, respectively, whereas wildtype mice did not develop PIN. Immunohistochemical analysis revealed decreased levels of irradiation-mediated apoptosis in homozygous and heterozygous mice when compared to wildtype counterparts, and this aligned with observed differences in apoptosis-related gene expression. Abstract We previously demonstrated that the Trp53-R270H mutation can drive prostate cancer (CaP) initiation using the FVB.129S4 (Trp53tm3Tyj/wt); FVB.129S (Nkx3-1tm3(cre)Mmswt) genetically engineered mouse model (GEM). We now validate this finding in a different model (B6.129S4-Trp53tm3.1Tyj/J mice) and use RNA-sequencing (RNA-Seq) to identify genes which may contribute to Trp53 R270H-mediated prostate carcinogenesis. Wildtype (Trp53WT/WT), heterozygous (Trp53R270H/WT), and homozygous mice (Trp53R270H/R270H) were exposed to 5 Gy irradiation to activate and stabilize p53, and thereby enhance our ability to identify differences in transcriptional activity between the three groups of mice. Mouse prostates were harvested 6 h post-irradiation and processed for histological/immunohistochemistry (IHC) analysis or were snap-frozen for RNA extraction and transcriptome profiling. IHC analyses determined that presence of the Trp53-R270H mutation impacts apoptosis (lower caspase 3 activity) but not cell proliferation (Ki67). RNA-Seq analysis identified 1378 differentially expressed genes, including wildtype p53 target genes (E.g., Cdkn1a, Bax, Bcl2, Kras, Mdm2), p53 gain-of-function (GOF)-related genes (Mgmt, Id4), and CaP-related genes (Cav-1, Raf1, Kras). Further understanding the mechanisms which contribute to prostate carcinogenesis could allow for the development of improved preventive methods, diagnostics, and treatments for CaP.
Collapse
|
8
|
Differential Expression of the Androgen Receptor, Splice Variants and Relaxin 2 in Renal Cancer. Life (Basel) 2021; 11:life11080731. [PMID: 34440475 PMCID: PMC8402134 DOI: 10.3390/life11080731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 12/09/2022] Open
Abstract
Background: The role of the androgen receptor (AR) in renal cell carcinoma (RCC) is unclear. We aimed to analyze the expression of AR and its splice variants (SVs) and their correlation with relaxin 2 (RLN2) and cytokines in RCC. Methods: We investigated the expression of RLN2 and AR variants in 25 clear cell RCC (ccRCC) and 9 papillary (pRCC) tumor tissues and the corresponding controls using quantitative PCR and serum RLN2, testosterone and cytokine levels in matched samples using ELISA and chemiluminescent immunometric assay, respectively. Results: ccRCC tissues but not pRCC tissues more frequently expressed AR and the SVs than did normal tissues. All pRCC samples expressed more AR than did ccRCC samples. The highest expression of all AR variants except AR-V12 was found in low-stage tumors, with dominant expression of AR-V7. In males in the ccRCC cohort, the expression of AR-FL, AR-V1 and AR-V3 was significantly correlated with that of RLN2. The secretion pattern of proinflammatory IL-6 was higher in ccRCC than in pRCC. Conclusions: The results highlight additional molecular differences between ccRCC and pRCC, suggesting the influence of external factors on the whole kidney or genetic predispositions to developing certain types of renal cancer, and may support further pathological analysis and studies of targeted hormone therapy.
Collapse
|
9
|
Martin B, Gabris B, Barakat AF, Henry BL, Giannini M, Reddy RP, Wang X, Romero G, Salama G. Relaxin reverses maladaptive remodeling of the aged heart through Wnt-signaling. Sci Rep 2019; 9:18545. [PMID: 31811156 PMCID: PMC6897890 DOI: 10.1038/s41598-019-53867-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 10/30/2019] [Indexed: 11/09/2022] Open
Abstract
Healthy aging results in cardiac structural and electrical remodeling that increases susceptibility to cardiovascular diseases. Relaxin, an insulin-like hormone, suppresses atrial fibrillation, inflammation and fibrosis in aged rats but the mechanisms-of-action are unknown. Here we show that relaxin treatment of aged rats reverses pathological electrical remodeling (increasing Nav1.5 expression and localization of Connexin43 to intercalated disks) by activating canonical Wnt signaling. In isolated adult ventricular myocytes, relaxin upregulated Nav1.5 (EC50 = 1.3 nM) by a mechanism inhibited by the addition of Dickkopf-1. Furthermore, relaxin increased the levels of connexin43, Wnt1, and cytosolic and nuclear β-catenin. Treatment with Wnt1 or CHIR-99021 (a GSK3β inhibitor) mimicked the relaxin effects. In isolated fibroblasts, relaxin blocked TGFβ-induced collagen elevation in a Wnt dependent manner. These findings demonstrate a close interplay between relaxin and Wnt-signaling resulting in myocardial remodeling and reveals a fundamental mechanism of great therapeutic potential.
Collapse
Affiliation(s)
- Brian Martin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Beth Gabris
- Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Amr F Barakat
- Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Brian L Henry
- Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Marianna Giannini
- Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Scuola Superiore Sant' Anna, Institute of Life Sciences, Pisa, Italy
| | - Rajiv P Reddy
- Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Xuewen Wang
- Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Guillermo Romero
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Guy Salama
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA. .,Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
10
|
Thanasupawat T, Glogowska A, Nivedita-Krishnan S, Wilson B, Klonisch T, Hombach-Klonisch S. Emerging roles for the relaxin/RXFP1 system in cancer therapy. Mol Cell Endocrinol 2019; 487:85-93. [PMID: 30763603 DOI: 10.1016/j.mce.2019.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023]
Abstract
A role for the hormone relaxin in cancer was described well before the receptor was identified. Relaxin predominantly increases the growth and invasive potential in cancers of different origins. However, relaxin was also shown to promote cell differentiation and to act in a dose-and time-dependent manner in different cancer cell models used. Following the discovery of the relaxin like family peptide receptor 1 (RXFP1) as the cellular receptor for RLN1 and RLN2, research has focussed on the ligand interaction with the large extracellular domain of RXFP1 and resulting molecular signaling mechanisms. RXFP1 activation mediates anti-apoptotic functions, angiogenesis and chemoresistance in cancer cells. This minireview summarizes the known biological functions of RXFP1 activation in different cancer entities in-vitro and in-vivo and outlines possible mechanisms to therapeutically address the relaxin-RXFP1 system in cancer cells.
