1
|
Androgen Glucuronidation in Mice: When, Where, and How. BIOLOGY 2022; 11:biology11030403. [PMID: 35336777 PMCID: PMC8945853 DOI: 10.3390/biology11030403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Hormone metabolism can vary from one species to another. In humans, specific UDP-glucuronosyltransferase (UGT) enzymes transform androgens (the male hormones) into glucuronide derivatives, which are easier to eliminate. Whether a similar mechanism also takes place in mice has never been ascertained. This study aimed at addressing this question. Organs and pure Ugt2b enzymes from mice were assayed for their ability to transform several androgens into their glucuronide derivatives. Results show that, as in humans, both murine organs and enzymes are reactive with androgen molecules, and glucuronide derivatives are formed with substrate-, organ- and enzyme-specific manner. In conclusion, these observations revealed that glucuronosyltransferase enzymes from mice works in a similar manner as their human counterparts. Abstract Glucuronidation, catalyzed by UDP-glucuronosyltransferase UGT2B enzymes, is a major inactivating and elimination pathway for androgen hormones in humans. Whether Ugt2b enzymes from mice are also reactive with these hormones have never been investigated. The present study aimed at evaluating the capability of murine tissues and Ugt2b enzymes to glucuronidated androgens. The 7 murine Ugt2b (Ugt2b1, 2b5, 2b34, 2b35, 2b36, 2b37 and 2b38) enzymes were cloned and stably expressed into HEK293 cells. In vitro glucuronidation assays were performed with microsomal proteins or homogenates from mice tissues (liver, kidney, intestine, adipose, testis, prostate, epididymis, bulbo, seminal vesicle, mammary glands, uterus, and ovary) and from Ugt2b-HEK293 cells. Male and female livers, as well as male kidneys, are the major sites for androgen glucuronidation in mice. The male liver is highly efficient at glucuronidation of dihydrotestosterone (DHT) and testosterone and is enriched in Ugt2b1 and 2b5 enzymes. Androsterone and 3α-Diol are conjugated in the male kidney through an Ugt2b37-dependent process. Interestingly, castration partially abolished hepatic Ugt2b1 expression and activity, while Ugt2b37 was totally repressed. DHT injection partially corrected these changes. In conclusion, these observations revealed the substrate- and tissue-specific manner in which murine Ugt2b enzymes conjugate androgens. They also evidence how androgens modulate their own glucuronide conjugation in mice.
Collapse
|
2
|
Ho V, Pasquet R, Luo S, Chen G, Goss P, Tu D, Lazarus P, Richardson H. Variation in the UGT2B17 genotype, exemestane metabolism and menopause-related toxicities in the CCTG MAP.3 trial. Breast Cancer Res Treat 2020; 183:705-716. [PMID: 32715442 DOI: 10.1007/s10549-020-05812-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/15/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To examine associations between the UGT2B17 gene deletion and exemestane metabolites, and commonly reported side effects (fatigue, hot flashes, and joint pain) among postmenopausal women participating in the MAP.3 chemoprevention trial. METHODS The analytical samples for the UGT2B17 analysis comprised 1752 women on exemestane and 1721 women on placebo; the exemestane metabolite analysis included 1360 women on exemestane with one-year serum samples. Both the UGT2B17 gene deletion and metabolites were measured in blood. The metabolites were conceptualized as a ratio (17-DHE-Gluc:17-DHE). Symptoms were assessed using the CTCAE v4.0 at approximately 1-year intervals. Log-binomial regression was used to examine the associations between UGT2B17 deletion, exemestane metabolites and each side effect at 1 and up to 5-year follow-up, adjusting for potential confounders. RESULTS Among individuals on exemestane with the UGT2B17 gene deletion (i.e., lower detoxification), a higher risk of severe fatigue (RR = 2.59 95% CI: 1.14-5.89) was observed at up to 5-year follow-up. Among individuals on placebo, those with the UGT2B17 gene deletion had a higher risk of any fatigue (RR = 1.39, 95% CI: 1.02-1.89) at year 1. A lower metabolite ratio (poor detoxification) was associated with a higher risk of any fatigue, hot flashes and joint pain at year 1 (fatigue: RR = 1.89, 95% CI: 1.16-3.09; hot flashes: RR = 1.77, 95% CI: 1.40-2.24; joint pain: RR = 2.05, 95% CI: 1.35-3.12); similar associations were observed at 5-year follow-up. CONCLUSION Variation in the metabolism of exemestane through the UGT2B17-mediated pathway is associated with subsequent risk of commonly reported symptoms in MAP.3.
Collapse
Affiliation(s)
- Vikki Ho
- Department of Social and Preventive Medicine, University of Montreal, 850 rue Saint-Denis, Tour Saint-Antoine, 3rd Floor, S03.412, Montréal, QC, H2X0A9, Canada.,Health Innovation and Evaluation Hub, University of Montreal Hospital Research Centre (CRCHUM), 850 rue Saint-Denis, Tour Saint-Antoine, 3rd Floor, S03.424, Montréal, QC, H2X0A9, Canada
| | - Romain Pasquet
- Department of Social and Preventive Medicine, University of Montreal, 850 rue Saint-Denis, Tour Saint-Antoine, 3rd Floor, S03.412, Montréal, QC, H2X0A9, Canada
| | - Shaman Luo
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, 412 E. Spokane Falls Blvd., PBS 431, Spokane, WA, 99202-2131, USA
| | - Gang Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, 412 E. Spokane Falls Blvd., PBS 431, Spokane, WA, 99202-2131, USA
| | - Paul Goss
- Harvard Medical School, MGH Cancer Center, Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA
| | - Dongsheng Tu
- Division Canadian Cancer Trials Group, Queen's University, 10 Stuart Street, Kingston, ON, K7L 3N6, Canada.,Department of Public Health Sciences, Cancer Research Institute, Queen's University, 10 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, 412 E. Spokane Falls Blvd., PBS 431, Spokane, WA, 99202-2131, USA
| | - Harriet Richardson
- Divisions of Canadian Cancer Trials Group and Cancer Care and Epidemiology, Cancer Research Institute, Queen's University, 10 Stuart Street, Room 220, Kingston, ON, K7L 3N6, Canada.
| | | |
Collapse
|
3
|
Vaillancourt J, Turcotte V, Caron P, Villeneuve L, Lacombe L, Pouliot F, Lévesque É, Guillemette C. Glucuronidation of Abiraterone and Its Pharmacologically Active Metabolites by UGT1A4, Influence of Polymorphic Variants and Their Potential as Inhibitors of Steroid Glucuronidation. Drug Metab Dispos 2019; 48:75-84. [DOI: 10.1124/dmd.119.088229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/08/2019] [Indexed: 11/22/2022] Open
|
4
|
Meech R, Hu DG, McKinnon RA, Mubarokah SN, Haines AZ, Nair PC, Rowland A, Mackenzie PI. The UDP-Glycosyltransferase (UGT) Superfamily: New Members, New Functions, and Novel Paradigms. Physiol Rev 2019; 99:1153-1222. [DOI: 10.1152/physrev.00058.2017] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UDP-glycosyltransferases (UGTs) catalyze the covalent addition of sugars to a broad range of lipophilic molecules. This biotransformation plays a critical role in elimination of a broad range of exogenous chemicals and by-products of endogenous metabolism, and also controls the levels and distribution of many endogenous signaling molecules. In mammals, the superfamily comprises four families: UGT1, UGT2, UGT3, and UGT8. UGT1 and UGT2 enzymes have important roles in pharmacology and toxicology including contributing to interindividual differences in drug disposition as well as to cancer risk. These UGTs are highly expressed in organs of detoxification (e.g., liver, kidney, intestine) and can be induced by pathways that sense demand for detoxification and for modulation of endobiotic signaling molecules. The functions of the UGT3 and UGT8 family enzymes have only been characterized relatively recently; these enzymes show different UDP-sugar preferences to that of UGT1 and UGT2 enzymes, and to date, their contributions to drug metabolism appear to be relatively minor. This review summarizes and provides critical analysis of the current state of research into all four families of UGT enzymes. Key areas discussed include the roles of UGTs in drug metabolism, cancer risk, and regulation of signaling, as well as the transcriptional and posttranscriptional control of UGT expression and function. The latter part of this review provides an in-depth analysis of the known and predicted functions of UGT3 and UGT8 enzymes, focused on their likely roles in modulation of levels of endogenous signaling pathways.
Collapse
Affiliation(s)
- Robyn Meech
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, 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, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Siti Nurul Mubarokah
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Alex Z. Haines
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Pramod C. Nair
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Andrew Rowland
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, 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, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|
5
|
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
|
6
|
Grant DJ, Chen Z, Howard LE, Wiggins E, De Hoedt A, Vidal AC, Carney ST, Squires J, Magyar CE, Huang J, Freedland SJ. UDP-glucuronosyltransferases and biochemical recurrence in prostate cancer progression. BMC Cancer 2017; 17:463. [PMID: 28673330 PMCID: PMC5496250 DOI: 10.1186/s12885-017-3463-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/28/2017] [Indexed: 12/31/2022] Open
Abstract
Background Uridine 5′-diphosphate-glucuronosyltransferase 2B (UGT2B) genes code for enzymes that catalyze the clearance of testosterone, dihydrotestosterone (DHT), and DHT metabolites in the prostate basal and luminal tissue. The expression of the UGT2B15, UGT2B17, and UGT2B28 enzymes has not been evaluated in prostate tissue samples from hormone therapy-naïve patients. Methods We determined the expression of UGT2B15, UGT2B17, and UGT2B28 enzymes in 190 prostate tissue samples from surgical specimens of a multiethnic cohort of patients undergoing radical prostatectomy at the Durham Veterans Affairs Medical Center. The association between each protein’s percent positive and H-score, a weighted score of staining intensity, and the risk of biochemical recurrence (BCR) was tested using separate Cox proportional hazards models. In an exploratory analysis, UGT2B17 total positive and H-score were divided at the median and we tested the association between UGT2B17 group and risk of BCR. Results The median follow-up for all patients was 118 months (IQR: 85-144). Of 190, 83 (44%) patients developed BCR. We found no association between UGT2B15 or UGT2B28 and risk of BCR. However, there was a trend for an association between UGT2B17 and BCR (HR = 1.01, 95% CI 1.00-1.02, p = 0.11), though not statistically significant. Upon further investigation, we found that patients with UGT2B17 higher levels of expression had a significant increased risk of BCR on univariable analysis (HR = 1.57, 95% CI 1.02-2.43, p = 0.041), although this association was attenuated in the multivariable model (HR = 1.50, 95% CI 0.94-2.40, p = 0.088). Conclusions Our findings suggest that UGT2B17 overexpression may be associated with a significant increased risk of BCR. These results are consistent with previous reports which showed UGT2B17 significantly expressed in advanced prostate cancer including prostate tumor metastases.
