1
|
Livie C, Lee LCY, Alimussina M, Conceicao C, Leung CH, McNeilly J, Ahmed SF, Syme C, Smith K, Johnston S. A liquid chromatography-tandem mass spectrometry method for the analysis of dehydroepiandrosterone sulphate (DHEAs) in serum and plasma with comparison to an immunoassay method in a neonatal population. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1220:123615. [PMID: 36906955 DOI: 10.1016/j.jchromb.2023.123615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 01/14/2023] [Accepted: 01/21/2023] [Indexed: 02/02/2023]
Abstract
The measurement of dehydroepiandrosterone-sulphate (DHEAs) is an important second-line test to aid in the diagnosis of premature adrenarche, peripubertal gynaecomastia in males and in identifying the source of elevated androgens in females. Historically, DHEAs has been measured by immunoassay platforms which are prone to poor sensitivity and more importantly poor specificity. The aim was to develop an LC-MSMS method for the measurement of DHEAs in human plasma and serum, develop an in-house paediatric (<6 year old) reference limit and compare the performance against the Abbott Alinity DHEAs immunoassay method. Following pre-treatment with an internal standard, samples were loaded onto EVOLUTE® EXPRESS ABN plate. Analytes were separated with reverse-phase chromatography using ACQUITY® UPLC® HSS T3 2.1 mm × 50 mm, 1.8 μm column. Mass spectrometry detection was performed using a Waters® Xevo TQ-XS in electrospray negative mode. For the paediatric reference range, samples were collected from an inpatient setting (age ≤ 6 years old) with no evidence of adrenal dysfunction or history of/current steroid use. The method comparison was performed using samples from this cohort aged between 0 and 52 weeks. The assay demonstrated linearity up to 15 µmol/L (r2 > 0.99) with a functional sensitivity of 0.1 µmol/L. Accuracy results revealed a mean bias of 0.7% (-14% to 15%) when compared against the NEQAS EQA LC-MSMS consensus mean (n = 48). The paediatric reference limit was calculated as ≤ 2.3 µmol/L (95% C.I. 1.4 to 3.8 µmol/L) for ≤ 6 year olds (n = 38). Comparison of neonatal (<52 weeks) DHEAs with the Abbott Alinity revealed that the immunoassay ran at a 166% positive bias (n = 24) which appeared to lessen with increasing age. Described is a robust LC-MSMS method for the measurement of plasma or serum DHEAs validated against internationally recognised protocols. Comparison of paediatric samples of <52 weeks against an immunoassay platform demonstrated that in the immediate new-born period results generated from the LC-MSMS method offer superior specificity than an immunoassay platform.
Collapse
Affiliation(s)
- Craig Livie
- Department of Clinical Biochemistry, Glasgow Royal Infirmary, Glasgow, UK.
| | - Louisa C Y Lee
- Department of Clinical Biochemistry, Royal Alexandra Hospital, Paisley, UK
| | - Malika Alimussina
- Developmental Endocrinology Research Group, University of Glasgow, Royal Hospital for Children (RHC), Glasgow, UK
| | - Carina Conceicao
- Department of Clinical Biochemistry, Glasgow Royal Infirmary, Glasgow, UK
| | - Chui Ha Leung
- Department of Clinical Biochemistry, Glasgow Royal Infirmary, Glasgow, UK
| | - Jane McNeilly
- Department of Clinical Biochemistry, Queen Elizabeth University Hospital, Glasgow, UK
| | - Syed Faisal Ahmed
- Developmental Endocrinology Research Group, University of Glasgow, Royal Hospital for Children (RHC), Glasgow, UK
| | - Charlotte Syme
- Department of Clinical Biochemistry, Forth Valley Royal Hospital, Falkirk, UK
| | - Karen Smith
- Department of Clinical Biochemistry, Glasgow Royal Infirmary, Glasgow, UK
| | - Susan Johnston
- Department of Clinical Biochemistry, Glasgow Royal Infirmary, Glasgow, UK
| |
Collapse
|
2
|
Chan KQ, Stewart C, Chester N, Hamzah SH, Yusof A. The effect of Eurycoma Longifolia on the regulation of reproductive hormones in young males. Andrologia 2021; 53:e14001. [PMID: 33559971 DOI: 10.1111/and.14001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/13/2021] [Accepted: 01/17/2021] [Indexed: 11/30/2022] Open
Abstract
Eurycoma longifolia supplementation increases testosterone levels in humans via activation of the hypothalamic-pituitary-gonadal axis and/or the hypothalamic-pituitary-adrenal axis mainly in older adults and nonhealthy populations. This study aimed to assess the impact of Eurycoma longifolia on the hypothalamic-pituitary-gonadal and hypothalamic-pituitary-adrenal axes in healthy young males since this might promote functional testosterone prowess. Thirty-two males (24.4 ± 4.7 years; 1.74 ± 0.07 m; 73.7 ± 8.4 kg) in a placebo-controlled, double-blind, matched-paired study received 600 mg/day Eurycoma longifolia or placebo for two weeks. Blood analysis using repeated measures analysis of variance showed significant interaction and time effects for testosterone (F1,30 = 9.04, p = .005), free testosterone (F1,30 = 7.13, p = .012) and estradiol (F1,30 = 8.07, p = .008) levels in favour of the treatment group, while luteinising hormone, follicle-stimulating hormone and sexual hormone-binding globulin did not. The lack of changes in luteinising hormone and follicle-stimulating hormone levels suggests that a lesser role played by Eurycoma longifolia in activating the hypothalamic-pituitary-gonadal axis in the young adults. The raised testosterone level may be due to a greater rate of hormone production via the hypothalamic-pituitary-adrenal axis. The supplementation of Eurycoma longifolia for two weeks demonstrates steroidogenic effects on young men were dose-related. Consequently, the raised testosterone following Eurycoma longifolia supplementations could benefit muscle and strength gain in young adults.
Collapse
Affiliation(s)
- Kai Quin Chan
- Department of Sport Science, Faculty of Applied Sciences, Tunku Abdul Rahman University College, Jalan Genting Kelang, Kuala Lumpur, Malaysia.,School of Sport and Exercise Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK.,Centre for Sport and Exercise Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Claire Stewart
- School of Sport and Exercise Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Neil Chester
- School of Sport and Exercise Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Sareena H Hamzah
- Centre for Sport and Exercise Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Ashril Yusof
- Centre for Sport and Exercise Sciences, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
3
|
Abstract
The adrenal gland is a source of sex steroid precursors, and its activity is particularly relevant during fetal development and adrenarche. Following puberty, the synthesis of androgens by the adrenal gland has been considered of little physiologic importance. Dehydroepiandrosterone (DHEA) and its sulfate, DHEAS, are the major adrenal androgen precursors, but they are biologically inactive. The second most abundant unconjugated androgen produced by the human adrenals is 11β-hydroxyandrostenedione (11OHA4). 11-Ketotestosterone, a downstream metabolite of 11OHA4 (which is mostly produced in peripheral tissues), and its 5α-reduced product, 11-ketodihydrotestosterone, are bioactive androgens, with potencies equivalent to those of testosterone and dihydrotestosterone. These adrenal-derived androgens all share an oxygen atom on carbon 11, so we have collectively termed them 11-oxyandrogens. Over the past decade, these androgens have emerged as major components of several disorders of androgen excess, such as congenital adrenal hyperplasia, premature adrenarche and polycystic ovary syndrome, as well as in androgen-dependent tumours, such as castration-resistant prostate cancer. Moreover, in contrast to the more extensively studied, traditional androgens, circulating concentrations of 11-oxyandrogens do not demonstrate an age-dependent decline. This Review focuses on the rapidly expanding knowledge regarding the implications of 11-oxyandrogens in human physiology and disease.
Collapse
Affiliation(s)
- Adina F Turcu
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA.
| | - Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - William E Rainey
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Nakamura Y, Ise K, McNamara KM, Azmahani A, Sato S, Fujishima F, Joh K, Suzuki H, Mitsuzuka K, Arai Y, Takahashi H, Sasano H. The expression of sex steroid receptors and sex steroid–synthesizing/metabolizing enzymes in metastasized lymph nodes of prostate cancer. Hum Pathol 2019; 84:124-132. [DOI: 10.1016/j.humpath.2018.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/16/2022]
|
5
|
Da Silva MHA, De Souza DB. Current evidence for the involvement of sex steroid receptors and sex hormones in benign prostatic hyperplasia. Res Rep Urol 2019; 11:1-8. [PMID: 30662879 PMCID: PMC6327899 DOI: 10.2147/rru.s155609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is a pathology that affects 50% of men over 50 years of age and 90% of men develop BPH in their eighth decade of life. In 2018, more than 1 billion men will be affected by this disease worldwide. However, the progression of BPH is highly complex and has been debated and studied for approximately four decades. Recent studies indicate that BPH can originate from the alteration of different hormone synthesis pathways, and that it is also linked to the function of hormone receptors. There is a close relationship between the progression of BPH and sexual hormones, such as progesterone, testosterone, dihydrotestosterone, and estrogen. The focus of this study was to characterize the interactions of these hormones and investigate the direct or indirect role of each sex hormone receptor in the progression of BPH. Although several studies have described the effects of these hormones on BPH, no conclusions have been drawn regarding their role in disease progression. Here, we present a literature review on the sexual receptors possibly involved in the progression of BPH.
