1
|
Meng T, Jung D, Cai XH, Lu ZQ, Yu JB, Qi TY, Meng FY, Ruan MZ, Duan JX. Characterization of AST-001 non-clinical pharmacokinetics: A novel selective AKR1C3-activated prodrug in mice, rats, and cynomolgus monkeys. Biopharm Drug Dispos 2024; 45:83-92. [PMID: 38492211 DOI: 10.1002/bdd.2385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/03/2024] [Accepted: 01/29/2024] [Indexed: 03/18/2024]
Abstract
AST-001 is a chemically synthesized inactive nitrogen mustard prodrug that is selectively cleaved to a cytotoxic aziridine (AST-2660) via aldo-keto reductase family 1 member C3 (AKR1C3). The purpose of this study was to investigate the pharmacokinetics and tissue distribution of the prodrug, AST-001, and its active metabolite, AST-2660, in mice, rats, and monkeys. After single and once daily intravenous bolus doses of 1.5, 4.5, and 13.5 mg/kg AST-001 to Sprague-Dawley rats and once daily 1 h intravenous infusions of 0.5, 1.5, and 4.5 mg/kg AST-001 to cynomolgus monkeys, AST-001 exhibited dose-dependent pharmacokinetics and reached peak plasma levels at the end of the infusion. No significant accumulation and gender differences were observed after 7 days of repeated dosing. In rats, the half-life of AST-001 was dose independent and ranged from 4.89 to 5.75 h. In cynomolgus monkeys, the half-life of AST-001 was from 1.66 to 5.56 h and increased with dose. In tissue distribution studies conducted in Sprague-Dawley rats and in liver cancer PDX models in female athymic nude mice implanted with LI6643 or LI6280 HepG2-GFP tumor fragments, AST-001 was extensively distributed to selected tissues. Following a single intravenous dose, AST-001 was not excreted primarily as the prodrug, AST-001 or the metabolite AST-2660 in the urine, feces, and bile. A comprehensive analysis of the preclinical data and inter-species allometric scaling were used to estimate the pharmacokinetic parameters of AST-001 in humans and led to the recommendation of a starting dose of 5 mg/m2 in the first-in-human dose escalation study.
Collapse
Affiliation(s)
- Teng Meng
- Ascentawits Pharmaceuticals, Ltd, Shenzhen, China
| | - Donald Jung
- Ascentawits Pharmaceuticals, Ltd, Shenzhen, China
| | | | | | - Ji-Bing Yu
- Ascentawits Pharmaceuticals, Ltd, Shenzhen, China
| | - Tian-Yang Qi
- Ascentawits Pharmaceuticals, Ltd, Shenzhen, China
| | | | | | | |
Collapse
|
2
|
Feng Y, Huang Z, Ma X, Zong X, Wu CY, Lee RH, Lin HW, Hamblin MR, Zhang Q. Activation of testosterone-androgen receptor mediates cerebrovascular protection by photobiomodulation treatment in photothrombosis-induced stroke rats. CNS Neurosci Ther 2024; 30:e14574. [PMID: 38421088 PMCID: PMC10851319 DOI: 10.1111/cns.14574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 03/02/2024] Open
Abstract
RATIONALE Numerous epidemiological studies have reported a link between low testosterone levels and an increased risk of cerebrovascular disease in men. However, there is ongoing controversy surrounding testosterone replacement therapy due to potential side effects. PBMT has been demonstrated to improve cerebrovascular function and promote testosterone synthesis in peripheral tissues. Despite this, the molecular mechanisms that could connect PBMT with testosterone and vascular function in the brain of photothrombosis (PT)-induced stroke rats remain largely unknown. METHODS We measured behavioral performance, cerebral blood flow (CBF), vascular permeability, and the expression of vascular-associated and apoptotic proteins in PT-induced stroke rats treated with flutamide and seven consecutive days of PBM treatment (350 mW, 808 nM, 2 min/day). To gain further insights into the mechanism of PBM on testosterone synthesis, we used testosterone synthesis inhibitors to study their effects on bEND.3 cells. RESULTS We showed that PT stroke caused a decrease in cerebrovascular testosterone concentration, which was significantly increased by 7-day PBMT (808 nm, 350 mW/cm2 , 42 J/cm2 ). Furthermore, PBMT significantly increased cerebral blood flow (CBF) and the expression of vascular-associated proteins, while inhibiting vascular permeability and reducing endothelial cell apoptosis. This ultimately mitigated behavioral deficits in PT stroke rats. Notably, treatment with the androgen receptor antagonist flutamide reversed the beneficial effects of PBMT. Cellular experiments confirmed that PBMT inhibited cell apoptosis and increased vascular-associated protein expression in brain endothelial cell line (bEnd.3) subjected to oxygen-glucose deprivation (OGD). However, these effects were inhibited by flutamide. Moreover, mechanistic studies revealed that PBMT-induced testosterone synthesis in bEnd.3 cells was partly mediated by 17β-hydroxysteroid dehydrogenase 5 (17β-HSD5). CONCLUSIONS Our study provides evidence that PBMT attenuates cerebrovascular injury and behavioral deficits associated with testosterone/AR following ischemic stroke. Our findings suggest that PBMT may be a promising alternative approach for managing cerebrovascular diseases.
Collapse
Affiliation(s)
- Yu Feng
- Department of NeurologyLouisiana State University Health Sciences CenterShreveportLouisianaUSA
| | - Zhihai Huang
- Department of NeurologyLouisiana State University Health Sciences CenterShreveportLouisianaUSA
| | - Xiaohui Ma
- Department of NeurologyLouisiana State University Health Sciences CenterShreveportLouisianaUSA
| | - Xuemei Zong
- Department of NeurologyLouisiana State University Health Sciences CenterShreveportLouisianaUSA
| | - Celeste Yin‐Chieh Wu
- Department of NeurologyLouisiana State University Health Sciences CenterShreveportLouisianaUSA
| | - Reggie Hui‐Chao Lee
- Department of NeurologyLouisiana State University Health Sciences CenterShreveportLouisianaUSA
| | - Hung Wen Lin
- Department of NeurologyLouisiana State University Health Sciences CenterShreveportLouisianaUSA
| | - Michael R. Hamblin
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Quanguang Zhang
- Department of NeurologyLouisiana State University Health Sciences CenterShreveportLouisianaUSA
| |
Collapse
|
3
|
Wang YJ, Xie XL, Liu HQ, Tian H, Jiang XY, Zhang JN, Chen SX, Liu T, Wang SL, Zhou X, Jin XX, Liu SM, Jiang HQ. Prostaglandin F 2α synthase promotes oxaliplatin resistance in colorectal cancer through prostaglandin F 2α-dependent and F 2α-independent mechanism. World J Gastroenterol 2023; 29:5452-5470. [PMID: 37900995 PMCID: PMC10600807 DOI: 10.3748/wjg.v29.i39.5452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Oxaliplatin (Oxa) is the first-line chemotherapy drug for colorectal cancer (CRC), and Oxa resistance is crucial for treatment failure. Prostaglandin F2α synthase (PGF2α) (PGFS), an enzyme that catalyzes the production of PGF2α, is involved in the proliferation and growth of a variety of tumors. However, the role of PGFS in Oxa resistance in CRC remains unclear. AIM To explore the role and related mechanisms of PGFS in mediating Oxa resistance in CRC. METHODS The PGFS expression level was examined in 37 pairs of CRC tissues and paracancerous tissues at both the mRNA and protein levels. Overexpression or knockdown of PGFS was performed in CRC cell lines with acquired Oxa resistance (HCT116-OxR and HCT8-OxR) and their parental cell lines (HCT116 and HCT8) to assess its influence on cell proliferation, chemoresistance, apoptosis, and DNA damage. For determination of the underlying mechanisms, CRC cells were examined for platinum-DNA adducts and reactive oxygen species (ROS) levels in the presence of a PGFS inhibitor or its products. RESULTS Both the protein and mRNA levels of PGFS were increased in the 37 examined CRC tissues compared to the adjacent normal tissues. Oxa induced PGFS expression in the parental HCT116 and HCT8 cells in a dose-dependent manner. Furthermore, overexpression of PGFS in parental CRC cells significantly attenuated Oxa-induced proliferative suppression, apoptosis, and DNA damage. In contrast, knockdown of PGFS in Oxa-resistant HCT116 and HCT8 cells (HCT116-OxR and HCT8-OxR) accentuated the effect of Oxa treatment in vitro and in vivo. The addition of the PGFS inhibitor indomethacin enhanced the cytotoxicity caused by Oxa. Treatment with the PGFS-catalyzed product PGF2α reversed the effect of PGFS knockdown on Oxa sensitivity. Interestingly, PGFS inhibited the formation of platinum-DNA adducts in a PGF2α-independent manner. PGF2α exerts its protective effect against DNA damage by reducing ROS levels. CONCLUSION PGFS promotes resistance to Oxa in CRC via both PGF2α-dependent and PGF2α-independent mechanisms.
Collapse
Affiliation(s)
- Yi-Jun Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Xiao-Li Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Hong-Qun Liu
- Liver Unit, University of Calgary, Calgary T1W0K6, Canada
| | - Hui Tian
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Xiao-Yu Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Jiu-Na Zhang
- Department of Gastroenterology, The Affiliated Hospital of Hebei Engineering University, Handan 056000, Hebei Province, China
| | - Sheng-Xiong Chen
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Ting Liu
- Department of Gastroenterology, The First Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Shu-Ling Wang
- Department of Gastroenterology, The First Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Xue Zhou
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Xiao-Xu Jin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Shi-Mao Liu
- Department of Gastroenterology, Hebei Youfu Hospital, Shijiazhuang 050000, Hebei Province, China
| | - Hui-Qing Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
4
|
Maddeboina K, Jonnalagadda SK, Morsy A, Duan L, Chhonker YS, Murry DJ, Penning TM, Trippier PC. Aldo-Keto Reductase 1C3 Inhibitor Prodrug Improves Pharmacokinetic Profile and Demonstrates In Vivo Efficacy in a Prostate Cancer Xenograft Model. J Med Chem 2023; 66:9894-9915. [PMID: 37428858 DOI: 10.1021/acs.jmedchem.3c00732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Aldo-keto reductase 1C3 (AKR1C3) is overexpressed in castration-resistant prostate cancer where it acts to drive proliferation and aggressiveness by producing androgens. The reductive action of the enzyme leads to chemoresistance development against various clinical antineoplastics across a range of cancers. Herein, we report the continued optimization of selective AKR1C3 inhibitors and the identification of 5r, a potent AKR1C3 inhibitor (IC50 = 51 nM) with >1216-fold selectivity for AKR1C3 over closely related isoforms. Due to the cognizance of the poor pharmacokinetics associated with free carboxylic acids, a methyl ester prodrug strategy was pursued. The prodrug 4r was converted to free acid 5r in vitro in mouse plasma and in vivo. The in vivo pharmacokinetic evaluation revealed an increase in systemic exposure and increased the maximum 5r concentration compared to direct administration of the free acid. The prodrug 4r demonstrated a dose-dependent effect to reduce the tumor volume of 22Rv1 prostate cancer xenografts without observed toxicity.