Collapse
Affiliation(s)
- Delores J Grant
- Department of Biological and Biomedical Science, Cancer Research Program, North Carolina Central University, Julius L. Chambers Biomedical/Biotechnology Research Institute, 1801 Fayetteville Street, Durham, NC, 27707, USA.
| | - Zinan Chen
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, 2424 Erwin Road, Suite 1102 Hock Plaza, Box 2721, Durham, NC, 27710, USA
| | - Lauren E Howard
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, 2424 Erwin Road, Suite 1102 Hock Plaza, Box 2721, Durham, NC, 27710, USA
| | - Emily Wiggins
- Durham Veterans Administration Medical Center, 508 Fulton St, Durham, NC, 27705, USA
| | - Amanda De Hoedt
- Durham Veterans Administration Medical Center, 508 Fulton St, Durham, NC, 27705, USA
| | - Adriana C Vidal
- Cedars-Sinai Health System, Center for Integrated Research on Cancer and Lifestyle, Cancer Genetics and Prevention Program, Surgery, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Skyla T Carney
- Department of Biological and Biomedical Science, Cancer Research Program, North Carolina Central University, Julius L. Chambers Biomedical/Biotechnology Research Institute, 1801 Fayetteville Street, Durham, NC, 27707, USA
| | - Jill Squires
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, The David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, CHS 14-112, Los Angeles, CA, 90095, USA
| | - Clara E Magyar
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, The David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, CHS 14-112, Los Angeles, CA, 90095, USA
| | - Jiaoti Huang
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, The David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, CHS 14-112, Los Angeles, CA, 90095, USA
| | - Stephen J Freedland
- Cedars-Sinai Health System, Center for Integrated Research on Cancer and Lifestyle, Cancer Genetics and Prevention Program, Surgery, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| |
Collapse
|
7
|
Dam PTM, Jang YJ, Park JI, Choi BC, Song SJ, Chun SY. Regulation of uridine diphosphate-glucuronosyltransferase 2B15 expression during ovulation in the rat. Endocr J 2017; 64:605-612. [PMID: 28442641 DOI: 10.1507/endocrj.ej16-0469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Uridine diphosphate-glucuronosyltransferase 2B15 (UGT2B15) conjugates 5α-androstane-3α, 17β-diol (3α-diol) to 3α-diol glucuronide (3α-diol G) in steroid target tissues. The present study investigated the regulation of UGT2B15 expression during the ovulatory process in the rat. Real-time PCR analysis revealed that treatment of immature rats with equine chorionic gonadotropin followed by human chorionic gonadotropin transiently stimulated UGT2B15 gene expression in granulosa cells of preovulatory follicles within 6 h. The progesterone receptor antagonist RU486 suppressed the gonadotropin-induced UGT2B15 expression. The expression of UGT2B15 and the levels of 3α-diol G were transiently increased by luteinizing hormone (LH) treatment in cultured preovulatory follicles. The LH-stimulated UGT2B15 mRNA level in cultured preovulatory follicles was inhibited by inhibitors of adenylyl cyclase, phosphoinositide 3-kinase and mitogen-activated protein kinase. Furthermore, a vitamin D receptor agonist (calcitriol) suppressed the LH-stimulated UGT2B15 expression in a dose-dependent manner. Taken together, these results indicate that gonadotropins transiently stimulate UGT2B15 expression and activity in preovulatory follicles, and UGT2B15 mRNA levels are regulated by the progesterone receptor and vitamin D receptor.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Chorionic Gonadotropin/pharmacology
- Enzyme Induction/drug effects
- Female
- Fertility Agents, Female/pharmacology
- Gene Expression Regulation, Developmental/drug effects
- Glucuronosyltransferase/antagonists & inhibitors
- Glucuronosyltransferase/chemistry
- Glucuronosyltransferase/genetics
- Glucuronosyltransferase/metabolism
- Gonadotropins/metabolism
- Granulosa Cells/cytology
- Granulosa Cells/drug effects
- Granulosa Cells/metabolism
- Luteinizing Hormone/pharmacology
- Luteolytic Agents/pharmacology
- Mifepristone/pharmacology
- Ovulation/drug effects
- Ovulation/metabolism
- Protein Kinase Inhibitors/pharmacology
- Rats, Sprague-Dawley
- Receptors, Calcitriol/agonists
- Receptors, Calcitriol/antagonists & inhibitors
- Receptors, Calcitriol/metabolism
- Receptors, Progesterone/agonists
- Receptors, Progesterone/antagonists & inhibitors
- Receptors, Progesterone/metabolism
- Signal Transduction/drug effects
- Tissue Culture Techniques
Collapse
Affiliation(s)
- Phuong T M Dam
- School of Biological Sciences and Technology, Faculty of Life Science, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - You-Jee Jang
- School of Biological Sciences and Technology, Faculty of Life Science, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Jae-Il Park
- Animal Facility of Aging Science, Korea Basic Science Institute, Gwangju 61186, Republic of Korea
| | - Bum-Chae Choi
- Center for Recurrent Miscarriage and Infertility, Creation and Love Women's Hospital, Gwangju 61917, Republic of Korea
| | - Sang-Jin Song
- Center for Recurrent Miscarriage and Infertility, Creation and Love Women's Hospital, Gwangju 61917, Republic of Korea
| | - Sang-Young Chun
- School of Biological Sciences and Technology, Faculty of Life Science, Chonnam National University, Gwangju 500-757, Republic of Korea
| |
Collapse
|
8
|
Braadland PR, Grytli HH, Ramberg H, Katz B, Kellman R, Gauthier-Landry L, Fazli L, Krobert KA, Wang W, Levy FO, Bjartell A, Berge V, Rennie PS, Mellgren G, Mælandsmo GM, Svindland A, Barbier O, Taskén KA. Low β₂-adrenergic receptor level may promote development of castration resistant prostate cancer and altered steroid metabolism. Oncotarget 2016; 7:1878-94. [PMID: 26646591 PMCID: PMC4811504 DOI: 10.18632/oncotarget.6479] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 11/21/2015] [Indexed: 11/25/2022] Open
Abstract
The underlying mechanisms responsible for the development of castration-resistant prostate cancer (CRPC) in patients who have undergone androgen deprivation therapy are not fully understood. This is the first study to address whether β2-adrenergic receptor (ADRB2)- mediated signaling may affect CRPC progression in vivo. By immunohistochemical analyses, we observed that low levels of ADRB2 is associated with a more rapid development of CRPC in a Norwegian patient cohort. To elucidate mechanisms by which ADRB2 may affect CRPC development, we stably transfected LNCaP cells with shRNAs to mimic low and high expression of ADRB2. Two UDP-glucuronosyltransferases, UGT2B15 and UGT2B17, involved in phase II metabolism of androgens, were strongly downregulated in two LNCaP shADRB2 cell lines. The low-ADRB2 LNCaP cell lines displayed lowered glucuronidation activities towards androgens than high-ADRB2 cells. Furthermore, increased levels of testosterone and enhanced androgen responsiveness were observed in LNCaP cells expressing low level of ADRB2. Interestingly, these cells grew faster than high-ADRB2 LNCaP cells, and sustained their low glucuronidation activity in castrated NOD/SCID mice. ADRB2 immunohistochemical staining intensity correlated with UGT2B15 staining intensity in independent TMA studies and with UGT2B17 in one TMA study. Similar to ADRB2, we show that low levels of UGT2B15 are associated with a more rapid CRPC progression. We propose a novel mechanism by which ADRB2 may affect the development of CRPC through downregulation of UGT2B15 and UGT2B17.
Collapse
Affiliation(s)
- Peder Rustøen Braadland
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Helene Hartvedt Grytli
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Håkon Ramberg
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Betina Katz
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Ralf Kellman
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Louis Gauthier-Landry
- Laboratory of Molecular Pharmacology, CHU-Québec Research Center and Faculty of Pharmacy, Laval University, Québec, Canada
| | - Ladan Fazli
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Kurt Allen Krobert
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway.,K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Wanzhong Wang
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Finn Olav Levy
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway.,K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anders Bjartell
- Department of Urology, Skåne University Hospital, Malmø, Sweden.,Department of Clinical Sciences Malmø, Division of Urological Cancers, Lund University, Lund, Sweden
| | - Viktor Berge
- Department of Urology, Oslo University Hospital, Oslo, Norway
| | - Paul S Rennie
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Gunnar Mellgren
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Institute for Pharmacy, Faculty of Health Science, University of Tromsø, Tromsø, Norway
| | - Aud Svindland
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, CHU-Québec Research Center and Faculty of Pharmacy, Laval University, Québec, Canada
| | - Kristin Austlid Taskén
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
9
|
Zimmer BM, Howell ME, Wei Q, Ma L, Romsdahl T, Loughman EG, Markham JE, Seravalli J, Barycki JJ, Simpson MA. Loss of exogenous androgen dependence by prostate tumor cells is associated with elevated glucuronidation potential. HORMONES & CANCER 2016; 7:260-71. [PMID: 27307252 PMCID: PMC4955861 DOI: 10.1007/s12672-016-0268-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/08/2016] [Indexed: 12/20/2022]
Abstract
Prostate epithelial cells control the potency and availability of androgen hormones in part by inactivation and elimination. UDP-glucose dehydrogenase (UGDH) catalyzes the NAD(+)-dependent oxidation of UDP-glucose to UDP-glucuronate, an essential precursor for androgen inactivation by the prostate glucuronidation enzymes UGT2B15 and UGT2B17. UGDH expression is androgen stimulated, which increases the production of UDP-glucuronate and fuels UGT-catalyzed glucuronidation. In this study, we compared the glucuronidation potential and its impact on androgen-mediated gene expression in an isogenic LNCaP model for androgen-dependent versus castration-resistant prostate cancer. Despite significantly lower androgen-glucuronide output, LNCaP 81 castration-resistant tumor cells expressed higher levels of UGDH, UGT2B15, and UGT2B17. However, the magnitude of androgen-activated UGDH and prostate-specific antigen (PSA) expression, as well as the androgen receptor (AR)-dependent repression of UGT2B15 and UGT2B17, was blunted several-fold in these cells. Consistent with these results, the ligand-activated binding of AR to the PSA promoter and subsequent transcriptional activation were also significantly reduced in castration-resistant cells. Analysis of the UDP-sugar pools and flux through pathways downstream of UDP-glucuronate production revealed that these glucuronidation precursor metabolites were channeled through proteoglycan and glycosaminoglycan biosynthetic pathways, leading to increased surface expression of Notch1. Knockdown of UGDH diminished Notch1 and increased glucuronide output. Overall, these results support a model in which the aberrant partitioning of UDP-glucuronate and other UDP-sugars into alternative pathways during androgen deprivation contributes to the loss of prostate tumor cell androgen sensitivity by promoting altered cell surface proteoglycan expression.