Collapse
Affiliation(s)
- Thatchawan Thanasupawat
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Aleksandra Glogowska
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Sai Nivedita-Krishnan
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Brian Wilson
- Department of Biology, Acadia University, Wolfville, Nova Scotia, Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
11
|
Valkovic AL, Bathgate RA, Samuel CS, Kocan M. Understanding relaxin signalling at the cellular level. Mol Cell Endocrinol 2019; 487:24-33. [PMID: 30592984 DOI: 10.1016/j.mce.2018.12.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/19/2018] [Accepted: 12/22/2018] [Indexed: 02/07/2023]
Abstract
The peptide hormone relaxin mediates many biological actions including anti-fibrotic, vasodilatory, angiogenic, anti-inflammatory, anti-apoptotic, and organ protective effects across a range of tissues. At the cellular level, relaxin binds to the G protein-coupled receptor relaxin family peptide receptor 1 (RXFP1) to activate a variety of downstream signal transduction pathways. This signalling cascade is complex and also varies in diverse cellular backgrounds. Moreover, RXFP1 signalling shows crosstalk with other receptors to mediate some of its physiological functions. This review summarises known signalling pathways induced by acute versus chronic treatment with relaxin across a range of cell types, it describes RXFP1 crosstalk with other receptors, signalling pathways activated by other ligands targeting RXFP1, and it also outlines physiological relevance of RXFP1 signalling outputs. Comprehensive understanding of the mechanism of relaxin actions in fibrosis, vasodilation, as well as organ protection, will further support relaxin's clinical potential.
Collapse
Affiliation(s)
- Adam L Valkovic
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Ross Ad Bathgate
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, 3800, Australia
| | - Martina Kocan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
12
|
Wang W, Chen ZX, Guo DY, Tao YX. Regulation of prostate cancer by hormone-responsive G protein-coupled receptors. Pharmacol Ther 2018; 191:135-147. [PMID: 29909235 DOI: 10.1016/j.pharmthera.2018.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/01/2018] [Indexed: 11/27/2022]
Abstract
Regulation of prostate cancer by androgen and androgen receptor (AR), and blockade of AR signaling by AR antagonists and steroidogenic enzyme inhibitors have been extensively studied. G protein-coupled receptors (GPCRs) are a family of membrane receptors that regulate almost all physiological processes. Nearly 40% of FDA-approved drugs in the market target GPCRs. A variety of GPCRs that mediate reproductive function have been demonstrated to be involved in the regulation of prostate cancer. These GPCRs include gonadotropin-releasing hormone receptor, luteinizing hormone receptor, follicle-stimulating hormone receptor, relaxin receptor, ghrelin receptor, and kisspeptin receptor. We highlight here GPCR regulation of prostate cancer by these GPCRs. Further therapeutic approaches targeting these GPCRs for the treatment of prostate cancer are summarized.
Collapse
Affiliation(s)
- Wei Wang
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Zhao-Xia Chen
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Dong-Yu Guo
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA.
| |
Collapse
|
13
|
Schneider JA, Logan SK. Revisiting the role of Wnt/β-catenin signaling in prostate cancer. Mol Cell Endocrinol 2018; 462:3-8. [PMID: 28189566 PMCID: PMC5550366 DOI: 10.1016/j.mce.2017.02.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/14/2016] [Accepted: 02/07/2017] [Indexed: 12/18/2022]
Abstract
The androgen receptor (AR) is a widely accepted therapeutic target in prostate cancer and multiple studies indicate that the AR and Wnt/β-catenin pathways intersect. Recent genome-wide analysis of prostate cancer metastases illustrate the importance of the Wnt/β-catenin pathway in prostate cancer and compel us to reexamine the interaction of the AR and Wnt/β-catenin signaling pathways. This review includes newer areas of interest such as non-canonical Wnt signaling and the role of Wnts in prostate cancer stem cells. The effort to develop Wnt modulating therapeutics, both biologics and small molecules, is also discussed.
Collapse
Affiliation(s)
- Jeffrey A Schneider
- Departments of Urology, Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, United States
| | - Susan K Logan
- Departments of Urology, Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, United States.
| |
Collapse
|
14
|
Ivell R, Agoulnik AI, Anand‐Ivell R. Relaxin-like peptides in male reproduction - a human perspective. Br J Pharmacol 2017; 174:990-1001. [PMID: 27933606 PMCID: PMC5406299 DOI: 10.1111/bph.13689] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/10/2016] [Accepted: 12/05/2016] [Indexed: 12/26/2022] Open
Abstract
The relaxin family of peptide hormones and their cognate GPCRs are becoming physiologically well-characterized in the cardiovascular system and particularly in female reproductive processes. Much less is known about the physiology and pharmacology of these peptides in male reproduction, particularly as regards humans. H2-relaxin is involved in prostate function and growth, while insulin-like peptide 3 (INSL3) is a major product of the testicular Leydig cells and, in the adult, appears to modulate steroidogenesis and germ cell survival. In the fetus, INSL3 is a key hormone expressed shortly after sex determination and is responsible for the first transabdominal phase of testicular descent. Importantly, INSL3 is becoming a very useful constitutive biomarker reflecting both fetal and post-natal development. Nothing is known about roles for INSL4 in male reproduction and only very little about relaxin-3, which is mostly considered as a brain peptide, or INSL5. The former is expressed at very low levels in the testes, but has no known physiology there, whereas the INSL5 knockout mouse does exhibit a testicular phenotype with mild effects on spermatogenesis, probably due to a disruption of glucose homeostasis. INSL6 is a major product of male germ cells, although it is relatively unexplored with regard to its physiology or pharmacology, except that in mice disruption of the INSL6 gene leads to a disruption of spermatogenesis. Clinically, relaxin analogues may be useful in the control of prostate cancer, and both relaxin and INSL3 have been considered as sperm adjuvants for in vitro fertilization. LINKED ARTICLES This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc.
Collapse
Affiliation(s)
- Richard Ivell
- School of BiosciencesUniversity of NottinghamNottinghamLE12 5RDUK
- School of Veterinary and Medical SciencesUniversity of NottinghamNottinghamLE12 5RDUK
| | - Alexander I Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of MedicineFlorida International UniversityMiamiFLUSA
| | | |
Collapse
|
15
|
Domińska K, Ochędalski T, Kowalska K, Matysiak-Burzyńska ZE, Płuciennik E, Piastowska-Ciesielska AW. Interaction between angiotensin II and relaxin 2 in the progress of growth and spread of prostate cancer cells. Int J Oncol 2016; 48:2619-28. [PMID: 27035428 DOI: 10.3892/ijo.2016.3458] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/18/2016] [Indexed: 11/06/2022] Open
Abstract
Deregulation of locally secreted hormones, such as angiotensin II (Ang II) and relaxin 2 (RLN2), has been linked to a higher risk of select cancers or a poor prognosis in patients. In this study, for the first time a common effect of Ang II and RLN2 in relation to various aspects of prostate cancer development and metastasis are presented. Four independent colorimetric assays were used to analyze cell viability and proliferation. The changes of cell adhesion to extracellular matrix proteins and invasion/aggressiveness ability of prostate cancer cells (LNCaP, PC3) before and after peptides treatment, were also investigated. The findings suggest that the both investigated systems, have an impact on cell growth/division or spread, to some degree via overlapping signal transduction pathways. Intermediate or sometimes poorer results were achieved by using a combination of both hormones than when each was used individually. It seems that Ang II and RLN2 can play a significant role in increasing the aggressiveness of prostate tumors by up-regulating BIRC5 expression and MMP-2 and MMP-9 secretion. In addition, we speculate that Ang II and RLN2 are involved in the transition from the androgen-dependent to the androgen-independent phenotype via modulation of the expression of androgen receptors.