Collapse
Affiliation(s)
- Krishnaiah Maddeboina
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Sravan K Jonnalagadda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Ahmed Morsy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Ling Duan
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yashpal S Chhonker
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Daryl J Murry
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| |
Collapse
|
5
|
Gao D, Liu R, Lv Y, Feng Y, Hong F, Xu X, Hu J, He A, Yang Y. A novel ferroptosis-related gene signature for predicting prognosis in multiple myeloma. Front Oncol 2023; 13:999688. [PMID: 36845727 PMCID: PMC9950937 DOI: 10.3389/fonc.2023.999688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/26/2023] [Indexed: 02/12/2023] Open
Abstract
Background Multiple myeloma (MM) is a highly malignant hematological tumor with a poor overall survival (OS). Due to the high heterogeneity of MM, it is necessary to explore novel markers for the prognosis prediction for MM patients. Ferroptosis is a form of regulated cell death, playing a critical role in tumorigenesis and cancer progression. However, the predictive role of ferroptosis-related genes (FRGs) in MM prognosis remains unknown. Methods This study collected 107 FRGs previously reported and utilized the least absolute shrinkage and selection operator (LASSO) cox regression model to construct a multi-genes risk signature model upon FRGs. The ESTIMATE algorithm and immune-related single-sample gene set enrichment analysis (ssGSEA) were carried out to evaluate immune infiltration level. Drug sensitivity was assessed based on the Genomics of Drug Sensitivity in Cancer database (GDSC). Then the synergy effect was determined with Cell counting kit-8 (CCK-8) assay and SynergyFinder software. Results A 6-gene prognostic risk signature model was constructed, and MM patients were divided into high and low risk groups. Kaplan-Meier survival curves showed that patients in the high risk group had significantly reduced OS compared with patients in the low risk group. Besides, the risk score was an independent predictor for OS. Receiver operating characteristic (ROC) curve analysis confirmed the predictive capacity of the risk signature. Combination of risk score and ISS stage had better prediction performance. Enrichment analysis revealed immune response, MYC, mTOR, proteasome and oxidative phosphorylation were enriched in high risk MM patients. We found high risk MM patients had lower immune scores and immune infiltration levels. Moreover, further analysis found that MM patients in high risk group were sensitive to bortezomib and lenalidomide. At last, the results of the in vitro experiment showed that ferroptosis inducers (RSL3 and ML162) may synergistically enhance the cytotoxicity of bortezomib and lenalidomide against MM cell line RPMI-8226. Conclusion This study provides novel insights into roles of ferroptosis in MM prognosis prediction, immune levels and drug sensitivity, which complements and improves current grading systems.
Collapse
Affiliation(s)
- Dandan Gao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yang Lv
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yuandong Feng
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Fei Hong
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xuezhu Xu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jinsong Hu
- Department of Cell Biology and Genetics, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China,National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China,*Correspondence: Aili He, ; Yun Yang,
| | - Yun Yang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China,*Correspondence: Aili He, ; Yun Yang,
| |
Collapse
|
6
|
Barnard M, Mostaghel EA, Auchus RJ, Storbeck KH. The role of adrenal derived androgens in castration resistant prostate cancer. J Steroid Biochem Mol Biol 2020; 197:105506. [PMID: 31672619 PMCID: PMC7883395 DOI: 10.1016/j.jsbmb.2019.105506] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/17/2019] [Accepted: 10/22/2019] [Indexed: 01/02/2023]
Abstract
Castration resistant prostate cancer (CRPC) remains androgen dependant despite castrate levels of circulating testosterone following androgen deprivation therapy, the first line of treatment for advanced metstatic prostate cancer. CRPC is characterized by alterations in the expression levels of steroidgenic enzymes that enable the tumour to derive potent androgens from circulating adrenal androgen precursors. Intratumoral androgen biosynthesis leads to the localized production of both canonical androgens such as 5α-dihydrotestosterone (DHT) as well as less well characterized 11-oxygenated androgens, which until recently have been overlooked in the context of CRPC. In this review we discuss the contribution of both canonical and 11-oxygenated androgen precursors to the intratumoral androgen pool in CRPC. We present evidence that CRPC remains androgen dependent and discuss the alterations in steroidogenic enzyme expression and how these affect the various pathways to intratumoral androgen biosynthesis. Finally we summarize the current treatment strategies for targeting adrenal derived androgen biosynthesis.
Collapse
Affiliation(s)
- Monique Barnard
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Elahe A Mostaghel
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA; Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Richard J Auchus
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
7
|
Penning TM. AKR1C3 (type 5 17β-hydroxysteroid dehydrogenase/prostaglandin F synthase): Roles in malignancy and endocrine disorders. Mol Cell Endocrinol 2019; 489:82-91. [PMID: 30012349 PMCID: PMC6422768 DOI: 10.1016/j.mce.2018.07.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/12/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022]
Abstract
Aldo-Keto-Reductase 1C3 (type 5 17β-hydroxysteroid dehydrogenase (HSD)/prostaglandin (PG) F2α synthase) is the only 17β-HSD that is not a short-chain dehydrogenase/reductase. By acting as a 17-ketosteroid reductase, AKR1C3 produces potent androgens in peripheral tissues which activate the androgen receptor (AR) or act as substrates for aromatase. AKR1C3 is implicated in the production of androgens in castration-resistant prostate cancer (CRPC) and polycystic ovarian syndrome; and is implicated in the production of aromatase substrates in breast cancer. By acting as an 11-ketoprostaglandin reductase, AKR1C3 generates 11β-PGF2α to activate the FP receptor and deprives peroxisome proliferator activator receptorγ of its putative PGJ2 ligands. These growth stimulatory signals implicate AKR1C3 in non-hormonal dependent malignancies e.g. acute myeloid leukemia (AML). AKR1C3 moonlights by acting as a co-activator of the AR and stabilizes ubiquitin ligases. AKR1C3 inhibitors have been used clinically for CRPC and AML and can be used to probe its pluripotency.
Collapse
Affiliation(s)
- Trevor M Penning
- Department of Systems Pharmacology and Translational Therapeutics and Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, 1315 BRBII/III 421 Curie Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
8
|
Miyazaki Y, Teramoto Y, Shibuya S, Goto T, Okasho K, Mizuno K, Uegaki M, Yoshikawa T, Akamatsu S, Kobayashi T, Ogawa O, Inoue T. Consecutive Prostate Cancer Specimens Revealed Increased Aldo⁻Keto Reductase Family 1 Member C3 Expression with Progression to Castration-Resistant Prostate Cancer. J Clin Med 2019; 8:E601. [PMID: 31052459 PMCID: PMC6571723 DOI: 10.3390/jcm8050601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 01/03/2023] Open
Abstract
Aldo-keto reductase family 1 member C3 (AKR1C3) is an enzyme in the steroidogenesis pathway, especially in formation of testosterone and dihydrotestosterone, and is believed to have a key role in promoting prostate cancer (PCa) progression, particularly in castration-resistant prostate cancer (CRPC). This study aims to compare the expression level of AKR1C3 between benign prostatic epithelium and cancer cells, and among hormone-naïve prostate cancer (HNPC) and CRPC from the same patients, to understand the role of AKR1C3 in PCa progression. Correlation of AKR1C3 immunohistochemical expression between benign and cancerous epithelia in 134 patient specimens was analyzed. Additionally, correlation between AKR1C3 expression and prostate-specific antigen (PSA) progression-free survival (PFS) after radical prostatectomy was analyzed. Furthermore, we evaluated the consecutive prostate samples derived from 11 patients both in the hormone-naïve and castration-resistant states. AKR1C3 immunostaining of cancer epithelium was significantly stronger than that of the benign epithelia in patients with localized HNPC (p < 0.0001). High AKR1C3 expression was an independent factor of poor PSA PFS (p = 0.032). Moreover, AKR1C3 immunostaining was significantly stronger in CRPC tissues than in HNPC tissues in the same patients (p = 0.0234). Our findings demonstrate that AKR1C3 is crucial in PCa progression.
Collapse
Affiliation(s)
- Yu Miyazaki
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Yuki Teramoto
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto 606-8507, Japan.
| | - Shinsuke Shibuya
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto 606-8507, Japan.
| | - Takayuki Goto
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Kosuke Okasho
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Kei Mizuno
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Masayuki Uegaki
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Takeshi Yoshikawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Shusuke Akamatsu
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Takahiro Inoue
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| |
Collapse
|
9
|
Evans K, Duan J, Pritchard T, Jones CD, McDermott L, Gu Z, Toscan CE, El-Zein N, Mayoh C, Erickson SW, Guo Y, Meng F, Jung D, Rathi KS, Roberts KG, Mullighan CG, Shia CS, Pearce T, Teicher BA, Smith MA, Lock RB. OBI-3424, a Novel AKR1C3-Activated Prodrug, Exhibits Potent Efficacy against Preclinical Models of T-ALL. Clin Cancer Res 2019; 25:4493-4503. [PMID: 31015346 DOI: 10.1158/1078-0432.ccr-19-0551] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/19/2019] [Accepted: 04/17/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE OBI-3424 is a highly selective prodrug that is converted by aldo-keto reductase family 1 member C3 (AKR1C3) to a potent DNA-alkylating agent. OBI-3424 has entered clinical testing for hepatocellular carcinoma and castrate-resistant prostate cancer, and it represents a potentially novel treatment for acute lymphoblastic leukemia (ALL). EXPERIMENTAL DESIGN We assessed AKR1C3 expression by RNA-Seq and immunoblotting, and evaluated the in vitro cytotoxicity of OBI-3424. We investigated the pharmacokinetics of OBI-3424 in mice and nonhuman primates, and assessed the in vivo efficacy of OBI-3424 against a large panel of patient-derived xenografts (PDX). RESULTS AKR1C3 mRNA expression was significantly higher in primary T-lineage ALL (T-ALL; n = 264) than B-lineage ALL (B-ALL; n = 1,740; P < 0.0001), and OBI-3424 exerted potent cytotoxicity against T-ALL cell lines and PDXs. In vivo, OBI-3424 significantly prolonged the event-free survival (EFS) of nine of nine ALL PDXs by 17.1-77.8 days (treated/control values 2.5-14.0), and disease regression was observed in eight of nine PDXs. A significant reduction (P < 0.0001) in bone marrow infiltration at day 28 was observed in four of six evaluable T-ALL PDXs. The importance of AKR1C3 in the in vivo response to OBI-3424 was verified using a B-ALL PDX that had been lentivirally transduced to stably overexpress AKR1C3. OBI-3424 combined with nelarabine resulted in prolongation of mouse EFS compared with each single agent alone in two T-ALL PDXs. CONCLUSIONS OBI-3424 exerted profound in vivo efficacy against T-ALL PDXs derived predominantly from aggressive and fatal disease, and therefore may represent a novel treatment for aggressive and chemoresistant T-ALL in an AKR1C3 biomarker-driven clinical trial.