Collapse
Affiliation(s)
- Brenna M Zimmer
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA
| | - Michelle E Howell
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA
| | - Qin Wei
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA
| | - Linlin Ma
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA
| | - Trevor Romsdahl
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA
| | - Eileen G Loughman
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA
| | - Jennifer E Markham
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA
| | - Javier Seravalli
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA
| | - Joseph J Barycki
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA
| | - Melanie A Simpson
- Department of Biochemistry, University of Nebraska, 1901 Vine Street, Lincoln, NE, 68588-0664, USA.
| |
Collapse
|
10
|
Lopez SM, Agoulnik AI, Zhang M, Peterson LE, Suarez E, Gandarillas GA, Frolov A, Li R, Rajapakshe K, Coarfa C, Ittmann MM, Weigel NL, Agoulnik IU. Nuclear Receptor Corepressor 1 Expression and Output Declines with Prostate Cancer Progression. Clin Cancer Res 2016; 22:3937-49. [PMID: 26968201 DOI: 10.1158/1078-0432.ccr-15-1983] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 02/19/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Castration therapy in advanced prostate cancer eventually fails and leads to the development of castration-resistant prostate cancer (CRPC), which has no cure. Characteristic features of CRPC can be increased androgen receptor (AR) expression and altered transcriptional output. We investigated the expression of nuclear receptor corepressor 1 (NCOR1) in human prostate and prostate cancer and the role of NCOR1 in response to antiandrogens. EXPERIMENTAL DESIGN NCOR1 protein levels were compared between matched normal prostate and prostate cancer in 409 patient samples. NCOR1 knockdown was used to investigate its effect on bicalutamide response in androgen-dependent prostate cancer cell lines and transcriptional changes associated with the loss of NCOR1. NCOR1 transcriptional signature was also examined in prostate cancer gene expression datasets. RESULTS NCOR1 protein was detected in cytoplasm and nuclei of secretory epithelial cells in normal prostate. Both cytoplasmic and nuclear NCOR1 protein levels were lower in prostate cancer than in normal prostate. Prostate cancer metastases show significant decrease in NCOR1 transcriptional output. Inhibition of LNCaP cellular proliferation by bicalutamide requires NCOR1. NCOR1-regulated genes suppress cellular proliferation and mediate bicalutamide resistance. In the mouse, NCOR1 is required for bicalutamide-dependent regulation of a subset of the AR target genes. CONCLUSIONS In summary, we demonstrated that NCOR1 function declines with prostate cancer progression. Reduction in NCOR1 levels causes bicalutamide resistance in LNCaP cells and compromises response to bicalutamide in mouse prostate in vivo Clin Cancer Res; 22(15); 3937-49. ©2016 AACR.
Collapse
Affiliation(s)
- Sandra M Lopez
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida. Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexander I Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida. Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas
| | - Manqi Zhang
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Leif E Peterson
- Center for Biostatistics, Houston Methodist Research Institute, Houston, Texas
| | - Egla Suarez
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Gregory A Gandarillas
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Anna Frolov
- Dan L. Duncan Cancer Center-Biostatistics, Baylor College of Medicine, Houston, Texas
| | - Rile Li
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Christian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas. Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Nancy L Weigel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Irina U Agoulnik
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas. Biomolecular Sciences Institute, School of Integrated Science and Humanity, Florida international University, Miami, Florida.
| |
Collapse
|
11
|
Dates CR, Fahmi T, Pyrek SJ, Yao-Borengasser A, Borowa-Mazgaj B, Bratton SM, Kadlubar SA, Mackenzie PI, Haun RS, Radominska-Pandya A. Human UDP-Glucuronosyltransferases: Effects of altered expression in breast and pancreatic cancer cell lines. Cancer Biol Ther 2016; 16:714-23. [PMID: 25996841 DOI: 10.1080/15384047.2015.1026480] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Increased aerobic glycolysis and de novo lipid biosynthesis are common characteristics of invasive cancers. UDP-glucuronosyltransferases (UGTs) are phase II drug metabolizing enzymes that in normal cells possess the ability to glucuronidate these lipids and speed their excretion; however, de-regulation of these enzymes in cancer cells can lead to an accumulation of bioactive lipids, which further fuels cancer progression. We hypothesize that UGT2B isoform expression is down-regulated in cancer cells and that exogenous re-introduction of these enzymes will reduce lipid content, change the cellular phenotype, and inhibit cancer cell proliferation. In this study, steady-state mRNA levels of UGT isoforms from the 2B family were measured using qPCR in 4 breast cancer and 5 pancreatic cancer cell lines. Expression plasmids for UGT2B isoforms known to glucuronidate cellular lipids, UGT2B4, 2B7, and 2B15 were transfected into MCF-7 and Panc-1 cells, and the cytotoxic effects of these enzymes were analyzed using trypan blue exclusion, annexin V/PI staining, TUNEL assays, and caspase-3 immunohistochemistry. There was a significant decrease in cell proliferation and a significant increase in the number of dead cells after transfection with each of the 3 UGT isoforms in both cell lines. Cellular lipids were also found to be significantly decreased after transfection. The results presented here support our hypothesis and emphasize the important role UGTs can play in cellular proliferation and lipid homeostasis. Evaluating the effect of UGT expression on the lipid levels in cancer cell lines can be relevant to understanding the complex regulation of cancer cells, identifying the roles of UGTs as "lipid-controllers" in cellular homeostasis, and illustrating their suitability as targets for future clinical therapy development.
Collapse
|
12
|
Qin X, Liu M, Wang X. New insights into the androgen biotransformation in prostate cancer: A regulatory network among androgen, androgen receptors and UGTs. Pharmacol Res 2016; 106:114-122. [PMID: 26926093 DOI: 10.1016/j.phrs.2016.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 01/15/2023]
Abstract
Androgen, as one kind of steroid hormones, is pivotal in the hormone-sensitive cancer, such as prostate cancer (PCa). The synthesis, elimination, and bioavailability of androgen in prostate cells have been proved to be a main cause of the carcinogenesis, maintenance and deterioration of PCa. This review illustrates the outlines of androgen biotransformation, and further discusses the different enzymes, especially UDP-glucuronyltransferases (UGTs) embedded in both benign and malignant prostate cells, which catalyze the reactions. Although many inhibitors of the enzymes responsible for the synthesis of androgens have been developed into drugs to fight against PCa, the elimination procedures metabolized by the UGTs are less emphasized. Thus the regulatory network among androgen, androgen receptors (AR) and UGTs is carefully reviewed in this article, indicating the determinant effects of UGTs on prostatic androgens and the regulation of AR. Finally, the hypothesis is also put forward that the regulators of UGTs may be developed to accelerate the androgen elimination and benefit PCa therapy.
Collapse
Affiliation(s)
- Xuan Qin
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China; Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, TX, USA
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
13
|
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
|
14
|
Gauthier-Landry L, Bélanger A, Barbier O. Multiple roles for UDP-glucuronosyltransferase (UGT)2B15 and UGT2B17 enzymes in androgen metabolism and prostate cancer evolution. J Steroid Biochem Mol Biol 2015; 145:187-92. [PMID: 24861263 DOI: 10.1016/j.jsbmb.2014.05.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 05/13/2014] [Accepted: 05/16/2014] [Indexed: 11/20/2022]
Abstract
In the prostate, approximately 50% of androgens are from adrenal steroids, mainly dehydroepiandrosterone (DHEA), its sulfate and androstenedione. These compounds are converted first into testosterone, and then into the active hormone dihydrotestosterone (DHT). After having activated the androgen receptor (AR), DHT is reduced into androstane-3α-DIOL (3α-DIOL) and androsterone (ADT), which are subsequently converted into 2 inactive and easily excretable metabolites: 3α-DIOL-17glucuronide (3α-DIOL-17G) and ADT-3glucuronide (ADT-3G). The formation of these last derivatives through the glucuronidation reaction involves 2 UDP-glucuronosyltransferase (UGT) enzymes, namely UGT2B15 and UGT2B17. The present review article aims at providing a comprehensive view of the physiological and pharmacological importance of these 2 enzymes for the control of androgen homeostasis. We will resume: (i) how UGT2B15 and UGT2B17 contribute to androgen elimination; (ii) how their glucuronidation capacity influences the androgen signaling pathway in prostate cells; (iii) how they contribute to the anti-proliferative properties of AR antagonists in prostate cancer cells; and (iv) how AR and its spliced variants regulate the UGT2B15 and/or UGT2B17 genes expression. Finally, whether the unexploited AR-UGT axis could serve as a prognostic maker or a pharmacological target for novel therapeutics in the treatment of prostate cancer is also discussed. This article is part of a special issue entitled 'Essential role of DHEA'.