Collapse
|
6
|
Screening, synthesis, crystal structure, and molecular basis of 6-amino-4-phenyl-1,4-dihydropyrano[2,3-c]pyrazole-5-carbonitriles as novel AKR1C3 inhibitors. Bioorg Med Chem 2018; 26:5934-5943. [DOI: 10.1016/j.bmc.2018.10.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/15/2018] [Accepted: 10/30/2018] [Indexed: 01/17/2023]
|
7
|
Rege J, Turcu AF, Kasa-Vubu JZ, Lerario AM, Auchus GC, Auchus RJ, Smith JM, White PC, Rainey WE. 11-Ketotestosterone Is the Dominant Circulating Bioactive Androgen During Normal and Premature Adrenarche. J Clin Endocrinol Metab 2018; 103:4589-4598. [PMID: 30137510 PMCID: PMC6226603 DOI: 10.1210/jc.2018-00736] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/14/2018] [Indexed: 01/18/2023]
Abstract
CONTEXT Adrenarche refers to the rise of dehydroepiandrosterone sulfate (DHEA-S) associated with the development of a functional adrenal zona reticularis. Clinical features of adrenarche include onset of body odor, axillary hair, and pubic hair, which reflect increased androgen action. An early rise in adrenal androgens, or premature adrenarche (PremA), is a risk factor for adverse metabolic profiles in adolescence and adulthood. The bioactive androgens associated with adrenarche and PremA remain poorly understood. The adrenal gland is a potential source of testosterone (T) and the 11-oxygenated derivatives 11β-hydroxytestosterone (11OHT) and 11-ketotestosterone (11KT). OBJECTIVE The objective of this study was to characterize the adrenal androgen biome contributing to adrenarche and PremA. PARTICIPANTS AND METHODS With the use of mass spectrometry, 19 steroids including the 11-oxygenated derivatives of T were measured in sera obtained from girls with PremA (n = 37; 4 to 7 years) and age-matched girls (n = 83; 4 to 10 years). RESULTS In reference population girls, dehydroepiandrosterone, DHEA-S, androstenediol-3-sulfate, T, and 11KT all increased at the onset of adrenarche (6 to 8 years) and beyond (9 to 10 years) (P < 0.05 vs younger subjects 4 to 5 years). T, 11OHT, and 11KT were further elevated in PremA vs age-matched girls (P < 0.001). Circulating concentrations of 11KT during adrenarche and PremA exceeded those of T and 11OHT (11KT > T ≥ 11OHT). Androgen receptor activity and nuclear translocation studies demonstrated that 11KT is a potent androgen similar to T. CONCLUSIONS Our findings suggest that 11KT is the dominant bioactive androgen in children during adrenarche and PremA. Its androgenic capacity suggests that it may be responsible for the phenotypic changes seen in these phenomena.
Collapse
Affiliation(s)
- Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | - Antonio M Lerario
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gabriela C Auchus
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | | | - Perrin C White
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Correspondence and Reprint Requests: William E. Rainey, PhD, Departments of Molecular and Integrative Physiology and Internal Medicine, University of Michigan, 1150 West Medical Center Drive, 2560C Medical Science Research Building II, Ann Arbor, Michigan 48109-5622. E-mail:
| |
Collapse
|
8
|
Morais-Santos M, Werneck-Gomes H, Campolina-Silva GH, Santos LC, Mahecha GAB, Hess RA, Oliveira CA. Basal Cells Show Increased Expression of Aromatase and Estrogen Receptor α in Prostate Epithelial Lesions of Male Aging Rats. Endocrinology 2018; 159:723-732. [PMID: 29121167 DOI: 10.1210/en.2017-00773] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/01/2017] [Indexed: 12/16/2022]
Abstract
Besides androgens, estrogen signaling plays a key role in normal development and pathologies of the prostate. Irreversible synthesis of estrogens from androgens is catalyzed by aromatase. Interestingly, animals lacking aromatase do not develop cancer or prostatitis, whereas those with overexpression of aromatase and, consequently, high estrogen levels develop prostatitis and squamous metaplasia via estrogen receptor 1 (ERα). Even with this evidence, the aromatase expression in the prostate is controversial. Moreover, little is known about the occurrence of age-dependent variation of aromatase and its association with histopathological changes commonly found in advanced age, a knowledge gap that is addressed herein. For this purpose, the immunoexpression of aromatase was evaluated in the prostatic complex of young adult to senile Wistar rats. ERα was also investigated, to extend our understanding of estrogen responsiveness in the prostate. Moderate cytoplasmic immunoreactivity for aromatase was detected in the glandular epithelium. Eventually, some basal cells showed intense staining for aromatase. The expression pattern for aromatase appeared similar in the normal epithelium when young and senile rats were compared; this result was corroborated by Western blotting. Conversely, in senile rats, there was an increase in the frequency of basal cells intensely stained for aromatase, which appeared concentrated in areas of intraepithelial proliferation and prostatitis. These punctual areas also presented increased ERα positivity. Together, these findings suggest a plausible source for hormonal imbalance favoring estrogen production, which, by acting through ERα, may favor the development of prostatic lesions commonly found in advanced age.
Collapse
Affiliation(s)
- Mônica Morais-Santos
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Animal Biology, Universidade Federal de Viçosa,, Viçosa, Minas Gerais, Brazil
| | - Hipácia Werneck-Gomes
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriel H Campolina-Silva
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Leticia C Santos
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Germán A B Mahecha
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rex A Hess
- Department of Comparative Biosciences, University of Illinois, Urbana, Illinois
| | - Cleida A Oliveira
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
9
|
Leimgruber C, Quintar AA, Peinetti N, Scalerandi MV, Nicola JP, Miano JM, Maldonado CA. Testosterone Rescues the De-Differentiation of Smooth Muscle Cells Through Serum Response Factor/Myocardin. J Cell Physiol 2017; 232:2806-2817. [PMID: 27861881 DOI: 10.1002/jcp.25679] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/07/2016] [Indexed: 12/23/2022]
Abstract
Prostatic smooth muscle cells (pSMCs) differentiation is a key factor for prostatic homeostasis, with androgens exerting multiple effects on these cells. Here, we demonstrated that the myodifferentiator complex Srf/Myocd is up-regulated by testosterone in a dose-dependent manner in primary cultures of rat pSMCs, which was associated to the increase in Acta2, Cnn1, and Lmod1 expressions. Blocking Srf or Myocd by siRNAs inhibited the myodifferentiator effect of testosterone. While LPS led to a dedifferentiated phenotype in pSMCs, characterized by down-regulation of Srf/Myocd and smooth muscle cell (SMC)-restricted genes, endotoxin treatment on Myocd-overexpressing cells did not result in phenotypic alterations. Testosterone at a physiological dose was able to restore the muscular phenotype by normalizing Srf/Myocd expression in inflammation-induced dedifferentiated pSMCs. Moreover, the androgen reestablished the proliferation rate and IL-6 secretion increased by LPS. These results provide novel evidence regarding the myodifferentiating role of testosterone on SMCs by modulating Srf/Myocd. Thus, androgens preserve prostatic SMC phenotype, which is essential to maintain the normal structure and function of the prostate. J. Cell. Physiol. 232: 2806-2817, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Carolina Leimgruber
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Amado A Quintar
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nahuel Peinetti
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María V Scalerandi
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Juan P Nicola
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Cristina A Maldonado
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
10
|
Abstract
INTRODUCTION AKR1C3 is a drug target in hormonal and hormonal independent malignancies and acts as a major peripheral 17β-hydroxysteroid dehydrogenase to yield the potent androgens testosterone and dihydrotestosterone, and as a prostaglandin (PG) F synthase to produce proliferative ligands for the PG FP receptor. AKR1C3 inhibitors may have distinct advantages over existing therapeutics for the treatment of castration resistant prostate cancer, breast cancer and acute myeloid leukemia. Area covered: This article reviews the patent literature on AKR1C3 inhibitors using SciFinder which identified inhibitors in the following chemical classes: N-phenylsulfonyl-indoles, N-(benzimidazoylylcarbonyl)- N-(indoylylcarbonyl)- and N-(pyridinepyrrolyl)- piperidines, N-benzimidazoles and N-benzindoles, repurposed nonsteroidal antiinflammatory drugs (indole acetic acids, N-phenylanthranilates and aryl propionic acids), isoquinolines, and nitrogen and sulfur substituted estrenes. The article evaluates inhibitor AKR potency, specificity, efficacy in cell-based and xenograft models and clinical utility. The advantage of bifunctional compounds that either competitively inhibit AKR1C3 and block its androgen receptor (AR) coactivator function or act as AKR1C3 inhibitors and direct acting AR antagonists are discussed. Expert opinion: A large number of potent and selective inhibitors of AKR1C3 have been described however, preclinical optimization, is required before their benefit in human disease can be assessed.
