1
|
Ghosh D. Structures and Functions of Human Placental Aromatase and Steroid Sulfatase, Two Key Enzymes in Estrogen Biosynthesis. Steroids 2023; 196:109249. [PMID: 37207843 DOI: 10.1016/j.steroids.2023.109249] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/21/2023]
Abstract
Cytochrome P450 aromatase (AROM) and steroid sulfatase (STS) are the two key enzymes for the biosynthesis of estrogens in human, and maintenance of the critical balance between androgens and estrogens. Human AROM, an integral membrane protein of the endoplasmic reticulum, is a member of the cytochrome P450 superfamily. It is the only enzyme to catalyze the conversion of androgens with non-aromatic A-rings to estrogens characterized by the aromatic A-ring. Human STS, also an integral membrane protein of the endoplasmic reticulum, is a Ca2+-dependent enzyme that catalyzes the hydrolysis of sulfate esters of estrone and dehydroepiandrosterone to the unconjugated steroids, the precursors of the most potent forms of estrogens and androgens, namely, 17β-estradiol, 16α,17β-estriol, testosterone and dihydrotestosterone. Expression of these steroidogenic enzymes locally within organs and tissues of the endocrine, reproductive, and central nervous systems is the key for maintaining high levels of the reproductive steroids. The enzymes have been drug targets for the prevention and treatment of diseases associated with steroid hormone excesses, especially in breast, endometrial and prostate malignancies. Both enzymes have been the subjects of vigorous research for the past six decades. In this article, we review the important findings on their structure-function relationships, specifically, the work that began with unravelling of the closely guarded secrets, namely, the 3-D structures, active sites, mechanisms of action, origins of substrate specificity and the basis of membrane integration. Remarkably, these studies were conducted on the enzymes purified in their pristine forms from human placenta, the discarded and their most abundant source. The purification, assay, crystallization, and structure determination methodologies are described. Also reviewed are their functional quaternary organizations, post-translational modifications and the advancements made in the structure-guided inhibitor design efforts. Outstanding questions that still remain open are summarized in closing.
Collapse
Affiliation(s)
- Debashis Ghosh
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210.
| |
Collapse
|
2
|
Ghosh D. Structure of human placental steroid sulfatase at 2.0 angstrom resolution: Catalysis, quaternary association, and a secondary ligand site. J Steroid Biochem Mol Biol 2023; 227:106228. [PMID: 36427797 DOI: 10.1016/j.jsbmb.2022.106228] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022]
Abstract
Human placental estrone (E1)/dehydroepiandrosterone (DHEA) sulfatase (human placental steroid sulfatase; hSTS) is an integral membrane protein of the endoplasmic reticulum. This Ca2+-dependent enzyme catalyzes the hydrolysis of sulfate esters of E1 and DHEA to yield the respective unconjugated steroids, which then act as precursors for the biosynthesis of 17β-estradiol (E2) and dihydrotestosterone (DHT), respectively, the most potent forms of estrogens and androgens. hSTS is a key enzyme for the local production of E2 and DHT in the breast and the prostate. The enzyme is known to be responsible for maintaining high levels of estrogens in the breast tumor cells. The crystal structure of hSTS purified from human placenta has previously been reported at 2.6 Å resolution. Here we present the structure of hSTS determined at the superior 2.0 Å resolution bringing new clarity to the atomic architecture of the active site. The molecular basis of catalysis and steroid-protein interaction are revisited in light of the new data. We also reexamine the enzyme's quaternary association and its implication on the membrane integration. A secondary ligand binding pocket at the intermolecular interface and adjacent to the active site access channel, buried into the gill of the mushroom-shaped molecule, has been identified. Its role as well as that of a phosphate ion bound to an exposed histidine side chain are examined from the structure-function perspective. Higher resolution data also aids in the tracing of an important loop missing in the previous structure.
Collapse
Affiliation(s)
- Debashis Ghosh
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
3
|
Karakus E, Schmid A, Leiting S, Fühler B, Schäffler A, Jakob T, Geyer J. Role of the Steroid Sulfate Uptake Transporter Soat (Slc10a6) in Adipose Tissue and 3T3-L1 Adipocytes. Front Mol Biosci 2022; 9:863912. [PMID: 35573729 PMCID: PMC9095825 DOI: 10.3389/fmolb.2022.863912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/24/2022] [Indexed: 11/22/2022] Open
Abstract
In addition to the endocrine and paracrine systems, peripheral tissues such as gonads, skin, and adipose tissue are involved in the intracrine mechanisms responsible for the formation of sex steroids via the transformation of dehydroepiandrosterone and dehydroepiandrosterone sulfate (DHEA/DHEAS) into potent androgenic and estrogenic hormones. Numerous studies have examined the relationship between overweight, central obesity, and plasma levels of DHEA and DHEAS. The sodium-dependent organic anion transporter Soat (Slc10a6) is a plasma membrane uptake transporter for sulfated steroids. Significantly increased expression of Slc10a6 mRNA has been previously described in organs and tissues of lipopolysaccharide (LPS)-treated mice, including white adipose tissue. These findings suggest that Soat plays a role in the supply of steroids in peripheral target tissues. The present study aimed to investigate the expression of Soat in adipocytes and its role in adipogenesis. Soat expression was analyzed in mouse white intra-abdominal (WAT), subcutaneous (SAT), and brown (BAT) adipose tissue samples and in murine 3T3-L1 adipocytes. In addition, adipose tissue mass and size of the adipocytes were analyzed in wild-type and Slc10a6−/− knockout mice. Soat expression was detected in mouse WAT, SAT, and BAT using immunofluorescence. The expression of Slc10a6 mRNA was significantly higher in 3T3-L1 adipocytes than that of preadipocytes and was significantly upregulated by exposure to lipopolysaccharide (LPS). Slc10a6 mRNA levels were also upregulated in the adipose tissue of LPS-treated mice. In Slc10a6−/− knockout mice, adipocytes increased in size in the WAT and SAT of female mice and in the BAT of male mice, suggesting adipocyte hypertrophy. The serum levels of adiponectin, resistin, and leptin were comparable in wild-type and Slc10a6−/− knockout mice. The treatment of 3T3-L1 adipocytes with DHEA significantly reduced lipid accumulation, while DHEAS did not have a significant effect. However, following LPS-induced Soat upregulation, DHEAS also significantly inhibited lipid accumulation in adipocytes. In conclusion, Soat-mediated import of DHEAS and other sulfated steroids could contribute to the complex pathways of sex steroid intracrinology in adipose tissues. Although in cell cultures the Soat-mediated uptake of DHEAS appears to reduce lipid accumulation, in Slc10a6−/− knockout mice, the Soat deletion induced adipocyte hyperplasia through hitherto unknown mechanisms.
Collapse
Affiliation(s)
- Emre Karakus
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Germany
| | - Andreas Schmid
- Department of Internal Medicine III, Giessen University Hospital, Justus Liebig University, Giessen, Germany
| | - Silke Leiting
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Germany
| | - Bärbel Fühler
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Germany
| | - Andreas Schäffler
- Department of Internal Medicine III, Giessen University Hospital, Justus Liebig University, Giessen, Germany
| | - Thilo Jakob
- Department of Dermatology and Allergology, Giessen University Hospital, Justus Liebig University, Giessen, Germany
| | - Joachim Geyer
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Germany
- *Correspondence: Joachim Geyer,
| |
Collapse
|
4
|
Chatterton RT. Functions of dehydroepiandrosterone in relation to breast cancer. Steroids 2022; 179:108970. [PMID: 35122788 DOI: 10.1016/j.steroids.2022.108970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/17/2022] [Accepted: 01/31/2022] [Indexed: 11/20/2022]
Abstract
Although DHEA sulfate (DS) is the most abundant steroid in the circulation, breast fluid contains an approximately 80-fold greater concentration than serum. Transport of DS into cells requires organic anion transporting polypeptides (OATPs), which are specific for cell type, cell location, and substrate, but may have a broader specificity for housekeeping functions. Specific classes, which may be modified by soluble factors including neutral steroids, have been identified in the breast. After transport, DS may be cleaved to DHEA by ubiquitous sulfatases, which may be modified by the cell milieu, or DHEA may enter by diffusion. Synthesis from cholesterol does not occur because CYP17B12 and cytochrome b5 are lacking in breast tissues. Case-control studies reveal a positive association of serum DS with risk of breast cancer. The association is even greater with DHEA, particularly in postmenopausal women with HR + invasive tumors. Metabolites of DHEA, androstenedione and testosterone, are associated with breast cancer but DHEA is likely to have an independent role as well. Mechanisms by which DHEA may promote breast cancer relate to its effect in increasing circulating IGF-I, by inhibiting the suppressive effect of glucocorticoids, and by promoting retention of pre-adipocytes with aromatase activity. In addition, DHEA may interact with the G-protein coupled receptor GPER for stimulation of miR-21 and subsequent activation of the MAPK pathway. DHEA also has antitumor properties that relate to stimulation of immunity, suppression of inflammation, and elevation of adipose tissue adiponectin synthesis. The net effect may depend on the which factors predominate.