Collapse
Affiliation(s)
- Louis Gauthier-Landry
- Laboratory of Molecular Pharmacology, CHU de Québec Research Centre, and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Alain Bélanger
- CHU de Québec Research Centre, and the Faculty of Medicine, Laval University, Québec, Canada
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, CHU de Québec Research Centre, and the Faculty of Pharmacy, Laval University, Québec, Canada.
| |
Collapse
|
15
|
Mostaghel EA. Beyond T and DHT - novel steroid derivatives capable of wild type androgen receptor activation. Int J Biol Sci 2014; 10:602-13. [PMID: 24948873 PMCID: PMC4062953 DOI: 10.7150/ijbs.8844] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 04/23/2014] [Indexed: 12/16/2022] Open
Abstract
While androgen deprivation therapy (ADT) remains the primary treatment for metastatic prostate cancer (PCa), castration does not eliminate androgens from the prostate tumor microenvironment, and residual intratumoral androgens are implicated in nearly every mechanism by which androgen receptor (AR)-mediated signaling promotes castration-resistant disease. The uptake and intratumoral (intracrine) conversion of circulating adrenal androgens such as dehydroepiandrosterone sulfate (DHEA-S) to steroids capable of activating the wild type AR is a recognized driver of castration resistant prostate cancer (CRPC). However, less well-characterized adrenal steroids, including 11-deoxcorticosterone (DOC) and 11beta-hydroxyandrostenedione (11OH-AED) may also play a previously unrecognized role in promoting AR activation. In particular, recent data demonstrate that the 5α-reduced metabolites of DOC and 11OH-AED are activators of the wild type AR. Given the well-recognized presence of SRD5A activity in CRPC tissue, these observations suggest that in the low androgen environment of CRPC, alternative sources of 5α-reduced ligands may supplement AR activation normally mediated by the canonical 5α-reduced agonist, 5α-DHT. Herein we review the emerging data that suggests a role for these alternative steroids of adrenal origin in activating the AR, and discuss the enzymatic pathways and novel downstream metabolites mediating these effects. We conclude by discussing the potential implications of these findings for CRPC progression, particularly in context of new agents such as abiraterone and enzalutamide which target the AR-axis for prostate cancer therapy.
Collapse
Affiliation(s)
- Elahe A Mostaghel
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle WA, USA
| |
Collapse
|
16
|
Grosse L, Pâquet S, Caron P, Fazli L, Rennie PS, Bélanger A, Barbier O. Androgen Glucuronidation: An Unexpected Target for Androgen Deprivation Therapy, with Prognosis and Diagnostic Implications. Cancer Res 2013; 73:6963-71. [DOI: 10.1158/0008-5472.can-13-1462] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
17
|
Kpoghomou MA, Soatiana JE, Kalembo FW, Bishwajit G, Sheng W. UGT2B17 Polymorphism and Risk of Prostate Cancer: A Meta-Analysis. ISRN ONCOLOGY 2013; 2013:465916. [PMID: 24106614 PMCID: PMC3782843 DOI: 10.1155/2013/465916] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 08/04/2013] [Indexed: 01/02/2023]
Abstract
Objective. Recent studies on the association between uridine diphosphosglucuronosyltransferases (UGTs) 2B17 polymorphism and risk of prostate cancer (PCa) showed inconclusive results. To clarify this possible association, we conducted a meta-analysis of published studies. Methods. We searched the published literature from PubMed, Embase, Google Scholar, and China National Knowledge Infrastructure (CNKI). According to our inclusion criteria, studies that observed the association between UGT2B17 polymorphism and PCa risk were included. The principal outcome measure was the adjusted odds ratio (OR) with 95% confidence interval (CI) for the risk of PCa associated with UGT2B17 polymorphism. Results. A total of 6 studies with 7,029 subjects (3,839 cases and 3,190 controls) were eligible for inclusion in the meta-analysis. Overall, there was a significant association between UGT2B17 polymorphism and increased risk of prostate cancer (OR = 1.74, 95% CI 1.14-2.64, P < 0.001). Similar results were found in the subgroup analyses by ethnicity and types of controls. Conclusion. This meta-analysis demonstrates that UGT2B17 polymorphism is associated with prostate cancer susceptibility, and it contributes to the increased risk of prostate cancer.
Collapse
Affiliation(s)
- Marce-Amara Kpoghomou
- Department of Epidemiology and Biostatistics, School of Public Health, Tong Ji Medical College, 13 Hang Kong Road, Wuhan 430030, China
| | | | | | | | | |
Collapse
|
18
|
Wu Y, Godoy A, Azzouni F, Wilton JH, Ip C, Mohler JL. Prostate cancer cells differ in testosterone accumulation, dihydrotestosterone conversion, and androgen receptor signaling response to steroid 5α-reductase inhibitors. Prostate 2013; 73:1470-82. [PMID: 23813697 PMCID: PMC3999702 DOI: 10.1002/pros.22694] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/08/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND Blocking 5α-reductase-mediated testosterone conversion to dihydrotestosterone (DHT) with finasteride or dutasteride is the driving hypothesis behind two prostate cancer prevention trials. Factors affecting intracellular androgen levels and the androgen receptor (AR) signaling axis need to be examined systematically in order to fully understand the outcome of interventions using these drugs. METHODS The expression of three 5α-reductase isozymes, as determined by immunohistochemistry and qRT-PCR, was studied in five human prostate cancer cell lines. Intracellular testosterone and DHT were analyzed using mass spectrometry. A luciferase reporter assay and AR-regulated genes were used to evaluate the modulation of AR activity. RESULTS Prostate cancer cells were capable of accumulating testosterone to a level 15-50 times higher than that in the medium. The profile and expression of 5α-reductase isozymes did not predict the capacity to convert testosterone to DHT. Finasteride and dutasteride were able to depress testosterone uptake in addition to lowering intracellular DHT. The inhibition of AR activity following drug treatment often exceeded the expected response due to reduced availability of DHT. The ability to maintain high intracellular testosterone might compensate for the shortage of DHT. CONCLUSIONS The biological effect of finasteride or dutasteride appears to be complex and may depend on the interplay of several factors, which include testosterone turnover, enzymology of DHT production, ability to use testosterone and DHT interchangeably, and propensity of cells for off-target AR inhibitory effect.
Collapse
Affiliation(s)
- Yue Wu
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
- Departmentof Urology, Roswell Park Cancer Institute, Buffalo, New York
| | - Alejandro Godoy
- Departmentof Urology, Roswell Park Cancer Institute, Buffalo, New York
- Departmentof Physiology, Pontificia Universidad Católicade Chile, Santiago de Chile
| | - Faris Azzouni
- Departmentof Urology, Roswell Park Cancer Institute, Buffalo, New York
| | - John H. Wilton
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Clement Ip
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
| | - James L. Mohler
- Departmentof Urology, Roswell Park Cancer Institute, Buffalo, New York
- Department of Urology, University at Buffalo School of Medicine and Biotechnology, Buffalo, New York
- Correspondence to: James L. Mohler, MD, Department of Urology, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263.
| |
Collapse
|
19
|
Coviello AD, Haring R, Wellons M, Vaidya D, Lehtimäki T, Keildson S, Lunetta KL, He C, Fornage M, Lagou V, Mangino M, Onland-Moret NC, Chen B, Eriksson J, Garcia M, Liu YM, Koster A, Lohman K, Lyytikäinen LP, Petersen AK, Prescott J, Stolk L, Vandenput L, Wood AR, Zhuang WV, Ruokonen A, Hartikainen AL, Pouta A, Bandinelli S, Biffar R, Brabant G, Cox DG, Chen Y, Cummings S, Ferrucci L, Gunter MJ, Hankinson SE, Martikainen H, Hofman A, Homuth G, Illig T, Jansson JO, Johnson AD, Karasik D, Karlsson M, Kettunen J, Kiel DP, Kraft P, Liu J, Ljunggren Ö, Lorentzon M, Maggio M, Markus MRP, Mellström D, Miljkovic I, Mirel D, Nelson S, Morin Papunen L, Peeters PHM, Prokopenko I, Raffel L, Reincke M, Reiner AP, Rexrode K, Rivadeneira F, Schwartz SM, Siscovick D, Soranzo N, Stöckl D, Tworoger S, Uitterlinden AG, van Gils CH, Vasan RS, Wichmann HE, Zhai G, Bhasin S, Bidlingmaier M, Chanock SJ, De Vivo I, Harris TB, Hunter DJ, Kähönen M, Liu S, Ouyang P, Spector TD, van der Schouw YT, Viikari J, Wallaschofski H, McCarthy MI, Frayling TM, Murray A, Franks S, Järvelin MR, de Jong FH, Raitakari O, Teumer A, Ohlsson C, Murabito JM, Perry JRB. A genome-wide association meta-analysis of circulating sex hormone-binding globulin reveals multiple Loci implicated in sex steroid hormone regulation. PLoS Genet 2012; 8:e1002805. [PMID: 22829776 PMCID: PMC3400553 DOI: 10.1371/journal.pgen.1002805] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 05/19/2012] [Indexed: 01/28/2023] Open
Abstract
Sex hormone-binding globulin (SHBG) is a glycoprotein responsible for the transport and biologic availability of sex steroid hormones, primarily testosterone and estradiol. SHBG has been associated with chronic diseases including type 2 diabetes (T2D) and with hormone-sensitive cancers such as breast and prostate cancer. We performed a genome-wide association study (GWAS) meta-analysis of 21,791 individuals from 10 epidemiologic studies and validated these findings in 7,046 individuals in an additional six studies. We identified twelve genomic regions (SNPs) associated with circulating SHBG concentrations. Loci near the identified SNPs included SHBG (rs12150660, 17p13.1, p = 1.8 × 10(-106)), PRMT6 (rs17496332, 1p13.3, p = 1.4 × 10(-11)), GCKR (rs780093, 2p23.3, p = 2.2 × 10(-16)), ZBTB10 (rs440837, 8q21.13, p = 3.4 × 10(-09)), JMJD1C (rs7910927, 10q21.3, p = 6.1 × 10(-35)), SLCO1B1 (rs4149056, 12p12.1, p = 1.9 × 10(-08)), NR2F2 (rs8023580, 15q26.2, p = 8.3 × 10(-12)), ZNF652 (rs2411984, 17q21.32, p = 3.5 × 10(-14)), TDGF3 (rs1573036, Xq22.3, p = 4.1 × 10(-14)), LHCGR (rs10454142, 2p16.3, p = 1.3 × 10(-07)), BAIAP2L1 (rs3779195, 7q21.3, p = 2.7 × 10(-08)), and UGT2B15 (rs293428, 4q13.2, p = 5.5 × 10(-06)). These genes encompass multiple biologic pathways, including hepatic function, lipid metabolism, carbohydrate metabolism and T2D, androgen and estrogen receptor function, epigenetic effects, and the biology of sex steroid hormone-responsive cancers including breast and prostate cancer. We found evidence of sex-differentiated genetic influences on SHBG. In a sex-specific GWAS, the loci 4q13.2-UGT2B15 was significant in men only (men p = 2.5 × 10(-08), women p = 0.66, heterogeneity p = 0.003). Additionally, three loci showed strong sex-differentiated effects: 17p13.1-SHBG and Xq22.3-TDGF3 were stronger in men, whereas 8q21.12-ZBTB10 was stronger in women. Conditional analyses identified additional signals at the SHBG gene that together almost double the proportion of variance explained at the locus. Using an independent study of 1,129 individuals, all SNPs identified in the overall or sex-differentiated or conditional analyses explained ~15.6% and ~8.4% of the genetic variation of SHBG concentrations in men and women, respectively. The evidence for sex-differentiated effects and allelic heterogeneity highlight the importance of considering these features when estimating complex trait variance.