Collapse
Affiliation(s)
- Trevor M Penning
- a Center of Excellence in Environmental Toxicology & Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine , University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
11
|
Vitku J, Kolatorova L, Hampl R. Occurrence and reproductive roles of hormones in seminal plasma. Basic Clin Androl 2017; 27:19. [PMID: 29046808 PMCID: PMC5640966 DOI: 10.1186/s12610-017-0062-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/16/2017] [Indexed: 12/13/2022] Open
Abstract
Only 2-5% of seminal fluid is composed of spermatozoa, while the rest is seminal plasma. The seminal plasma is a rich cocktail of organic and inorganic compounds including hormones, serving as a source of nutrients for sperm development and maturation, protecting them from infection and enabling them to overcome the immunological and chemical environment of the female reproductive tract. In this review, a survey of the hormones found in human seminal plasma, with particular emphasis on reproductive hormones is provided. Their participation in fertilization is discussed including their indispensable role in ovum fertilization. The origin of individual hormones found in seminal plasma is discussed, along with differences in the concentrations in seminal plasma and blood plasma. A part of review is devoted to methods of measurement, emphasising particular instances in which they differ from measurement in blood plasma. These methods include separation techniques, overcoming the matrix effect and current ways for end-point measurement, focusing on so called hyphenated techniques as a combination of chromatographic separation and mass spectrometry. Finally, the informative value of their determination as markers of male fertility disorders (impaired spermatogenesis, abnormal sperm parameters, varicocele) is discussed, along with instances where measuring their levels in seminal plasma is preferable to measurement of levels in blood plasma.
Collapse
Affiliation(s)
- Jana Vitku
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 8, 116 94 Prague, Czech Republic
| | - Lucie Kolatorova
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 8, 116 94 Prague, Czech Republic
| | - Richard Hampl
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 8, 116 94 Prague, Czech Republic
| |
Collapse
|
12
|
Mendonca BB, Gomes NL, Costa EMF, Inacio M, Martin RM, Nishi MY, Carvalho FM, Tibor FD, Domenice S. 46,XY disorder of sex development (DSD) due to 17β-hydroxysteroid dehydrogenase type 3 deficiency. J Steroid Biochem Mol Biol 2017; 165:79-85. [PMID: 27163392 DOI: 10.1016/j.jsbmb.2016.05.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/04/2016] [Accepted: 05/05/2016] [Indexed: 12/14/2022]
Abstract
17β-hydroxysteroid dehydrogenase 3 deficiency consists of a defect in the last phase of steroidogenesis, in which androstenedione is converted into testosterone and estrone into estradiol. External genitalia range from female-like to atypical genitalia and most affected males are raised as females. Virilization in subjects with 17β-HSD3 deficiency occurs at the time of puberty and several of them change to male social sex. In male social sex patients, testes can be safely maintained, as long as they are positioned inside the scrotum The phenotype of 46,XY DSD due to 17β-HSD3 deficiency is extremely variable and clinically indistinguishable from other causes of 46,XY DSD such as partial androgen insensitivity syndrome and 5α-reductase 2 deficiency. Laboratory diagnosis is based on a low testosterone/androstenedione ratio due to high serum levels of androstenedione and low levels of testosterone. The disorder is caused by a homozygous or compound heterozygous mutations in the HSD17B3 gene that encodes the 17β-HSD3 isoenzyme leading to an impairment of the conversion of 17-keto into 17-hydroxysteroids. Molecular genetic testing confirms the diagnosis and provides the orientation for genetic counseling. Our proposal in this article is to review the previously reported cases of 17β-HSD3 deficiency adding our own cases.
Collapse
Affiliation(s)
- Berenice B Mendonca
- Developmental Endocrinology Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Internal Medicine Department, Medical School, University of São Paulo, Brazil.
| | - Nathalia Lisboa Gomes
- Developmental Endocrinology Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Internal Medicine Department, Medical School, University of São Paulo, Brazil
| | - Elaine M F Costa
- Developmental Endocrinology Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Internal Medicine Department, Medical School, University of São Paulo, Brazil
| | - Marlene Inacio
- Developmental Endocrinology Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Internal Medicine Department, Medical School, University of São Paulo, Brazil
| | - Regina M Martin
- Developmental Endocrinology Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Internal Medicine Department, Medical School, University of São Paulo, Brazil
| | - Mirian Y Nishi
- Developmental Endocrinology Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Internal Medicine Department, Medical School, University of São Paulo, Brazil
| | | | - Francisco Denes Tibor
- Urology Division, Surgery Department, Medical School, University of São Paulo, Brazil
| | - Sorahia Domenice
- Developmental Endocrinology Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Internal Medicine Department, Medical School, University of São Paulo, Brazil
| |
Collapse
|
13
|
Rege J, Karashima S, Lerario AM, Smith JM, Auchus RJ, Kasa-Vubu JZ, Sasano H, Nakamura Y, White PC, Rainey WE. Age-dependent Increases in Adrenal Cytochrome b5 and Serum 5-Androstenediol-3-sulfate. J Clin Endocrinol Metab 2016; 101:4585-4593. [PMID: 27623070 PMCID: PMC5155691 DOI: 10.1210/jc.2016-2864] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Adrenal production of dehydroepiandrosterone sulfate (DHEA-S) increases throughout childhood owing to expansion of the zona reticularis (ZR). ZR features cells with a steroidogenic phenotype distinct from that of the adjacent zona fasciculata, with higher expression of cytochrome b5 type A (CYB5A) and steroid sulfotransferase type 2A1 but decreased 3β-hydroxysteroid dehydrogenase type 2 (HSD3B2). In addition to DHEA-S, three adrenal Δ5-steroid sulfates could provide additional tools to define adrenal maturation. OBJECTIVE This study sought to simultaneously measure serum levels of four adrenal Δ5-steroid sulfates, pregnenolone sulfate (Preg-S), 17α-hydroxypregnenolone sulfate (17OHPreg-S), DHEA-S, and 5-androstenediol-3-sulfate (Adiol-S) as a function of age and relate their production to the age-dependent adrenal localization of CYB5A. PARTICIPANTS AND METHODS Δ5-steroid sulfates were quantified by liquid chromatography-tandem mass spectrometry in sera from 247 normal children (129 males,118 females) age 1.5-18 y and 42 adults (20 males, 22 females). Immunofluorescence localized HSD3B2 and CYB5A in normal adrenal glands from subjects age 2-35 y. Finally, HAC15 adrenocortical cells were transduced with lentiviral short hairpin RNA to suppress CYB5A expression. RESULTS Of the Δ5-steroid sulfates quantified, DHEA-S was most abundant. Adiol-S increased in parallel with DHEA-S. Steroid ratios (17OHPreg-S/DHEA-S) suggested increases in 17,20-lyase activity during childhood. Immunofluorescence analysis showed age-related increases in ZR CYB5A immunoreactivity. Furthermore, silencing CYB5A in HAC15 adrenocortical cells significantly reduced DHEA-S and Adiol-S production. CONCLUSION Adiol-S shows a similar age-related increase to that of DHEA-S. This likely results from the childhood expansion of CYB5A-expressing ZR, which enhances 17,20-lyase activity and the production of DHEA-S and Adiol-S.
Collapse
Affiliation(s)
- Juilee Rege
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| | - Shigehiro Karashima
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| | - Antonio M Lerario
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| | - Joshua M Smith
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| | - Richard J Auchus
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| | - Josephine Z Kasa-Vubu
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| | - Hironobu Sasano
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| | - Yasuhiro Nakamura
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| | - Perrin C White
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| | - William E Rainey
- Department of Molecular and Integrative Physiology (J.R., S.K., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Internal Medicine (A.M.L., R.J.A.), University of Michigan, Ann Arbor, Michigan 48109; Division of Pediatric Endocrinology (J.M.S.), Specially for Children, Austin, Texas 78723; Department of Pediatrics (J.Z.K.-V.), University of Michigan, Ann Arbor, Michigan 48109; Department of Pathology (H.S., Y.N.), Tohoku University School of Medicine, Sendai, 980-8575 Japan; Division of Pathology, Faculty of Medicine (Y.N.), Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan; and Department of Pediatrics (P.C.W.), University of Texas Southwestern Medical Center, Dallas, Texas 75235
| |
Collapse
|
14
|
Cabeza M, Sánchez-Márquez A, Garrido M, Silva A, Bratoeff E. Recent Advances in Drug Design and Drug Discovery for Androgen- Dependent Diseases. Curr Med Chem 2016; 23:792-815. [PMID: 26861003 PMCID: PMC5412001 DOI: 10.2174/0929867323666160210125642] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 12/28/2015] [Accepted: 02/09/2016] [Indexed: 11/22/2022]
Abstract
This article summarizes the importance of different targets such as 5α-reductase, 17β-HSD, CYP17A, androgen receptor and protein kinase A for the treatment of prostate cancer and benign prostatic hyperplasia. It is a well known fact that dihydrotestosterone (DHT) is associated with the development of androgen-dependent afflictions. At the present time, several research groups are attempting to develop new steroidal and non-steroidal molecules with the purpose of inhibiting the synthesis and biological response of DHT. This review also discusses the most recent studies reported in the literature that describe the therapeutic potential of novel compounds, as well as the new drugs, principally inhibitors of 5α-reductase.