Collapse
Affiliation(s)
- Kathryn Evans
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - JianXin Duan
- Ascentawits Pharmaceuticals, Ltd, Nanshan Shenzhen, China
| | - Tara Pritchard
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Connor D Jones
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Lisa McDermott
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Zhaohui Gu
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Cara E Toscan
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Narimanne El-Zein
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | | | - Yuelong Guo
- RTI International, Research Triangle Park, North Carolina
| | - Fanying Meng
- Ascentawits Pharmaceuticals, Ltd, Nanshan Shenzhen, China
| | - Donald Jung
- Ascentawits Pharmaceuticals, Ltd, Nanshan Shenzhen, China
| | - Komal S Rathi
- Division of Oncology and Center for Childhood Cancer Research, Department of Biomedical and Health Informatics and Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kathryn G Roberts
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles G Mullighan
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | | | | | | | - Richard B Lock
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Sydney, Sydney, Australia.
| |
Collapse
|
10
|
Penning TM, Wangtrakuldee P, Auchus RJ. Structural and Functional Biology of Aldo-Keto Reductase Steroid-Transforming Enzymes. Endocr Rev 2019; 40:447-475. [PMID: 30137266 PMCID: PMC6405412 DOI: 10.1210/er.2018-00089] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/05/2018] [Indexed: 12/19/2022]
Abstract
Aldo-keto reductases (AKRs) are monomeric NAD(P)(H)-dependent oxidoreductases that play pivotal roles in the biosynthesis and metabolism of steroids in humans. AKR1C enzymes acting as 3-ketosteroid, 17-ketosteroid, and 20-ketosteroid reductases are involved in the prereceptor regulation of ligands for the androgen, estrogen, and progesterone receptors and are considered drug targets to treat steroid hormone-dependent malignancies and endocrine disorders. In contrast, AKR1D1 is the only known steroid 5β-reductase and is essential for bile-acid biosynthesis, the generation of ligands for the farnesoid X receptor, and the 5β-dihydrosteroids that have their own biological activity. In this review we discuss the crystal structures of these AKRs, their kinetic and catalytic mechanisms, AKR genomics (gene expression, splice variants, polymorphic variants, and inherited genetic deficiencies), distribution in steroid target tissues, roles in steroid hormone action and disease, and inhibitor design.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania
| | - Phumvadee Wangtrakuldee
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine and Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, Michigan
| |
Collapse
|
11
|
Storbeck KH, Mostaghel EA. Canonical and Noncanonical Androgen Metabolism and Activity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:239-277. [PMID: 31900912 DOI: 10.1007/978-3-030-32656-2_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Androgens are critical drivers of prostate cancer. In this chapter we first discuss the canonical pathways of androgen metabolism and their alterations in prostate cancer progression, including the classical, backdoor and 5α-dione pathways, the role of pre-receptor DHT metabolism, and recent findings on oncogenic splicing of steroidogenic enzymes. Next, we discuss the activity and metabolism of non-canonical 11-oxygenated androgens that can activate wild-type AR and are less susceptible to glucuronidation and inactivation than the canonical androgens, thereby serving as an under-recognized reservoir of active ligands. We then discuss an emerging literature on the potential non-canonical role of androgen metabolizing enzymes in driving prostate cancer. We conclude by discussing the potential implications of these findings for prostate cancer progression, particularly in context of new agents such as abiraterone and enzalutamide, which target the AR-axis for prostate cancer therapy, including mechanisms of response and resistance and implications of these findings for future therapy.
Collapse
Affiliation(s)
- Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Elahe A Mostaghel
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA. .,Department of Medicine, University of Washington, Seattle, WA, USA. .,Geriatric Research, Education and Clinical Center S-182, VA Puget Sound Health Care System, Seattle, WA, USA.
| |
Collapse
|
12
|
Hu DG, Marri S, McKinnon RA, Mackenzie PI, Meech R. Deregulation of the Genes that Are Involved in Drug Absorption, Distribution, Metabolism, and Excretion in Hepatocellular Carcinoma. J Pharmacol Exp Ther 2018; 368:363-381. [PMID: 30578287 DOI: 10.1124/jpet.118.255018] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/19/2018] [Indexed: 12/25/2022] Open
Abstract
Genes involved in drug absorption, distribution, metabolism, and excretion (ADME) are called ADME genes. Currently, 298 genes that encode phase I and II drug metabolizing enzymes, transporters, and modifiers are designated as ADME genes by the PharmaADME Consortium. ADME genes are highly expressed in the liver and their levels can be influenced by liver diseases such as hepatocellular carcinoma (HCC). In this study, we obtained RNA-sequencing and microRNA (miRNA)-sequencing data from 371 HCC patients via The Cancer Genome Atlas liver hepatocellular carcinoma project and performed ADME gene-targeted differential gene expression analysis and expression correlation analysis. Two hundred thirty-three of the 298 ADME genes (78%) were expressed in HCC. Of these genes, almost one-quarter (58 genes) were significantly downregulated, while only 6% (15) were upregulated in HCC relative to healthy liver. Moreover, one-half (14/28) of the core ADME genes (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2E1, CYP3A4, NAT1, NAT2, UGT2B7, SLC22A1, SLCO1B1, and SLCO1B3) were downregulated. In addition, about one-half of the core ADME genes were positively correlated with each other and were also positively (AHR, ARNT, HNF4A, PXR, CAR, PPARA, and RXRA) or negatively (PPARD and PPARG) correlated with transcription factors known as ADME modifiers. Finally, we show that most miRNAs known to regulate core ADME genes are upregulated in HCC. Collectively, these data reveal 1) an extensive transcription factor-mediated ADME coexpression network in the liver that efficiently coordinates the metabolism and elimination of endogenous and exogenous compounds; and 2) a widespread deregulation of this network in HCC, most likely due to deregulation of both transcriptional and post-transcriptional (miRNA) pathways.
Collapse
Affiliation(s)
- Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer (D.G.H., R.A.M., P.I.M., R.M.), and Department of Molecular Medicine and Pathology (S.M.), Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Shashikanth Marri
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer (D.G.H., R.A.M., P.I.M., R.M.), and Department of Molecular Medicine and Pathology (S.M.), Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Ross A McKinnon
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer (D.G.H., R.A.M., P.I.M., R.M.), and Department of Molecular Medicine and Pathology (S.M.), Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Peter I Mackenzie
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer (D.G.H., R.A.M., P.I.M., R.M.), and Department of Molecular Medicine and Pathology (S.M.), Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Robyn Meech
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer (D.G.H., R.A.M., P.I.M., R.M.), and Department of Molecular Medicine and Pathology (S.M.), Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|
13
|
Prekovic S, van den Broeck T, Linder S, van Royen ME, Houtsmuller AB, Handle F, Joniau S, Zwart W, Claessens F. Molecular underpinnings of enzalutamide resistance. Endocr Relat Cancer 2018; 25:R545–R557. [PMID: 30306781 DOI: 10.1530/erc-17-0136] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Prostate cancer (PCa) is among the most common adult malignancies, and the second leading cause of cancer-related death in men. As PCa is hormone dependent, blockade of the androgen receptor (AR) signaling is an effective therapeutic strategy for men with advanced metastatic disease. The discovery of enzalutamide, a compound that effectively blocks the AR axis and its clinical application has led to a significant improvement in survival time. However, the effect of enzalutamide is not permanent, and resistance to treatment ultimately leads to development of lethal disease, for which there currently is no cure. This review will focus on the molecular underpinnings of enzalutamide resistance, bridging the gap between the preclinical and clinical research on novel therapeutic strategies for combating this lethal stage of prostate cancer.
Collapse
Affiliation(s)
- S Prekovic
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - T van den Broeck
- Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - S Linder
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M E van Royen
- Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
- Erasmus Optical Imaging Centre, Erasmus MC, Rotterdam, The Netherlands
| | - A B Houtsmuller
- Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
- Erasmus Optical Imaging Centre, Erasmus MC, Rotterdam, The Netherlands
| | - F Handle
- Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - S Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - W Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - F Claessens
- Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Deb S, Pham S, Ming DS, Chin MY, Adomat H, Hurtado-Coll A, Gleave ME, Guns EST. Characterization of Precursor-Dependent Steroidogenesis in Human Prostate Cancer Models. Cancers (Basel) 2018; 10:cancers10100343. [PMID: 30241348 PMCID: PMC6210088 DOI: 10.3390/cancers10100343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/17/2018] [Accepted: 09/17/2018] [Indexed: 11/17/2022] Open
Abstract
Castration-resistant prostate tumors acquire the independent capacity to generate androgens by upregulating steroidogenic enzymes or using steroid precursors produced by the adrenal glands for continued growth and sustainability. The formation of steroids was measured by liquid chromatography-mass spectrometry in LNCaP and 22Rv1 prostate cancer cells, and in human prostate tissues, following incubation with steroid precursors (22-OH-cholesterol, pregnenolone, 17-OH-pregnenolone, progesterone, 17-OH-progesterone). Pregnenolone, progesterone, 17-OH-pregnenolone, and 17-OH-progesterone increased C21 steroid (5-pregnan-3,20-dione, 5-pregnan-3,17-diol-20-one, 5-pregnan-3-ol-20-one) formation in the backdoor pathway, and demonstrated a trend of stimulating dihydroepiandrosterone or its precursors in the backdoor pathway in LNCaP and 22Rv1 cells. The precursors differentially affected steroidogenic enzyme messenger RNA (mRNA) expressions in the cell lines. The steroidogenesis following incubation of human prostate tissue with 17-OH-pregnenolone and progesterone produced trends similar to those observed in cell lines. Interestingly, the formation of C21 steroids from classical pathway was not stimulated but backdoor pathway steroids (e.g., 5-pregnan-3,20-dione, 5-pregnan-3-ol-20-one) were elevated following incubations with prostate tissues. Overall, C21 steroids were predominantly formed in the classical as well as backdoor pathways, and steroid precursors induced a diversion of steroidogenesis to the backdoor pathway in both cell lines and human prostate tissue, and influenced adaptive steroidogenesis to form C21 steroids.
Collapse
Affiliation(s)
- Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA.
| | - Steven Pham
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Dong-Sheng Ming
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Mei Yieng Chin
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Hans Adomat
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Antonio Hurtado-Coll
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Martin E Gleave
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| | - Emma S Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| |
Collapse
|
15
|
Study on the structure, vibrational analysis and molecular docking of fluorophenyl derivatives using FT-IR and density functional theory computations. J Mol Struct 2018. [DOI: 10.1016/j.molstruc.2018.03.070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
16
|
Schiffer L, Arlt W, Storbeck KH. Intracrine androgen biosynthesis, metabolism and action revisited. Mol Cell Endocrinol 2018; 465:4-26. [PMID: 28865807 PMCID: PMC6565845 DOI: 10.1016/j.mce.2017.08.016] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/28/2017] [Accepted: 08/28/2017] [Indexed: 12/19/2022]
Abstract
Androgens play an important role in metabolic homeostasis and reproductive health in both men and women. Androgen signalling is dependent on androgen receptor activation, mostly by testosterone and 5α-dihydrotestosterone. However, the intracellular or intracrine activation of C19 androgen precursors to active androgens in peripheral target tissues of androgen action is of equal importance. Intracrine androgen synthesis is often not reflected by circulating androgens but rather by androgen metabolites and conjugates. In this review we provide an overview of human C19 steroid biosynthesis including the production of 11-oxygenated androgens, their transport in circulation and uptake into peripheral tissues. We conceptualise the mechanisms of intracrinology and review the intracrine pathways of activation and inactivation in selected human tissues. The contribution of liver and kidney as organs driving androgen inactivation and renal excretion are also highlighted. Finally, the importance of quantifying androgen metabolites and conjugates to assess intracrine androgen production is discussed.