Collapse
Affiliation(s)
- Andrea D. Coviello
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Section of Endocrinology, Diabetes, and Nutrition, Boston University School of Medicine, Boston, Massachusetts, United States of America
- National Heart, Lung, and Blood Institute's The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Robin Haring
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine, Ernst-Moritz-Arndt University of Greifswald, Greifswald, Germany
| | - Melissa Wellons
- Department of Medicine and Department of Obstetrics and Gynecology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Dhananjay Vaidya
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital and University of Tampere School of Medicine, Tampere, Finland
| | - Sarah Keildson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Chunyan He
- Department of Public Health, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana, United States of America
| | - Myriam Fornage
- University of Texas Health Sciences Center at Houston, Houston, Texas, United States of America
| | - Vasiliki Lagou
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - N. Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Brian Chen
- Program on Genomics and Nutrition and the Center for Metabolic Disease Prevention, School of Public Health, University of California Los Angeles, Los Angeles, California, United States of America
| | - Joel Eriksson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Melissa Garcia
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Yong Mei Liu
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
| | - Annemarie Koster
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Kurt Lohman
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital and University of Tampere School of Medicine, Tampere, Finland
| | - Ann-Kristin Petersen
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jennifer Prescott
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Consortium of Healthy Aging, Rotterdam, The Netherlands
| | - Liesbeth Vandenput
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andrew R. Wood
- Genetics of Complex Traits, Peninsula Medical School, University of Exeter, Exeter, United Kingdom
| | - Wei Vivian Zhuang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Aimo Ruokonen
- Institute of Diagnostics, University of Oulu, Oulu, Finland
| | | | - Anneli Pouta
- National Institute for Health and Welfare and Institute of Health Sciences, University of Oulu, Oulu, Finland
| | | | - Reiner Biffar
- Department of Prosthetic Dentistry, Gerostomatology, and Dental Materials, University of Greifswald, Greifswald, Germany
| | - Georg Brabant
- Experimental and Clinical Endocrinology, University of Lübeck, Lübeck, Germany
| | - David G. Cox
- Cancer Research Center of Lyon, INSERM U1052, Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, United Kingdom
| | - Yuhui Chen
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Steven Cummings
- California Pacific Medical Center, San Francisco, California, United States of America
| | - Luigi Ferrucci
- Longitudinal Studies Section, Clinical Research Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Marc J. Gunter
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, United Kingdom
| | - Susan E. Hankinson
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Massachusetts, United States of America
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Hannu Martikainen
- Department of Obstetrics and Gynecology, University Hospital of Oulu, Oulu, Finland
| | - Albert Hofman
- Netherlands Consortium of Healthy Aging, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - John-Olov Jansson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andrew D. Johnson
- National Heart, Lung, and Blood Institute's The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - David Karasik
- Hebrew SeniorLife Institute for Aging Research and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department of Clinical Sciences and Department of Orthopaedics, Lund University, Malmö, Sweden
| | - Johannes Kettunen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Douglas P. Kiel
- Hebrew SeniorLife Institute for Aging Research and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Jingmin Liu
- Women's Health Initiative Clinical Coordinating Center, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Östen Ljunggren
- Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - Mattias Lorentzon
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marcello Maggio
- Department of Internal Medicine and Biomedical Sciences, Section of Geriatrics, University of Parma, Parma, Italy
| | | | - Dan Mellström
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Iva Miljkovic
- University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Daniel Mirel
- Gene Environment Initiative, Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, Massachusetts, United States of America
| | - Sarah Nelson
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Laure Morin Papunen
- Department of Obstetrics and Gynecology, University Hospital of Oulu, Oulu, Finland
| | - Petra H. M. Peeters
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Leslie Raffel
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Martin Reincke
- Medizinische Klinik and Poliklinik IV, Ludwig-Maximilians University, Munich, Germany
| | - Alex P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Kathryn Rexrode
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Consortium of Healthy Aging, Rotterdam, The Netherlands
| | - Stephen M. Schwartz
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - David Siscovick
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Nicole Soranzo
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Doris Stöckl
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Obstetrics and Gynaecology, Ludwig-Maximilians-University, Munich, Germany
| | - Shelley Tworoger
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Consortium of Healthy Aging, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Carla H. van Gils
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ramachandran S. Vasan
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- National Heart, Lung, and Blood Institute's The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - H.-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Klinikum Großhadern, Munich, Germany
| | - Guangju Zhai
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- Discipline of Genetics, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Shalender Bhasin
- Section of Endocrinology, Diabetes, and Nutrition, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Martin Bidlingmaier
- Medizinische Klinik and Poliklinik IV, Ludwig-Maximilians University, Munich, Germany
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Immaculata De Vivo
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - David J. Hunter
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and University of Tampere School of Medicine, Tampere, Finland
| | - Simin Liu
- Program on Genomics and Nutrition, Department of Epidemiology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Pamela Ouyang
- Division of Cardiology, Johns Hopkins Bayview Medical Center, Baltimore, Maryland, United States of America
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Yvonne T. van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jorma Viikari
- Department of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Henri Wallaschofski
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine, Ernst-Moritz-Arndt University of Greifswald, Greifswald, Germany
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Timothy M. Frayling
- Genetics of Complex Traits, Peninsula Medical School, University of Exeter, Exeter, United Kingdom
| | - Anna Murray
- Genetics of Complex Traits, Peninsula Medical School, University of Exeter, Exeter, United Kingdom
| | - Steve Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Marjo-Riitta Järvelin
- Department of Biostatistics and Epidemiology, School of Public Health, MRC-HPA Centre for Environment and Health, Faculty of Medicine, Imperial College London, London, United Kingdom
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- National Institute of Health and Welfare, University of Oulu, Oulu, Finland
| | - Frank H. de Jong
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Olli Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital and Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joanne M. Murabito
- National Heart, Lung, and Blood Institute's The Framingham Heart Study, Framingham, Massachusetts, United States of America
- Section of General Internal Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - John R. B. Perry
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- Genetics of Complex Traits, Peninsula Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
20
|
Chakraborty SK, Basu NK, Jana S, Basu M, Raychoudhuri A, Owens IS. Protein kinase Cα and Src kinase support human prostate-distributed dihydrotestosterone-metabolizing UDP-glucuronosyltransferase 2B15 activity. J Biol Chem 2012; 287:24387-96. [PMID: 22532564 DOI: 10.1074/jbc.m111.335067] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Because human prostate-distributed UDP-glucuronosyltransferase (UGT) 2B15 metabolizes 5α-dihydrotestosterone (DHT) and 3α-androstane-5α,17β-diol metabolite, we sought to determine whether 2B15 requires regulated phosphorylation similar to UGTs already analyzed. Reversible down-regulation of 2B15-transfected COS-1 cells following curcumin treatment and irreversible inhibition by calphostin C, bisindolylmaleimide, or röttlerin treatment versus activation by phorbol 12-myristate 13-acetate indicated that 2B15 undergoes PKC phosphorylation. Mutation of three predicted PKC and two tyrosine kinase sites in 2B15 caused 70-100 and 80-90% inactivation, respectively. Anti-UGT-1168 antibody trapped 2B15-His-containing co-immunoprecipitates of PKCα in 130-140- and >150-kDa complexes by gradient SDS-PAGE analysis. Complexes bound to WT 2B15-His remained intact during electrophoresis, whereas 2B15-His mutants at phosphorylation sites differentially dissociated. PKCα siRNA treatment inactivated >50% of COS-1 cell-expressed 2B15. In contrast, treatment of 2B15-transfected COS-1 cells with the Src-specific activator 1,25-dihydroxyvitamin D(3) enhanced activity; treatment with the Src-specific PP2 inhibitor or Src siRNA inhibited >50% of the activity. Solubilized 2B15-His-transfected Src-free fibroblasts subjected to in vitro [γ-(33)P]ATP-dependent phosphorylation by PKCα and/or Src, affinity purification, and SDS gel analysis revealed 2-fold more radiolabeling of 55-58-kDa 2B15-His by PKCα than by Src; labeling was additive for combined kinases. Collectively, the evidence indicates that 2B15 requires regulated phosphorylation by both PKCα and Src, which is consistent with the complexity of synthesis and metabolism of its major substrate, DHT. Whether basal cells import or synthesize testosterone for transport to luminal cells for reduction to DHT by 5α-steroid reductase 2, comparatively low-activity luminal cell 2B15 undergoes a complex pattern of regulated phosphorylation necessary to maintain homeostatic DHT levels to support occupation of the androgen receptor for prostate-specific functions.