Collapse
Affiliation(s)
- Marisa Cabeza
- Departamento De Sistemas Biológicos, Universidad Autónoma Metropolitana- Xochimilco Calzada Del Hueso No. 1100, México, D.F., C.P. 04960, México.
| | | | | | | | | |
Collapse
|
15
|
Liu TT, Grubisha MJ, Frahm KA, Wendell SG, Liu J, Ricke WA, Auchus RJ, DeFranco DB. Opposing Effects of Cyclooxygenase-2 (COX-2) on Estrogen Receptor β (ERβ) Response to 5α-Reductase Inhibition in Prostate Epithelial Cells. J Biol Chem 2016; 291:14747-60. [PMID: 27226548 DOI: 10.1074/jbc.m115.711515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Indexed: 11/06/2022] Open
Abstract
Current pharmacotherapies for symptomatic benign prostatic hyperplasia (BPH), an androgen receptor-driven, inflammatory disorder affecting elderly men, include 5α-reductase (5AR) inhibitors (i.e. dutasteride and finasteride) to block the conversion of testosterone to the more potent androgen receptor ligand dihydrotestosterone. Because dihydrotestosterone is the precursor for estrogen receptor β (ERβ) ligands, 5AR inhibitors could potentially limit ERβ activation, which maintains prostate tissue homeostasis. We have uncovered signaling pathways in BPH-derived prostate epithelial cells (BPH-1) that are impacted by 5AR inhibition. The induction of apoptosis and repression of the cell adhesion protein E-cadherin by the 5AR inhibitor dutasteride requires both ERβ and TGFβ. Dutasteride also induces cyclooxygenase type 2 (COX-2), which functions in a negative feedback loop in TGFβ and ERβ signaling pathways as evidenced by the potentiation of apoptosis induced by dutasteride or finasteride upon pharmacological inhibition or shRNA-mediated ablation of COX-2. Concurrently, COX-2 positively impacts ERβ action through its effect on the expression of a number of steroidogenic enzymes in the ERβ ligand metabolic pathway. Therefore, effective combination pharmacotherapies, which have included non-steroidal anti-inflammatory drugs, must take into account biochemical pathways affected by 5AR inhibition and opposing effects of COX-2 on the tissue-protective action of ERβ.
Collapse
Affiliation(s)
- Teresa T Liu
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Melanie J Grubisha
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Krystle A Frahm
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Stacy G Wendell
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Jiayan Liu
- Division of Metabolism, Endocrinology, and Diabetes, Departments of Internal Medicine and Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan 48109, and
| | - William A Ricke
- Department of Urology, University of Wisconsin, Madison, Wisconsin 53705
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, Departments of Internal Medicine and Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan 48109, and
| | - Donald B DeFranco
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260,
| |
Collapse
|
16
|
Which play a more important role in the development of large-sized prostates (≥80 ml), androgen receptors or oestrogen receptors? A comparative study. Int Urol Nephrol 2015; 48:325-33. [PMID: 26685888 DOI: 10.1007/s11255-015-1181-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/30/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To compare the expression levels of androgen receptor (AR), oestrogen receptor α (ERα)and oestrogen receptor β (ERβ) in human prostate with various degrees of benign hyperplasia. MATERIALS AND METHODS Forty-five BPH patients undergoing transurethral plasmakinetic resection of the prostate were divided into three 15-people groups of participants with small (<40 ml), medium (40-80 ml) and large (>80 ml) prostates. Patient blood samples were collected for measuring serum oestradiol (E2), total testosterone (TT), follicle-stimulating and luteinizing hormone by radioimmunoassay. Postoperative prostate specimens were collected to detect AR, ERα and ERβ expression by immunoblotting and immunohistochemical staining. RESULTS There were significantly lower serum E2 levels in the patients with large prostates compared to the patients with small prostates (P < 0.05) and medium prostates (P < 0.01). A high TT and E2 serum ratio was observed in patients with large-sized prostates (P < 0.05), but there were no obvious differences in the serum TT levels between the three groups. Prostate AR expression in patients with large- (P < 0.05) and medium-sized prostates (P < 0.01) was high compared to patients with small-sized prostates. Prostate ERα expression in patients with large prostates was low compared to patients with small- and medium-sized prostates (P < 0.01). The prostate ERβ expression did not differ between the three groups (P > 0.05). CONCLUSIONS Increased AR and decreased ERα expression in the prostate suggested that AR might be more important than ERα in stimulating prostate proliferation in patients with large-sized prostates. Patients with small- and large-sized prostates might require different management procedures involving selective oestrogen receptor modulators and selective androgen receptor modulators, respectively.
Collapse
|
17
|
Abstract
The human adult adrenal cortex is composed of the zona glomerulosa (zG), zona fasciculata (zF), and zona reticularis (zR), which are responsible for production of mineralocorticoids, glucocorticoids, and adrenal androgens, respectively. The final completion of cortical zonation in humans does not occur until puberty with the establishment of the zR and its production of adrenal androgens; a process called adrenarche. The maintenance of the adrenal cortex involves the centripetal displacement and differentiation of peripheral Sonic hedgehog-positive progenitors cells into zG cells that later transition to zF cells and subsequently zR cells.
Collapse
Affiliation(s)
- Yewei Xing
- Internal Medicine, Medical School, University of Michigan, 109 Zina Pitcher Place, 1860 BSRB, Ann Arbor, MI 48109, USA
| | - Antonio M Lerario
- Internal Medicine, Medical School, University of Michigan, 109 Zina Pitcher Place, 1860 BSRB, Ann Arbor, MI 48109, USA
| | - William Rainey
- Internal Medicine, Medical School, University of Michigan, 109 Zina Pitcher Place, 1860 BSRB, Ann Arbor, MI 48109, USA; Department of Molecular & Integrative Physiology, University of Michigan, 2560D MSRB II, 1150 W. Medical Center Dr., Ann Arbor, MI 48109-5622, USA
| | - Gary D Hammer
- Endocrine Oncology Program, Center for Organogenesis, University of Michigan, 109 Zina Pitcher Place, 1528 BSRB, Ann Arbor, MI 48109-2200, USA.
| |
Collapse
|
18
|
Labrie F. Combined blockade of testicular and locally made androgens in prostate cancer: a highly significant medical progress based upon intracrinology. J Steroid Biochem Mol Biol 2015; 145:144-56. [PMID: 24925260 DOI: 10.1016/j.jsbmb.2014.05.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/15/2014] [Accepted: 05/20/2014] [Indexed: 10/25/2022]
Abstract
Recently two drugs, namely the antiandrogen MDV-3100 and the inhibitor of 17α-hydroxylase abiraterone have been accepted by the FDA for the treatment of castration-resistant prostate cancer (CRPC) with or without previous chemotherapy, with a prolongation of overall survival of 2.2-4.8 months. While medical (GnRH agonist) or surgical castration reduces the serum levels of testosterone by about 97%, an important concentration of testosterone and dihydrotestosterone remains in the prostate and activates the androgen receptor (AR), thus offering an explanation for the positive data obtained in CRPC. In fact, explanation of the response observed with MDV-3100 or enzalutamide in CRPC is essentially a blockade of the action or formation of intraprostatic androgens. In addition to the inhibition of the action or formation of androgens made locally by the mechanisms of intracrinology, increased AR levels and AR mutations can be involved, especially in very advanced disease. Future developments look at more efficient inhibitors of the action or formation of intraprostatic androgens and starting treatment earlier when blockade of androgens can exert long-term control and even cure prostate cancer treated at a stage before the appearance of metastases. This article is part of a Special Issue entitled 'Essential role of DHEA'.