Collapse
Affiliation(s)
- Robert T Chatterton
- Department of Obstetrics and Gynecology and the Robert H Lurie Comprehensive Cancer Center of Northwestern, Northwestern University Feinberg Medical School. Chicago, IL 60911, USA.
| |
Collapse
|
5
|
Häussler S, Sadri H, Ghaffari MH, Sauerwein H. Symposium review: Adipose tissue endocrinology in the periparturient period of dairy cows. J Dairy Sci 2022; 105:3648-3669. [PMID: 35181138 DOI: 10.3168/jds.2021-21220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/05/2022] [Indexed: 12/17/2022]
Abstract
The involvement of adipose tissue (AT) in metabolism is not limited to energy storage but turned out to be much more complex. We now know that in addition to lipid metabolism, AT is important in glucose homeostasis and AA metabolism and also has a role in inflammatory processes. With the discovery of leptin in 1994, the concept of AT being able to secrete messenger molecules collectively termed as adipokines, and acting in an endo-, para-, and autocrine manner emerged. Moreover, based on its asset of receptors, many stimuli from other tissues reaching AT via the bloodstream can also elicit distinct responses and thus integrate AT as a control element in the regulatory circuits of the whole body's functions. The protein secretome of human differentiated adipocytes was described to comprise more than 400 different proteins. However, in dairy cows, the characterization of the physiological time course of adipokines in AT during the transition from pregnancy to lactation is largely limited to the mRNA level; for the protein level, the analytical methods are limited and available assays often lack sound validation. In addition to proteinaceous adipokines, small compounds such as steroids can also be secreted from AT. Due to the lipophilic nature of steroids, they are stored in AT, but during the past years, AT became also known as being able to metabolize and even to generate steroid hormones de novo. In high-yielding dairy cows, AT is substantially mobilized due to increased energy requirements related to lactation. As to whether the steroidogenic system in AT is affected and may change during the common loss of body fat is largely unknown. Moreover, most research about AT in transition dairy cows is based on subcutaneous AT, whereas other depots have scarcely been investigated. This contribution aims to review the changes in adipokine mRNA and-where available-protein expression with time relative to calving in high-yielding dairy cows at different conditions, including parity, body condition, diet, specific feed supplements, and health disorders. In addition, the review provides insights into steroidogenic pathways in dairy cows AT, and addresses differences between fat depots where possible.
Collapse
Affiliation(s)
- Susanne Häussler
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - Hassan Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 516616471 Tabriz, Iran
| | - Morteza H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
6
|
Bone C, Squires EJ. The Uptake and Deconjugation of Androstenone Sulfate in the Adipose Tissue of the Boar. Animals (Basel) 2021; 11:ani11113158. [PMID: 34827890 PMCID: PMC8614246 DOI: 10.3390/ani11113158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022] Open
Abstract
Boars express high testicular levels of sulfotransferase enzymes, and consequently, the boar taint causing compound androstenone predominantly circulates as a steroid sulfate. Androstenone sulfate is suspected to function as a steroid reservoir that can be deconjugated to provide a source of free androstenone for accumulation. Therefore, the purpose of this study was to characterize the uptake and deconjugation of androstenone sulfate in the adipose tissue of the boar. Real-time PCR was used to quantify the expression of steroid sulfatase (STS) and several organic anion transporting polypeptides (OATPs) in the adipose tissue. Additionally, [3H]-androstenone sulfate was incubated with adipocytes or supernatant from homogenized fat to assess steroid uptake and conversion, respectively. A positive correlation existed between OATP-B expression and androstenone sulfate uptake (r = 0.86, p = 0.03), as well as between STS expression and androstenone sulfate conversion (r = 0.76, p < 0.001). Moreover, fat androstenone concentrations were positively correlated (r = 0.85, p < 0.001) with androstenone sulfate conversion and tended to increase with STS expression in early maturing boars. This suggests that androstenone sulfate uptake and deconjugation are mediated by OATP-B and STS, respectively, which may influence the development of boar taint in early maturing animals.
Collapse
|
7
|
Emami N, Moini A, Yaghmaei P, Akbarinejad V, Shahhoseini M, Alizadeh A. Differences in expression of genes related to steroidgenesis in abdominal subcutaneous adipose tissue of pregnant women with and without PCOS; a case control study. BMC Pregnancy Childbirth 2021; 21:490. [PMID: 34233642 PMCID: PMC8261994 DOI: 10.1186/s12884-021-03957-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND It was reported that steroid-related gene expressions in the adipose tissue (AT) of women differ between women affected with polycystic ovary syndrome (PCOS) and non-PCOS. Although association between PCOS in mother and offspring's health is a crucial issue, there are few studies focusing on AT of pregnant women suffering from PCOS. Our objectives were to determine the differences between mRNA expression levels of key steroid-converting enzymes in abdominal subcutaneous AT of pregnant women afflicted with PCOS and non-PCOS. METHODS Twelve pregnant women with PCOS (case) and thirty six non-PCOS pregnant women (control) (1:3 ratio; age- and BMI-matched) undergoing cesarean section were enrolled for the present study. Expressions of fifteen genes related to steriodogenesis in abdominal subcutaneous AT were investigated using quantitative real-time PCR. RESULTS No significant differences were detected with respect to age, BMI (prior pregnancy and at delivery day), gestational period and parity among pregnant women with PCOS and non-PCOS. Most of the sex steroid-converting genes except 17β-Hydroxysteroid dehydrogenases2 (17BHSD2), were highly expressed on the day of delivery in subcutaneous AT. Women with PCOS showed significantly higher mRNA levels of steroidgenic acute regulator (STAR; P < 0.001), cytochrome P450 monooxygenase (CYP11A1; P < 0.05), 17α-hydroxylase (CYP17A1; P < 0.05), and 11β-Hydroxysteroid dehydrogenase (11BHSD1 and 11BHSD2; P < 0.05). The expression of steroid 21-hydroxylase (CYP21) in non-PCOS was fourfold higher than those of women with PCOS (P < 0.001). There were no significant differences between relative expression of aromatase cytochrome P450 (CYP19A1), 3β-hydroxysteroid dehydrogenase (3BHSD1 and 3BHSD2), and 17BHSD family (1, 3, 5, 7, and 12) between the two groups. CONCLUSION The expression levels of genes related to sex steroids metabolism were similar to age-matched and BMI- matched pregnant non-PCOS and pregnant women with PCOS at delivery day. However, the alterations in gene expressions involved in glucocorticoids and mineralocorticoids metabolism were shown. It is necessary to point out that further studies regarding functional activity are required. More attention should be given to AT of pregnant women with PCOS that was previously ignored.
Collapse
Affiliation(s)
- Neda Emami
- Department of Biology, Faculty of Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ashraf Moini
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,Breast Disease Research Center (BDRC), Tehran University of Medical Sciences, Tehran, Iran.,Department of Gynecology and Obstetrics, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehreh Yaghmaei
- Department of Biology, Faculty of Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Maryam Shahhoseini
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran. .,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran. .,Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - AliReza Alizadeh
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
8
|
Hetemäki N, Mikkola TS, Tikkanen MJ, Wang F, Hämäläinen E, Turpeinen U, Haanpää M, Vihma V, Savolainen-Peltonen H. Adipose tissue estrogen production and metabolism in premenopausal women. J Steroid Biochem Mol Biol 2021; 209:105849. [PMID: 33610799 DOI: 10.1016/j.jsbmb.2021.105849] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/03/2021] [Accepted: 02/07/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Although the ovaries produce the majority of estrogens in women before menopause, estrogen is also synthesized in peripheral tissues such as adipose tissue (AT). The typical female AT distribution, concentrated in subcutaneous and femoro-gluteal regions, is estrogen-mediated, but the significance of estrogen synthesis in AT of premenopausal women is poorly understood. DESIGN AND METHODS Serum and subcutaneous and visceral AT homogenates from 28 premenopausal women undergoing non-malignant surgery were analyzed for estrone, estradiol, and serum estrone sulfate (E1S) concentrations with liquid chromatography-tandem mass spectrometry. Isotopic precursors were used to measure enzyme activities of estrone-producing steroid sulfatase and estradiol-producing 17β-hydroxysteroid dehydrogenases (17β-HSD). Messenger RNA (mRNA) expression levels of genes for estrogen-metabolizing enzymes were analyzed using real-time reverse transcription quantitative polymerase chain reaction. RESULTS While estradiol was the predominant circulating active estrogen, estrone dominated in AT, with a higher concentration in visceral than subcutaneous AT (median, 2657 vs 1459 pmol/kg; P = 0.002). Both AT depots converted circulating E1S to estrone, and estrone to estradiol. Median levels of estrone were five to ten times higher in subcutaneous and visceral AT than in serum (P < 0.001) and the estradiol level in visceral AT was 1.3 times higher than in serum (P < 0.005). The local estrone concentration in visceral AT correlated positively with mRNA expression of estrone-producing enzyme aromatase (r = 0.65, P = 0.003). Waist circumference correlated positively with increased estradiol production in subcutaneous AT (r = 0.60, P = 0.039). CONCLUSIONS Premenopausal AT demonstrated high estrogenic enzyme activity and considerable local estrogen concentrations. This may be a factor promoting female-typical AT distribution in premenopausal women.