Collapse
Affiliation(s)
- Sunit K Chakraborty
- Section on Genetic Disorders of Drug Metabolism, Program on Developmental Endocrinology and Genetics, NICHD, National Institutes of Health, Bethesda, Maryland 20892-1830, USA
| | | | | | | | | | | |
Collapse
|
21
|
Hu DG, Mackenzie PI. Forkhead Box Protein A1 Regulates UDP-Glucuronosyltransferase 2B15 Gene Transcription in LNCaP Prostate Cancer Cells. Drug Metab Dispos 2010; 38:2105-9. [DOI: 10.1124/dmd.110.035436] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
22
|
Hu DG, Gardner-Stephen D, Severi G, Gregory PA, Treloar J, Giles GG, English DR, Hopper JL, Tilley WD, Mackenzie PI. A Novel Polymorphism in a Forkhead Box A1 (FOXA1) Binding Site of the Human UDP Glucuronosyltransferase 2B17 Gene Modulates Promoter Activity and Is Associated with Altered Levels of Circulating Androstane-3α,17β-diol Glucuronide. Mol Pharmacol 2010; 78:714-22. [DOI: 10.1124/mol.110.065953] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
23
|
Karatzas A, Giannatou E, Tzortzis V, Gravas S, Aravantinos E, Moutzouris G, Melekos M, Tsezou A. Genetic polymorphisms in the UDP-glucuronosyltransferase 1A1 (UGT1A1) gene and prostate cancer risk in Caucasian men. Cancer Epidemiol 2010; 34:345-9. [DOI: 10.1016/j.canep.2010.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 02/14/2010] [Accepted: 02/23/2010] [Indexed: 10/19/2022]
|
24
|
Huang D, Casale GP, Tian J, Lele SM, Pisarev VM, Simpson MA, Hemstreet GP. Udp-glucose dehydrogenase as a novel field-specific candidate biomarker of prostate cancer. Int J Cancer 2010; 126:315-27. [PMID: 19676054 DOI: 10.1002/ijc.24820] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Uridine diphosphate (UDP)-glucose dehydrogenase (UGDH) catalyzes the oxidation of UDP-glucose to yield UDP-glucuronic acid, a precursor for synthesis of glycosaminoglycans and proteoglycans that promote aggressive prostate cancer (PC) progression. The purpose of our study was to determine if the UGDH expression in normal appearing acini (NAA) from cancerous glands is a candidate biomarker for PC field disease/effect assayed by quantitative fluorescence imaging analysis (QFIA). A polyclonal antibody to UGDH was titrated to saturation binding and fluorescent microscopic images acquired from fixed, paraffin-embedded tissue slices were quantitatively analyzed. Specificity of the assay was confirmed by Western blot analysis and competitive inhibition of tissue labeling with the recombinant UGDH. Reproducibility of the UGDH measurements was high within and across analytical runs. Quantification of UGDH by QFIA and Reverse-Phase Protein Array analysis were strongly correlated (r = 0.97), validating the QFIA measurements. Analysis of cancerous acini (CA) and NAA from PC patients vs. normal acini (NA) from noncancerous controls (32 matched pairs) revealed significant (p < 0.01) differences, with CA (increased) vs. NA, NAA (decreased) vs. NA and CA (increased) vs. NAA. Areas under the Receiver Operating Characteristic curves were 0.68 (95% CI: 0.59-0.83) for NAA and 0.71 (95% CI: 0.59-0.83) for CA (both vs. NA). These results support the UGDH content in prostatic acini as a novel candidate biomarker that may complement the development of a multi-biomarker panel for detecting PC within the tumor adjacent field on a histologically normal biopsy specimen.
Collapse
Affiliation(s)
- Dali Huang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Trottier J, El Husseini D, Perreault M, Pâquet S, Caron P, Bourassa S, Verreault M, Inaba TT, Poirier GG, Bélanger A, Guillemette C, Trauner M, Barbier O. The human UGT1A3 enzyme conjugates norursodeoxycholic acid into a C23-ester glucuronide in the liver. J Biol Chem 2009; 285:1113-21. [PMID: 19889628 DOI: 10.1074/jbc.m109.073908] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Norursodeoxycholic acid (norUDCA) exhibits efficient anti-cholestatic properties in an animal model of sclerosing cholangitis. norUDCA is eliminated as a C(23)-ester glucuronide (norUDCA-23G) in humans. The present study aimed at identifying the human UDP-glucuronosyltransferase (UGT) enzyme(s) involved in hepatic norUDCA glucuronidation and at evaluating the consequences of single nucleotide polymorphisms in the coding region of UGT genes on norUDCA-23G formation. The effects of norUDCA on the formation of the cholestatic lithocholic acid-glucuronide derivative and of rifampicin on hepatic norUDCA glucuronidation were also explored. In vitro glucuronidation assays were performed with microsomes from human tissues (liver and intestine) and HEK293 cells expressing human UGT enzymes and variant allozymes. UGT1A3 was identified as the major hepatic UGT enzyme catalyzing the formation of norUDCA-23G. Correlation studies using samples from a human liver bank (n = 16) indicated that the level of UGT1A3 protein is a strong determinant of in vitro norUDCA glucuronidation. Analyses of the norUDCA-conjugating activity by 11 UGT1A3 variant allozymes identified three phenotypes with high, low, and intermediate capacity. norUDCA is also identified as a competitive inhibitor for the hepatic formation of the pro-cholestatic lithocholic acid-glucuronide derivative, whereas norUDCA glucuronidation is weakly stimulated by rifampicin. This study identifies human UGT1A3 as the major enzyme for the hepatic norUDCA glucuronidation and supports that some coding polymorphisms affecting the conjugating activity of UGT1A3 in vitro may alter the pharmacokinetic properties of norUDCA in cholestasis treatment.
Collapse
Affiliation(s)
- Jocelyn Trottier
- Laboratory of Molecular Pharmacology, CHUQ Research Center, and the Faculty of Pharmacy, Laval University, Québec, Québec G1V 4G2, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Ménard V, Eap O, Harvey M, Guillemette C, Lévesque Ã. Copy-number variations (CNVs) of the human sex steroid metabolizing genesUGT2B17andUGT2B28and their associations with aUGT2B15functional polymorphism. Hum Mutat 2009; 30:1310-9. [DOI: 10.1002/humu.21054] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
Wei Q, Galbenus R, Raza A, Cerny RL, Simpson MA. Androgen-stimulated UDP-glucose dehydrogenase expression limits prostate androgen availability without impacting hyaluronan levels. Cancer Res 2009; 69:2332-9. [PMID: 19244115 DOI: 10.1158/0008-5472.can-08-3083] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
UDP-glucose dehydrogenase (UGDH) oxidizes UDP-glucose to UDP-glucuronate, an essential precursor for production of hyaluronan (HA), proteoglycans, and xenobiotic glucuronides. High levels of HA turnover in prostate cancer are correlated with aggressive progression. UGDH expression is high in the normal prostate, although HA accumulation is virtually undetectable. Thus, its normal role in the prostate may be to provide precursors for glucuronosyltransferase enzymes, which inactivate and solubilize androgens by glucuronidation. In this report, we quantified androgen dependence of UGDH, glucuronosyltransferase, and HA synthase expression. Androgen-dependent and androgen-independent human prostate cancer cell lines were used to test the effects of UGDH manipulation on tumor cell growth, HA production, and androgen glucuronidation. Dihydrotestosterone (DHT) increased UGDH expression approximately 2.5-fold in androgen-dependent cells. However, up-regulation of UGDH did not affect HA synthase expression or enhance HA production. Mass spectrometric analysis showed that DHT was converted to a glucuronide, DHT-G, at a 6-fold higher level in androgen-dependent cells relative to androgen-independent cells. The increased solubilization and elimination of DHT corresponded to slower cellular growth kinetics, which could be reversed in androgen-dependent cells by treatment with a UDP-glucuronate scavenger. Collectively, these results suggest that dysregulated expression of UGDH could promote the development of androgen-independent tumor cell growth by increasing available levels of intracellular androgen.
Collapse
Affiliation(s)
- Qin Wei
- Department of Biochemistry and Chemistry, University of Nebraska, Lincoln, Nebraska, USA
| | | | | | | | | |
Collapse
|
29
|
Genome-wide copy-number-variation study identified a susceptibility gene, UGT2B17, for osteoporosis. Am J Hum Genet 2008; 83:663-74. [PMID: 18992858 DOI: 10.1016/j.ajhg.2008.10.006] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 10/01/2008] [Accepted: 10/09/2008] [Indexed: 11/24/2022] Open
Abstract
Osteoporosis, a highly heritable disease, is characterized mainly by low bone-mineral density (BMD), poor bone geometry, and/or osteoporotic fractures (OF). Copy-number variation (CNV) has been shown to be associated with complex human diseases. The contribution of CNV to osteoporosis has not been determined yet. We conducted case-control genome-wide CNV analyses, using the Affymetrix 500K Array Set, in 700 elderly Chinese individuals comprising 350 cases with homogeneous hip OF and 350 matched controls. We constructed a genomic map containing 727 CNV regions in Chinese individuals. We found that CNV 4q13.2 was strongly associated with OF (p = 2.0 x 10(-4), Bonferroni-corrected p = 0.02, odds ratio = 1.73). Validation experiments using PCR and electrophoresis, as well as real-time PCR, further identified a deletion variant of UGT2B17 in CNV 4q13.2. Importantly, the association between CNV of UGT2B17 and OF was successfully replicated in an independent Chinese sample containing 399 cases with hip OF and 400 controls. We further examined this CNV's relevance to major risk factors for OF (i.e., hip BMD and femoral-neck bone geometry) in both Chinese (689 subjects) and white (1000 subjects) samples and found consistently significant results (p = 5.0 x 10(-4) -0.021). Because UGT2B17 encodes an enzyme catabolizing steroid hormones, we measured the concentrations of serum testosterone and estradiol for 236 young Chinese males and assessed their UGT2B17 copy number. Subjects without UGT2B17 had significantly higher concentrations of testosterone and estradiol. Our findings suggest the important contribution of CNV of UGT2B17 to the pathogenesis of osteoporosis.