Collapse
Affiliation(s)
- Kamila Domińska
- Department of Comparative Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Tomasz Ochędalski
- Department of Comparative Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Karolina Kowalska
- Department of Comparative Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
| | | | - Elżbieta Płuciennik
- Department of Molecular Cancerogenesis, Medical University of Lodz, 90-752 Lodz, Poland
| | | |
Collapse
|
16
|
E. Livermore K, Munkley J, J. Elliott D. Androgen receptor and prostate cancer. AIMS MOLECULAR SCIENCE 2016. [DOI: 10.3934/molsci.2016.2.280] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
17
|
Pimenta MT, Francisco RAR, Silva RP, Porto CS, Lazari MFM. Relaxin affects cell organization and early and late stages of spermatogenesis in a coculture of rat testicular cells. Andrology 2015; 3:772-86. [PMID: 26041439 DOI: 10.1111/andr.12056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/15/2015] [Accepted: 04/30/2015] [Indexed: 01/03/2023]
Abstract
Relaxin and its receptor RXFP1 are co-expressed in Sertoli cells, and relaxin can stimulate proliferation of Sertoli cells. In this study, we investigated a role of relaxin in spermatogenesis, using a short-term culture of testicular cells of the rat that allowed differentiation of spermatogonia to spermatids. Sertoli, germ, and peritubular myoid cells were the predominant cell types in the culture. Sertoli and germ cells expressed RXFP1. Cultures were incubated without (control) or with 0.5% fetal bovine serum (FBS) or 100 ng/mL H2 relaxin (RLN) for 2 days. Cell organization, number, and differentiation were analyzed after 2 (D2), 5 (D5) or 8 (D8) days of culturing. Although the proportion of germ cells decayed from D2 to D5, the relative contribution of HC, 1C, 2C, and 4C germ cell populations remained constant in the control group during the whole culture. RLN did not affect the proportion of germ cell populations compared with control, but increased gene and/or protein expression of the undifferentiated and differentiated spermatogonia markers PLZF and c-KIT, and of the post-meiotic marker Odf2 in D5. RLN favored organization of cells in tubule-like structures, the arrangement of myoid cells around the tubules, arrangement of c-KIT-positive spermatogonia at the basal region of the tubules, and expression of the cell junction protein β-catenin close to the plasma membrane region. Knockdown of relaxin with small interfering RNA (siRNA) reduced expression of β-catenin at the cell junctions, and shifted its expression to the nucleus. We propose that relaxin may affect spermatogenesis by modulating spermatogonial self renewal and favoring cell contact.
Collapse
Affiliation(s)
- M T Pimenta
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - R A R Francisco
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - R P Silva
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - C S Porto
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - M F M Lazari
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| |
Collapse
|
18
|
Neschadim A, Summerlee AJS, Silvertown JD. Targeting the relaxin hormonal pathway in prostate cancer. Int J Cancer 2014; 137:2287-95. [PMID: 25043063 DOI: 10.1002/ijc.29079] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 07/07/2014] [Indexed: 11/11/2022]
Abstract
Targeting the androgen signalling pathway has long been the hallmark of anti-hormonal therapy for prostate cancer. However, development of androgen-independent prostate cancer is an inevitable outcome to therapies targeting this pathway, in part, owing to the shift from cancer dependence on androgen signalling for growth in favor of augmentation of other cellular pathways that provide proliferation-, survival- and angiogenesis-promoting signals. This review focuses on the role of the hormone relaxin in the development and progression of prostate cancer, prior to and after the onset of androgen independence, as well as its role in cancers of other reproductive tissues. As the body of literature expands, examining relaxin expression in cancerous tissues and its role in a growing number of in vitro and in vivo cancer models, our understanding of the important involvement of this hormone in cancer biology is becoming clearer. Specifically, the pleiotropic functions of relaxin affecting cell growth, angiogenesis, blood flow, cell migration and extracellular matrix remodeling are examined in the context of cancer progression. The interactions and intercepts of the intracellular signalling pathways of relaxin with the androgen pathway are explored in the context of progression of castration-resistant and androgen-independent prostate cancers. We provide an overview of current anti-hormonal therapeutic treatment options for prostate cancer and delve into therapeutic approaches and development of agents aimed at specifically antagonizing relaxin signalling to curb tumor growth. We also discuss the rationale and challenges utilizing such agents as novel anti-hormonals in the clinic, and their potential to supplement current therapeutic modalities.
Collapse
Affiliation(s)
- Anton Neschadim
- Armour Therapeutics Inc., Toronto, 124 Orchard View Blvd, Toronto, ON, Canada
| | | | - Joshua D Silvertown
- Armour Therapeutics Inc., Toronto, 124 Orchard View Blvd, Toronto, ON, Canada
| |
Collapse
|
19
|
Neschadim A, Pritzker LB, Pritzker KPH, Branch DR, Summerlee AJS, Trachtenberg J, Silvertown JD. Relaxin receptor antagonist AT-001 synergizes with docetaxel in androgen-independent prostate xenografts. Endocr Relat Cancer 2014; 21:459-71. [PMID: 24812057 DOI: 10.1530/erc-14-0088] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Androgen hormones and the androgen receptor (AR) pathway are the main targets of anti-hormonal therapies for prostate cancer. However, resistance inevitably develops to treatments aimed at the AR pathway resulting in androgen-independent or hormone-refractory prostate cancer (HRPC). Therefore, there is a significant unmet need for new, non-androgen anti-hormonal strategies for the management of prostate cancer. We demonstrate that a relaxin hormone receptor antagonist, AT-001, an analog of human H2 relaxin, represents a first-in-class anti-hormonal candidate treatment designed to significantly curtail the growth of androgen-independent human prostate tumor xenografts. Chemically synthesized AT-001, administered subcutaneously, suppressed PC3 xenograft growth by up to 60%. AT-001 also synergized with docetaxel, standard first-line chemotherapy for HRPC, to suppress tumor growth by more than 98% in PC3 xenografts via a mechanism involving the downregulation of hypoxia-inducible factor 1 alpha and the hypoxia-induced response. Our data support developing AT-001 for clinical use as an anti-relaxin hormonal therapy for advanced prostate cancer.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Binding, Competitive
- Blotting, Western
- Cell Proliferation/drug effects
- Docetaxel
- Drug Synergism
- Humans
- Immunoenzyme Techniques