Collapse
Affiliation(s)
- Natalia Hetemäki
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland; Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Tomi S Mikkola
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland; Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Matti J Tikkanen
- Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland; Heart and Lung Center, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Feng Wang
- Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Esa Hämäläinen
- Department of Clinical Chemistry, University of Helsinki, FIN-00029 HUS, Helsinki, Finland
| | - Ursula Turpeinen
- HUSLAB, Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Mikko Haanpää
- HUSLAB, Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Veera Vihma
- Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland; Department of General Practice and Primary Health Care, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Hanna Savolainen-Peltonen
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland; Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland.
| |
Collapse
|
9
|
Lightning TA, Gesteira TF, Mueller JW. Steroid disulfates - Sulfation double trouble. Mol Cell Endocrinol 2021; 524:111161. [PMID: 33453296 DOI: 10.1016/j.mce.2021.111161] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/24/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
Sulfation pathways have recently come into the focus of biomedical research. For steroid hormones and related compounds, sulfation represents an additional layer of regulation as sulfated steroids are more water-soluble and tend to be biologically less active. For steroid diols, an additional sulfation is possible, carried out by the same sulfotransferases that catalyze the first sulfation step. The steroid disulfates that are formed are the focus of this review. We discuss both their biochemical production as well as their putative biological function. Steroid disulfates have also been linked to various clinical conditions in numerous untargeted metabolomics studies. New analytical techniques exploring the biosynthetic routes of steroid disulfates have led to novel insights, changing our understanding of sulfation in human biology. They promise a bright future for research into sulfation pathways, hopefully too for the diagnosis and treatment of several associated diseases.
Collapse
Affiliation(s)
- Thomas Alec Lightning
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Tarsis F Gesteira
- College of Optometry, University of Houston, Houston, TX, USA; Optimvia, LLC, Batavia, OH, USA
| | - Jonathan Wolf Mueller
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.
| |
Collapse
|
10
|
Pork Production with Entire Males: Directions for Control of Boar Taint. Animals (Basel) 2020; 10:ani10091665. [PMID: 32947846 PMCID: PMC7552340 DOI: 10.3390/ani10091665] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Castration of male piglets has traditionally been carried out to control boar taint, but animal welfare concerns about surgical castration has brought this practice under scrutiny. In addition, castration decreases growth performance and increases the environmental impact of pork production, so alternatives to castration are needed to control boar taint. In this review, we summarize the current knowledge on boar taint metabolism and outline some key areas that require further study. We also describe some opportunities for controlling the boar taint problem and propose that by defining the differences in metabolic processes and the genetic variations that can lead to boar taint in individual pigs, we can design effective custom solutions for boar taint. Abstract Boar taint is caused by the accumulation of androstenone and skatole and other indoles in the fat; this is regulated by the balance between synthesis and degradation of these compounds and can be affected by a number of factors, including environment and management practices, sexual maturity, nutrition, and genetics. Boar taint can be controlled by immunocastration, but this practice has not been accepted in some countries. Genetics offers a long-term solution to the boar taint problem via selective breeding or genome editing. A number of short-term strategies to control boar taint have been proposed, but these can have inconsistent effects and there is too much variability between breeds and individuals to implement a blanket solution for boar taint. Therefore, we propose a precision livestock management approach to developing solutions for controlling taint. This involves determining the differences in metabolic processes and the genetic variations that cause boar taint in specific groups of pigs and using this information to design custom treatments based on the cause of boar taint. Genetic, proteomic or metabolomic profiling can then be used to identify and implement effective solutions for boar taint for specific populations of animals.
Collapse
|
11
|
Bulant J, Hill M, Velíková M, Yamamotová A, Martásek P, Papežová H. Changes of BMI, steroid metabolome and psychopathology in patients with anorexia nervosa during hospitalization. Steroids 2020; 153:108523. [PMID: 31622616 DOI: 10.1016/j.steroids.2019.108523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/10/2019] [Accepted: 10/05/2019] [Indexed: 12/19/2022]
Abstract
Anorexia nervosa (AN) is associated with various alterations including the dysfunction of the HPA axis and consequently the hypercortisolemia and deficit in sex hormones but the comprehensive evaluation of changes in circulating steroids during the hospitalization of AN patients is lacking. We investigated the effect of realimentation of women with AN during hospitalization on 45 circulating steroids, the relationships between BMI, its change during hospitalization and physical activity, on one side and initial levels and their changes for two adipokines, circulating steroids, anorexia-specific (hunger, appetite and satiety), and anorexia non-specific symptoms (anxiety, depression fatigue, sleep, and body pain) on the other side. We included 33 women with anorexia who were hospitalized for 38(35, 44) days (median with quartiles). The increase of BMI from the initial value 15.2 (13.2, 16.6) kg/m2 was 1.69 (1.37, 2.66) kg/m2. The patients with more severe anorexia showed higher activity in 7β-, and 16α-hydroxylation of androgen precursors, which declined during hospitalization. Otherwise, the 7α-hydroxylation activity is higher in AN patients with less severe malnutrition and the ratio of 5-androstene-3β,7α,17β-triol to 5-androstene-3β,7β,17β-triol increased during the realimentation. Our data allow to speculate that the intensive 7β-, and 16α- and possibly also the 7α-hydroxylation of C19 Δ5 steroids participate in the pathophysiology of anorexia by additional catabolism of substrates available for synthesis of active androgens and estrogens. However, the question remains whether the synthetic analogues of 7α/β- and 16α-hydroxy-steroids prevent the catabolism of the sex steroid precursors, or further activate the "energy wasting" mitochondrial thermogenic metabolism.
Collapse
Affiliation(s)
- Josef Bulant
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic; Institute of Endocrinology, Prague, Czech Republic; Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| | - Martin Hill
- Institute of Endocrinology, Prague, Czech Republic.
| | | | - Anna Yamamotová
- Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Pavel Martásek
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic; Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec, Czech Republic.
| | - Hana Papežová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| |
Collapse
|
12
|
Amer SA, Alzanati NG, Warren A, Tarbox R, Khan R. Excess androgen production in subcutaneous adipose tissue of women with polycystic ovarian syndrome is not related to insulin or LH. J Endocrinol 2019; 241:JOE-18-0674.R1. [PMID: 30802211 DOI: 10.1530/joe-18-0674] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 02/25/2019] [Indexed: 01/08/2023]
Abstract
The purpose of this study was to investigate androgen production and the role of insulin and LH in its regulation in subcutaneous adipose tissue (SAT) of women with polycystic ovarian syndrome (PCOS). Protein and mRNA expression of androgen synthesis enzymes (Cytochrome P450 17A1 [CYP17A1] and Aldo-keto reductase 1C3 [AKR1C3]) were measured in SAT biopsies from women with PCOS, diagnosed according to the Rotterdam criteria (n=15) and healthy controls (n=15). Cultured mature adipocytes (differentiated from SAT biopsies) were treated with insulin ± phosphoinositol-3-kinase inhibitor (LY294002) or LH ± insulin. CYP17A1 and AKR1C3 mRNA expression and testosterone concentrations were measured in treated and untreated adipocyte cultures. AKR1C3 mRNA was significantly (P<0.001) greater in PCOS versus non-PCOS SAT, but CYP17A1 was not significantly different between the two groups. AKR1C3 and CYP17A1 protein expression was not significantly different in PCOS versus non-PCOS SAT. In untreated adipocyte cultures, CYP17A1, AKR1C3 and testosterone levels were significantly higher in the PCOS versus the non-PCOS groups. Addition of insulin increased AKR1C3 mRNA and testosterone levels, but not CYP17A1 mRNA in non-PCOS with no effect on PCOS adipocytes. The stimulatory effects of insulin were not inhibited by LY294002. Addition of LH increased CYP17A1, AKR1C3 and testosterone in non-PCOS adipocytes with no effect in PCOS adipocytes. In conclusion, SAT of women with PCOS produces excess androgen, which may contribute to PCOS-related hyperandrogenaemia. This SAT androgen excess is independent of obesity and is not directly stimulated by insulin or LH.
Collapse
Affiliation(s)
- Saad Aks Amer
- S Amer, Division of Medical Sciences and Graduate Entry Medicine, University of Nottingham School of Health Sciences, Derby, United Kingdom of Great Britain and Northern Ireland
| | - Nadia G Alzanati
- N Alzanati, Division of Medical Sciences and Graduate Entry Medicine , University of Nottingham School of Health Sciences, Derby, United Kingdom of Great Britain and Northern Ireland
| | - Avril Warren
- A Warren, Division of Medical Sciences and Graduate Entry Medicine , University of Nottingham School of Medicine, Derby, United Kingdom of Great Britain and Northern Ireland
| | - Rebecca Tarbox
- R Tarbox, Division of Medical Sciences and Graduate Entry Medicine , University of Nottingham School of Health Sciences, Derby, United Kingdom of Great Britain and Northern Ireland
| | - Raheela Khan
- R Khan, Division of Medical Sciences and Graduate Entry Medicine , University of Nottingham School of Medicine, Derby, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
13
|
Nyce JW. Detection of a novel, primate-specific 'kill switch' tumor suppression mechanism that may fundamentally control cancer risk in humans: an unexpected twist in the basic biology of TP53. Endocr Relat Cancer 2018; 25:R497-R517. [PMID: 29941676 PMCID: PMC6106910 DOI: 10.1530/erc-18-0241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
Abstract
The activation of TP53 is well known to exert tumor suppressive effects. We have detected a primate-specific adrenal androgen-mediated tumor suppression system in which circulating DHEAS is converted to DHEA specifically in cells in which TP53 has been inactivated DHEA is an uncompetitive inhibitor of glucose-6-phosphate dehydrogenase (G6PD), an enzyme indispensable for maintaining reactive oxygen species within limits survivable by the cell. Uncompetitive inhibition is otherwise unknown in natural systems because it becomes irreversible in the presence of high concentrations of substrate and inhibitor. In addition to primate-specific circulating DHEAS, a unique, primate-specific sequence motif that disables an activating regulatory site in the glucose-6-phosphatase (G6PC) promoter was also required to enable function of this previously unrecognized tumor suppression system. In human somatic cells, loss of TP53 thus triggers activation of DHEAS transport proteins and steroid sulfatase, which converts circulating DHEAS into intracellular DHEA, and hexokinase which increases glucose-6-phosphate substrate concentration. The triggering of these enzymes in the TP53-affected cell combines with the primate-specific G6PC promoter sequence motif that enables G6P substrate accumulation, driving uncompetitive inhibition of G6PD to irreversibility and ROS-mediated cell death. By this catastrophic 'kill switch' mechanism, TP53 mutations are effectively prevented from initiating tumorigenesis in the somatic cells of humans, the primate with the highest peak levels of circulating DHEAS. TP53 mutations in human tumors therefore represent fossils of kill switch failure resulting from an age-related decline in circulating DHEAS, a potentially reversible artifact of hominid evolution.