Collapse
|
30
|
Bao BY, Chuang BF, Wang Q, Sartor O, Balk SP, Brown M, Kantoff PW, Lee GSM. Androgen receptor mediates the expression of UDP-glucuronosyltransferase 2 B15 and B17 genes. Prostate 2008; 68:839-48. [PMID: 18302198 PMCID: PMC2703184 DOI: 10.1002/pros.20749] [Citation(s) in RCA: 60] [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] [Indexed: 11/07/2022]
Abstract
BACKGROUND Enhanced androgen receptor (AR) activity by increased testosterone availability may play important roles in prostate cancer progressing to castration resistant state. Comparison of expression profiles in androgen dependent and independent prostate tumors demonstrated a marked increase of the expression of UDP-glucuronosyltransferase 2B15 (UGT2B15), an androgen catabolic enzyme. We investigated mechanisms controlling the differential expression of UGT2B15 and B17 in response to androgen treatments. METHODS Gene expression was determined by RT-PCR. The association of AR with UGT2B15/B17 genes was determined by Chromatin immuno-precipitation (CHIP). RNA interference was used to knock-down gene expression. RESULTS UGT2B15 and B17 genes were not expressed in AR negative prostate cancer cell lines, PC3 and DU145, while they were expressed in AR positive cell lines, LNCaP, LNCaP-abl (an androgen independent LNCaP sub-line), and VCaP. The expression levels of UGT2B15/B17 were up-regulated in LNCaP-abl comparing to those in LNCaP. These results suggest the requirement of AR for the expression of UGT2B15/B17. Treatment with DHT down-regulated the expression of UGT2B15/B17 in LNCaP in a time and dose dependent manner and this down-regulation was competitively antagonized by flutamide and bicalutimide, suggesting a pathway mediated by AR. Further CHIP experiments demonstrated the direct interaction of AR with the promoter regions of UGT2B15/B17 genes. Knocking down AR expression in LNCaP significantly reduced the expression of UGT2B15/B17 and completely inhibited the DHT-induced down-regulation of UGT2B15/B17 genes. CONCLUSIONS We demonstrated that UGT2B15 and B17 are primary androgen-regulated genes and AR is required for both their basal expression and their androgen-regulated expression.
Collapse
Affiliation(s)
- Bo-Ying Bao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Bin-Fay Chuang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Qianben Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Oliver Sartor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Steven P. Balk
- Cancer Biology Program, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Philip W. Kantoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gwo-Shu Mary Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Correspondence to: Gwo-Shu Mary Lee, Department of Medical Oncology, D710B, 44 Binney Street, Boston, MA 02115. E-mail:
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Despite clinical and experimental evidence that show androgens are important in prostate carcinogenesis, epidemiologic studies of serum androgens have been inconclusive. In this review, we summarize the current state of the literature and provide insights and direction for epidemiologic research on androgens and prostate cancer. RECENT FINDINGS To date, data on serum androgens in prostate cancer remain inconclusive. Large studies on variants in some androgen-metabolizing genes [SRD5A2, CYP17A1, and hydroxysteroid dehydrogenase (HSD)17B1] do not show a convincing links to prostate cancer, though there are insufficient data to draw conclusions on other genes related to androgen metabolism, including UDP-glycosyltransferases (UGT), sulfotransferases (SULT), CYP3A, and estrogen-related genes. There is some evidence, although controversial, suggesting that select variants may confer risk to certain subtypes of prostate cancer. The most notable finding in 2007 is the highly reproducible link between the chromosome 8q24 risk region and prostate cancer susceptibility. SUMMARY Besides the link between the 8q24 region and prostate cancer risk, population studies do not convincingly show that polymorphisms in androgen metabolism genes are associated with prostate cancer risk. Large epidemiologic studies with comprehensive gene coverage and reliable exposure data are needed to clarify further the role of androgens and their related genes in prostate cancer.
Collapse
Affiliation(s)
- Lisa W Chu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
32
|
Nelson EC, Rodriguez RL, Dawson K, Galvez AF, Evans CP. The Interaction of Genetic Polymorphisms With Lifestyle Factors: Implications for the Dietary Prevention of Prostate Cancer. Nutr Cancer 2008; 60:301-12. [DOI: 10.1080/01635580701745319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Eric C. Nelson
- a Department of Urology , University of California at Davis , Sacramento, California, USA
| | - Raymond L. Rodriguez
- b Center for Excellence in Nutritional Genomics , University of California at Davis , California, USA
| | - Kevin Dawson
- b Center for Excellence in Nutritional Genomics , University of California at Davis , California, USA
| | - Alfredo F. Galvez
- b Center for Excellence in Nutritional Genomics , University of California at Davis , California, USA
| | - Christopher P. Evans
- a Department of Urology , University of California at Davis , Sacramento, California, USA
| |
Collapse
|
33
|
Olsson M, Lindström S, Häggkvist B, Adami HO, Bälter K, Stattin P, Ask B, Rane A, Ekström L, Grönberg H. The UGT2B17 gene deletion is not associated with prostate cancer risk. Prostate 2008; 68:571-5. [PMID: 18247404 DOI: 10.1002/pros.20700] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Deletion polymorphism of the UDP-glucuronosyltransferase 2B17 (UGT2B17) gene has been associated with an increased prostate cancer risk in two previous independent studies. Here we determine the risk in a large-scale population-based case-control study. METHODS Genotyping was conducted with a 5'-nuclease activity assay to distinguish those with one or two UGT2B17 gene copies (ins/del and ins/ins) from individuals homozygous for the deletion (del/del) allele. RESULTS In contrast to previous findings, no association between the UGT2B17 deletion polymorphism and prostate cancer risk was found. Furthermore the UGT2B17 gene deletion did not affect the risk for prostate cancer specific death. CONCLUSION The UGT2B17 deletion polymorphism does not play a major role in prostate cancer susceptibility as previously indicated.
Collapse
Affiliation(s)
- Mats Olsson
- Department of Urology, Karolinska University Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Barbier O, Bélanger A. Inactivation of androgens by UDP-glucuronosyltransferases in the human prostate. Best Pract Res Clin Endocrinol Metab 2008; 22:259-70. [PMID: 18471784 DOI: 10.1016/j.beem.2008.01.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
In the human prostate, dihydrotestosterone (DHT) -- the natural androgen having the highest affinity for the androgen receptor -- is not released directly into the systemic circulation from peripheral target tissues but it is rather converted in situ into two metabolites which have a low affinity for the androgen receptor: androsterone (ADT) and androstane-3alpha,17beta-diol (3alpha-DIOL). Several clinical observations indicate that these two androgen metabolites are further inactivated in the prostate by glucuronidation. In the human, the family of UDP-glucuronosyltransferase (UGT) enzymes comprises 18 members in three subfamilies: UGT1A, UGT2A and UGT2B. Identification of the substrates for each member has revealed that three UGT2B enzymes are mainly responsible for DHT, ADT and 3alpha-DIOL glucuronidation: UGT2B7, UGT2B15 and UGT2B17. Tissue distribution and cellular localization of UGT2B transcripts and proteins clearly indicate that only UGT2B15 and UGT2B17 are expressed in the prostate. Using the human prostate carcinoma LNCaP cell line, it was shown that UGT2B expression and activity are negatively regulated by several factors, including androgens. On the other hand, inhibition of UGT2B115/17 expression by small interfering RNA (siRNA) resulted in an induced response to DHT of androgen-receptor target genes such as PSA, KLK4, NKX3.1, TMPRSS2, SLC16A6 and VEGF. It is suggested that the conjugating activity of UGT enzymes in androgen target tissues is a mechanism for modulating the action of steroids and/or protecting the tissues from deleterious high concentrations of androgens.
Collapse
Affiliation(s)
- Olivier Barbier
- Laboratory of Molecular Endocrinology and Oncology, Laval University Hospital Research Center, Laval University, Québec, Canada
| | | |
Collapse
|
35
|
Pelletier G. Expression of steroidogenic enzymes and sex-steroid receptors in human prostate. Best Pract Res Clin Endocrinol Metab 2008; 22:223-8. [PMID: 18471781 DOI: 10.1016/j.beem.2008.02.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Identification of the cell types expressing the steroidogenic enzymes and sex steroid receptors in the human prostate has recently been performed using immunocytochemistry and in-situ hybridization. The enzymes 3beta-hydroxysteroid dehydrogenase (3beta-HSD), which converts dehydroepiandrosterone (DHEA) into androstenedione, and type 5 17beta-HSD, which catalyzes the reduction of androstenedione to testosterone, have been localized in basal cells of alveoli as well as in stromal cells and endothelial cells of blood vessels. On the other hand, type-2 5alpha-reductase, which converts testosterone into the most potent androgen dihydrotestosterone (DHT), has been mostly observed in the luminal cells in alveoli. Aromatase, which converts testosterone into estradiol, has also been found to be expressed in the luminal cells of the alveoli as well as in stromal cells. Androgen receptor (AR) has been localized in luminal cell nuclei of alveoli and a large number of stromal cells, while estrogen receptor beta has been detected in both basal and luminal cells in alveoli and also in stromal cells.
Collapse
Affiliation(s)
- Georges Pelletier
- Oncology and Molecular Endocrinology Laboratory Research Center, Laval University Hospital Research Center (CRCHUL) and Laval University, Quebec, Canada.
| |
Collapse
|
36
|
Chouinard S, Yueh MF, Tukey RH, Giton F, Fiet J, Pelletier G, Barbier O, Bélanger A. Inactivation by UDP-glucuronosyltransferase enzymes: the end of androgen signaling. J Steroid Biochem Mol Biol 2008; 109:247-53. [PMID: 18467088 DOI: 10.1016/j.jsbmb.2008.03.016] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Conjugation by UDP-Glucuronosyltransferase (UGT) is the major pathway of androgen metabolism and elimination in the human. High concentrations of glucuronide conjugates of androsterone (ADT) and androstane-3alpha,17beta-diol (3alpha-diol) are present in circulation and several studies over the last 30 years have concluded that the serum levels of these metabolites might reflect the androgen metabolism in several tissues, including the liver and androgen target tissues. Three UGT2B enzymes are responsible for the conjugation of DHT and its metabolites ADT and 3alpha-diol: UGT2B7, B15 and B17. UGT2B7 is expressed in the liver and skin whereas UGT2B15 and B17 were found in the liver, prostate and skin. Very specific antibodies against each UGT2B enzyme have been obtained and used for immunohistochemical studies in the human prostate. It was shown that UGT2B17 is expressed in basal cells whereas UGT2B15 is only localized in luminal cells, where it inactivates DHT. By using LNCaP cells, we have also demonstrated that the expression and activity of UGT2B15 and B17 are modulated by several endogenous prostate factors including androgen. Finally, to study the physiological role of UGT2B enzymes, transgenic mice bearing the human UGT2B15 gene were recently obtained. A decrease in reproductive tissue weight from transgenic animals compared to those from control animals was observed. In conclusion, the conjugation by UGT2B7, B15 and B17, which represents a non-reversible step in androgen metabolism, is an important means by which androgens are regulated locally. It is also postulated that UGT enzymes protect the tissue from deleteriously high concentrations of active androgen.