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Receptors, Androgen/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, Peptide/antagonists & inhibitors
- Taxoids/pharmacology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Anton Neschadim
- Armour Therapeutics Inc., 124 Orchard View Boulevard, Toronto, Ontario, Canada Rna Diagnostics Inc., 595 Bay Street, Suite 1204, Toronto, Ontario, Canada Departments of Laboratory Medicine and Pathobiology Surgery, University of Toronto, Toronto, Ontario, Canada Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada Departments of Medicine Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada Division of Advanced Diagnostics - Infection and Immunity, Toronto General Research Institute (TGRI), University Health Network, Toronto, Ontario, Canada Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada Departments of Surgery and Medical Imaging, University of Toronto, Toronto, Ontario, Canada Division of Urology, Department of Surgical Oncology Prostate Centre, Princess Margaret Hospital Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
20
|
Clark EL, Hadjimichael C, Temperley R, Barnard A, Fuller-Pace FV, Robson CN. p68/DdX5 supports β-catenin & RNAP II during androgen receptor mediated transcription in prostate cancer. PLoS One 2013; 8:e54150. [PMID: 23349811 PMCID: PMC3547877 DOI: 10.1371/journal.pone.0054150] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 12/07/2012] [Indexed: 11/18/2022] Open
Abstract
The DEAD box RNA helicase p68 (Ddx5) is an important androgen receptor (AR) transcriptional co-activator in prostate cancer (PCa) and is over-expressed in late stage disease. β-Catenin is a multifunctional protein with important structural and signalling functions which is up-regulated in PCa and similar to p68, interacts with the AR to co-activate expression of AR target genes. Importantly, p68 forms complexes with nuclear β-Catenin and promotes gene transcription in colon cancer indicating a functional interplay between these two proteins in cancer progression. In this study, we explore the relationship of p68 and β-Catenin in PCa to assess their potential co-operation in AR-dependent gene expression, which may be of importance in the development of castrate resistant prostate cancer (CRPCa). We use immunoprecipitation to demonstrate a novel interaction between p68 and β-Catenin in the nucleus of PCa cells, which is androgen dependent in LNCaP cells but androgen independent in a hormone refractory derivative of the same cell line (representative of the CRPCa disease type). Enhanced AR activity is seen in androgen-dependent luciferase reporter assays upon transient co-transfection of p68 and β-Catenin as an additive effect, and p68-depleted Chromatin-Immunoprecipitation (ChIP) showed a decrease in the recruitment of the AR and β-Catenin to androgen responsive promoter regions. In addition, we found p68 immunoprecipitated with the processive and non-processive form of RNA polymerase II (RNAP II) and show p68 recruited to elongating regions of the AR mediated PSA gene, suggesting a role for p68 in facilitating RNAP II transcription of AR mediated genes. These results suggest p68 is important in facilitating β-Catenin and AR transcriptional activity in PCa cells.
Collapse
Affiliation(s)
- Emma L. Clark
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Richard Temperley
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Amy Barnard
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Frances V. Fuller-Pace
- Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Craig N. Robson
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
- * E-mail:
| |
Collapse
|
21
|
Muñoz-Moreno L, Arenas MI, Schally AV, Fernández-Martínez AB, Zarka E, González-Santander M, Carmena MJ, Vacas E, Prieto JC, Bajo AM. Inhibitory effects of antagonists of growth hormone-releasing hormone on growth and invasiveness of PC3 human prostate cancer. Int J Cancer 2012; 132:755-65. [PMID: 22777643 DOI: 10.1002/ijc.27716] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/11/2012] [Accepted: 06/22/2012] [Indexed: 01/28/2023]
Abstract
New approaches are needed to the therapy of advanced prostate cancer. This study determined the effect of growth hormone-releasing hormone (GHRH) antagonists, JMR-132 and JV-1-38 on growth of PC3 tumors as well as on angiogenesis and metastasis through the evaluation of various factors that contribute largely to the progression of prostate cancer. Human PC3 androgen-independent prostate cancer cells were injected subcutaneously into nude mice. The treatment with JMR-132 (10 μg/day) or JV-1-38 (20 μg/day) lasted 41 days. We also evaluated the effects of JMR-132 and JV-1-38 on proliferation, cell adhesion and migration in PC-3 cells in vitro. Several techniques (Western blot, reverse transcription polymerase chain reaction, immunohistochemistry, ELISA and zymography) were used to evaluate the expression levels of GHRH receptors and its splice variants, GHRH, vascular endothelial growth factor (VEGF), hypoxia inducible factor (HIF)-1α, metalloproteinases (MMPs) -2 and -9, β-catenin and E-cadherin. GHRH antagonists suppressed the proliferation of PC-3 cells in vitro and significantly inhibited growth of PC3 tumors. After treatment with these analogues, we found an increase in expression of GHRH receptor accompanied by a decrease of GHRH levels, a reduction in both VEGF and HIF-1α expression and in active forms of MMP-2 and MMP-9, a significant increase in levels of membrane-associated β-catenin and a significant decline in E-cadherin. These results support that the blockade of GHRH receptors can modulate elements involved in angiogenesis and metastasis. Consequently, GHRH antagonists could be considered as suitable candidates for therapeutic trials in the management of androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Laura Muñoz-Moreno
- Molecular Neuroendocrinology Unit, Department of Biochemistry and Molecular Biology, University of Alcalá, Alcalá de Henares, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wray J, Hartmann C. WNTing embryonic stem cells. Trends Cell Biol 2011; 22:159-68. [PMID: 22196214 DOI: 10.1016/j.tcb.2011.11.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 11/08/2011] [Accepted: 11/14/2011] [Indexed: 01/02/2023]
Abstract
Embryonic stem cells (ESCs) - undifferentiated cells originating from preimplantation stage embryos - have prolonged self-renewal capacity and are pluripotent. Activation of the canonical Wnt pathway is implicated in maintenance of and exit from the pluripotent state. Recent findings demonstrate that the essential mediator of canonical Wnt signaling, β-catenin, is dispensable for ESC maintenance; however, its activation inhibits differentiation through derepression of T cell factor 3 (Tcf3)-bound genes. Wnt agonists are useful in deriving ESCs from recalcitrant mouse strains and the rat and in nuclear reprogramming of somatic stem cells. We discuss recent advances in our understanding of the role of canonical Wnt signaling in the regulation of ESC self-renewal and how its manipulation can improve pluripotent ESC derivation and maintenance.