Collapse
|
14
|
Piccinato CA, Malvezzi H, Gibson DA, Saunders PTK. SULFATION PATHWAYS: Contribution of intracrine oestrogens to the aetiology of endometriosis. J Mol Endocrinol 2018; 61:T253-T270. [PMID: 30030390 DOI: 10.1530/jme-17-0297] [Citation(s) in RCA: 7] [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: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022]
Abstract
Endometriosis is an incurable hormone-dependent inflammatory disease that causes chronic pelvic pain and infertility characterized by implantation and growth of endometrial tissue outside the uterine cavity. Symptoms have a major impact on the quality of life of patients resulting in socioeconomic, physical and psychological burdens. Although the immune system and environmental factors may play a role in the aetiology of endometriosis, oestrogen dependency is still considered a hallmark of the disorder. The impact of oestrogens such as oestrone and particularly, oestradiol, on the endometrium or endometriotic lesions may be mediated by steroids originating from ovarian steroidogenesis or local intra-tissue production (intracrinology) dependent upon the expression and activity of enzymes that regulate oestrogen biosynthesis and metabolism. Two key pathways have been implicated: while there is contradictory data on the participation of the aromatase enzyme (encoded by CYP19A1), there is increasing evidence that the steroid sulphatase pathway plays a role in both the aetiology and pathology of endometriosis. In this review, we consider the evidence related to the pathways leading to oestrogen accumulation in endometriotic lesions and how this might inform the development of new therapeutic strategies to treat endometriosis without causing the undesirable side effects of current regimes that suppress ovarian hormone production.
Collapse
Affiliation(s)
| | - Helena Malvezzi
- Hospital Israelita Albert Einstein, São Paulo, São Paulo, Brazil
| | - Douglas A Gibson
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
15
|
Simitsidellis I, Saunders PTK, Gibson DA. Androgens and endometrium: New insights and new targets. Mol Cell Endocrinol 2018; 465:48-60. [PMID: 28919297 DOI: 10.1016/j.mce.2017.09.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/08/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022]
Abstract
Androgens are synthesised in both the ovary and adrenals in women and play an important role in the regulation of female fertility, as well as in the aetiology of disorders such as polycystic ovarian syndrome, endometriosis and endometrial cancer. The endometrium is an androgen target tissue and the impact of AR-mediated effects has been studied using human endometrial tissue samples and rodent models. In this review we highlight recent evidence that endometrial androgen biosynthesis and intracrine action is important in preparation of a tissue microenvironment that can support implantation and establishment of pregnancy. The impact of androgens on endometrial cell proliferation, in repair of the endometrial wound at the time of menstruation and in endometrial disorders is discussed. Future directions for research focused on AR function as a therapeutic target are considered.
Collapse
Affiliation(s)
- Ioannis Simitsidellis
- Medical Research Council Centre for Inflammation Research, The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Philippa T K Saunders
- Medical Research Council Centre for Inflammation Research, The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Douglas A Gibson
- Medical Research Council Centre for Inflammation Research, The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
16
|
Nguyen TV, Wu M, Lew J, Albaugh MD, Botteron KN, Hudziak JJ, Fonov VS, Collins DL, Campbell BC, Booij L, Herba C, Monnier P, Ducharme S, McCracken JT. Dehydroepiandrosterone impacts working memory by shaping cortico-hippocampal structural covariance during development. Psychoneuroendocrinology 2017; 86:110-121. [PMID: 28946055 PMCID: PMC5659912 DOI: 10.1016/j.psyneuen.2017.09.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 11/18/2022]
Abstract
Existing studies suggest that dehydroepiandrosterone (DHEA) may be important for human brain development and cognition. For example, molecular studies have hinted at the critical role of DHEA in enhancing brain plasticity. Studies of human brain development also support the notion that DHEA is involved in preserving cortical plasticity. Further, some, though not all, studies show that DHEA administration may lead to improvements in working memory in adults. Yet these findings remain limited by an incomplete understanding of the specific neuroanatomical mechanisms through which DHEA may impact the CNS during development. Here we examined associations between DHEA, cortico-hippocampal structural covariance, and working memory (216 participants [female=123], age range 6-22 years old, mean age: 13.6 +/-3.6 years, each followed for a maximum of 3 visits over the course of 4 years). In addition to administering performance-based, spatial working memory tests to these children, we also collected ecological, parent ratings of working memory in everyday situations. We found that increasingly higher DHEA levels were associated with a shift toward positive insular-hippocampal and occipito-hippocampal structural covariance. In turn, DHEA-related insular-hippocampal covariance was associated with lower spatial working memory but higher overall working memory as measured by the ecological parent ratings. Taken together with previous research, these results support the hypothesis that DHEA may optimize cortical functions related to general attentional and working memory processes, but impair the development of bottom-up, hippocampal-to-cortical connections, resulting in impaired encoding of spatial cues.
Collapse
Affiliation(s)
- Tuong-Vi Nguyen
- Department of Psychiatry, McGill University, Montreal, QC, H3A1A1, Canada; Department of Obstetrics-Gynecology, McGill University Health Center, Montreal, QC, H4A 3J1, Canada; Research Institute of the McGill University Health Center, Montreal, QC, H4A 3J1, Canada.
| | - Mia Wu
- Department of Psychology, McGill University, Montreal, QC, H4A 3J1, Canada
| | - Jimin Lew
- Department of Psychology, McGill University, Montreal, QC, H4A 3J1, Canada
| | - Matthew D Albaugh
- Department of Psychology, University of Vermont, College of Medicine, Burlington, VT, 05405, USA
| | - Kelly N Botteron
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA; Brain Development Cooperative Group, United States
| | - James J Hudziak
- Department of Psychology, University of Vermont, College of Medicine, Burlington, VT, 05405, USA; Brain Development Cooperative Group, United States
| | - Vladimir S Fonov
- McConnell Brain imaging Centre, Montreal Neurological Institute, Montreal, QC, H3A 2B4, Canada
| | - D Louis Collins
- McConnell Brain imaging Centre, Montreal Neurological Institute, Montreal, QC, H3A 2B4, Canada
| | - Benjamin C Campbell
- Department of Anthropology, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Linda Booij
- Department of Psychiatry, McGill University, Montreal, QC, H3A1A1, Canada; Department of Psychology, Concordia University, Montreal, QC, H4B 1R6, Canada; CHU Sainte Justine Hospital Research Centre, University of Montreal, Montreal, QC, H3T1C5, Canada
| | - Catherine Herba
- CHU Sainte Justine Hospital Research Centre, University of Montreal, Montreal, QC, H3T1C5, Canada; Department of Psychology, Université du Québec à Montréal, Montreal, QC, Canada
| | - Patricia Monnier
- Department of Obstetrics-Gynecology, McGill University Health Center, Montreal, QC, H4A 3J1, Canada; Research Institute of the McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Simon Ducharme
- Department of Psychiatry, McGill University, Montreal, QC, H3A1A1, Canada; McConnell Brain imaging Centre, Montreal Neurological Institute, Montreal, QC, H3A 2B4, Canada; Department of Neurology & Neurosurgery, McGill University, Montreal, QC, H3A 1A1, Canada
| | - James T McCracken
- Brain Development Cooperative Group, United States; Department of Child and Adolescent Psychiatry, University of California in Los Angeles, Los Angeles, CA, 90024, USA
| |
Collapse
|
17
|
Hetemäki N, Savolainen-Peltonen H, Tikkanen MJ, Wang F, Paatela H, Hämäläinen E, Turpeinen U, Haanpää M, Vihma V, Mikkola TS. Estrogen Metabolism in Abdominal Subcutaneous and Visceral Adipose Tissue in Postmenopausal Women. J Clin Endocrinol Metab 2017; 102:4588-4595. [PMID: 29029113 DOI: 10.1210/jc.2017-01474] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/21/2017] [Indexed: 02/13/2023]
Abstract
CONTEXT In postmenopausal women, adipose tissue (AT) levels of estrogens exceed circulating concentrations. Although increased visceral AT after menopause is related to metabolic diseases, little is known about differences in estrogen metabolism between different AT depots. OBJECTIVE We compared concentrations of and metabolic pathways producing estrone and estradiol in abdominal subcutaneous and visceral AT in postmenopausal women. DESIGN, SETTING, PATIENTS, AND INTERVENTIONS AT and serum samples were obtained from 37 postmenopausal women undergoing surgery for nonmalignant gynecological reasons. Serum and AT estrone, estradiol, and serum estrone sulfate (E1S) concentrations were quantitated using liquid chromatography-tandem mass spectrometry. Activity of steroid sulfatase and reductive 17β-hydroxysteroid dehydrogenase enzymes was measured using radiolabeled precursors. Messenger RNA (mRNA) expression of estrogen-converting enzymes was analyzed by real-time reverse transcription quantitative polymerase chain reaction. RESULTS Estrone concentration was higher in visceral than subcutaneous AT (median, 928 vs 706 pmol/kg; P = 0.002) and correlated positively with body mass index (r = 0.46; P = 0.011). Both AT depots hydrolyzed E1S to estrone, and visceral AT estrone and estradiol concentrations correlated positively with serum E1S. Compared with visceral AT, subcutaneous AT produced more estradiol from estrone (median rate of estradiol production, 1.02 vs 0.57 nmol/kg AT/h; P = 0.004). In visceral AT, the conversion of estrone to estradiol increased with waist circumference (r = 0.65; P = 0.022), and estradiol concentration correlated positively with mRNA expression of HSD17B7 (r = 0.76; P = 0.005). CONCLUSIONS Both estrone and estradiol production in visceral AT increased with adiposity, but estradiol was produced more effectively in subcutaneous fat. Both AT depots produced estrone from E1S. Increasing visceral adiposity could increase overall estrogen exposure in postmenopausal women.