Collapse
Affiliation(s)
- Sarah Chouinard
- Oncology and Molecular Endocrinology Research Center, Laval University Medical Center (CHUL) and Laval University, Quebec G1V 4G2, Canada
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Activators of the farnesoid X receptor negatively regulate androgen glucuronidation in human prostate cancer LNCAP cells. Biochem J 2008; 410:245-53. [PMID: 17988216 DOI: 10.1042/bj20071136] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Androgens are major regulators of prostate cell growth and physiology. In the human prostate, androgens are inactivated in the form of hydrophilic glucuronide conjugates. These metabolites are formed by the two human UGT2B15 [UGT (UDP-glucuronosyltransferase) 2B15] and UGT2B17 enzymes. The FXR (farnesoid X receptor) is a bile acid sensor controlling hepatic and/or intestinal cholesterol, lipid and glucose metabolism. In the present study, we report the expression of FXR in normal and cancer prostate epithelial cells, and we demonstrate that its activation by chenodeoxycholic acid or GW4064 negatively interferes with the levels of UGT2B15 and UGT2B17 mRNA and protein in prostate cancer LNCaP cells. FXR activation also causes a drastic reduction of androgen glucuronidation in these cells. These results point out activators of FXR as negative regulators of androgen-conjugating UGT expression in the prostate. Finally, the androgen metabolite androsterone, which is also an activator of FXR, dose-dependently reduces the glucuronidation of androgens catalysed by UGT2B15 and UGT2B17 in an FXR-dependent manner in LNCaP cells. In conclusion, the present study identifies for the first time the activators of FXR as important regulators of androgen metabolism in human prostate cancer cells.
Collapse
|
38
|
Kaeding J, Bélanger J, Caron P, Verreault M, Bélanger A, Barbier O. Calcitrol (1α,25-dihydroxyvitamin D3) inhibits androgen glucuronidation in prostate cancer cells. Mol Cancer Ther 2008; 7:380-90. [DOI: 10.1158/1535-7163.mct-07-0455] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Chouinard S, Barbier O, Bélanger A. UDP-glucuronosyltransferase 2B15 (UGT2B15) and UGT2B17 Enzymes Are Major Determinants of the Androgen Response in Prostate Cancer LNCaP Cells. J Biol Chem 2007; 282:33466-33474. [PMID: 17848572 DOI: 10.1074/jbc.m703370200] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Uridine diphosphate-glucuronosyltransferase 2 (UGT2)B15 and B17 enzymes conjugate dihydrotestosterone (DHT) and its metabolites androstane-3alpha, 17beta-diol (3alpha-DIOL) and androsterone (ADT). The presence of UGT2B15/B17 in the epithelial cells of the human prostate has been clearly demonstrated, and significant 3alpha-DIOL glucuronide and ADT-glucuronide concentrations have been detected in this tissue. The human androgen-dependent cancer cell line, LNCaP, expresses UGT2B15 and -B17 and is also capable of conjugating androgens. To assess the impact of these two genes in the inactivation of androgens in LNCaP cells, their expression was inhibited using RNA interference. The efficient inhibitory effects of a UGT2B15/B17 small interfering RNA (siRNA) probe was established by the 70% reduction of these UGT mRNA levels, which was further confirmed at the protein levels. The glucuronidation of dihydrotestosterone (DHT), 3alpha-DIOL, and ADT by LNCaP cell homogenates was reduced by more than 75% in UGT2B15/B17 siRNA-transfected LNCaP cells when compared with cells transfected with a non-target probe. In UGT2B15/B17-deficient LNCaP cells, we observe a stronger response to DHT than in control cells, as determined by cell proliferation and expression of eight known androgen-sensitive genes. As expected, the amounts of DHT in cell culture media from control cells were significantly lower than that from UGT2B15/B17 siRNA-treated cells, which was caused by a higher conversion to its corresponding glucuronide derivative. Taken together these data support the idea that UGT2B15 and -B17 are critical enzymes for the local inactivation of androgens and that glucuronidation is a major determinant of androgen action in prostate cells.
Collapse
Affiliation(s)
- Sarah Chouinard
- Oncology and Molecular Endocrinology Research Center, CHUL Research Center, Québec G1V 4G2, Canada; Faculty of Medicine, Laval University, Québec, G1K 7P4, Canada
| | - Olivier Barbier
- Oncology and Molecular Endocrinology Research Center, CHUL Research Center, Québec G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Québec G1K 7P4, Canada
| | - Alain Bélanger
- Oncology and Molecular Endocrinology Research Center, CHUL Research Center, Québec G1V 4G2, Canada; Faculty of Medicine, Laval University, Québec, G1K 7P4, Canada.
| |
Collapse
|
40
|
Caillier B, Lépine J, Tojcic J, Ménard V, Perusse L, Bélanger A, Barbier O, Guillemette C. A pharmacogenomics study of the human estrogen glucuronosyltransferase UGT1A3. Pharmacogenet Genomics 2007; 17:481-95. [PMID: 17558304 DOI: 10.1097/fpc.0b013e32806d87a4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
UGT1A3 is one of the most efficient at conjugating estrone, a precursor for biosynthesis of estradiol in peripheral tissues. We established the genetic mechanisms that might contribute to individual variation in UGT1A3 expression and activity. UGT1A3 first exon and 5'-flanking regions were sequenced in 249 Caucasians. We identified 17 polymorphisms, among them seven regulatory and 10 exonic polymorphisms with six leading to amino-acid changes. Luciferase reporter assays, site-directed mutagenesis and electrophoretic mobility shift assays using hepatoma HepG2 cells were carried out to show functionality of variant promoters. Reduced transcriptional activity was associated with all six variant promoters (two-fold; P<0.001). One of the potential mechanisms would involve the -148 T>C and -581 C>T variations that modulate gene function by affecting hepatocyte nuclear factor-1alpha and hepatocyte nuclear factor-4alpha binding, respectively. Then, estrone-conjugating activity was assessed with 11 heterologously expressed allozymes. Three phenotypes were observed; UGT1A3*1, *2 (WR, VA) and *3 (WR) with high intrinsic clearance values; UGT1A3*5 (QR, WR), *7 (FI), *9 (WR, ML), *10 (VA) and *11 (WR, VA and MI) had intermediate CLint (2X-10X lower vs. *1), whereas UGT1A3*4 (RW), *6 (WR, VA, MV) and *8 (AV) had low CLint (>10X lower vs. *1). Diplotype analyses indicate that 20.1% of individuals carry two alleles affecting UGT1A3 expression and/or activity. This study did not investigate genotype-phenotype association, but raise the possibility that genetically determined variation might contribute to variability in the inactivation of estrone by UGT1A3 and subsequent changes in lifetime exposure to estrogens potentially modifying risk of cancer.
Collapse
Affiliation(s)
- Bertrand Caillier
- Laboratory of Pharmacogenomics, Oncology and Molecular Endocrinology Research Center, CHUQ Research Center and Faculty of Pharmacy, Laval University, Québec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Park J, Chen L, Ratnashinge L, Sellers TA, Tanner JP, Lee JH, Dossett N, Lang N, Kadlubar FF, Ambrosone CB, Zachariah B, Heysek RV, Patterson S, Pow-Sang J. Deletion polymorphism of UDP-glucuronosyltransferase 2B17 and risk of prostate cancer in African American and Caucasian men. Cancer Epidemiol Biomarkers Prev 2006; 15:1473-8. [PMID: 16896035 DOI: 10.1158/1055-9965.epi-06-0141] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
PURPOSE UDP-glucuronosyltransferases (UGT) are a family of enzymes that glucuronidate many endogenous chemicals, including androgens. This makes them more hydrophilic, alters biological activity, and facilitates their excretion. A deletion polymorphism in the UGT2B17 gene was recently described that was associated with a reduced rate of glucuronidation in vivo. The purpose of this study was to determine if the deletion polymorphism is associated with susceptibility to prostate cancer. MATERIALS AND METHODS UGT2B17 expression was determined by reverse transcription-PCR of pathologically normal prostate tissues (n = 5). In a case-control study with 420 patients with incident primary prostate cancer (127 African Americans and 293 Caucasians) and 487 controls (120 African Americans and 367 Caucasians), the frequency of UGT2B17 deletion polymorphism in genomic DNA was compared between cases and controls with PCR analysis. RESULTS UGT2B17 mRNA was detected only in individuals with at least one UGT2B17 allele. The frequency of the null genotype was present in 0.11 and 0.12 of Caucasian and African American controls, respectively. When all subjects were considered, a significant association was found between the UGT2B17 deletion polymorphism and prostate cancer risk [odds ratio (OR), 1.7; 95% confidence interval (95% CI), 1.2-2.6]. There was an increase in prostate cancer risk among individuals with UGT2B17 deletion polymorphism in Caucasians (OR, 1.9; 95% CI, 1.2-3.0) but not in African Americans (OR, 1.3; 95% CI, 0.6-2.7). CONCLUSIONS These results suggest that the UGT2B17 enzyme may play a role in the metabolism of androgens in prostate tissue and that the UGT2B17 deletion polymorphism is associated with prostate cancer risk.
Collapse
Affiliation(s)
- Jong Park
- Division of Cancer Prevention and Control, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The uridine diphosphoglucuronosyltransferases (UGTs) belong to a superfamily of enzymes that catalyse the glucuronidation of numerous endobiotics and xenobiotics. Several human hepatic and extrahepatic UGT isozymes have been characterized with respect to their substrate specificity, tissue expression and gene structure. Genetic polymorphisms have been identified for almost all the UGT family members. A wide variety of anticancer drugs, dietary chemopreventives and carcinogens are known to be conjugated by members of both UGT1A and UGT2B subfamilies. This review examines in detail each UGT isozyme known to be associated with cancer and carcinogenesis. The cancer-related substrates for several UGTs are summarized, and the functionally relevant genetic polymorphisms of UGTs are reviewed. A number of genotype-phenotype association studies have been carried out to characterize the role of UGT pharmacogenetics in several types of cancer, and these examples are discussed here. In summary, this review focuses on the role of the human UGT genetic polymorphisms in carcinogenesis, chemoprevention and cancer risk.
Collapse
Affiliation(s)
- S Nagar
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA 19140, USA.
| | | |
Collapse
|