Collapse
Affiliation(s)
- Jason Wray
- University College London, Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | | |
Collapse
|
23
|
Vinall RL, Mahaffey CM, Davis RR, Luo Z, Gandour-Edwards R, Ghosh PM, Tepper CG, de Vere White RW. Dual blockade of PKA and NF-κB inhibits H2 relaxin-mediated castrate-resistant growth of prostate cancer sublines and induces apoptosis. Discov Oncol 2011; 2:224-38. [PMID: 21789713 DOI: 10.1007/s12672-011-0076-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
We previously demonstrated that H2 relaxin (RLN2) facilitates castrate-resistant (CR) growth of prostate cancer (CaP) cells through PI3K/Akt/β-catenin-mediated activation of the androgen receptor (AR) pathway. As inhibition of this pathway caused only ~50% reduction in CR growth, the goal of the current study was to identify additional RLN2-activated pathways that contribute to CR growth. Next-generation sequencing-based transcriptome and gene ontology analyses comparing LNCaP stably transfected with RLN2 versus LNCaP-vector identified differential expression of genes associated with cell proliferation (12.7% of differentially expressed genes), including genes associated with the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) and nuclear factor-kappaB (NF-κB) pathways. Subsequent molecular analyses confirmed that the cAMP/PKA and NF-κB pathways play a role in facilitating H2 relaxin-mediated CR growth of CaP cells. Inhibition of PKA-attenuated RLN2-mediated AR activity inhibited proliferation and caused a small but significant increase in apoptosis. Combined inhibition of the PKA and NF-κB signaling pathways via inhibition of PKA and Akt induced significant apoptosis and dramatically reduced clonogenic potential, outperforming docetaxel, the standard of care treatment for CR CaP. Immunohistochemical analysis of tissue microarrays in combination with multispectral quantitative imaging comparing RLN2 levels in patients with benign prostatic hyperplasia (BPH), prostatic intraepithelial neoplasia, and CaP determined that RLN2 is significantly upregulated in CaP vs BPH (p = 0.002). The combined data indicate RLN2 overexpression is frequent in CaP patients and provides a growth advantage to CaP cells. A near-complete inhibition of RLN2-induced CR growth can be achieved by simultaneous blockade of both pathways.
Collapse
Affiliation(s)
- Ruth L Vinall
- Department of Urology, University of California, Davis, School of Medicine and Cancer Center, 4860 Y Street, Suite 3500, Sacramento, CA 95817, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Chun JY, Tummala R, Nadiminty N, Lou W, Liu C, Yang J, Evans CP, Zhou Q, Gao AC. Andrographolide, an herbal medicine, inhibits interleukin-6 expression and suppresses prostate cancer cell growth. Genes Cancer 2011; 1:868-76. [PMID: 21442031 DOI: 10.1177/1947601910383416] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 08/06/2010] [Accepted: 08/12/2010] [Indexed: 01/02/2023] Open
Abstract
Elevated interleukin-6 (IL-6), a major mediator of the inflammatory response, has been implicated in androgen receptor (AR) activation, cellular growth and differentiation, plays important roles in the development and progression of prostate cancer, and is a potential target in cancer therapy. Through drug screening using human prostate cancer cells expressing IL-6 autocrine loop, we found that andrographolide, a diterpenoid lactone isolated from a traditional Chinese and Indian medicinal plant Andrographis paniculata, could inhibit IL-6 expression and suppress IL-6-mediated signals. Andrographolide inhibits IL-6 expression at both mRNA and protein levels in a dose-dependent manner. Andrographolide suppresses both IL-6 autocrine loop- and paracrine loop-induced cell signaling including Stat3 and Erk phosphorylation. Furthermore, andrographolide inhibits cell viability and induces apoptotic cell death in both androgen-stimulated and castration-resistant human prostate cancer cells without causing significant toxicity to normal immortalized prostate epithelial cells. Moreover, treatment of andrographolide to mice bearing castration-resistant DU145 human prostate tumors that express constitutive IL-6 autocrine loop significantly suppresses tumor growth. Taken together, these results demonstrate that andrographolide could be developed as a therapeutic agent to treat both androgen-stimulated and castration-resistant prostate cancer possibly by suppressing IL-6 expression and IL-6-induced signaling.
Collapse
Affiliation(s)
- Jae Yeon Chun
- Department of Urology, University of California at Davis, Davis, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Schütz SV, Schrader AJ, Zengerling F, Genze F, Cronauer MV, Schrader M. Inhibition of glycogen synthase kinase-3β counteracts ligand-independent activity of the androgen receptor in castration resistant prostate cancer. PLoS One 2011; 6:e25341. [PMID: 21980429 PMCID: PMC3183056 DOI: 10.1371/journal.pone.0025341] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 09/01/2011] [Indexed: 11/19/2022] Open
Abstract
In order to generate genomic signals, the androgen receptor (AR) has to be transported into the nucleus upon androgenic stimuli. However, there is evidence from in vitro experiments that in castration-resistant prostate cancer (CRPC) cells the AR is able to translocate into the nucleus in a ligand-independent manner. The recent finding that inhibition of the glycogen-synthase-kinase 3β (GSK-3β) induces a rapid nuclear export of the AR in androgen-stimulated prostate cancer cells prompted us to analyze the effects of a GSK-3β inhibition in the castration-resistant LNCaP sublines C4-2 and LNCaP-SSR. Both cell lines exhibit high levels of nuclear AR in the absence of androgenic stimuli. Exposure of these cells to the maleimide SB216763, a potent GSK-3β inhibitor, resulted in a rapid nuclear export of the AR even under androgen-deprived conditions. Moreover, the ability of C4-2 and LNCaP-SSR cells to grow in the absence of androgens was diminished after pharmacological inhibition of GSK-3β in vitro. The ability of SB216763 to modulate AR signalling and function in CRPC in vivo was additionally demonstrated in a modified chick chorioallantoic membrane xenograft assay after systemic delivery of SB216763. Our data suggest that inhibition of GSK-3β helps target the AR for export from the nucleus thereby diminishing the effects of mislocated AR in CRPC cells. Therefore, inhibition of GSK-3β could be an interesting new strategy for the treatment of CRPC.
Collapse
|
26
|
Feng H, Cheng ASL, Tsang DP, Li MS, Go MY, Cheung YS, Zhao GJ, Ng SS, Lin MC, Yu J, Lai PB, To KF, Sung JJY. Cell cycle-related kinase is a direct androgen receptor-regulated gene that drives β-catenin/T cell factor-dependent hepatocarcinogenesis. J Clin Invest 2011; 121:3159-75. [PMID: 21747169 DOI: 10.1172/jci45967] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 05/18/2011] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide. It is more prevalent in men than women. Related to this, recent genetic studies have revealed a causal role for androgen receptor (AR) in hepatocarcinogenesis, but the underlying molecular mechanism remains unclear. Here, we used genome-wide location and functional analyses to identify a critical mediator of AR signaling - cell cycle-related kinase (CCRK) - that drives hepatocarcinogenesis via a signaling pathway dependent on β-catenin and T cell factor (TCF). Ligand-bound AR activated CCRK transcription and protein expression via direct binding to the androgen-responsive element of the CCRK promoter in human HCC cell lines. In vitro analyses showed that CCRK was critical in human cell lines for AR-induced cell cycle progression, hepatocellular proliferation, and malignant transformation. Ectopic expression of CCRK in immortalized human liver cells activated β-catenin/TCF signaling to stimulate cell cycle progression and to induce tumor formation, as shown in both xenograft and orthotopic models. Conversely, knockdown of CCRK decreased HCC cell growth, and this could be rescued by constitutively active β-catenin or TCF. In primary human HCC tissue samples, AR, CCRK, and β-catenin were concordantly overexpressed in the tumor cells. Furthermore, CCRK overexpression correlated with the tumor staging and poor overall survival of patients. Our results reveal a direct AR transcriptional target, CCRK, that promotes hepatocarcinogenesis through the upregulation of β-catenin/TCF signaling.