Collapse
Affiliation(s)
- Natalia Hetemäki
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Finland
- Folkhälsan Research Center, University of Helsinki, Finland
| | - Hanna Savolainen-Peltonen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Finland
- Folkhälsan Research Center, University of Helsinki, Finland
| | - Matti J Tikkanen
- Folkhälsan Research Center, University of Helsinki, Finland
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Feng Wang
- Folkhälsan Research Center, University of Helsinki, Finland
| | - Hanna Paatela
- Folkhälsan Research Center, University of Helsinki, Finland
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Finland
| | | | | | | | - Veera Vihma
- Folkhälsan Research Center, University of Helsinki, Finland
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Tomi S Mikkola
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Finland
- Folkhälsan Research Center, University of Helsinki, Finland
| |
Collapse
|
18
|
Hennig K, Antignac JP, Bichon E, Morvan ML, Miran I, Delaloge S, Feunteun J, Le Bizec B. Steroid hormone profiling in human breast adipose tissue using semi-automated purification and highly sensitive determination of estrogens by GC-APCI-MS/MS. Anal Bioanal Chem 2017; 410:259-275. [PMID: 29147745 DOI: 10.1007/s00216-017-0717-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/22/2017] [Accepted: 10/18/2017] [Indexed: 01/08/2023]
Abstract
Body mass index is a known breast cancer risk factor due to, among other mechanisms, adipose-derived hormones. We developed a method for steroid hormone profiling in adipose tissue to evaluate healthy tissue around the tumor and define new biomarkers for cancer development. A semi-automated sample preparation method based on gel permeation chromatography and subsequent derivatization with trimethylsilyl (TMS) is presented. Progestagens and androgens were determined by GC-EI-MS/MS (LOQ 0.5 to 10 ng/g lipids). For estrogen measurement, a highly sensitive GC-APCI-MS/MS method was developed to reach the required lower limits of detection (0.05 to 0.1 ng/g lipids in matrix, 100-200 fg on column for pure standards). The combination of the two methods allows the screening of 27 androgens and progestagens and 4 estrogens from a single sample. Good accuracies and repeatabilities were achieved for each compound class at their respective limit of detection. The method was applied to determine steroid hormone profiles in adipose tissue of 51 patients, collected both at proximity and distant to the tumor. Out of the 31 tested steroid hormones, 14 compounds were detected in human samples. Pregnenolone, 17-hydroxypregnenolone, dehydroepiandrosterone (DHEA), and androstendione accounted together for 80% of the observed steroid hormone profiles, whereas the estrogens accounted for only 1%. These profiles did not differ based on sampling location, except for ß-estradiol; steroid hormone conversions from androgens to estrogens that potentially take place in adipose or tumoral tissue might not be detectable due a factor 100 difference in concentration of for example DHEA and ß-estradiol. Graphical Abstract Schematic overview of the determination of steroid hormones and metabolites in adipose tissue in proximity and distal to the tumor.
Collapse
Affiliation(s)
- Kristin Hennig
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France
| | - Jean Philippe Antignac
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France.
| | - Emmanuelle Bichon
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France
| | - Marie-Line Morvan
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France
| | - Isabelle Miran
- UMR981 INSERM, Gustave Roussy, Paris-Saclay University, 94805, Villejuif, France
| | - Suzette Delaloge
- Breast Cancer Group, Gustave Roussy Cancer Campus, 94805, Villejuif, France
| | - Jean Feunteun
- UMR8200 CNRS, Gustave Roussy, Paris-Saclay University, 94805, Villejuif, France
| | - Bruno Le Bizec
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France
| |
Collapse
|
19
|
Kulle AE, Reinehr T, Simic-Schleicher G, Hornig NC, Holterhus PM. Determination of 17OHPreg and DHEAS by LC-MS/MS: Impact of Age, Sex, Pubertal Stage, and BMI on the Δ5 Steroid Pathway. J Clin Endocrinol Metab 2017; 102:232-241. [PMID: 27809697 DOI: 10.1210/jc.2016-2849] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/01/2016] [Indexed: 02/13/2023]
Abstract
BACKGROUND Dehydroepiandrosterone sulfate (DHEAS) and 17-hydroxypregnenolone (17OHPreg) are important for understanding the Δ5 pathway (e.g., in adrenarche and obesity). Although mass spectrometry has become the state-of-the-art method for quantifying steroids, there are few comprehensive age-, sex-, and pubertal stage-specific reference ranges for children. AIMS To develop a sensitive and reliable ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method for simultaneous quantification of DHEAS and 17OHPreg and to establish entire age-, sex- and pubertal stage-specific reference ranges in children. METHODS A total of 684 children, 453 (243 female, 210 male) with normal body mass index (BMI; <90th) and 231 (132 female, 99 male) obese subjects (>97th), were categorized into 11 age groups, and age- and Tanner stage (PH)-specific reference ranges were determined. RESULTS The limit of detection was 0.05 nmol/L for 17OHPreg and 0.5 nmol/L for DHEAS. Levels of both steroids declined after the neonatal period. Comparisons with RIA assays (Siemens, Munich, Germany) (DHEAS) and an in-house kit (17OHPreg) revealed 0.95 and 0.93, respectively, as coefficients of determination. Although DHEAS-generally higher in boys-increased continuously starting at 3 to 6 years, 17OHPreg remained largely constant. In obese patients, both were significantly elevated, also in part after alignment to Tanner stages (PH). CONCLUSIONS UPLC-MS/MS is sensitive and reliable for quantifying DHEAS and 17OHPreg. Our data support differential maturation of CYP17 during adrenarche with successively increasing 17,20-lyase activity but largely constant 17α-hydroxylation activity. Endocrine interpretation of 17OHPreg and DHEAS must consider differential patterns for age, sex, pubertal stage, and BMI.
Collapse
Affiliation(s)
- Alexandra E Kulle
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Thomas Reinehr
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten/Herdecke, Datteln, Germany; and
| | | | - Nadine C Hornig
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Paul-Martin Holterhus
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
20
|
Piccinato CA, Neme RM, Torres N, Sanches LR, Derogis PBMC, Brudniewski HF, Rosa E Silva JC, Ferriani RA. Effects of steroid hormone on estrogen sulfotransferase and on steroid sulfatase expression in endometriosis tissue and stromal cells. J Steroid Biochem Mol Biol 2016; 158:117-126. [PMID: 26723541 DOI: 10.1016/j.jsbmb.2015.12.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 11/23/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022]
Abstract
Endometriosis is an estrogen-dependent disease that afflicts about 10% of women in their reproductive age, causing severe pain and infertility. The potential roles of female steroid hormones in modulating key estrogen-metabolizing enzymes, steroid sulfatase (STS) and estrogen sulfotransferase (SULT1E1), were investigated. The expression of STS and SULT1E1 mRNA in biopsy samples (n=78) of superficial and deep endometriotic lesions, eutopic endometrium of women with endometriosis and endometrium from control patients were compared according to the menstrual cycle phase. Increased STS gene expression was detected in superficial and deep-infiltrating lesions and a reduced SULT1E1 expression was also observed in the eutopic endometrium relative to the superficial lesions. Additionally, a significantly positive correlation was detected between STS and SULT1E1 mRNA expression levels in biopsy specimens collected from the endometriosis patients, and not in control individuals. The actions of female steroid hormones on SULT1E1 and STS expression were evidenced in endometriosis, revealed by increased expression levels in the luteal phase of the cycle. There was an increased STS expression in primary eutopic and ectopic endometrial stromal cells treated with estradiol and progesterone (representative of the luteal phase, n=3). Although an increased STS mRNA expression was observed in hormone-induced endometrial stromal cells in vitro, no difference could be detected between the hormone treatment groups in estradiol formation from estradiol sulfate measured by LC-MS-MS. Interestingly, a greater expression of STS was observed in stromal cells from eutopic endometrium with an agreement in estradiol formation originated from estradiol sulfate. The differential regulation of STS and SULT1E1 could provide insights for novel studies of the therapeutic use of STS inhibitors.