Collapse
Affiliation(s)
- Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Karl-Heinz Storbeck
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
17
|
Xia D, Lai DV, Wu W, Webb ZD, Yang Q, Zhao L, Yu Z, Thorpe JE, Disch BC, Ihnat MA, Jayaraman M, Dhanasekaran DN, Stratton KL, Cookson MS, Fung KM, Lin HK. Transition from androgenic to neurosteroidal action of 5α-androstane-3α, 17β-diol through the type A γ-aminobutyric acid receptor in prostate cancer progression. J Steroid Biochem Mol Biol 2018; 178:89-98. [PMID: 29155210 DOI: 10.1016/j.jsbmb.2017.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/10/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
Abstract
Androgen ablation is the standard of care prescribed to patients with advanced or metastatic prostate cancer (PCa) to slow down disease progression. Unfortunately, a majority of PCa patients under androgen ablation progress to castration-resistant prostate cancer (CRPC). Several mechanisms including alternative intra-prostatic androgen production and androgen-independent androgen receptor (AR) activation have been proposed for CRPC progression. Aldo-keto reductase family 1 member C3 (AKR1C3), a multi-functional steroid metabolizing enzyme, is specifically expressed in the cytoplasm of PCa cells; and positive immunoreactivity of the type A γ-aminobutyric acid receptor (GABAAR), an ionotropic receptor and ligand-gated ion channel, is detected on the membrane of PCa cells. We studied a total of 72 radical prostatectomy cases by immunohistochemistry, and identified that 21 cases exhibited positive immunoreactivities for both AKR1C3 and GABAAR. In the dual positive cancer cases, AKR1C3 and GABAAR subunit α1 were either expressed in the same cells or in neighboring cells. Among several possible substrates, AKR1C3 reduces 5α-dihydrotesterone (DHT) to form 5α-androstane-3α, 17β-diol (3α-diol). 3α-diol is a neurosteroid that acts as a positive allosteric modulator of the GABAAR in the central nervous system (CNS). We examined the hypothesis that 3α-diol-regulated pathological effects in the prostate are GABAAR-dependent, but are independent of the AR. In GABAAR-positive, AR-negative human PCa PC-3 cells, 3α-diol significantly stimulated cell growth in culture and the in ovo chorioallantoic membrane (CAM) xenograft model. 3α-diol also up-regulated expression of the epidermal growth factor (EGF) family of growth factors and activation of EGF receptor (EGFR) and Src as measured by quantitative polymerase chain reaction and immunoblotting, respectively. Inclusion of GABAAR antagonists reversed 3α-diol-stimulated tumor cell growth, expression of EGF family members, and activation of EGFR and Src to the level observed in untreated cells. Results from the present study suggest that 3α-diol may act as an alternative intra-prostatic neurosteroid that activates AR-independent PCa progression. The involvement of AKR1C3-mediated steroid metabolisms in modulating GABAAR activation and promoting PCa progression requires continued studies.
Collapse
Affiliation(s)
- Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Doan V Lai
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Weijuan Wu
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zachary D Webb
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Qing Yang
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lichao Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zhongxin Yu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jessica E Thorpe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma College of Pharmacy, OKC, OK 73117, USA
| | - Bryan C Disch
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma College of Pharmacy, OKC, OK 73117, USA
| | - Michael A Ihnat
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma College of Pharmacy, OKC, OK 73117, USA
| | | | - Danny N Dhanasekaran
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kelly L Stratton
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael S Cookson
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kar-Ming Fung
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pathology, Veterans Affairs Medical Center, Oklahoma City, Oklahoma, OK 73104, USA
| | - Hsueh-Kung Lin
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
18
|
Xin Z, Pu L, Gao W, Wang Y, Wei J, Shi T, Yao Z, Guo C. Riboflavin deficiency induces a significant change in proteomic profiles in HepG2 cells. Sci Rep 2017; 7:45861. [PMID: 28367977 PMCID: PMC5377456 DOI: 10.1038/srep45861] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/03/2017] [Indexed: 01/07/2023] Open
Abstract
Riboflavin deficiency is widespread in many regions over the world, especially in underdeveloped countries. In this study, we investigated the effects of riboflavin deficiency on protein expression profiles in HepG2 cells in order to provide molecular information for the abnormalities induced by riboflavin deficiency. HepG2 cells were cultured in media containing different concentrations of riboflavin. Changes of cell viability and apoptosis were assessed. A comparative proteomic analysis was performed using a label-free shotgun method with LC-MS/MS to investigate the global changes of proteomic profiles in response to riboflavin deficiency. Immunoblotting test was used to validate the results of proteomic approach. The cell viability and apoptosis tests showed that riboflavin was vital in maintaining the cytoactivity of HepG2 cells. The label-free proteomic analysis revealed that a total of 37 proteins showing differential expression (±2 fold, p < 0.05) were identified after riboflavin deficiency. Bioinformatics analysis indicated that the riboflavin deficiency caused an up-regulation of Parkinson's disease pathway, steroid catabolism, endoplasmic reticulum stress and apoptotic process, while the fatty acid metabolism, tricarboxylic citrate cycle, oxidative phosphorylation and iron metabolism were down-regulated. These findings provide a molecular basis for the elucidation of the effects caused by riboflavin deficiency.
Collapse
Affiliation(s)
- Zhonghao Xin
- Institute of Health and Environmental Medicine, Tianjin, 300050, China
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Lingling Pu
- Institute of Health and Environmental Medicine, Tianjin, 300050, China
| | - Weina Gao
- Institute of Health and Environmental Medicine, Tianjin, 300050, China
| | - Yawen Wang
- Institute of Health and Environmental Medicine, Tianjin, 300050, China
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Jingyu Wei
- Institute of Health and Environmental Medicine, Tianjin, 300050, China
| | - Tala Shi
- Institute of Health and Environmental Medicine, Tianjin, 300050, China
| | - Zhanxin Yao
- Institute of Health and Environmental Medicine, Tianjin, 300050, China
| | - Changjiang Guo
- Institute of Health and Environmental Medicine, Tianjin, 300050, China
| |
Collapse
|
19
|
Wadhwa B, Dumbre R. Achieving resistance specificity in prostate cancer. Chem Biol Interact 2016; 260:243-247. [PMID: 27720870 DOI: 10.1016/j.cbi.2016.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/03/2016] [Indexed: 11/24/2022]
Abstract
Prostate (CaP) cancer is the second-leading cause of cancer-related mortality in men in Western societies. Androgen receptor (AR) signaling is a critical survival pathway for prostate cancer cells, and androgen-deprivation therapy (ADT) remains the principal treatment for patients with locally advanced and metastatic disease. Although a majority of patients initially respond to ADT, most will eventually develop castrate resistance. The recent discovery that AR signaling persists during systemic castration via intratumoral production of androgens led to the development of novel anti-androgen therapies. Although these therapies effectively palliate symptoms and prolong life, metastatic castration-resistant prostate cancer remains incurable. Recent studies suggest that epithelial plasticity, which covers a range of changes in differentiation and cell behavior, with full epithelial integrity at one end and epithelial-mesenchymal Transition (EMT) as the full realization of a plasticity is regulated by microRNAs (miRNAs). MicroRNAs are involved in human tumourigenesis and are aberrantly expressed in CaP cell lines, xenografts and clinical tissues and is associated with enhanced survival signaling, proliferation, migration, invasion, integrin-mediated adhesion, EMT, and drug resistance. Due to the oncogenic or tumor suppressive properties of CaP-related miRNAs, they are likely to be of clinical use as therapeutic targets for prostate cancer treatment in the near future. This review summarizes our current understanding of CaP and castration-recurrent CaP (CR-CaP) to earlier studies that characterized ADT and the molecular mechanisms that facilitate the transition from androgen-stimulated CaP to CR-CaP.
Collapse
Affiliation(s)
- Bhumika Wadhwa
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India; Cancer Pharmacology Division, Indian Institute of Integrative Medicine, CSIR, Jammu 180001, India.
| | - Rashmi Dumbre
- Centre for Biotechnology, Pravara Institute of Medical Sciences, Loni, India
| |
Collapse
|
20
|
Adeniji A, Uddin MJ, Zang T, Tamae D, Wangtrakuldee P, Marnett LJ, Penning TM. Discovery of (R)-2-(6-Methoxynaphthalen-2-yl)butanoic Acid as a Potent and Selective Aldo-keto Reductase 1C3 Inhibitor. J Med Chem 2016; 59:7431-44. [PMID: 27486833 PMCID: PMC5149398 DOI: 10.1021/acs.jmedchem.6b00160] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Type 5 17β-hydroxysteroid dehydrogenase, aldo-keto reductase 1C3 (AKR1C3) converts Δ(4)-androstene-3,17-dione and 5α-androstane-3,17-dione to testosterone (T) and 5α-dihydrotestosterone, respectively, in castration resistant prostate cancer (CRPC). In CRPC, AKR1C3 is implicated in drug resistance, and enzalutamide drug resistance can be surmounted by indomethacin a potent inhibitor of AKR1C3. We examined a series of naproxen analogues and find that (R)-2-(6-methoxynaphthalen-2-yl)butanoic acid (in which the methyl group of R-naproxen was replaced by an ethyl group) acts as a potent AKR1C3 inhibitor that displays selectivity for AKR1C3 over other AKR1C enzymes. This compound was devoid of inhibitory activity on COX isozymes and blocked AKR1C3 mediated production of T and induction of PSA in LNCaP-AKR1C3 cells as a model of a CRPC cell line. R-Profens are substrate selective COX-2 inhibitors and block the oxygenation of endocannabinoids and in the context of advanced prostate cancer R-profens could inhibit intratumoral androgen synthesis and act as analgesics for metastatic disease.
Collapse
Affiliation(s)
- Adegoke Adeniji
- Department of Systems Pharmacology and Translational Therapeutics and the Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, 1315 BRBII/III, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104-6160, United States
| | - Md. Jashim Uddin
- Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Tianzhu Zang
- Department of Systems Pharmacology and Translational Therapeutics and the Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, 1315 BRBII/III, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104-6160, United States
| | - Daniel Tamae
- Department of Systems Pharmacology and Translational Therapeutics and the Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, 1315 BRBII/III, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104-6160, United States
| | - Phumvadee Wangtrakuldee
- Department of Systems Pharmacology and Translational Therapeutics and the Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, 1315 BRBII/III, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104-6160, United States
| | - Lawrence J. Marnett
- Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Trevor M. Penning
- Department of Systems Pharmacology and Translational Therapeutics and the Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, 1315 BRBII/III, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104-6160, United States
| |
Collapse
|
21
|
Zhang A, Zhang J, Plymate S, Mostaghel EA. Classical and Non-Classical Roles for Pre-Receptor Control of DHT Metabolism in Prostate Cancer Progression. Discov Oncol 2016; 7:104-13. [PMID: 26797685 DOI: 10.1007/s12672-016-0250-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/05/2016] [Indexed: 12/22/2022] Open
Abstract
Androgens play an important role in prostate cancer (PCa) development and progression. Accordingly, androgen deprivation therapy remains the front-line treatment for locally recurrent or advanced PCa, but patients eventually relapse with the lethal form of the disease termed castration resistant PCa (CRPC). Importantly, castration does not eliminate androgens from the prostate tumor microenvironment which is characterized by elevated tissue androgens that are well within the range capable of activating the androgen receptor (AR). In this mini-review, we discuss emerging data that suggest a role for the enzymes mediating pre-receptor control of dihydrotestosterone (DHT) metabolism, including AKR1C2, HSD17B6, HSD17B10, and the UGT family members UGT2B15 and UGT2B17, in controlling intratumoral androgen levels, and thereby influencing PCa progression. We review the expression of steroidogenic enzymes involved in this pathway in primary PCa and CRPC, the activity and regulation of these enzymes in PCa experimental models, and the impact of genetic variation in genes mediating pre-receptor DHT metabolism on PCa risk. Finally, we discuss recent data that suggests several of these enzymes may also play an unrecognized role in CRPC progression separate from their role in androgen inactivation.