Collapse
Affiliation(s)
- Hai Feng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Li Z, Feng S, Lopez V, Elhammady G, Anderson ML, Kaftanovskaya EM, Agoulnik AI. Uterine cysts in female mice deficient for caveolin-1 and insulin-like 3 receptor RXFP2. Endocrinology 2011; 152:2474-82. [PMID: 21467199 PMCID: PMC3100621 DOI: 10.1210/en.2010-1015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gene mutations of insulin-like 3 (INSL3) peptide or its G protein-coupled receptor RXFP2 (relaxin family peptide receptor 2) lead to cryptorchidism. The role of INSL3 in adult females is less known, although INSL3 expression has been described in female reproductive organs. Caveolin-1 (CAV1), the main component of caveoli cell membrane invaginations, has been shown to play an important role in epithelial organization and stromal-epithelial interactions. We created a null allele of Cav1 mice by deleting its second exon through embryonic stem cell targeting. Immunohistochemical analysis demonstrated that CAV1 expression was primarily localized to endothelial blood vessel cells and the myometrium uterus, whereas the strongest expression of Rxfp2 was detected in the endometrial epithelium. By 12 months of age approximately 18% of Cav1-/- females developed single or multiple dilated endometrial cysts lined by a flattened, simple low epithelium. A deficiency for Rxfp2 on Cav1-deficient background led to more than a 2-fold increase in the incidence of uterine cysts (54-58%). Appearance of cysts led to a severe disorganization of uterine morphology. We have found that the cysts had an increased expression of β-catenin and estrogen receptor β in endometrial stromal and epithelial cells and increased epithelial proliferation. An analysis of simple dilated cysts in human patients for CAV1 expression did not show appreciable differences with control regardless of menstrual phase, suggesting an involvement of additional factors in human disease. The results of this study suggest a novel synergistic role of INSL3/RXFP2 and CAV1 in structural maintenance of the uterus.
Collapse
Affiliation(s)
- Zhen Li
- Department of Human Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Wu X, Daniels G, Shapiro E, Xu K, Huang H, Li Y, Logan S, Greco MA, Peng Y, Monaco ME, Melamed J, Lepor H, Grishina I, Lee P. LEF1 identifies androgen-independent epithelium in the developing prostate. Mol Endocrinol 2011; 25:1018-26. [PMID: 21527502 DOI: 10.1210/me.2010-0513] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Lymphoid enhancer-binding factor (LEF)1 is a major mediator and a target in canonical Wnt/β-catenin pathway. Interactions between the androgen receptor (AR) and canonical Wnt pathways have been implicated in the development of the genitourinary organs. Here, we investigated the localization and role of LEF1-positive cells during development of the prostate gland in human and in the murine model. We show that during human prostate development, LEF1 is restricted to the basal epithelial layer of the urogenital sinus. During mouse development, Lef1 is also present in the urogenital mesenchyme in addition to the basal epithelial layer of the urogenital sinus. In the course of elongation and branching of the prostatic ducts, Lef1 is localized to the proliferating epithelium at the distal tips of the buds. Notably, during branching morphogenesis, domains of Lef1 and AR are mutually exclusive. We further employed the TOPGAL reporter strain to examine the dynamics of Wnt signaling in the context of prostate regression upon a 7-d treatment with a competitive AR inhibitor, bicalutamide. We found that Wnt/Lef1-positive basal cells are not dependent upon androgen for survival. Furthermore, upon bicalutamide treatment, Wnt/Lef1-positive basal progenitors repopulated the luminal compartment. We conclude that Wnt/Lef1 activity identifies an androgen-independent population of prostate progenitors, which is important for embryonic development and organ maintenance and regeneration in the adult.
Collapse
Affiliation(s)
- Xinyu Wu
- Department of Urology, New York University School of Medicine, 423 East 23rd Street, New York, New York 10010, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ivell R, Kotula-Balak M, Glynn D, Heng K, Anand-Ivell R. Relaxin family peptides in the male reproductive system--a critical appraisal. Mol Hum Reprod 2010; 17:71-84. [DOI: 10.1093/molehr/gaq086] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
30
|
Hombach-Klonisch S, Bialek J, Radestock Y, Truong A, Agoulnik AI, Fiebig B, Willing C, Weber E, Hoang-Vu C, Klonisch T. INSL3 has tumor-promoting activity in thyroid cancer. Int J Cancer 2010; 127:521-31. [PMID: 19950223 DOI: 10.1002/ijc.25068] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The functional role of INSL3 and its receptor RXFP2 in carcinogenesis is largely unknown. We have previously demonstrated (pro-)cathepsin-L as a target of INSL3 in human thyroid cancer cells facilitating penetration of tumor cells through elastin matrices. We demonstrate the expression of RXFP2 in human thyroid tissues and in mouse follicular thyroid epithelial cells using Cre-recombinase transgene driven by Rxfp2 promoter. Recombinant and secreted INSL3 increased the motility of thyroid carcinoma (TC) cells in an autocrine/paracrine manner. This effect required the presence of RXFP2. We identified S100A4 as a novel INSL3 target molecule and showed that S100A4 facilitated INSL3-induced enhanced motility. Stable transfectants of the human follicular TC cell line FTC-133 expressing and secreting bioactive human INSL3 displayed enhanced anchorage-independent growth in soft agar assays. Xenotransplant experiments in nude mice showed that INSL3, but not EGFP-mock transfectants, developed fast-growing and highly vascularized xenografts. We used human umbilical vein endothelial cells in capillary tube formation assays to demonstrate increased 2-dimensional tube formations induced by recombinant human INSL3 and human S100A4 comparable to the effect of vascular endothelial growth factor used as positive control. We conclude that INSL3 is a powerful and multifunctional promoter of tumor growth and angiogenesis in human thyroid cancer cell xenografts. INSL3 actions involve RXFP2 activation and the secretion of S100A4 and (pro-)cathepsin-L.