Collapse
Affiliation(s)
- Carla A Piccinato
- Hospital Israelita Albert Einstein, São Paulo, SP 05652-900, Brazil; Department of Gynaecology and Obstetrics, School of Medicine of Ribeirão Preto, Universidade de São Paulo, Brazil.
| | - Rosa M Neme
- Hospital Israelita Albert Einstein, São Paulo, SP 05652-900, Brazil; Centro de Endometriose São Paulo, Av. República do Líbano, 460 São Paulo, SP 04502-000, Brazil.
| | - Natália Torres
- Hospital Israelita Albert Einstein, São Paulo, SP 05652-900, Brazil.
| | - Lívia Renta Sanches
- Department of Clinical Pathology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | | | - Heloísa F Brudniewski
- Hospital Israelita Albert Einstein, São Paulo, SP 05652-900, Brazil; Centro de Endometriose São Paulo, Av. República do Líbano, 460 São Paulo, SP 04502-000, Brazil.
| | - Júlio C Rosa E Silva
- Department of Gynaecology and Obstetrics, School of Medicine of Ribeirão Preto, Universidade de São Paulo, Brazil.
| | - Rui A Ferriani
- Department of Gynaecology and Obstetrics, School of Medicine of Ribeirão Preto, Universidade de São Paulo, Brazil.
| |
Collapse
|
21
|
Paatela H, Wang F, Vihma V, Savolainen-Peltonen H, Mikkola TS, Turpeinen U, Hämäläinen E, Jauhiainen M, Tikkanen MJ. Steroid sulfatase activity in subcutaneous and visceral adipose tissue: a comparison between pre- and postmenopausal women. Eur J Endocrinol 2016; 174:167-75. [PMID: 26553725 DOI: 10.1530/eje-15-0831] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/06/2015] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Adipose tissue is an important extragonadal site for steroid hormone biosynthesis. After menopause, estrogens are synthesized exclusively in peripheral tissues from circulating steroid precursors, of which the most abundant is dehydroepiandrosterone sulfate (DHEAS). Our aim was to study activity of steroid sulfatase, an enzyme hydrolyzing DHEAS, and expression of steroid-converting enzyme genes in subcutaneous and visceral adipose tissue derived from pre- and postmenopausal women. DESIGN Serum and paired abdominal subcutaneous and visceral adipose tissue samples were obtained from 18 premenopausal and seven postmenopausal women undergoing elective surgery for non-malignant reasons in Helsinki University Central Hospital. METHODS To assess steroid sulfatase activity, radiolabeled DHEAS was incubated in the presence of adipose tissue homogenate and the liberated dehydroepiandrosterone (DHEA) was measured. Gene mRNA expressions were analyzed by quantitative RT-PCR. Serum DHEAS, DHEA, and estrogen concentrations were determined by liquid chromatography-tandem mass spectrometry. RESULTS Steroid sulfatase activity was higher in postmenopausal compared to premenopausal women in subcutaneous (median 379 vs 257 pmol/kg tissue per hour; P=0.006) and visceral (545 vs 360 pmol/kg per hour; P=0.004) adipose tissue. Visceral fat showed higher sulfatase activity than subcutaneous fat in premenopausal (P=0.035) and all (P=0.010) women. The mRNA expression levels of two estradiol-producing enzymes, aromatase and 17β-hydroxysteroid dehydrogenase type 12, were higher in postmenopausal than in premenopausal subcutaneous adipose tissue. CONCLUSIONS Steroid sulfatase activity in adipose tissue was higher in postmenopausal than in premenopausal women suggesting that DHEAS, derived from the circulation, could be more efficiently utilized in postmenopausal adipose tissue for the formation of biologically active sex hormones.
Collapse
Affiliation(s)
- Hanna Paatela
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Feng Wang
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Veera Vihma
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Hanna Savolainen-Peltonen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Tomi S Mikkola
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Ursula Turpeinen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Esa Hämäläinen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Matti Jauhiainen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Matti J Tikkanen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| |
Collapse
|
22
|
Li J, Papadopoulos V, Vihma V. Steroid biosynthesis in adipose tissue. Steroids 2015; 103:89-104. [PMID: 25846979 DOI: 10.1016/j.steroids.2015.03.016] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/20/2015] [Accepted: 03/24/2015] [Indexed: 12/25/2022]
Abstract
Tissue-specific expression of steroidogenic enzymes allows the modulation of active steroid levels in a local manner. Thus, the measurement of local steroid concentrations, rather than the circulating levels, has been recognized as a more accurate indicator of the steroid action within a specific tissue. Adipose tissue, one of the largest endocrine tissues in the human body, has been established as an important site for steroid storage and metabolism. Locally produced steroids, through the enzymatic conversion from steroid precursors delivered to adipose tissue, have been proven to either functionally regulate adipose tissue metabolism, or quantitatively contribute to the whole body's steroid levels. Most recently, it has been suggested that adipose tissue may contain the steroidogenic machinery necessary for the initiation of steroid biosynthesis de novo from cholesterol. This review summarizes the evidence indicating the presence of the entire steroidogenic apparatus in adipose tissue and discusses the potential roles of local steroid products in modulating adipose tissue activity and other metabolic parameters.
Collapse
Affiliation(s)
- Jiehan Li
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Department of Biochemistry, McGill University, Montreal, Canada
| | - Vassilios Papadopoulos
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Department of Biochemistry, McGill University, Montreal, Canada.
| | - Veera Vihma
- Folkhälsan Research Center, Helsinki, Finland; University of Helsinki and Helsinki University Central Hospital, Heart and Lung Center, Helsinki, Finland.
| |
Collapse
|
23
|
Mueller JW, Gilligan LC, Idkowiak J, Arlt W, Foster PA. The Regulation of Steroid Action by Sulfation and Desulfation. Endocr Rev 2015; 36:526-63. [PMID: 26213785 PMCID: PMC4591525 DOI: 10.1210/er.2015-1036] [Citation(s) in RCA: 285] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/21/2015] [Indexed: 12/14/2022]
Abstract
Steroid sulfation and desulfation are fundamental pathways vital for a functional vertebrate endocrine system. After biosynthesis, hydrophobic steroids are sulfated to expedite circulatory transit. Target cells express transmembrane organic anion-transporting polypeptides that facilitate cellular uptake of sulfated steroids. Once intracellular, sulfatases hydrolyze these steroid sulfate esters to their unconjugated, and usually active, forms. Because most steroids can be sulfated, including cholesterol, pregnenolone, dehydroepiandrosterone, and estrone, understanding the function, tissue distribution, and regulation of sulfation and desulfation processes provides significant insights into normal endocrine function. Not surprisingly, dysregulation of these pathways is associated with numerous pathologies, including steroid-dependent cancers, polycystic ovary syndrome, and X-linked ichthyosis. Here we provide a comprehensive examination of our current knowledge of endocrine-related sulfation and desulfation pathways. We describe the interplay between sulfatases and sulfotransferases, showing how their expression and regulation influences steroid action. Furthermore, we address the role that organic anion-transporting polypeptides play in regulating intracellular steroid concentrations and how their expression patterns influence many pathologies, especially cancer. Finally, the recent advances in pharmacologically targeting steroidogenic pathways will be examined.
Collapse
Affiliation(s)
- Jonathan W Mueller
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Lorna C Gilligan
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jan Idkowiak
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Wiebke Arlt
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Paul A Foster
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
24
|
Tchernof A, Mansour MF, Pelletier M, Boulet MM, Nadeau M, Luu-The V. Updated survey of the steroid-converting enzymes in human adipose tissues. J Steroid Biochem Mol Biol 2015; 147:56-69. [PMID: 25448733 DOI: 10.1016/j.jsbmb.2014.11.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 11/10/2014] [Accepted: 11/12/2014] [Indexed: 12/26/2022]
Abstract
Over the past decade, adipose tissues have been increasingly known for their endocrine properties, that is, their ability to secrete a number of adipocytokines that may exert local and/or systemic effects. In addition, adipose tissues have long been recognized as significant sites for steroid hormone transformation and action. We hereby provide an updated survey of the many steroid-converting enzymes that may be detected in human adipose tissues, their activities and potential roles. In addition to the now well-established role of aromatase and 11β-hydroxysteroid dehydrogenase (HSD) type 1, many enzymes have been reported in adipocyte cell lines, isolated mature cells and/or preadipocytes. These include 11β-HSD type 2, 17β-HSDs, 3β-HSD, 5α-reductases, sulfatases and glucuronosyltransferases. Some of these enzymes are postulated to bear relevance for adipose tissue physiology and perhaps for the pathophysiology of obesity. This elaborate set of steroid-converting enzymes in the cell types of adipose tissue deserves further scientific attention. Our work on 20α-HSD (AKR1C1), 3α-HSD type 3 (AKR1C2) and 17β-HSD type 5 (AKR1C3) allowed us to clarify the relevance of these enzymes for some aspects of adipose tissue function. For example, down-regulation of AKR1C2 expression in preadipocytes seems to potentiate the inhibitory action of dihydrotestosterone on adipogenesis in this model. Many additional studies are warranted to assess the impact of intra-adipose steroid hormone conversions on adipose tissue functions and chronic conditions such as obesity, diabetes and cancer.