Collapse
Affiliation(s)
- Ailin Zhang
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, MS D5-380, Seattle, WA, 98109, USA
| | - Jiawei Zhang
- School of Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Stephen Plymate
- Department of Medicine, University of Washington, Seattle, WA, 98104, USA
| | - Elahe A Mostaghel
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, MS D5-380, Seattle, WA, 98109, USA.
| |
Collapse
|
22
|
Doig CL, Battaglia S, Khanim FL, Bunce CM, Campbell MJ. Knockdown of AKR1C3 exposes a potential epigenetic susceptibility in prostate cancer cells. J Steroid Biochem Mol Biol 2016; 155:47-55. [PMID: 26429394 PMCID: PMC5391256 DOI: 10.1016/j.jsbmb.2015.09.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 09/24/2015] [Accepted: 09/26/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND The aldo-keto reductase 1C3 (AKR1C3) has been heavily implicated in the propagation of prostate malignancy. AKR1C3 protein is elevated within prostate cancer tissue, it contributes to the formation of androgens and downstream stimulation of the androgen receptor (AR). Elevated expression of AKR1C3 is also reported in acute myeloid leukemia but the target nuclear receptors have been identified as members of the peroxisome-proliferator activated receptor (PPARs) subfamily. Thus, AKR1C3 cancer biology is likely to be tissue dependent and hormonally linked to the availability of ligands for both the steroidogenic and non-steroidogenic nuclear receptors. METHODS In the current study we investigated the potential for AKR1C3 to regulate the availability of prostaglandin-derived ligands for PPARg mainly, prostaglandin J2 (PGJ2). Using prostate cancer cell lines with stably reduced AKR1C3 levels we examined the impact of AKR1C3 upon proliferation mediated by PPAR ligands. RESULTS These studies revealed knockdown of AKR1C3 had no effect upon the sensitivity of androgen receptor independent prostate cancer cells towards PPAR ligands. However, the reduction of levels of AKR1C3 was accompanied by a significantly reduced mRNA expression of a range of HDACs, transcriptional co-regulators, and increased sensitivity towards SAHA, a clinically approved histone deacetylase inhibitor. CONCLUSIONS These results suggest a hitherto unidentified link between AKR1C3 levels and the epigenetic status in prostate cancer cells. This raises an interesting possibility of a novel rational to target AKR1C3, the utilization of AKRIC3 selective inhibitors in combination with HDAC inhibition as part of novel epigenetic therapies in androgen deprivation therapy recurrent prostate cancer.
Collapse
Affiliation(s)
- Craig L Doig
- Centre for Endocrinology Diabetes & Metabolism, School of Clinical & Experimental Medicine, University of Birmingham, Edgbaston B15 2TT, UK.
| | - Sebastiano Battaglia
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Farhat L Khanim
- School of Biosciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | | | - Moray J Campbell
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
23
|
Frycz BA, Murawa D, Borejsza-Wysocki M, Wichtowski M, Spychała A, Marciniak R, Murawa P, Drews M, Jagodziński PP. Transcript level of AKR1C3 is down-regulated in gastric cancer. Biochem Cell Biol 2015; 94:138-46. [PMID: 27019068 DOI: 10.1139/bcb-2015-0096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Steroid hormones have been shown to play a role in gastric carcinogenesis. Large amounts of steroid hormones are locally produced in the peripheral tissues of both genders. Type 5 of 17β-hydroxysteroid dehydrogenase, encoded by the AKR1C3 gene, plays a pivotal role in both androgen and estrogen metabolism, and its expression was found to be deregulated in different cancers. In this study we measured AKR1C3 transcript and protein levels in nontumoral and primary tumoral gastric tissues, and evaluated their association with some clinicopathological features of gastric cancer (GC). We found decreased levels of AKR1C3 transcript (p < 0.0001) and protein (p = 0.0021) in GC tissues compared with the adjacent, apparently histopathologically normal, mucosa. Lower levels of AKR1C3 transcript were observed in diffuse and intestinal types of GC, whereas AKR1C3 protein levels were decreased in tumors with multisite localization, in diffuse histological type, T3, T4, and G3 grades. We also determined the effect of the histone deacetylase inhibitor sodium butyrate (NaBu) on AKR1C3 expression in EPG 85-257 and HGC-27 GC cell lines. We found that NaBu elevates the levels of both AKR1C3 transcript and protein in the cell lines we investigated. Together, our results suggest that decreased expression of AKR1C3 may be involved in development of GC and can be restored by NaBu.
Collapse
Affiliation(s)
- Bartosz Adam Frycz
- a Department of Biochemistry and Molecular Biology, University of Medical Sciences, Poznań, Poland
| | - Dawid Murawa
- b First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, Poznań, Poland.,c Regional Specialist Hospital, Research and Development Centre, Wrocław, Poland
| | - Maciej Borejsza-Wysocki
- d Department of General, Endocrinological Surgery and Gastroenterological Oncology, University of Medical Sciences, Poznań, Poland
| | - Mateusz Wichtowski
- b First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, Poznań, Poland
| | - Arkadiusz Spychała
- b First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, Poznań, Poland
| | - Ryszard Marciniak
- d Department of General, Endocrinological Surgery and Gastroenterological Oncology, University of Medical Sciences, Poznań, Poland
| | - Paweł Murawa
- b First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, Poznań, Poland
| | - Michał Drews
- d Department of General, Endocrinological Surgery and Gastroenterological Oncology, University of Medical Sciences, Poznań, Poland
| | - Paweł Piotr Jagodziński
- a Department of Biochemistry and Molecular Biology, University of Medical Sciences, Poznań, Poland
| |
Collapse
|
24
|
Penning TM. Mechanisms of drug resistance that target the androgen axis in castration resistant prostate cancer (CRPC). J Steroid Biochem Mol Biol 2015; 153:105-13. [PMID: 26032458 PMCID: PMC4568163 DOI: 10.1016/j.jsbmb.2015.05.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/23/2015] [Accepted: 05/25/2015] [Indexed: 12/19/2022]
Abstract
Castrate resistant prostate cancer (CRPC) is the fatal-form of prostate cancer and remains androgen dependent. The reactivation of the androgen axis occurs due to adaptive intratumoral androgen biosynthesis which can be driven by adrenal androgens and/or by changes in the androgen receptor (AR) including AR gene amplification. These mechanisms are targeted with P450c17 inhibitors e.g., abiraterone acetate and AR super-antagonists e.g., enzalutamide, respectively. Clinical experience indicates that with either agent an initial response is followed by drug resistance and the patient clinically progresses on these agents. This article reviews the mechanisms of intrinsic and acquired drug resistance that target the androgen axis and how this might be surmounted.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160, USA.
| |
Collapse
|
25
|
AKR1C3 is a biomarker of sensitivity to PR-104 in preclinical models of T-cell acute lymphoblastic leukemia. Blood 2015; 126:1193-202. [PMID: 26116659 DOI: 10.1182/blood-2014-12-618900] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 06/20/2015] [Indexed: 12/20/2022] Open
Abstract
PR-104, a phosphate ester of the nitrogen mustard prodrug PR-104A, has shown evidence of efficacy in adult leukemia clinical trials. Originally designed to target hypoxic cells, PR-104A is independently activated by aldo-keto-reductase 1C3 (AKR1C3). The aim of this study was to test whether AKR1C3 is a predictive biomarker of in vivo PR-104 sensitivity. In a panel of 7 patient-derived pediatric acute lymphoblastic leukemia (ALL) xenografts, PR-104 showed significantly greater efficacy against T-lineage ALL (T-ALL) than B-cell-precursor ALL (BCP-ALL) xenografts. Single-agent PR-104 was more efficacious against T-ALL xenografts compared with a combination regimen of vincristine, dexamethasone, and l-asparaginase. Expression of AKR1C3 was significantly higher in T-ALL xenografts compared with BCP-ALL, and correlated with PR-104/PR-104A sensitivity in vivo and in vitro. Overexpression of AKR1C3 in a resistant BCP-ALL xenograft resulted in dramatic sensitization to PR-104 in vivo. Testing leukemic blasts from 11 patients confirmed that T-ALL cells were more sensitive than BCP-ALL to PR-104A in vitro, and that sensitivity correlated with AKR1C3 expression. Collectively, these results indicate that PR-104 shows promise as a novel therapy for relapsed/refractory T-ALL, and that AKR1C3 expression could be used as a biomarker to select patients most likely to benefit from such treatment in prospective clinical trials.
Collapse
|
26
|
Swart AC, Storbeck KH. 11β-Hydroxyandrostenedione: Downstream metabolism by 11βHSD, 17βHSD and SRD5A produces novel substrates in familiar pathways. Mol Cell Endocrinol 2015; 408:114-23. [PMID: 25542845 DOI: 10.1016/j.mce.2014.12.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 01/23/2023]
Abstract
11β-Hydroxyandrostenedione (11OHA4), a major C19 steroid produced by the adrenal, was first reported in the 1950s. Initially the subject of numerous studies, interest dwindled due to the apparent lack of physiological function and, by the end of the century, 11OHA4 was no longer considered as an adrenal C19 steroid. Our recent studies, however, showed that 11OHA4 is the precursor to novel active androgens which include 11-ketodihydrotestosterone (11KDHT) which has been implicated in prostate cancer, thereby renewing interest in 11OHA4. In this paper we review the biosynthesis and downstream metabolism of 11OHA4. We discuss the extra-adrenal biosynthesis of 11OHA4 in humans and in other species, highlighting the well-documented role of 11OHA4 in the testes of male fish in which the steroid functions as an active androgen. Finally, we discuss the physiological relevance of 11OHA4 metabolism in castration resistant prostate cancer and outline future prospects.
Collapse
Affiliation(s)
- Amanda C Swart
- Department of Biochemistry, University of Stellenbosch, Stellenbosch 7602, South Africa.
| | - Karl-Heinz Storbeck
- Department of Biochemistry, University of Stellenbosch, Stellenbosch 7602, South Africa.
| |
Collapse
|
27
|
Mizokami A, Namiki M. Reconsideration of progression to CRPC during androgen deprivation therapy. J Steroid Biochem Mol Biol 2015; 145:164-71. [PMID: 24717975 DOI: 10.1016/j.jsbmb.2014.03.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 03/19/2014] [Accepted: 03/28/2014] [Indexed: 02/02/2023]
Abstract
Androgen blockade-naïve prostate cancer (PCa) develops into CRPC during androgen deprivation therapy (ADT) by various genetic actions. The androgen-AR signaling axis plays a key role in this development. PCa cells mainly adapt themselves to the environment of lower androgen concentrations and change into androgen-hypersensitive cells or androgen-independent cells. Androgens of adrenal origin and their metabolites synthesized in the microenvironment in an intracrine/paracrine fashion act on surviving PCa cells and secrete prostate specific antigen (PSA). Total androgen deprivation (TAD) (castration, antiandrogen, and CYP17A1 inhibitor) can become an effective therapeutic strategy concerning the androgen signaling axis-related pathway. However, it is important to ascertain whether elevation of serum PSA results from AR activation or from an androgen-independent tumor volume effect. Then, clinicians can judge it adequately using the imaging studies such as CT or bone scan as well as PSA and bone metabolic markers, an approach which is necessary to judge which treatment is most suitable for the CRPC patients. This article is part of a Special Issue entitled 'Essential role of DHEA'.
Collapse
Affiliation(s)
- Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Mikio Namiki
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan
| |
Collapse
|
28
|
Fokidis HB, Adomat HH, Kharmate G, Hosseini-Beheshti E, Guns ES, Soma KK. Regulation of local steroidogenesis in the brain and in prostate cancer: lessons learned from interdisciplinary collaboration. Front Neuroendocrinol 2015; 36:108-29. [PMID: 25223867 DOI: 10.1016/j.yfrne.2014.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 11/16/2022]
Abstract
Sex steroids play critical roles in the regulation of the brain and many other organs. Traditionally, researchers have focused on sex steroid signaling that involves travel from the gonads via the circulation to intracellular receptors in target tissues. This classic concept has been challenged, however, by the growing number of cases in which steroids are synthesized locally and act locally within diverse tissues. For example, the brain and prostate carcinoma were previously considered targets of gonadal sex steroids, but under certain circumstances, these tissues can upregulate their steroidogenic potential, particularly when circulating sex steroid concentrations are low. We review some of the similarities and differences between local sex steroid synthesis in the brain and prostate cancer. We also share five lessons that we have learned during the course of our interdisciplinary collaboration, which brought together neuroendocrinologists and cancer biologists. These lessons have important implications for future research in both fields.