Collapse
|
31
|
Thompson VC, Hurtado-Coll A, Turbin D, Fazli L, Lehman ML, Gleave ME, Nelson CC. Relaxin drives Wnt signaling through upregulation of PCDHY in prostate cancer. Prostate 2010; 70:1134-45. [PMID: 20503398 DOI: 10.1002/pros.21148] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Relaxin, a potent peptide hormone of the insulin-like family normally produced and secreted by the human prostate, is upregulated in castrate resistant prostate cancer progression. In various tissues, relaxin increases angiogenesis and cell motility through upregulation of vascular endothelial growth factor, matrix metalloproteases, and nitric oxide, and therefore maybe an attractive target for cancer therapeutics. METHODS To examine the role of relaxin in prostate cancer progression, LNCaP cells stably transfected with relaxin (LNCaP(RLN)) were used to form xenograft tumors, and microarray expression analysis was subsequently performed to determine novel pathways regulated by relaxin. Prostate cancer tissue microarrays from patient samples were stained by immunohistochemistry for further validation and correlation of the findings. RESULTS Expression analysis identified novel relaxin regulated pathways, including the ProtocadherinY (PCDHY)/Wnt pathway. PCDHY, which upregulates Wnt11, has previously been shown to stabilize beta-catenin, causing beta-catenin to translocate from the cytoplasmic membrane to the nucleus and initiate TCF-mediated signaling. LNCaP(RLN) xenografts exhibit increased PCDHY expression and increased cytoplasmic localization of beta-catenin, suggesting relaxin directs Wnt11 overexpression through PCDHY upregulation. Similarly, prostate cancer samples from patients who have undergone androgen ablation have increased Wnt11 expression, which is further upregulated in castrate resistant tissues. Like relaxin, Wnt11, and PCDHY are negatively regulated by androgens, and further analysis indicated that the overexpression of relaxin results in dysregulation of androgen-regulated genes. CONCLUSIONS These data suggest that prostate cancer cell motility and altered androgen receptor activity attributed to relaxin may be mediated in part by Wnt11.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Cadherins/genetics
- Cadherins/metabolism
- Cell Line, Tumor
- Humans
- Immunohistochemistry
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Oligonucleotide Array Sequence Analysis
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- RNA/chemistry
- RNA/genetics
- Receptors, G-Protein-Coupled/biosynthesis
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Peptide/biosynthesis
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Relaxin/biosynthesis
- Relaxin/genetics
- Relaxin/metabolism
- Signal Transduction
- Statistics, Nonparametric
- Transfection
- Transplantation, Heterologous
- Up-Regulation
- Wnt1 Protein/genetics
- Wnt1 Protein/metabolism
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Vanessa C Thompson
- The Vancouver Prostate Centre, and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | | | |
Collapse
|
32
|
Schütz SV, Cronauer MV, Rinnab L. Inhibition of glycogen synthase kinase-3beta promotes nuclear export of the androgen receptor through a CRM1-dependent mechanism in prostate cancer cell lines. J Cell Biochem 2010; 109:1192-200. [PMID: 20127713 DOI: 10.1002/jcb.22500] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The androgen receptor (AR) is a ligand-dependent transcription factor belonging to the steroid hormone receptor superfamily. Under normal conditions, in the absence of a ligand, the AR is localized to the cytoplasm and is actively transported into the nucleus upon binding of androgens. In advanced prostate cancer (PCa) cell lines, an increased sensitivity to dihydrotestosterone (DHT), enabling the cells to proliferate under sub-physiological levels of androgens, has been associated with increased stability and nuclear localization of the AR. There is experimental evidence that the glycogen synthase kinase-3beta (GSK-3beta), a multifunctional serine/threonine kinase is involved in estrogen and AR stability. As demonstrated in the following study by immunoprecipitation analysis, GSK-3beta binds to the AR forming complexes in the cytoplasm and in the nucleus. Furthermore, inhibition of GSK-3beta activity by pharmacological inhibitors like the maleimide SB216761, the chloromethyl-thienyl-ketone GSK-3 inhibitor VI or the aminopyrazol GSK-3 inhibitor XIII in cells grown in the presence of DHT triggered a rapid nuclear export of endogenous AR as well as of green fluorescent AR-EosFP. The nuclear export of AR following GSK-3beta inhibition could be blocked by leptomycin B suggesting a CRM1-dependent export mechanism. This assumption is supported by the localization of a putative CRM1 binding site at the C-terminus of the AR protein. The results suggest that GSK-3beta is an important element not only in AR stability but also significantly alters nuclear translocation of the AR, thereby modulating the androgenic response of human PCa cells.
Collapse
Affiliation(s)
- Stefanie V Schütz
- Institute of General Zoology and Endocrinology, Ulm University, Albert Einstein Allee 11, 89069 Ulm, Germany
| | | | | |
Collapse
|
33
|
Grisouard J, Mayer D. Specific involvement of glycogen synthase kinase-3 in the function and activity of sex steroid hormone receptors reveals the complexity of their regulation. J Steroid Biochem Mol Biol 2009; 117:87-92. [PMID: 19703560 DOI: 10.1016/j.jsbmb.2009.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 08/12/2009] [Accepted: 08/13/2009] [Indexed: 12/15/2022]
Abstract
Protein kinases represent key nodes for the integration of multiple intracellular signalling pathways, resulting in modulation of both ligand-dependent and ligand-independent mechanisms of sex steroid receptor (sSR) signalling cascades. The proline-directed Ser/Thr kinases including mitogen-activated protein kinases and cyclin dependent kinases were especially reported to contribute to the function and activity of sSRs. The relevant effects of these kinases are well-documented but the impact of glycogen synthase kinase-3 (GSK-3), another member of this kinase family, has been underestimated. Indeed, the specific role of GSK-3 regarding the different sSRs will help to understand further the complexity of sSR signalling. So far, AR and ERalpha were identified as GSK-3 substrates. Additionally, the docking properties of GSK-3 were demonstrated to play a crucial role in sSR signal transduction. Reciprocally, GSK-3 was described as a potential target of non-genomic effects of sSRs. Therefore, GSK-3 regulates and is regulated by sSRs. This review focuses on the emerging and promising involvements of GSK-3 regarding the signalling cascade of the respective sSRs. This review represents a necessary complement of information to highlight the importance of GSK-3 regarding sSR function and activity.
Collapse
Affiliation(s)
- Jean Grisouard
- Hormones and Signal Transduction Group, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany
| | | |
Collapse
|
34
|
Robertson BW, Chellaiah MA. Osteopontin induces beta-catenin signaling through activation of Akt in prostate cancer cells. Exp Cell Res 2009; 316:1-11. [PMID: 19850036 DOI: 10.1016/j.yexcr.2009.10.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Revised: 10/13/2009] [Accepted: 10/15/2009] [Indexed: 01/13/2023]
Abstract
Secretion of osteopontin (OPN) by cancer cells is a known mediator of tumorigenesis and cancer progression in both experimental and clinical studies. Our work demonstrates that OPN can activate Akt, an important step in cancer progression. Both ILK and PI3K are integral proteins in the OPN/Akt pathway, as inhibition of either kinase leads to a loss of OPN-mediated Akt activation. Subsequent to OPN-induced Akt activation, we observe inactivation of GSK-3beta, a regulator of beta-catenin. Osteopontin stimulation leads to an overall increase in beta-catenin protein levels with a resultant transfer of beta-catenin to the nucleus. Through the nuclear import of beta-catenin, OPN increases both the transcription and protein levels of MMP-7 and CD44, which are known TCF/LEF transcription targets. This work describes an important aspect of cancer progression induced by OPN.