Collapse
Affiliation(s)
- André Tchernof
- Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Canada; École de Nutrition, Université Laval, Québec, Canada; Endocrinologe et Néphrologie, Centre Hospitalier Universitaire de Québec, Québec, Canada.
| | - Mohamed Fouad Mansour
- Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Canada; Endocrinologe et Néphrologie, Centre Hospitalier Universitaire de Québec, Québec, Canada
| | - Mélissa Pelletier
- Endocrinologe et Néphrologie, Centre Hospitalier Universitaire de Québec, Québec, Canada
| | - Marie-Michèle Boulet
- Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Canada; École de Nutrition, Université Laval, Québec, Canada
| | - Mélanie Nadeau
- Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Canada
| | - Van Luu-The
- Endocrinologe et Néphrologie, Centre Hospitalier Universitaire de Québec, Québec, Canada
| |
Collapse
|
25
|
Munoz J, Wheler JJ, Kurzrock R. Androgen receptors beyond prostate cancer: an old marker as a new target. Oncotarget 2015; 6:592-603. [PMID: 25595907 PMCID: PMC4359241 DOI: 10.18632/oncotarget.2831] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/14/2014] [Indexed: 12/25/2022] Open
Abstract
Androgen receptors (ARs) play a critical role in the development of prostate cancer. Targeting ARs results in important salutary effects in this malignancy. Despite mounting evidence that ARs also participate in the pathogenesis and/or progression of diverse tumors, exploring the impact of hormonal manipulation of these receptors has not been widely pursued beyond prostate cancer. This review describes patterns of AR expression in a spectrum of cancers, and the potential to exploit this knowledge in the clinical therapeutic setting.
Collapse
Affiliation(s)
- Javier Munoz
- Hematology-Oncology, Banner MD Anderson Cancer Center, Gilbert, Arizona
| | - Jennifer J. Wheler
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Division and Hematology and Oncology, Moores Cancer Center, The University of California San Diego, La Jolla, California
| |
Collapse
|
26
|
Cho E, Montgomery RB, Mostaghel EA. Minireview: SLCO and ABC transporters: a role for steroid transport in prostate cancer progression. Endocrinology 2014; 155:4124-32. [PMID: 25147980 PMCID: PMC4298565 DOI: 10.1210/en.2014-1337] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Androgens play a critical role in the development and progression of prostate cancer (PCa), and androgen deprivation therapy via surgical or medical castration is front-line therapy for patients with advanced PCa. However, intratumoral testosterone levels are elevated in metastases from patients with castration-resistant disease, and residual intratumoral androgens have been implicated in mediating ligand-dependent mechanisms of androgen receptor activation. The source of residual tissue androgens present despite castration has not been fully elucidated, but proposed mechanisms include uptake and conversion of adrenal androgens, such as dehdroepiandrosterone to testosterone and dihydrotestosterone, or de novo androgen synthesis from cholesterol or progesterone precursors. In this minireview, we discuss the emerging evidence that suggests a role for specific transporters in mediating transport of steroids into or out of prostate cells, thereby influencing intratumoral androgen levels and PCa development and progression. We focus on the solute carrier and ATP binding cassette gene families, which have the most published data for a role in PCa-related steroid transport, and review the potential impact of genetic variation on steroid transport activity and PCa outcomes. Continued assessment of transport activity in PCa models and human tumor tissue is needed to better delineate the different roles these transporters play in physiologic and neoplastic settings, and in order to determine whether targeting the uptake of steroid substrates by specific transporters may be a clinically feasible therapeutic strategy.
Collapse
Affiliation(s)
- Eunpi Cho
- School of Medicine (E.C., R.B.M.), University of Washington, Seattle, Washington 98195; and Division of Clinical Research (E.A.M.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | | | | |
Collapse
|
27
|
O'Reilly MW, House PJ, Tomlinson JW. Understanding androgen action in adipose tissue. J Steroid Biochem Mol Biol 2014; 143:277-84. [PMID: 24787657 DOI: 10.1016/j.jsbmb.2014.04.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/14/2014] [Accepted: 04/20/2014] [Indexed: 12/15/2022]
Abstract
Androgens play an important role in regulation of body fat distribution in humans. They exert direct effects on adipocyte differentiation in a depot-specific manner, via the androgen receptor (AR), leading to modulation of adipocyte size and fat compartment expansion. Androgens also impact directly on key adipocyte functions including insulin signalling, lipid metabolism, fatty acid uptake and adipokine production. Androgen excess and deficiency have implications for metabolic health in both males and females, and these metabolic effects may be mediated through adipose tissue via effects on fat distribution, adipocyte function and lipolysis. Research into the field of androgen metabolism in human and animal adipose tissue has produced inconsistent results; it is important to take into account the sex-, depot- and organism-specific effects of androgens in fat. In general, studies point towards a stimulatory effect on lipolysis, with impairment of adipocyte differentiation, insulin signalling and adipokine generation. Observed effects are frequently gender-specific. Adipose tissue is an important organ of pre-receptor androgen metabolism, through which local androgen availability is rigorously controlled. Adipose androgen exposure is tightly controlled by isoenzymes of AKR1C, 5α-reductase and others, but regulation of the balance between generation and irreversible inactivation remains poorly understood. In particular, AKR1C2 and AKR1C3 are crucial in the regulation of local androgen bioavailability within adipose tissue. These isoforms control the balance between activation of androstenedione (A) to testosterone (T) by the 17β-hydroxysteroid dehydrogenase activity (17β-HSD) of AKR1C3, or inactivation of 5α-dihydrotestosterone (DHT) to 5α-androstane-3α,17β-diol by the 3α-hydroxysteroid dehydrogenase (3α-HSD) activity of AKR1C2. Most studies suggest that androgen inactivation is the predominant reaction in fat, particularly in the abdominal subcutaneous (SC) depot. Modulation of local adipose androgen availability may afford future therapeutic options to improve metabolic phenotype in disorders of androgen excess and deficiency.
Collapse
Affiliation(s)
- Michael W O'Reilly
- Centre for Endocrinology, Diabetes and Metabolism, School of Clinical and Experimental Medicine, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Philip J House
- Centre for Endocrinology, Diabetes and Metabolism, School of Clinical and Experimental Medicine, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Jeremy W Tomlinson
- Centre for Endocrinology, Diabetes and Metabolism, School of Clinical and Experimental Medicine, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
28
|
Campagnoli C, Berrino F, Venturelli E, Abbà C, Biglia N, Brucato T, Cogliati P, Danese S, Donadio M, Zito G, Pasanisi P. Metformin decreases circulating androgen and estrogen levels in nondiabetic women with breast cancer. Clin Breast Cancer 2014; 13:433-8. [PMID: 24267731 DOI: 10.1016/j.clbc.2013.08.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/08/2013] [Accepted: 08/26/2013] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Diabetic patients treated with metformin have a lower risk of developing BC or a better BC prognosis. Metformin might reduce cancer growth through direct antiproliferative effects or through indirect mechanisms, particularly the reduction of insulin. In a randomized study on nondiabetic BC patients in natural menopause with high testosterone levels, we observed a significant decrease in insulin and in testosterone levels with metformin 1500 mg/d compared with 1000 mg/d. We present the results of a new analysis of our study on the effect of metformin on the bioavailability of sex hormones. PATIENTS AND METHODS One hundred twenty-four eligible women were initially invited to take metformin 500 mg/d for 3 months. The 108 women who completed the first 3 months continued the study using 1000 mg/d for 1 month. The women were then randomized into 2 groups, and, for the subsequent 5 months, 1 group increased the dose to 1500 mg/d, and the other group continued with 1000 mg/d. RESULTS Ninety-six women completed the study, 43 receiving metformin 1500 mg/day, and 53 receiving 1000 mg/day. The women receiving 1500 mg/d showed a greater and significant reduction of free testosterone (-29%) and estradiol (-38%), a borderline significant reduction of estrone and insulin-like growth factor-1, and a nonsignificant reduction of androstenedione. They also showed a nonsignificant increase of dehydroepiandrosterone sulfate. CONCLUSION Metformin does not interfere with the production of dehydroepiandrosterone sulfate. Besides, it decreases estradiol levels, basically through the reduction of testosterone. These hormonal changes might have clinical relevance.
Collapse
Affiliation(s)
- Carlo Campagnoli
- Unit of Endocrinological Gynecology, Ospedale Sant'Anna di Torino, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kinoshita T, Honma S, Shibata Y, Yamashita K, Watanabe Y, Maekubo H, Okuyama M, Takashima A, Takeshita N. An innovative LC-MS/MS-based method for determining CYP 17 and CYP 19 activity in the adipose tissue of pre- and postmenopausal and ovariectomized women using 13C-labeled steroid substrates. J Clin Endocrinol Metab 2014; 99:1339-47. [PMID: 24456285 DOI: 10.1210/jc.2013-3715] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Does adipose tissue produce steroid hormones like an endocrine organ? OBJECT To clarify whether adipose tissue produces sex steroid hormone like an endocrine organ, we estimated several key steroid hormone levels, as well as CYP17 and CYP19 activity, in ovariectomized, pre- and postmenopausal women by liquid chromatography-tandem mass spectrometry (LC-MS/MS). SUBJECTS AND METHODS The subjects were 19 premenopausal (n = 12), postmenopausal (n = 4), and ovariectomized women (n = 3) aged 27-68 years. Serum, visceral adipose and sc adipose samples were taken from these subjects and stored at -70°C. The levels of cortisol, cortisone, progesterone (Prog), androstenedione, dehydroepiandrosterone, estrone, estradiol (E2), and T in serum and adipose tissue were estimated simultaneously by LC-MS/MS. CYP17 and CYP19 activity in tissues were assayed with the use of (13)C-labeled steroid precursors and LC-MS/MS-based estimation of the metabolites. RESULTS E2 and Prog levels in the sera of postmenopausal or ovariectomized women were less than 10% of those in premenopausal women. No marked variations were seen in other hormones. Estrone, androstenedione, dehydroepiandrosterone, and Prog levels in the visceral and sc tissues of postmenopausal and ovariectomized women were 9-60 times higher than those in serum, whereas E2 and T levels were 3- to 7-fold higher than those in serum, and cortisol and cortisone levels were 20% of those found for serum. CYP17 in adipose tissue was found to have 17-hydroxylase and 20,17-lyase activity, with each catalytic activity being essentially equal. Therefore, CYP17 in adipose tissue is of the testicular/ovarian type but not adrenal type, which has 17-hydroxylase activity dominant. The presence of CYP19 activity in adipose tissue was approximately 3% of CYP17. CONCLUSION Our findings suggest that adipose tissue acts as an endocrine organ, with CYP17 and CYP19 activity playing an essential role in sex steroid hormone biosynthesis.