Collapse
Affiliation(s)
- H Bobby Fokidis
- Department of Biology, Rollins College, Winter Park, FL 37289, USA; Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada.
| | - Hans H Adomat
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | | | | | - Emma S Guns
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada; Department of Urological Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kiran K Soma
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Brain Research Centre, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
29
|
Atsriku C, Hoffmann M, Moghaddam M, Kumar G, Surapaneni S. In vitrometabolism of a novel JNK inhibitor tanzisertib: interspecies differences in oxido-reduction and characterization of enzymes involved in metabolism. Xenobiotica 2014; 45:465-80. [DOI: 10.3109/00498254.2014.991367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
30
|
Joshi S, Nair N, Bedwal RS. Dietary zinc deficiency effects dorso-lateral and ventral prostate of Wistar rats: histological, biochemical and trace element study. Biol Trace Elem Res 2014; 161:91-100. [PMID: 25053558 DOI: 10.1007/s12011-014-0053-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/16/2014] [Indexed: 12/24/2022]
Abstract
Zinc deficiency has become a global problem affecting the developed and developing countries due to inhibitors in the diet which prevents its absorption or due to a very low concentration of bioavailable zinc in the diet. Being present in high concentration in the prostate and having diverse biological function, we investigated the effects of dietary zinc deficiency for 2 and 4 weeks on dorso-lateral and ventral prostate. Sixty prepubertal rats were divided into three groups: zinc control (ZC), pair fed (PF) and zinc deficient (ZD) and fed on 100 μg/g (zinc control and pair fed groups) and 1 μg/g (zinc deficient) diet. Zinc deficiency was associated with degenerative changes in dorso-lateral and ventral prostate as made evident by karyolysis, karyorhexis, cytoplasmolysis, loss of cellularisation, decreased intraluminar secretion and degeneration of fibromuscular stroma. In response, protein carbonyl, nitric oxide, acid phosphatase, 3β-hydroxysteroid dehydrogenase and 17β-hydroxysteroid dehydrogenase increased, exhibiting variable level of significance. Total protein and total zinc concentration in dorso-lateral and ventral prostate as well as in serum decreased (P < 0.001). Decrease (P < 0.001) was recorded in serum FSH and testosterone after 2 and 4 weeks of zinc deficiency. The changes were more prominent after 4 weeks of synthetic zinc deficient diet. The results indicate that zinc deficiency during prepubertal period affects the prostate structure, total protein concentration, enhanced protein carbonyl concentration, nitric oxide as well as acid phosphatase activities and impaired hydroxysteroid dehydrogenase activities. Evidently these changes could be attributed to dysfunction of dorso-lateral and ventral prostate after dietary zinc deficiency as well as impairment of metabolic and secretory activity, reduced gonadotropin levels by hypothalamus -hypophysial system which is indicative of a critical role of zinc in maintaining the prostate integrity.
Collapse
Affiliation(s)
- Sangeeta Joshi
- Cell Biology Laboratory, Department of Zoology, University of Rajasthan, Jaipur, 302055, Rajasthan, India
| | | | | |
Collapse
|
31
|
Carver BS. Strategies for targeting the androgen receptor axis in prostate cancer. Drug Discov Today 2014; 19:1493-7. [PMID: 25107669 DOI: 10.1016/j.drudis.2014.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 07/14/2014] [Indexed: 01/27/2023]
Abstract
Androgen receptor (AR) signaling plays a critical role in prostate cancer cell proliferation, survival, and differentiation. Therapeutic strategies targeting the androgen receptor have been developed for the treatment of metastatic hormone-naïve prostate cancer; however, despite effective targeting recent studies have demonstrated that during progression to a castrate-resistant phenotype there is restoration of AR target gene expression. On the basis of this observation, second-generation therapeutics have been developed to target AR in the castrate-resistant setting resulting in a survival benefit. In this review we will discuss the mechanisms promoting AR signaling and the development of second-generation therapeutics targeting AR in castrate-resistant prostate cancer.
Collapse
Affiliation(s)
- Brett S Carver
- Department of Surgery, Division of Urology and Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
32
|
Abstract
Prostate cancer is the second leading cause of death in adult males in the USA. Recent advances have revealed that the fatal form of this cancer, known as castration-resistant prostate cancer (CRPC), remains hormonally driven despite castrate levels of circulating androgens. CRPC arises as the tumor undergoes adaptation to low levels of androgens by either synthesizing its own androgens (intratumoral androgens) or altering the androgen receptor (AR). This article reviews the major routes to testosterone and dihydrotestosterone synthesis in CRPC cells and examines the enzyme targets and progress in the development of isoform-specific inhibitors that could block intratumoral androgen biosynthesis. Because redundancy exists in these pathways, it is likely that inhibition of a single pathway will lead to upregulation of another so that drug resistance would be anticipated. Drugs that target multiple pathways or bifunctional agents that block intratumoral androgen biosynthesis and antagonize the AR offer the most promise. Optimal use of enzyme inhibitors or AR antagonists to ensure maximal benefits to CRPC patients will also require application of precision molecular medicine to determine whether a tumor in a particular patient will be responsive to these treatments either alone or in combination.
Collapse
Affiliation(s)
- Trevor M Penning
- Perelman School of MedicineCenter of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, USA
| |
Collapse
|
33
|
Kikuchi A, Furutani T, Azami H, Watanabe K, Niimi T, Kamiyama Y, Kuromitsu S, Baskin-Bey E, Heeringa M, Ouatas T, Enjo K. In vitro and in vivo characterisation of ASP9521: a novel, selective, orally bioavailable inhibitor of 17β-hydroxysteroid dehydrogenase type 5 (17βHSD5; AKR1C3). Invest New Drugs 2014; 32:860-70. [DOI: 10.1007/s10637-014-0130-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 06/17/2014] [Indexed: 01/12/2023]
|
34
|
Expression of AKR1C3 and CNN3 as markers for detection of lymph node metastases in colorectal cancer. Clin Exp Med 2014; 15:333-41. [PMID: 24934327 PMCID: PMC4522272 DOI: 10.1007/s10238-014-0298-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/10/2014] [Indexed: 12/21/2022]
Abstract
The aim of the study was to identify a set of discriminating genes that could be used for the prediction of Lymph node (LN) metastasis in human colorectal cancer (CRC), and for this, we compared the whole genome profiles of two CRC cell lines (the primary cell line SW480 and its LN metastatic variant, SW620) and identified eight genes [S100 calcium-binding protein P; aldo–keto reductase family 1(AKR1), member B1 (aldose reductase; AKR1B1); AKR1, member C3 (AKR1C3); calponin 3, acidic; metastasis associated in colon cancer 1; hemoglobin, epsilon 1; trefoil factor 3; and FGGY carbohydrate kinase domain containing]. These genes were examined by quantitative RT-PCR in tissues and LNs in 14 CRC patients and 11 control patients. The level of AKR1C3 mRNA expression was significantly different between the Dukes’ stage A, B, and C groups and the control group (p < 0.05, p < 0.001, and p < 0.001) and was also significantly different between Dukes’ stage C and A or B groups (p < 0.05 and p < 0.001, respectively). The expression of CNN3 was significantly different between the Dukes’ stage C and B or control groups (p < 0.001 and p < 0.01, respectively). There were significant correlations between the expression levels of AKR1C3 and CNN3. AKR1C3 and CNN3 expressions are more accurate and suitable markers for the diagnosis of LN metastasis than the other six genes examined in this study.
Collapse
|
35
|
Abstract
The discovery of androgen dependence in prostate cancer in 1941 by Huggins and colleagues has remained the backbone for the treatment of this disease. However, although many patients initially respond to androgen depletion therapy, they almost invariably relapse and develop resistance with transition of the disease to a castration-resistant state. Over the past decade, the better understanding of the mechanisms that drive resistance to castration has led to the development of next-generation androgen receptor targeting agents such as abiraterone acetate and enzalutamide. This Review aims to revisit the discovery and evolution of androgen receptor targeting therapeutics for the treatment of advanced-stage prostate cancer over the years and to discuss the upcoming future and challenges in the treatment of this common cancer.
Collapse
|
36
|
Tian Y, Zhao L, Zhang H, Liu X, Zhao L, Zhao X, Li Y, Li J. AKR1C3 overexpression may serve as a promising biomarker for prostate cancer progression. Diagn Pathol 2014; 9:42. [PMID: 24571686 PMCID: PMC3939640 DOI: 10.1186/1746-1596-9-42] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 02/21/2014] [Indexed: 12/19/2022] Open
Abstract
Background Aldo-keto reductase family 1 member C3 (AKR1C3) is a key steroidogenic enzyme that is overexpressed in prostate cancer (PCa) and is associated with the development of castration-resistant prostate cancer (CRPC). The aim of this study was to investigate the correlation between the expression level of AKR1C3 and the progression of PCa. Methods Sixty human prostate needle biopsy tissue specimens and ten LNCaP xenografts from intact or castrated male mice were included in the study. The relationship between the level of AKR1C3 expression by immunohistochemistry and evaluation factors for PCa progression, including prostate-specific antigen (PSA), Gleason score (GS) and age, were analyzed. Results Low immunoreactivity of AKR1C3 was detected in normal prostate epithelium, benign prostatic hyperplasia (BPH) and prostatic intraepithelial neoplasia (PIN). Positive staining was gradually increased with an elevated GS in PCa epithelium and LNCaP xenografts in mice after castration. The Spearman’s r values (rs) of AKR1C3 to GS and PSA levels were 0.396 (P = 0.025) and -0.377 (P = 0.036), respectively, in PCa biopsies. The rs of AKR1C3 to age was 0.76 (P = 0.011). No statistically significant difference was found with other variables. Conclusion Our study suggests that the level of AKR. 1C3 expression is positively correlated with an elevated GS, indicating that AKR1C3 can serve as a promising biomarker for the progression of PCa. Virtual slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/7748245591110149.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yi Li
- College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin, China.
| | | |
Collapse
|
37
|
Rižner TL, Penning TM. Role of aldo-keto reductase family 1 (AKR1) enzymes in human steroid metabolism. Steroids 2014; 79:49-63. [PMID: 24189185 PMCID: PMC3870468 DOI: 10.1016/j.steroids.2013.10.012] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 10/16/2013] [Accepted: 10/24/2013] [Indexed: 12/30/2022]
Abstract
Human aldo-keto reductases AKR1C1-AKR1C4 and AKR1D1 play essential roles in the metabolism of all steroid hormones, the biosynthesis of neurosteroids and bile acids, the metabolism of conjugated steroids, and synthetic therapeutic steroids. These enzymes catalyze NADPH dependent reductions at the C3, C5, C17 and C20 positions on the steroid nucleus and side-chain. AKR1C1-AKR1C4 act as 3-keto, 17-keto and 20-ketosteroid reductases to varying extents, while AKR1D1 acts as the sole Δ(4)-3-ketosteroid-5β-reductase (steroid 5β-reductase) in humans. AKR1 enzymes control the concentrations of active ligands for nuclear receptors and control their ligand occupancy and trans-activation, they also regulate the amount of neurosteroids that can modulate the activity of GABAA and NMDA receptors. As such they are involved in the pre-receptor regulation of nuclear and membrane bound receptors. Altered expression of individual AKR1C genes is related to development of prostate, breast, and endometrial cancer. Mutations in AKR1C1 and AKR1C4 are responsible for sexual development dysgenesis and mutations in AKR1D1 are causative in bile-acid deficiency.