Collapse
|
19
|
Turcu A, Smith JM, Auchus R, Rainey WE. Adrenal androgens and androgen precursors-definition, synthesis, regulation and physiologic actions. Compr Physiol 2014; 4:1369-81. [PMID: 25428847 PMCID: PMC4437668 DOI: 10.1002/cphy.c140006] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The human adrenal produces more 19 carbon (C19) steroids, by mass, than either glucocorticoids or mineralocorticoids. However, the mechanisms regulating adrenal C19 steroid biosynthesis continue to represent one of the most intriguing mysteries of endocrine physiology. This review will discuss the C19 steroids synthesized by the human adrenal and the features within the adrenal that allow production of these steroids. Finally, we consider the effects of these steroids in normal physiology and disorders of adrenal C19 steroid excess.
Collapse
Affiliation(s)
- Adina Turcu
- Department of Internal Medicine, Division of Metabolism Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan; Department of Pediatrics, Division of Pediatric Endocrinology, University of Texas Southwestern Medical Center, Texas; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | | | | | | |
Collapse
|
20
|
Abstract
The discovery of medical castration with GnRH agonists in 1979 rapidly replaced surgical castration and high doses of estrogens for the treatment of prostate cancer. Soon afterwards, it was discovered that androgens were made locally in the prostate from the inactive precursor DHEA of adrenal origin, a mechanism called intracrinology. Taking into account these novel facts, combined androgen blockade (CAB) using a pure antiandrogen combined with castration in order to block the two sources of androgens was first published in 1982. CAB was the first treatment shown in randomized and placebo-controlled trials to prolong life in prostate cancer, even at the metastatic stage. Most importantly, the results recently obtained with the novel pure antiandrogen enzalutamide as well as with abiraterone, an inhibitor of 17α-hydroxylase in castration-resistant prostate cancer, has revitalized the CAB concept. The effects of CAB observed on survival of heavily pretreated patients further demonstrates the importance of the androgens made locally in the prostate and are a strong motivation to apply CAB to efficiently block all sources of androgens earlier at start of treatment and, even better, before metastasis occurs. The future of research in this field thus seems to be centered on the development of more potent blockers of androgens formation and action in order to obtain better results at the metastatic stage and, for the localized stage, reduce the duration of treatment required to achieve complete apoptosis and control of prostate cancer proliferation before it reaches the metastatic or noncurable stage.
Collapse
Affiliation(s)
- Fernand Labrie
- 2795 Laurier BoulevardSuite 500, Quebec City, Quebec, Canada G1V 4M7
| |
Collapse
|
21
|
Grubisha MJ, DeFranco DB. Local endocrine, paracrine and redox signaling networks impact estrogen and androgen crosstalk in the prostate cancer microenvironment. Steroids 2013; 78:538-41. [PMID: 23380371 PMCID: PMC3644803 DOI: 10.1016/j.steroids.2013.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Revised: 01/10/2013] [Accepted: 01/18/2013] [Indexed: 12/16/2022]
Abstract
Androgen receptor (AR) signaling is essential for the initial development and progression of prostate cancer (PCa) as well as the growth and survival of castration-resistant tumors. However, AR action may be opposed by estrogen receptor beta (ERß) that responds to androgen metabolites produced in the prostate. The balance between the activity of these two receptors is not only influenced by the steroidogenic capacity of the prostatic microenvironment but also by its redox status and local paracrine signals such as transforming growth factor-beta (TGF-ß). In this review, we highlight the studies that revealed select roles for AR and ERß in distinct compartments of the prostate cancer microenvironment. We also discuss new work that identified stromal-epithelial crosstalk through TGF-ß1 signaling that drives the production of reactive oxygen species in stromal cells thereby selectively limiting the anti-tumor activity of ERß in cancer cells. Therefore, any new therapeutic approaches that seek to limit AR but enhance ERß activity in PCa, must take into account potential adaptive changes in the tumor microenvironment that utilize paracrine signals and altered redox balance to divert local androgen metabolites towards AR at the expense of ERß.
Collapse
Affiliation(s)
- Melanie J. Grubisha
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 3051 Fifth Avenue, Pittsburgh, PA 15260 USA
| | - Donald B. DeFranco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 3051 Fifth Avenue, Pittsburgh, PA 15260 USA
- Corresponding Author: Donald B. DeFranco, tel: 412-624-4259, fax: 412-648-7029,
| |
Collapse
|
22
|
McNamara KM, Nakamura Y, Sasano H, Handelsman DJ, Simanainen U. Prostate epithelial AR inactivation leads to increased intraprostatic androgen synthesis. Prostate 2013; 73:316-27. [PMID: 22976924 DOI: 10.1002/pros.22570] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 07/10/2012] [Indexed: 11/06/2022]
Abstract
BACKGROUND Regulation of steroid synthesis within the prostate is not well understood. In this study, we examined androgen synthesis and metabolism in the mouse prostate. METHODS Using LC-MSMS steroid assays, immunohistochemistry and real-time PCR we examined the role of prostate epithelial AR in regulating 5αR expression and subsequent androgen metabolism by analyzing natural differences in epithelial AR expression between lobes as well as in the prostate epithelial AR knockout (PEARKO) mouse model. Subsequently, the role of intraprostatic androgen metabolism and epithelial AR in the generation and progression of prostate epithelial pathology was examined using long-term exogenous testosterone (T) + estradiol (E2) exposure. RESULTS Epithelial AR and 5αR2 expression as well as intraprostatic DHT followed the same lobe-specific pattern being lower in anterior than the other lobes (n = 6-8, P < 0.05). Lobe-specific 5αR2 expression was similar in PEARKO and wild-type (WT) prostate. However, PEARKO prostate had higher intraprostatic DHT content with significantly increased 5αR2 expression localized in abnormal epithelium. T + E2 treatment induced epithelial pathology was more common in PEARKO prostate compared to WT (20% vs. 2%), and was associated with increased 5αR2 expression (n = 6, P < 0.001). CONCLUSIONS We suggest that androgen synthesis via 5αR2 expression is driven by its own product (DHT) acting on adjacent stromal cells in a paracrine loop leading to increased in situ androgen levels in the PEARKO prostate. This may form part of a feed-forward loop that promotes the development of epithelial pathology.
Collapse
Affiliation(s)
- Keely M McNamara
- Andrology, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | | | | | | | | |
Collapse
|
23
|
Haag M, Hamann T, Kulle AE, Riepe FG, Blatt T, Wenck H, Holterhus PM, Peirano RI. Age and skin site related differences in steroid metabolism in male skin point to a key role of sebocytes in cutaneous hormone metabolism. DERMATO-ENDOCRINOLOGY 2012; 4:58-64. [PMID: 22870354 PMCID: PMC3408994 DOI: 10.4161/derm.19201] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hormone concentrations decline with aging. Up to now it was not clear, whether the decrease of hormone concentrations in blood samples are also present in cutaneous suction blister fluids, and whether skin from different anatomical sites shows different hormone concentrations. Analysis of suction blister fluids and paired blood samples from young (mean 27.8 y) and old (mean 62.6 y) male subjects by UPLC-MS/MS showed that DHEA concentration in blood samples was age-dependently significantly reduced, but increased in suction blister fluids, while androstenedione behaved in an opposite manner to DHEA. Testosterone decreased age-dependently in blood samples and in suction blister fluids. Regarding skin sites, DHEA was lower in samples from upper back compared with samples from the forearm. In contrast, the concentrations of androstenedione and testosterone were higher in samples from upper back. In vitro analyses showed that SZ95 sebocytes, but neither primary fibroblasts nor keratinocytes, were able to use DHEA as precursor for testosterone biosynthesis, which was confirmed by expression analysis of 3β-hydroxysteroiddehydrogenase in skin biopsies. In conclusion, we show an inverse pattern of DHEA and androstenedione concentrations in blood vs. suction blister fluids, highlighting age-dependent changes of dermal testosterone biosynthesis, and a stronger metabolism in young skin. Furthermore, sebocytes play a central role in cutaneous androgen metabolism.
Collapse
|
24
|
Mitsiades N, Sung CC, Schultz N, Danila DC, He B, Eedunuri VK, Fleisher M, Sander C, Sawyers CL, Scher HI. Distinct patterns of dysregulated expression of enzymes involved in androgen synthesis and metabolism in metastatic prostate cancer tumors. Cancer Res 2012; 72:6142-52. [PMID: 22971343 DOI: 10.1158/0008-5472.can-12-1335] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Androgen receptor (AR) signaling persists in castration-resistant prostate carcinomas (CRPC), because of several mechanisms that include increased AR expression and intratumoral androgen metabolism. We investigated the mechanisms underlying aberrant expression of transcripts involved in androgen metabolism in CRPC. We compared gene expression profiles and DNA copy number alteration (CNA) data from 29 normal prostate tissue samples, 127 primary prostate carcinomas (PCa), and 19 metastatic PCas. Steroidogenic enzyme transcripts were evaluated by quantitative reverse transcriptase PCR in PCa cell lines and circulating tumor cells (CTC) from CRPC patients. Metastatic PCas expressed higher transcript levels for AR and several steroidogenic enzymes, including SRD5A1, SRD5A3, and AKR1C3, whereas expression of SRD5A2, CYP3A4, CYP3A5, and CYP3A7 was decreased. This aberrant expression was rarely associated with CNAs. Instead, our data suggest distinct patterns of coordinated aberrant enzyme expression. Inhibition of AR activity by itself stimulated AKR1C3 expression. The aberrant expression of the steroidogenic enzyme transcripts was detected in CTCs from CRPC patients. In conclusion, our findings identify substantial interpatient heterogeneity and distinct patterns of dysregulated expression of enzymes involved in intratumoral androgen metabolism in PCa. These steroidogenic enzymes represent targets for complete suppression of systemic and intratumoral androgen levels, an objective that is supported by the clinical efficacy of the CYP17 inhibitor abiraterone. A comprehensive AR axis-targeting approach via simultaneous, frontline enzymatic blockade, and/or transcriptional repression of several steroidogenic enzymes, in combination with GnRH analogs and potent antiandrogens, would represent a powerful future strategy for PCa management.