Collapse
Affiliation(s)
- Brian W Robertson
- Department of Oncology and Diagnostic Sciences, Dental School, University of Maryland, Baltimore, MD 21201, USA
| | | |
Collapse
|
35
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2009; 16:260-77. [PMID: 19390324 DOI: 10.1097/med.0b013e32832c937e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Xu K, Shimelis H, Linn DE, Jiang R, Yang X, Sun F, Guo Z, Chen H, Li W, Chen H, Kong X, Melamed J, Fang S, Xiao Z, Veenstra TD, Qiu Y. Regulation of androgen receptor transcriptional activity and specificity by RNF6-induced ubiquitination. Cancer Cell 2009; 15:270-82. [PMID: 19345326 PMCID: PMC2848969 DOI: 10.1016/j.ccr.2009.02.021] [Citation(s) in RCA: 188] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Revised: 12/08/2008] [Accepted: 02/23/2009] [Indexed: 01/16/2023]
Abstract
The androgen receptor (AR) plays a critical role in prostate cancer. We have identified a ubiquitin E3 ligase, RNF6, as an AR-associated protein in a proteomic screen. RNF6 induces AR ubiquitination and promotes AR transcriptional activity. Specific knockdown of RNF6 or mutation of RNF6-induced ubiquitination acceptor sites on AR selectively alters expression of a subset of AR target genes and diminishes recruitment of AR and its coactivators to androgen-responsive elements present in the regulatory region of these genes. Furthermore, RNF6 is overexpressed in hormone-refractory human prostate cancer tissues and required for prostate cancer cell growth under androgen-depleted conditions. Our data suggest that RNF6-induced ubiquitination may regulate AR transcriptional activity and specificity through modulating cofactor recruitment.
Collapse
Affiliation(s)
- Kexin Xu
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Hermela Shimelis
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Douglas E. Linn
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Richeng Jiang
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Xi Yang
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Feng Sun
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Zhiyong Guo
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Hege Chen
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Wei Li
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Hegang Chen
- Department of Epidemiology & Preventive Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Xiangtian Kong
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Jonathan Melamed
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Shengyun Fang
- Medical Biotechnology Center, University of Maryland Biotechnology Institute, Baltimore, MD 21201
| | - Zhen Xiao
- Laboratory of Proteomics and Analytical Technologies, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702
| | - Timothy D. Veenstra
- Laboratory of Proteomics and Analytical Technologies, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702
| | - Yun Qiu
- Department of Pharmacology & Experimental Therapeutics and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
37
|
Kung HJ, Evans CP. Oncogenic activation of androgen receptor. Urol Oncol 2009; 27:48-52. [PMID: 19111798 DOI: 10.1016/j.urolonc.2008.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 06/17/2008] [Accepted: 06/20/2008] [Indexed: 12/29/2022]
Abstract
BACKGROUND There is considerable evidence implicating the aberrant activation or "reactivation" of androgen receptor in the course of androgen-ablation therapy as a potential cause for the development of castration-resistant prostate cancer. Several non-mutually exclusive mechanisms including the inappropriate activation of androgen receptor (AR) by non-steroids have been postulated. The present work is aimed to understand the role of neuropeptides released by neuroendocrine transdifferentiated prostate cancer cells in the aberrant activation of AR. OBJECTIVES The study was designed to study how neuropeptides such as gastrin-releasing peptide activate AR and to define the crucial signal pathways involved, in the hope to identify therapeutic targets. METHODS AND MATERIALS Androgen-dependent LNCaP cell line was used to study the effects of bombesin/gastrin-releasing peptide on the growth of the cell line and the transactivation of AR. The neuropeptide was either added to the media or introduced as a transgene in LNCaP cells to study its paracrine or autocrine effect on LNCaP growth under androgen-deprived conditions. The activation of AR was monitored by reporter assay, chromatin immunoprecipitation (ChIP) of AR, translocation into the nucleus and cDNA microarray of the AR response genes. RESULTS Bombesin/gastrin releasing peptides induce androgen-independent growth of LNCaP in vitro and in vivo. It does so by activating AR, which is accompanied by the activation of Src tyrosine kinase and its target c-myc oncogene. The bombesin or Src-activated AR induces an overlapping set of AR response genes as androgen, but they also a unique set of genes. Intriguingly, the Src-activated and androgen-bound ARs differ in their binding specificity toward AR response elements, indicating the receptors activated by these 2 mechanisms are not conformationally identical. Finally, Src inhibitor was shown to effectively block the activation of AR and the growth effects induced by bombesin. CONCLUSION The results showed that AR can be activated by neuropeptide, a ligand for G-protein coupled receptor, in the absence of androgen. The activation goes through Src-tyrosine kinase pathway, and tyrosine kinase inhibitor is a potentially useful adjunctive therapy during androgen ablation.
Collapse
Affiliation(s)
- Hsing-Jien Kung
- Department of Basic Sciences, University of California, Davis Cancer Center, Sacramento, CA 95817, USA.
| | | |
Collapse
|
38
|
Li Y, Wang Z, Kong D, Li R, Sarkar SH, Sarkar FH. Regulation of Akt/FOXO3a/GSK-3beta/AR signaling network by isoflavone in prostate cancer cells. J Biol Chem 2008; 283:27707-27716. [PMID: 18687691 PMCID: PMC2562074 DOI: 10.1074/jbc.m802759200] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 06/23/2008] [Indexed: 01/03/2023] Open
Abstract
We have previously shown that genistein could inhibit Akt activation and down-regulate AR (androgen receptor) and PSA (prostate-specific antigen) expression in prostate cancer (PCa) cells. However, pure genistein showed increased lymph node metastasis in an animal model, but such an adverse effect was not seen with isoflavone, suggesting that further mechanistic studies are needed for elucidating the role of isoflavone in PCa. It is known that FOXO3a and GSK-3beta, targets of Akt, regulate cell proliferation and apoptosis. Moreover, FOXO3a, GSK-3beta, and Src are AR regulators and regulate transactivation of AR, mediating the development and progression of PCa. Therefore, we investigated the molecular effects of isoflavone on the Akt/FOXO3a/GSK-3beta/AR signaling network in hormone-sensitive LNCaP and hormone-insensitive C4-2B PCa cells. We found that isoflavone inhibited the phosphorylation of Akt and FOXO3a, regulated the phosphorylation of Src, and increased the expression of GSK-3beta, leading to the down-regulation of AR and its target gene PSA. We also found that isoflavone inhibited AR nuclear translocation and promoted FOXO3a translocation to the nucleus. By electrophoretic mobility shift assay and chromatin immunoprecipitation assay, we found that isoflavone inhibited FOXO3a binding to the promoter of AR and increased FOXO3a binding to the p27(KIP1) promoter, resulting in the alteration of AR and p27(KIP1) expression, the inhibition of cell proliferation, and the induction of apoptosis in both androgen-sensitive and -insensitive PCa cells. These results suggest that isoflavone-induced inhibition of cell proliferation and induction of apoptosis are partly mediated through the regulation of the Akt/FOXO3a/GSK-3beta/AR signaling network. In conclusion, our data suggest that isoflavone could be useful for the prevention and/or treatment of PCa.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Zhiwei Wang
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Dejuan Kong
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Ran Li
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Sanila H Sarkar
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Fazlul H Sarkar
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201.
| |
Collapse
|