Collapse
Affiliation(s)
- Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Slovenia.
| | - Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
38
|
Zhao J, Xiang Y, Xiao C, Guo P, Wang D, Liu Y, Shen Y. AKR1C3 overexpression mediates methotrexate resistance in choriocarcinoma cells. Int J Med Sci 2014; 11:1089-97. [PMID: 25170291 PMCID: PMC4147634 DOI: 10.7150/ijms.9239] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/31/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Chemotherapy is typically used to treat choriocarcinoma, but a small proportion of tumors develop resistance to chemotherapy. Similarly, methotrexate (MTX) is a first-line chemotherapy used to treat choriocarcinoma; although ~30% of patients are drug-resistant for MTX mono-therapy. Thus, we sought to elucidate the mechanism of chemotherapeutic-resistance of MTX. METHODS RNA interference technology, colony formation, and MTT assays were used to investigate the role of aldo-keto reductase family 1, member C3 (AKR1C3) in MTX resistance in choriocarcinoma cells. RESULTS AKR1C3 expression was higher in JeG-3R cells compared to JeG-3 cells and targeted inhibition of AKR1C3 expression with shRNA suppresses growth of choriocarcinoma cells as measured by colony formation and MTT assays. Overexpression of AKR1C3 increased chemotherapeutic resistance in JeG-3 cells. Furthermore, AKR1C3 silencing increases sensitivity to MTX in JeG-3R choriocarcinoma cells. Increasing MTX sensitivity spears to be related to DNA damage induction by increased reactive oxygen species (ROS), apoptosis, and cell cycle arrest. CONCLUSIONS Data show that AKR1C3 is critical to the development of methotrexate resistance in choriocarcinoma and suggest that AKR1C3 may potentially serve as a therapeutic marker for this disease.
Collapse
Affiliation(s)
- Jing Zhao
- 1. Departments of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yang Xiang
- 1. Departments of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Changji Xiao
- 1. Departments of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Peng Guo
- 1. Departments of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Dan Wang
- 1. Departments of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Ying Liu
- 1. Departments of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yun Shen
- 2. Reproductive Health Centre, National Science Institute for Family Planning, Beijing, People's Republic of China
| |
Collapse
|
39
|
Kumagai J, Hofland J, Erkens-Schulze S, Dits NFJ, Steenbergen J, Jenster G, Homma Y, de Jong FH, van Weerden WM. Intratumoral conversion of adrenal androgen precursors drives androgen receptor-activated cell growth in prostate cancer more potently than de novo steroidogenesis. Prostate 2013; 73:1636-50. [PMID: 23996639 DOI: 10.1002/pros.22655] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Accepted: 01/23/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Despite an initial response to hormonal therapy, patients with advanced prostate cancer (PC) almost always progress to castration-resistant disease (CRPC). Although serum testosterone (T) is reduced by androgen deprivation therapy, intratumoral T levels in CRPC are comparable to those in prostate tissue of eugonadal men. These levels could originate from intratumoral conversion of adrenal androgens and/or from de novo steroid synthesis. However, the relative contribution of de novo steroidogenesis to AR-driven cell growth is unknown. METHODS The relative contribution of androgen biosynthetic pathways to activate androgen receptor (AR)-regulated cell growth and expression of PSA, FKBP5, and TMPRSS2 was studied at physiologically relevant levels of adrenal androgen precursors and intermediates of de novo androgen biosynthesis in human prostate cancer cell lines, PC346C, VCaP, and LNCaP. RESULTS In PC346C and VCaP, responses to pregnenolone and progesterone were absent or minimal, while large effects of adrenal androgen precursors were found. VCaP CRPC clones overexpressing CYP17A1 did not acquire an increased ability to use pregnenolone or progesterone to activate AR. In contrast, all precursors stimulated growth and gene expression in LNCaP cells, presumably resulting from the mutated AR in these cells. CONCLUSIONS Our data indicate that at physiological levels of T precursors PC cells can generally convert adrenal androgens, while de novo steroidogenesis is not generally possible in PC cells and is not able to support AR transactivation and PC growth.
Collapse
Affiliation(s)
- Jinpei Kumagai
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Piao YS, Wiesenfeld P, Sprando R, Arnold JT. TGFβ1 alters androgenic metabolites and hydroxysteroid dehydrogenase enzyme expression in human prostate reactive stromal primary cells: Is steroid metabolism altered by prostate reactive stromal microenvironment? J Steroid Biochem Mol Biol 2013; 138:206-13. [PMID: 23770322 PMCID: PMC3839662 DOI: 10.1016/j.jsbmb.2013.05.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/18/2013] [Accepted: 05/30/2013] [Indexed: 10/26/2022]
Abstract
The inflammatory tissue microenvironment can be an active promoter in preneoplastic cancer lesions. Altered steroid hormone metabolism as induced by the inflammatory microenvironment may contribute to epithelial cancer progression. Dehydroepiandrosterone sulfate (DHEAS) is the most abundant endogenous steroid hormone present in human serum and can be metabolized to DHEA, androgens and/or estrogens in peripheral tissues. We have previously reported that TGFβ1-induced reactive prostate stromal cells increase DHEA metabolism to active androgens and alter prostate cancer cell gene expression. While much of the focus on mechanisms of prostate cancer and steroid metabolism is in the epithelial cancer cells, this study focuses on TGFβ1-induced effects on DHEA metabolic pathways and enzymes in human prostate stromal cells. In DHEA-treated primary prostate stromal cells, TGFβ1 produced time- and dose-dependent increases in metabolism of DHEA to androstenedione and testosterone. Also TGFβ1-treated prostate stromal cells exhibited changes in the gene expression of enzymes involved in steroid metabolism including up-regulation of 3β hydroxysteroid dehydrogenase (HSD), and down-regulation of 17βHSD5, and 17βHSD2. These studies suggest that reactive prostate stroma and the inflammatory microenvironment may contribute to altered steroid metabolism and increased intratumoral androgens.
Collapse
Affiliation(s)
- Yun-shang Piao
- Division of Intramural Research, National Center for Complementary and Alternative Medicine, National Institutes of Health, Bethesda, MD 20892, United States
| | - Paddy Wiesenfeld
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, Food and Drug Administration, Laurel, MD 20708, United States
| | - Robert Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, Food and Drug Administration, Laurel, MD 20708, United States
| | - Julia T. Arnold
- Division of Intramural Research, National Center for Complementary and Alternative Medicine, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
41
|
Storbeck KH, Bloem LM, Africander D, Schloms L, Swart P, Swart AC. 11β-Hydroxydihydrotestosterone and 11-ketodihydrotestosterone, novel C19 steroids with androgenic activity: a putative role in castration resistant prostate cancer? Mol Cell Endocrinol 2013; 377:135-46. [PMID: 23856005 DOI: 10.1016/j.mce.2013.07.006] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 06/10/2013] [Accepted: 07/05/2013] [Indexed: 01/28/2023]
Abstract
Adrenal C19 steroids, dehydroepiandrostenedione (DHEA(S)) and androstenedione (A4), play a critical role in castration resistant prostate cancer (CRPC) as they are metabolised to dihydrotestosterone (DHT), via testosterone (T), or via the alternate 5α-dione pathway, bypassing T. Adrenal 11OHA4 metabolism in CRPC is, however, unknown. We present a novel pathway for 11OHA4 metabolism in CRPC leading to the production of 11ketoT (11KT) and novel 5α-reduced C19 steroids - 11OH-5α-androstanedione, 11keto-5α-androstanedione, 11OHDHT and 11ketoDHT (11KDHT). The pathway was validated in the androgen-dependent prostate cancer cell line, LNCaP. Androgen receptor (AR) transactivation studies showed that while 11KT and 11OHDHT act as a partial AR agonists, 11KDHT is a full AR agonist exhibiting similar activity to DHT at 1nM. Our data demonstrates that, while 11OHA4 has negligible androgenic activity, its metabolism to 11KT and 11KDHT yields androgenic compounds which may be implicated, together with A4 and DHEA(S), in driving CRPC in the absence of testicular T.
Collapse
Affiliation(s)
- Karl-Heinz Storbeck
- Department of Biochemistry, University of Stellenbosch, Stellenbosch 7600, South Africa
| | | | | | | | | | | |
Collapse
|
42
|
Yang L, Zhang J, Zhang S, Dong W, Lou X, Liu S. Quantitative evaluation of aldo-keto reductase expression in hepatocellular carcinoma (HCC) cell lines. GENOMICS PROTEOMICS & BIOINFORMATICS 2013; 11:230-40. [PMID: 23584128 PMCID: PMC4357791 DOI: 10.1016/j.gpb.2013.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 03/31/2013] [Accepted: 04/07/2013] [Indexed: 12/03/2022]
Abstract
The involvement of aldo–keto reductases (AKRs) in tumorigenesis is widely reported, but their roles in the pathological process are not generally recognized due to inconsistent measurements of their expression. To overcome this problem, we simultaneously employed real-time PCR to examine gene expression and multiple reaction monitoring (MRM) of mass spectrometry (MS) to examine the protein expression of AKRs in five different hepatic cell lines. These include one relatively normal hepatic cell line, L-02, and four hepatocellular carcinoma (HCC) cell lines, HepG2, HuH7, BEL7402 and SMMC7721. The results of real-time PCR showed that expression of genes encoding the AKR1C family members rather than AKR1A and AKR1B was associated with tumor, and most of genes encoding AKRs were highly expressed in HuH7. Similar observations were obtained through MRM. Different from HuH7, the protein abundance of AKR1A and AKR1B was relatively consistent among the other four hepatic cell lines, while protein expression of AKR1C varied significantly compared to L-02. Therefore, we conclude that the abundant distribution of AKR1C proteins is likely to be associated with liver tumorigenesis, and the AKR expression status in HuH7 is completely different from other liver cancer cell lines. This study, for the first time, provided both overall and quantitative information regarding the expression of AKRs at both mRNA and protein levels in hepatic cell lines. Our observations put the previous use of AKRs as a biomarker into question since it is only consistent with our data from HuH7. Furthermore, the data presented herein demonstrated that quantitative evaluation and comparisons within a protein family at both mRNA and protein levels were feasible using current techniques.
Collapse
Affiliation(s)
- Lei Yang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | | | | | |
Collapse
|
43
|
Zhang S, Wen B, Zhou B, Yang L, Cha C, Xu S, Qiu X, Wang Q, Sun H, Lou X, Zi J, Zhang Y, Lin L, Liu S. Quantitative analysis of the human AKR family members in cancer cell lines using the mTRAQ/MRM approach. J Proteome Res 2013; 12:2022-33. [PMID: 23544749 DOI: 10.1021/pr301153z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Members of human aldo-keto reductase (AKR) superfamily have been reported to be involved in cancer progression, whereas the final conclusion is not generally accepted. Herein, we propose a quantitative method to measure human AKR proteins in cells using mTRAQ-based multiple reaction monitoring (MRM). AKR peptides with multiple transitions were carefully selected upon tryptic digestion of the recombinant AKR proteins, while AKR proteins were identified by SDS-PAGE fractionation coupled with LC-MS/MS. Utilizing mTRAQ triplex labeling to produce the derivative peptides, calibration curves were generated using the mixed lysate as background, and no significantly different quantification of AKRs was elicited from the two sets of calibration curves under the mixed and single lysate as background. We employed this approach to quantitatively determine the 6 AKR proteins, AKR1A1, AKR1B1, AKR1B10, AKR1C1/C2, AKR1C3, and AKR1C4, in 7 different cancer cell lines and for the first time to obtain the absolute quantities of all the AKR proteins in each cell. The cluster plot revealed that AKR1A and AKR1B were widely distributed in most cancer cells with relatively stable abundances, whereas AKR1Cs were unevenly detected among these cells with diverse dynamic abundances. The AKR quantitative distribution in different cancer cells, therefore, may assist further exploration toward how the AKR proteins are involved in tumorigenesis.