Collapse
Affiliation(s)
- Nicholas Mitsiades
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
McNamara KM, Handelsman DJ, Simanainen U. The mouse as a model to investigate sex steroid metabolism in the normal and pathological prostate. J Steroid Biochem Mol Biol 2012; 131:107-21. [PMID: 22146616 DOI: 10.1016/j.jsbmb.2011.10.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 10/21/2011] [Accepted: 10/23/2011] [Indexed: 12/29/2022]
Abstract
Metabolism of sex steroids within the prostate is an important factor affecting its growth and pathology. Mouse models with genetic gain- and especially loss-of-function have characterised different steroid metabolic pathways and their contribution to prostate pathology. With reference to the human prostate, this review aims to summarize the steroidogenic pathways in the mouse prostate as the basis for using the mouse as a model for intraprostatic steroid signalling. In this review we summarize the current information for three main components of the steroid signalling pathway in the mouse prostate: circulating steroids, steroid receptors and steroidogenic enzymes with regard to signalling via androgen, estrogen, progesterone and glucocorticoid pathways. This review reveals many opportunities for characterisation steroid metabolism in various mouse models. The knowledge of steroid metabolism within prostate tissue and in a lobe (rodent)/region (human) specific manner, will give valuable information for future, novel hypotheses of intraprostatic control of steroid actions. This review summarizes knowledge of steroid metabolism in the mouse prostate and its relevance to the human.
Collapse
|
26
|
Abstract
Adrenarche is an endocrine developmental process whereby humans and select nonhuman primates increase adrenal output of a series of steroids, especially DHEA and DHEAS. The timing of adrenarche varies among primates, but in humans serum levels of DHEAS are seen to increase at around 6 years of age. This phenomenon corresponds with the development and expansion of the zona reticularis of the adrenal gland. The physiological phenomena that trigger the onset of adrenarche are still unknown; however, the biochemical pathways leading to this event have been elucidated in detail. There are numerous reviews examining the process of adrenarche, most of which have focused on the changes within the adrenal as well as the phenotypic results of adrenarche. This article reviews the recent and past studies that show the breadth of changes in the circulating steroid metabolome that occur during the process of adrenarche.
Collapse
Affiliation(s)
- Juilee Rege
- Department of Physiology, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | | |
Collapse
|
27
|
Adomat HH, Bains OS, Lubieniecka JM, Gleave ME, Guns ES, Grigliatti TA, Reid RE, Riggs KW. Validation of a sequential extraction and liquid chromatography-tandem mass spectrometric method for determination of dihydrotestosterone, androstanediol and androstanediol-glucuronide in prostate tissues. J Chromatogr B Analyt Technol Biomed Life Sci 2012; 902:84-95. [PMID: 22818945 DOI: 10.1016/j.jchromb.2012.06.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 06/19/2012] [Accepted: 06/24/2012] [Indexed: 11/17/2022]
Abstract
Androgens are key mediators of prostate development and function, a role that extends to the development of prostate diseases such as benign prostatic hyperplasia (BPH) and prostate cancer. In prostate, DHT is the major androgen and reduction and glucuronidation are the major metabolic pathways for DHT elimination. A streamlined method for quantitation of dihydrotestosterone (DHT), 5α-androstan-3α,17β-diol (3α-diol), and 3α-diol glucuronide (diol-gluc) was established and validated for use with archived prostate tissue specimens to facilitate examination of the roles of the underlying metabolism. This involved a sequential 70/30 hexane/ethyl acetate (hex/EtOAc) extraction of steroids, followed by an ethyl acetate extraction for diol-gluc. Derivatization of the hex/EtOAc fraction with2-fluoro-1-methylpyridinium p-toluene-4-sulfonate (FMP) was used to enhance sensitivity for hydroxyl steroids and liquid chromatography-tandem mass spectrometry (LC-MS/MS) was utilized for analysis of both fractions. The method was validated with calibration standards followed by recovery assessment from spiked samples of BPH and normal prostate. Lower limits of quantitation (LLOQ) were 50 pg/g, 20 pg/g and 100 pg/g for DHT, 3α-diol and diol-gluc, respectively for extracts from 50mg equivalents of tissue. Prepared samples were stable for up to three weeks at 4 °C and 37 °C. The method provides excellent sensitivity and selectivity for determination of tissue levels of DHT, 3α-diol, and diol-gluc. Furthermore, this protocol can easily be extended to other hydroxyl steroids, is relatively straightforward to perform and is an effective tool for assessing steroid levels in archived clinical prostate samples.
Collapse
Affiliation(s)
- Hans H Adomat
- The Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Bedoya DJ, Mitsiades N. Abiraterone acetate, a first-in-class CYP17 inhibitor, establishes a new treatment paradigm in castration-resistant prostate cancer. Expert Rev Anticancer Ther 2012; 12:1-3. [PMID: 22149426 DOI: 10.1586/era.11.196] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
29
|
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
|
30
|
El Kihel L. Oxidative metabolism of dehydroepiandrosterone (DHEA) and biologically active oxygenated metabolites of DHEA and epiandrosterone (EpiA)--recent reports. Steroids 2012; 77:10-26. [PMID: 22037250 DOI: 10.1016/j.steroids.2011.09.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 09/14/2011] [Accepted: 09/18/2011] [Indexed: 12/24/2022]
Abstract
Dehydroepiandrosterone (DHEA) is a multifunctional steroid with a broad range of biological effects in humans and animals. DHEA can be converted to multiple oxygenated metabolites in the brain and peripheral tissues. The mechanisms by which DHEA exerts its effects are not well understood. However, evidence that the effects of DHEA are mediated by its oxygenated metabolites has accumulated. This paper will review the panel of oxygenated DHEA metabolites (7, 16 and 17-hydroxylated derivatives) including a number of 5α-androstane derivatives, such as epiandrosterone (EpiA) metabolites. The most important aspects of the oxidative metabolism of DHEA in the liver, intestine and brain are described. Then, this article reviews the reported biological effects of oxygenated DHEA metabolites from recent findings with a specific focus on cancer, inflammatory and immune processes, osteoporosis, thermogenesis, adipogenesis, the cardiovascular system, the brain and the estrogen and androgen receptors.
Collapse
Affiliation(s)
- Laïla El Kihel
- Université de Caen Basse-Normandie, UFR des Sciences Pharmaceutiques, Centre d'Etudes et de Recherche sur le Médicament de Normandie, UPRES EA-4258, FR CNRS INC3M, Caen, France.
| |
Collapse
|
31
|
Simanainen U, McNamara K, Gao YR, McPherson S, Desai R, Jimenez M, Handelsman DJ. Anterior prostate epithelial AR inactivation modifies estrogen receptor expression and increases estrogen sensitivity. Am J Physiol Endocrinol Metab 2011; 301:E727-35. [PMID: 21750267 DOI: 10.1152/ajpendo.00580.2010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Androgens influence prostate growth and development, so androgen withdrawal can control progression of prostate diseases. Although estrogen treatment was originally used to induce androgen withdrawal, more recently direct estrogen effects on the prostate have been recognized, but the nature of androgen-estrogen interactions within the prostate remain poorly understood. To characterize androgen effects on estrogen sensitivity in the mouse prostate, we contrasted models of castration-induced androgen withdrawal in the prostate stromal and epithelial compartments with a prostate epithelial androgen receptor (AR) knockout (PEARKO) mouse model of selective epithelial AR inactivation. Castration markedly increased prostate epithelial estrogen receptor (ER)α immunoreactivity compared with very low ERα expression in intact males. Similarly, strong basal and luminal ERα expression was detected in PEARKO prostate of intact males, suggesting that epithelial AR activity regulated epithelial ERα expression. ERβ was strongly expressed in intact, castrated, and PEARKO prostate. However, strong clusters of epithelial ERβ positivity coincided with epithelial stratification in PEARKO prostate. In vivo estrogen sensitivity was increased in PEARKO males, with greater estradiol-induced prostate growth and epithelial proliferation leading to squamous metaplasia, featuring markedly increased epithelial proliferation, thickening, and keratinization compared with littermate controls. Our results suggest that ERα expression in the prostate epithelial cells is regulated by local, epithelia-specific, androgen-dependent mechanisms, and this imbalance in the AR- and ER-mediated signaling sensitizes the mature prostate to exogenous estrogens.