Collapse
Affiliation(s)
- Shenyan Zhang
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 101318, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Thyroid hormones (THs) may play a role in diseases other than hyper- and hypothyroidism. Several lines of evidence suggest tumor-promoting effects of TH and TH receptors. They are possibly mediated by phosphatidylinositol-3-kinase and MAPK and involve among others stimulation of angiogenesis via αvβ3. Thus, an increased risk for colon, lung, prostate, and breast cancer with lower TSH has been demonstrated in epidemiological studies, even suggesting a TH dose effect on cancer occurrence. Furthermore, higher TH levels were associated with an advanced clinical stage of breast and prostate cancer. In rodent models, TH stimulated growth and metastasis of tumor transplants, whereas hypothyroidism had opposite effects. In clinical studies of glioblastoma and head and neck cancer, hypothyroid patients showed longer survival than euthyroid patients. Also, patients with renal cell cancer that were treated with the tyrosine kinase inhibitor sunitinib and developed hypothyroidism in due course showed significantly longer survival than patients that remained euthyroid. Development of hypothyroidism was an independent predictor for survival in two studies. Yet, it is still possible that hypothyroidism is only a surrogate marker for treatment efficacy and does not positively influence treatment outcome by itself. Future cancer treatment studies, especially with substances that can induce hypothyroidism, should therefore be designed in a way that allows for an analysis of thyroid function status and its contribution on treatment outcome.
Collapse
Affiliation(s)
- Lars C Moeller
- Division of Laboratory Research, Department of Endocrinology and Metabolic Diseases, University of Duisburg-Essen, Hufelandstraße 55, 45127 Essen, Germany.
| | | |
Collapse
|
45
|
McNamara KM, Nakamura Y, Sasano H, Handelsman DJ, Simanainen U. Prostate epithelial AR inactivation leads to increased intraprostatic androgen synthesis. Prostate 2013; 73:316-27. [PMID: 22976924 DOI: 10.1002/pros.22570] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 07/10/2012] [Indexed: 11/06/2022]
Abstract
BACKGROUND Regulation of steroid synthesis within the prostate is not well understood. In this study, we examined androgen synthesis and metabolism in the mouse prostate. METHODS Using LC-MSMS steroid assays, immunohistochemistry and real-time PCR we examined the role of prostate epithelial AR in regulating 5αR expression and subsequent androgen metabolism by analyzing natural differences in epithelial AR expression between lobes as well as in the prostate epithelial AR knockout (PEARKO) mouse model. Subsequently, the role of intraprostatic androgen metabolism and epithelial AR in the generation and progression of prostate epithelial pathology was examined using long-term exogenous testosterone (T) + estradiol (E2) exposure. RESULTS Epithelial AR and 5αR2 expression as well as intraprostatic DHT followed the same lobe-specific pattern being lower in anterior than the other lobes (n = 6-8, P < 0.05). Lobe-specific 5αR2 expression was similar in PEARKO and wild-type (WT) prostate. However, PEARKO prostate had higher intraprostatic DHT content with significantly increased 5αR2 expression localized in abnormal epithelium. T + E2 treatment induced epithelial pathology was more common in PEARKO prostate compared to WT (20% vs. 2%), and was associated with increased 5αR2 expression (n = 6, P < 0.001). CONCLUSIONS We suggest that androgen synthesis via 5αR2 expression is driven by its own product (DHT) acting on adjacent stromal cells in a paracrine loop leading to increased in situ androgen levels in the PEARKO prostate. This may form part of a feed-forward loop that promotes the development of epithelial pathology.
Collapse
Affiliation(s)
- Keely M McNamara
- Andrology, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | | | | | | | | |
Collapse
|
46
|
Ni X, Suhail MM, Yang Q, Cao A, Fung KM, Postier RG, Woolley C, Young G, Zhang J, Lin HK. Frankincense essential oil prepared from hydrodistillation of Boswellia sacra gum resins induces human pancreatic cancer cell death in cultures and in a xenograft murine model. Altern Ther Health Med 2012; 12:253. [PMID: 23237355 PMCID: PMC3538159 DOI: 10.1186/1472-6882-12-253] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 12/11/2012] [Indexed: 12/22/2022]
Abstract
Background Regardless of the availability of therapeutic options, the overall 5-year survival for patients diagnosed with pancreatic cancer remains less than 5%. Gum resins from Boswellia species, also known as frankincense, have been used as a major ingredient in Ayurvedic and Chinese medicine to treat a variety of health-related conditions. Both frankincense chemical extracts and essential oil prepared from Boswellia species gum resins exhibit anti-neoplastic activity, and have been investigated as potential anti-cancer agents. The goals of this study are to identify optimal condition for preparing frankincense essential oil that possesses potent anti-tumor activity, and to evaluate the activity in both cultured human pancreatic cancer cells and a xenograft mouse cancer model. Methods Boswellia sacra gum resins were hydrodistilled at 78°C; and essential oil distillate fractions were collected at different durations (Fraction I at 0–2 h, Fraction II at 8–10 h, and Fraction III at 11–12 h). Hydrodistillation of the second half of gum resins was performed at 100°C; and distillate was collected at 11–12 h (Fraction IV). Chemical compositions were identified by gas chromatography–mass spectrometry (GC-MS); and total boswellic acids contents were quantified by high-performance liquid chromatography (HPLC). Frankincense essential oil-modulated pancreatic tumor cell viability and cytotoxicity were determined by colorimetric assays. Levels of apoptotic markers, signaling molecules, and cell cycle regulators expression were characterized by Western blot analysis. A heterotopic (subcutaneous) human pancreatic cancer xenograft nude mouse model was used to evaluate anti-tumor capability of Fraction IV frankincense essential oil in vivo. Frankincense essential oil-induced tumor cytostatic and cytotoxic activities in animals were assessed by immunohistochemistry. Results Longer duration and higher temperature hydrodistillation produced more abundant high molecular weight compounds, including boswellic acids, in frankincense essential oil fraactions. Human pancreatic cancer cells were sensitive to Fractions III and IV (containing higher molecular weight compounds) treatment with suppressed cell viability and increased cell death. Essential oil activated the caspase-dependent apoptotic pathway, induced a rapid and transient activation of Akt and Erk1/2, and suppressed levels of cyclin D1 cdk4 expression in cultured pancreatic cancer cells. In addition, Boswellia sacra essential oil Fraction IV exhibited anti-proliferative and pro-apoptotic activities against pancreatic tumors in the heterotopic xenograft mouse model. Conclusion All fractions of frankincense essential oil from Boswellia sacra are capable of suppressing viability and inducing apoptosis of a panel of human pancreatic cancer cell lines. Potency of essential oil-suppressed tumor cell viability may be associated with the greater abundance of high molecular weight compounds in Fractions III and IV. Although chemical component(s) responsible for tumor cell cytotoxicity remains undefined, crude essential oil prepared from hydrodistillation of Boswellia sacra gum resins might be a useful alternative therapeutic agent for treating patients with pancreatic adenocarcinoma, an aggressive cancer with poor prognosis.
Collapse
|
47
|
Tsao CK, Galsky MD, Small AC, Yee T, Oh WK. Targeting the androgen receptor signalling axis in castration-resistant prostate cancer (CRPC). BJU Int 2012; 110:1580-8. [DOI: 10.1111/j.1464-410x.2012.11445.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Schleutker J. Polymorphisms in androgen signaling pathway predisposing to prostate cancer. Mol Cell Endocrinol 2012; 360:25-37. [PMID: 21782882 DOI: 10.1016/j.mce.2011.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 06/30/2011] [Accepted: 07/01/2011] [Indexed: 11/30/2022]
Abstract
Prostate cancer is the most frequent male malignancy diagnosed in western countries and androgens are known to mediate key physiological processes in prostate tissue. Since endogenous androgens have long been considered to be risk factors for prostate cancer, genes involved in androgen biosynthesis and metabolism have been extensively studied. In this review, association of androgen pathway genes, their polymorphic sites and risk of prostate cancer in different ethnic backgrounds is addressed together with their use to predict susceptibility and clinical outcomes of prostate cancer patients. The effect of the polymorphisms seems vary in different patients, populations and ethnic backgrounds. To date it is evident that the association between androgen pathway gene polymorphisms and prostate cancer risk is complex and many of the results are characterized by irreproducibility, which can be attributed to a variety of biological, statistical and technical reasons. In the future, with increasing knowledge, developing technologies and new genomic biomarkers it likely becomes possible to better estimate the risk of prostate cancer, and distinguish indolent disease from aggressive based on molecular profiling, and the analysis of gene-gene and gene-environment interactions.
Collapse
Affiliation(s)
- Johanna Schleutker
- Institute of Biomedical Technology, University of Tampere, and Centre for Laboratory Medicine, Tampere University Hospital, Biokatu 8, 33520 Tampere, Finland.
| |
Collapse
|
49
|
Schulze JJ, Karypidis H, Ekström L. Basal and Regulatory Promoter Studies of the AKR1C3 Gene in Relation to Prostate Cancer. Front Pharmacol 2012; 3:151. [PMID: 22888320 PMCID: PMC3412290 DOI: 10.3389/fphar.2012.00151] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 07/17/2012] [Indexed: 11/22/2022] Open
Abstract
Background: Human 17β-hydroxysteroid dehydrogenase type 5 (17β-HSD5) formally known as aldo-keto reductase 1C3 (AKR1C3) play a major role in the formation and metabolism of androgens. The enzyme is highly expressed in the prostate gland and previous studies indicate that genetic variation in the AKR1C3 gene may influence the prostate volume and risk of prostate cancer. Aim: Here we aimed to further study the genetic regulation of AKR1C3 and its putative role in prostate cancer. Experiments: A previously identified promoter polymorphism (A>G, rs3763676) localized at −138 from the translational start site were studied in relation to prostate cancer in a Swedish population based case–control study including 176 patients diagnosed with prostate cancer and 161 controls. Moreover, we have studied the basal and androgen induced promoter activity of the AKR1C3 gene. Expression studies with AKR1C3 promoter reporter constructs were performed in HepG2 and DSL2 cells. Results: We found that carriers of the promoter A-allele had a borderline significant decreased risk of prostate cancer (OR = 0.59; 95% CI = 0.32–1.08). We also show that dihydrotestosterone (DHT) induced the promoter activity of the A-allele 2.2-fold (p = 0.048). Sp3 seem to play an important role in regulating the transcription activity of AKR1C3 and site-directed mutagenesis of a GC-box 78 base-pair upstream the ATG-site significantly inhibited the basal AKR1C3 promoter activity by 70%. Conclusion: These results further supports previous findings that the A>G promoter polymorphism may be functional and that AKR1C3 plays a critical role in prostate carcinogenesis. Our findings also show that the members of Sp family of transcription factors are important for the constitutive expression of AKR1C3 gene.
Collapse
Affiliation(s)
- Jenny J Schulze
- Department of Laboratory Medicine, Karolinska Institutet Stockholm, Sweden
| | | | | |
Collapse
|
50
|
Androgen Receptor Signaling Regulates Cell Growth and Vulnerability to Doxorubicin in Bladder Cancer. J Urol 2012; 188:276-86. [DOI: 10.1016/j.juro.2012.02.2554] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Indexed: 11/15/2022]
|