Collapse
Affiliation(s)
- Ulla Simanainen
- ANZAC Research Institute, University of Sydney, NSW, Australia
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Testosterone is the most abundant circulating androgen, and can be converted to dihydrotestosterone (DHT), a more potent androgen, by the 5α-reductase enzymes in target tissues. Current treatments for prostate cancer consist of reducing androgen levels by chemical or surgical castration or pure antiandrogen therapy that directly targets the androgen receptor (AR). Although these therapies reduce tumor burden and AR activity, the cancer inevitably recurs within 18-30 months. An approach targeting the androgen-AR axis at different levels could, therefore, improve the efficacy of prostate cancer therapy. Inhibition of 5α-reductase is one such approach; however, the two largest trials to investigate the use of the 5α-reductase inhibitors (5ARIs) finasteride and dutasteride in patients with prostate cancer have shown that, although the incidence of cancer was reduced by 5ARI treatment, those cancers that were detected were more aggressive than in patients treated with placebo. Thus, the best practice for using these drugs to prevent and treat prostate cancer remains unclear.
Collapse
|
33
|
Nicholson TM, Ricke WA. Androgens and estrogens in benign prostatic hyperplasia: past, present and future. Differentiation 2011; 82:184-99. [PMID: 21620560 DOI: 10.1016/j.diff.2011.04.006] [Citation(s) in RCA: 207] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 04/19/2011] [Accepted: 04/26/2011] [Indexed: 01/28/2023]
Abstract
Benign prostatic hyperplasia (BPH) and associated lower urinary tract symptoms (LUTS) are common clinical problems in urology. While the precise molecular etiology remains unclear, sex steroids have been implicated in the development and maintenance of BPH. Sufficient data exists linking androgens and androgen receptor pathways to BPH and use of androgen reducing compounds, such as 5α-reductase inhibitors which block the conversion of testosterone into dihydrotestosterone, are a component of the standard of care for men with LUTS attributed to an enlarged prostate. However, BPH is a multifactorial disease and not all men respond well to currently available treatments, suggesting factors other than androgens are involved. Testosterone, the primary circulating androgen in men, can also be metabolized via CYP19/aromatase into the potent estrogen, estradiol-17β. The prostate is an estrogen target tissue and estrogens directly and indirectly affect growth and differentiation of prostate. The precise role of endogenous and exogenous estrogens in directly affecting prostate growth and differentiation in the context of BPH is an understudied area. Estrogens and selective estrogen receptor modulators (SERMs) have been shown to promote or inhibit prostate proliferation signifying potential roles in BPH. Recent research has demonstrated that estrogen receptor signaling pathways may be important in the development and maintenance of BPH and LUTS; however, new models are needed to genetically dissect estrogen regulated molecular mechanisms involved in BPH. More work is needed to identify estrogens and associated signaling pathways in BPH in order to target BPH with dietary and therapeutic SERMs.
Collapse
Affiliation(s)
- Tristan M Nicholson
- University of Rochester School of Medicine & Dentistry, Rochester, NY, United States
| | | |
Collapse
|
34
|
Abstract
Estrogens and androgens have both been implicated as causes of benign prostatic hyperplasia (BPH). Although epidemiological data on an association between serum androgen concentrations and BPH are inconsistent, it is generally accepted that androgens play a permissive role in BPH pathogenesis. In clinical practice, inhibitors of 5α-reductase (which converts testosterone to the more potent androgen dihydrotestosterone) have proven effective in the management of BPH, confirming an essential role for androgens in BPH pathophysiology. To date, multiple lines of evidence support a role for estrogens in BPH pathogenesis. Studies of the two estrogen receptor (ER) subtypes have shed light on their differential functions in the human prostate; ERα and ERβ have proliferative and antiproliferative effects on prostate cells, respectively. Effects of estrogens on the prostate are associated with multiple mechanisms including apoptosis, aromatase expression and paracrine regulation via prostaglandin E2. Selective estrogen receptor modulators or other agents that can influence intraprostatic estrogen levels might conceivably be potential therapeutic targets for the treatment of BPH.
Collapse
|
35
|
Cabeza M, Trejo KV, González C, García P, Soriano J, Heuze Y, Bratoeff E. Steroidal 5α-reductase inhibitors using 4-androstenedione as substrate. J Enzyme Inhib Med Chem 2011; 26:712-9. [DOI: 10.3109/14756366.2010.548330] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Marisa Cabeza
- Department of Biological Systems and Animal Production, Metropolitan University-Xochimilco, Mexico D. F., Mexico
| | - Karla Valeria Trejo
- Department of Biological Systems and Animal Production, Metropolitan University-Xochimilco, Mexico D. F., Mexico
| | - Claudia González
- Department of Biological Systems and Animal Production, Metropolitan University-Xochimilco, Mexico D. F., Mexico
| | - Perla García
- Department of Pharmacy, Faculty of Chemistry, National University of Mexico City, Mexico D. F., Mexico
| | - Juan Soriano
- Department of Pathology of the General Hospital of Mexico, Mexico D. F
| | - Yvonne Heuze
- Department of Biological Systems and Animal Production, Metropolitan University-Xochimilco, Mexico D. F., Mexico
| | - Eugene Bratoeff
- Department of Pharmacy, Faculty of Chemistry, National University of Mexico City, Mexico D. F., Mexico
| |
Collapse
|
36
|
Arai S, Miyashiro Y, Shibata Y, Tomaru Y, Kobayashi M, Honma S, Suzuki K. Effect of castration monotherapy on the levels of adrenal androgens in cancerous prostatic tissues. Steroids 2011; 76:301-8. [PMID: 21147140 DOI: 10.1016/j.steroids.2010.12.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 10/30/2010] [Accepted: 12/03/2010] [Indexed: 12/01/2022]
Abstract
The mechanism accounting for the development of castration-resistant prostate cancer (CRPC) remains unclear. Studies in CRPC tissues suggest that, after androgen deprivation therapy (ADT), the adrenal androgens may be an important source of testosterone (T) and 5-alpha dihydrotestosterone (DHT) in CRPC tissues. To clarify the role of adrenal androgens in the prostatic tissues (prostatic tissue adrenal androgens) during ADT, we developed a high sensitive and specific quantification method for the levels of androgens in prostatic tissue using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Human prostatic tissues were purified using mixed-mode reversed-phase, strong anion exchange Oasis cartridges (Oasis MAX). Analysis of steroids was performed using LC-MS/MS after picolinic acid derivatization. The validation tests showed that our method of quantitative analysis was precise and sensitive enough for the quantification of dehydroepiandrosterone (DHEA), androstenedione, androstenediol, T, and DHT in the prostatic tissue. The levels of adrenal androgens in prostate cancer tissues after ADT were similar to those in untreated PCa. Especially, DHEA was the most existing androgen precursor in PCa tissues after ADT. The levels of DHEA were high in PCa tissues, irrespective of ADT. We assumed that DHEA played a significant role in the synthesis of T and DHT in PCa tissues after ADT.
Collapse
Affiliation(s)
- Seiji Arai
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Prostate cancer is the most hormone sensitive of all cancers. However, any hormonal therapeutic strategy must take into account the fact that two almost equivalent sources of androgens act in the prostate, namely testosterone of testicular origin, and the locally produced androgens testosterone and dihydrotestosterone (DHT) derived from dehydroepiandrosterone of adrenal origin. Combined androgen blockade--medical or surgical castration plus a pure antiandrogen--would, therefore, be the logical first-line treatment for prostate cancer, although castration or an antiandrogen alone is still chosen in the majority of cases. Although long-term control, or even cure, is possible when combined androgen blockade is used when the tumor is localized, resistance to treatment invariably develops in patients when start of treatment is delayed until the disease has become metastatic. This observation can be explained either by elevated levels of the androgen receptor, which can increase the response to low levels of androgens and also modify the response to antiandrogens; or by local biosynthesis of androgens. Research to identify new and more potent antiandrogens, as well as blockers of peripheral and adrenal androgen biosynthesis--such as abiraterone--could be of great importance.
Collapse
Affiliation(s)
- Fernand Labrie
- Research Center in Molecular Endocrinology, Oncology and Human Genomics, Laval University, Laurier Boulevard, Quebec City, QC G1V 4G2, Canada.
| |
Collapse
|
38
|
McNamara KM, Harwood DT, Simanainen U, Walters KA, Jimenez M, Handelsman DJ. Measurement of sex steroids in murine blood and reproductive tissues by liquid chromatography-tandem mass spectrometry. J Steroid Biochem Mol Biol 2010; 121:611-8. [PMID: 20144714 DOI: 10.1016/j.jsbmb.2010.02.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 01/22/2010] [Accepted: 02/02/2010] [Indexed: 11/15/2022]
Abstract
Accurate measurement of sex steroids is essential to evaluate mouse models for human reproductive development and disorders. The recent advent of liquid chromatography-tandem mass spectrometry (LC-MS/MS) assays that match the sensitivity of steroid immunoassay could overcome problems arising from the limited specificity of steroid immunoassay. In this current study we validate a LC-MS/MS assay for the measurement of key sex steroids from murine serum and reproductive tissues. The assay gave excellent dilutional linearity (r(2)> or =0.98) and reproducibility (CV< or =10% of replicate samples) in serum and reproductive tissues with sensitive quantitation limits; testosterone (T; 2pg), dihydrotestosterone (DHT; 10pg), 5alpha-androstane-3alpha,17beta-diol (3alphaDiol; 40pg), 5alpha-androstane-3beta,17beta-diol (3betaDiol; 40pg), estradiol (E2; 0.5pg) and estrone (E1; 0.3pg). Using 0.1mL sample, T was the only consistently detectable steroid (detection limit 20pg/ml) in both male and female mouse serum. In the testis, T and DHT were quantifiable as were both diols at relatively high levels. Prostatic T levels were low and DHT was determined to be the most abundant androgen in this tissue. Uterine and ovarian levels of E2, E1 and T were measurable, with levels varying according to estrous cycle stage. Hence, we demonstrate that this LC-MS/MS method has the sensitivity, specificity and multi-analyte capability to offer accurate steroid profiling in mouse serum and reproductive tissues.
Collapse
Affiliation(s)
- K M McNamara
- Andrology, ANZAC Research Institute, University of Sydney, Sydney, NSW 2139, Australia.
| | | | | | | | | | | |
Collapse
|
39
|
Corradi LS, Góes RM, Vilamaior PSL, Taboga SR. Increased androgen receptor and remodeling in the prostatic stroma after the inhibition of 5-alpha reductase and aromatase in gerbil ventral prostate. Microsc Res Tech 2010; 72:939-50. [PMID: 19484778 DOI: 10.1002/jemt.20740] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Prostate require high levels of steroidogenic enzymes such as 5alpha-reductase (5alpha-r) and Aromatase (Aro) for the formation of active steroids. Dihydrotestosterone (DHT), the prostate dominant androgen, is converted from testosterone (T) by the action of 5alpha-r. Aro provides an alternative pathway for estrogen, via T aromatization. Since prostatic maintenance is dependent on both reciprocal stromal-epithelial interaction and regulation by steroids, this study aimed to elucidate what the absence of 5alpha-r and Aro enzymes provokes in the prostate microenvironment after their long-term inhibition. Data obtained 1 day after the 30 consecutive days of enzymatic inhibition with Finasteride (5alpha-r inhibitor) and Letrozole (Aro inhibitor) demonstrated a marked stromal remodeling, with an increased deposition of extracellular matrix (ECM) proteins besides androgen receptor (AR) overexpression in the three phases of postnatal development analyzed. The subepithelial area of acini from ventral prostate presented collagen and reticular fibers accumulation, besides various altered and active fibroblasts. The AR content immunostaining was elevated after enzymatic inhibition therapy, mainly in the nuclei of epithelial cells. Similar data were observed in the ventral prostates even 21 days after the end of treatments. Results obtained following the long-term inhibition of 5alpha-r and Aro are relevant and highlight the actions of these enzymes as crucial not only for the maintenance of tissue architecture and ECM arrangement but also for androgen and AR function. The long-term absence of their action imposes a novel situation on the prostate from which its normal physiology could not be restored by the conclusion of the treatments.
Collapse
Affiliation(s)
- Lara S Corradi
- Campinas State University-UNICAMP, Department of Cell Biology-Institute of Biology, Campinas, São Paulo, CEP 15054-000, Brazil
| | | | | | | |
Collapse
|
40
|
Abstract
Of all cancers, prostate cancer is the most sensitive to hormones: it is thus very important to take advantage of this unique property and to always use optimal androgen blockade when hormone therapy is the appropriate treatment. A fundamental observation is that the serum testosterone concentration only reflects the amount of testosterone of testicular origin which is released in the blood from which it reaches all tissues. Recent data show, however, that an approximately equal amount of testosterone is made from dehydroepiandrosterone (DHEA) directly in the peripheral tissues, including the prostate, and does not appear in the blood. Consequently, after castration, the 95-97% fall in serum testosterone does not reflect the 40-50% testosterone (testo) and dihydrotestosterone (DHT) made locally in the prostate from DHEA of adrenal origin. In fact, while elimination of testicular androgens by castration alone has never been shown to prolong life in metastatic prostate cancer, combination of castration (surgical or medical with a gonadotropin-releasing hormone (GnRH) agonist) with a pure anti-androgen has been the first treatment shown to prolong life. Most importantly, when applied at the localized stage, the same combined androgen blockade (CAB) can provide long-term control or cure of the disease in more than 90% of cases. Obviously, since prostate cancer usually grows and metastasizes without signs or symptoms, screening with prostate-specific antigen (PSA) is absolutely needed to diagnose prostate cancer at an 'early' stage before metastasis occurs and the cancer becomes non-curable. While the role of androgens was believed to have become non-significant in cancer progressing under any form of androgen blockade, recent data have shown increased expression of the androgen receptor (AR) in treatment-resistant disease with a benefit of further androgen blockade. Since the available anti-androgens have low affinity for AR and cannot block androgen action completely, especially in the presence of increased AR levels, it becomes important to discover more potent and purely antagonistic blockers of AR. The data obtained with compounds under development are promising. While waiting for this (these) new anti-androgen(s), combined treatment with castration and a pure anti-androgen (bicalutamide, flutamide or nilutamide) is the only available and the best scientifically based means of treating prostate cancer by hormone therapy at any stage of the disease with the optimal chance of success and even cure in localized disease.
Collapse
Affiliation(s)
- Fernand Labrie
- Research Center in Molecular Endocrinology, Oncology and Human Genomics, Laval University and Laval University Hospital Research Center (CRCHUL), Quebec, Canada.
| |
Collapse
|
41
|
Alexaki VI, Charalampopoulos I, Panayotopoulou M, Kampa M, Gravanis A, Castanas E. Dehydroepiandrosterone protects human keratinocytes against apoptosis through membrane binding sites. Exp Cell Res 2009; 315:2275-83. [DOI: 10.1016/j.yexcr.2009.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Revised: 04/07/2009] [Accepted: 04/11/2009] [Indexed: 01/28/2023]
|
42
|
Vajda EG, López FJ, Rix P, Hill R, Chen Y, Lee KJ, O'Brien Z, Chang WY, Meglasson MD, Lee YH. Pharmacokinetics and pharmacodynamics of LGD-3303 [9-chloro-2-ethyl-1-methyl-3-(2,2,2-trifluoroethyl)-3H-pyrrolo-[3,2-f]quinolin-7(6H)-one], an orally available nonsteroidal-selective androgen receptor modulator. J Pharmacol Exp Ther 2008; 328:663-70. [PMID: 19017848 DOI: 10.1124/jpet.108.146811] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Selective androgen receptor modulators (SARMs) are a new class of molecules in development to treat a variety of diseases. SARMs maintain the beneficial effects of androgens, including increased muscle mass and bone density, while having reduced activity on unwanted side effects. The mechanisms responsible for the tissue-selective activity of SARMs are not fully understood, and the pharmacokinetic (PK)/pharmacodynamic (PD) relationships are poorly described. Tissue-specific compound distribution potentially could be a mechanism responsible for apparent tissue selectivity. We examined the PK/PD relationship of a novel SARM, LGD-3303 [9-chloro-2-ethyl-1-methyl-3-(2,2,2-trifluoroethyl)-3H-pyrrolo[3,2-f]quinolin-7(6H)-one], in a castrated rat model of androgen deficiency. LGD-3303 has potent activity on levator ani muscle but is a partial agonist on the preputial gland and ventral prostate. LGD-3303 never stimulated ventral prostate above intact levels despite increasing plasma concentrations of compound. Tissue-selective activity was maintained when LGD-3303 was dosed orally or by continuous infusion, two routes of administration with markedly different time versus exposure profiles. Despite the greater muscle activity relative to prostate activity, local tissue concentrations of LGD-3303 were higher in the prostate than in the levator ani muscle. LGD-3303 has SARM properties that are independent of its pharmacokinetic profile, suggesting that the principle mechanism for tissue-selective activity is the result of altered molecular interactions at the level of the androgen receptor.
Collapse
Affiliation(s)
- Eric G Vajda
- Discovery Research, Ligand Pharmaceuticals, Inc., San Diego, California, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|