Collapse
Affiliation(s)
- Toshihiko Kinoshita
- Department of Obstetrics and Gynecology (T.K., A.T., N.T.), Toho University Medical Center Sakura Hospital, Chiba 285-8741, Japan; Department of Analytical Research (S.H., Y.W., H.M., M.O.), ASKA Pharma Medical Co Ltd, Kawasaki 213-8522, Japan; Department of Urology (Y.S.), Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; and Department of Clinical Analysis (K.Y.), Tohoku Pharmaceutical University, Sendai 981-8558, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Rižner TL. Estrogen biosynthesis, phase I and phase II metabolism, and action in endometrial cancer. Mol Cell Endocrinol 2013; 381:124-39. [PMID: 23911898 DOI: 10.1016/j.mce.2013.07.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 05/30/2013] [Accepted: 07/22/2013] [Indexed: 12/21/2022]
Abstract
Endometrial cancer is the most common gynecological malignancy in the developed World. Based on their histopathology, clinical manifestation, and epidemiology, the majority of endometrial cancer cases can be divided into two groups: the more prevalent type 1 which is associated with unopposed estrogen exposure; and the less common type 2, which is usually not associated with hyper-estrogenic factors. This manuscript overviews the published data on the expression of genes encoding the estrogen biosynthetic enzymes, the phase I and phase II estrogen metabolic enzymes, and the estrogen receptors in endometrial cancer, at the mRNA, protein and enzyme activity levels. The potential role of altered expression of these enzymes and receptors in cancerous versus control endometrial tissue, and the implication of estrogens in tumor initiation and promotion, are discussed. Finally, based on the published data, a model of estrogen metabolism and actions is proposed for pre-cancerous and cancerous endometrial tissue, and the role of the estrogens in the progression of endometrial cancer from endometrial hyperplasia is suggested.
Collapse
Affiliation(s)
- Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia.
| |
Collapse
|
31
|
Blevins JK, Coxworth JE, Herndon JG, Hawkes K. Brief communication: Adrenal androgens and aging: Female chimpanzees (Pan troglodytes) compared with women. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2013; 151:643-8. [PMID: 23818143 PMCID: PMC4412270 DOI: 10.1002/ajpa.22300] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 04/29/2013] [Indexed: 11/08/2022]
Abstract
Ovarian cycling continues to similar ages in women and chimpanzees yet our nearest living cousins become decrepit during their fertile years and rarely outlive them. Given the importance of estrogen in maintaining physiological systems aside from fertility, similar ovarian aging in humans and chimpanzees combined with somatic aging differences indicates an important role for nonovarian estrogen. Consistent with this framework, researchers have nominated the adrenal androgen dehydroepiandrosterone (DHEA) and its sulfate (DHEAS), which can be peripherally converted to estrogen, as a biomarker of aging in humans and other primates. Faster decline in production of this steroid with age in chimpanzees could help explain somatic aging differences. Here, we report circulating levels of DHEAS in captive female chimpanzees and compare them with published levels in women. Instead of faster, the decline is slower in chimpanzees, but from a much lower peak. Levels reported for other great apes are lower still. These results point away from slowed decline but toward increased DHEAS production as one of the mechanisms underlying the evolution of human longevity.
Collapse
Affiliation(s)
- James K. Blevins
- Department of Anthropology, University of Utah, Salt Lake City, UT
- Department of Biology, Salt Lake Community College, Salt Lake City, UT
| | | | - James G. Herndon
- Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Kristen Hawkes
- Department of Anthropology, University of Utah, Salt Lake City, UT
| |
Collapse
|
32
|
Postmenopausal breast cancer, androgens, and aromatase inhibitors. Breast Cancer Res Treat 2013; 139:1-11. [PMID: 23572296 DOI: 10.1007/s10549-013-2505-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/25/2013] [Indexed: 02/06/2023]
Abstract
Recent data can help to better define the long debated relationship between androgens and breast cancer (BC) after menopause. We reviewed the available literature data on: the origin of androgens after menopause, the association between circulating androgens and BC incidence and recurrence, the relationship between circulating and intratumoral hormones, the prognostic significance of the presence of androgen receptors (ARs) in the different BC subtypes, the androgen effect on BC cell lines, and the relationship between androgens and aromatase inhibitors. Epidemiological, clinical, and preclinical data on the role of androgens and of ARs on estrogen receptor (ER)-negative BC are somewhat controversial. However, most preclinical studies suggest that activated ARs, when present, have a proliferative effect, particularly in HER2 expressing cell lines, due to the cross-talk between AR and HER2 pathways. As regards ER-positive BC, epidemiological studies associate androgen levels with increased incidence and risk of recurrences, whilst clinical studies associate the AR positivity with a better prognosis. Preclinical studies suggest that the action of androgens is bidirectional: mainly proliferative, because circulating androgens are the precursors of estrogens, but also anti-proliferative, because AR activation restrains ER activity. The relative increase of androgenic action that follows the blocking of androgen aromatization into estrogens by aromatase inhibitors (AIs), could contribute to their therapeutic efficacy in AR-positive cases. Available data, although defining a complex picture, suggest that circulating androgen levels are clinically relevant, particularly when AIs are used.
Collapse
|
33
|
Matsumoto J, Ariyoshi N, Ishii I, Kitada M. Functional characterization of seven single-nucleotide polymorphisms of the steroid sulfatase gene found in a Japanese population. J Hum Genet 2013; 58:267-72. [PMID: 23466819 DOI: 10.1038/jhg.2013.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Steroid sulfatase (STS) is an enzyme that hydrolyzes steroid sulfates such as dehydroepiandrosterone sulfate (DHEA-S) and estrone sulfate. STS has a key role in the synthesis of steroid hormones in placenta and breast cancer cells. Recently, we have identified six novel single-nucleotide polymorphisms (SNPs) and one nonsynonymous SNP (V476M) in the STS gene in a Japanese population. To clarify the effects of SNPs in the 5'-flanking region or 5' untranslated region on transcriptional activity, a reporter gene assay was conducted. In addition, DHEA-S desulfatase activity of a variant (Met at codon 476)-type enzyme was compared with that of the wild (Wd)-type enzyme in COS-1 cells. The transcriptional activities were significantly decreased (155A) and increased (-2837A and -1588C) in MCF-7 cells. On the other hand, no significant difference was found in expression levels of STS protein or specific activities of DHEA-S desulfation between Wd and the variant enzymes. This is the first report on the effects of various SNPs in the STS gene detected in Japanese healthy subjects.
Collapse
Affiliation(s)
- Jun Matsumoto
- Department of Clinical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | | | | | | |
Collapse
|
34
|
Montalcini T, Migliaccio V, Ferro Y, Gazzaruso C, Pujia A. Androgens for postmenopausal women's health? Endocrine 2012; 42:514-20. [PMID: 22581204 DOI: 10.1007/s12020-012-9692-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 04/30/2012] [Indexed: 12/24/2022]
Abstract
Obesity, metabolic syndrome, and diabetes are becoming a leading health concern in the developed Countries, due to their link to cardiovascular disease. These conditions are common in women in the post-menopausal period. Unfortunately, actual lifestyle change strategy fail to prevent cardiovascular events for several reasons, thus specific medications are needed. In addition, it was showed an increased cardiovascular diseases and breast cancer risk in postmenopausal women taking estrogens alone or with progestin, thus the optimal therapy for the prevention of chronic disease in women is still lacking. Androgens exert different actions on organs like adipose tissue, brain, bone, and on cardiovascular system. However, a debate still exists on the positive role of androgens on human health, especially in women. Furthermore, the vascular effects of androgens remain poorly understood and have been controversial for a long time. Sex hormones are important determinants of body composition. Aging is, often, accompanied by a decrease in free testosterone levels, a concomitant reduction in muscle mass and an increase in fat mass. Furthermore, numerous studies showed that total serum testosterone levels were inversely related to the atherosclerosis disease incidence in postmenopausal women. New therapeutic targets may, therefore, arise understanding how androgen could influence the fat distribution, the metabolic disease onset, the vascular reactivity and cardiovascular risk, in both sex.
Collapse
Affiliation(s)
- Tiziana Montalcini
- Clinical Nutrition Unit, Department of Medical and Surgical Science, University Magna Graecia, Catanzaro, Italy.
| | | | | | | | | |
Collapse
|
35
|
Adrenarche and middle childhood. HUMAN NATURE-AN INTERDISCIPLINARY BIOSOCIAL PERSPECTIVE 2012; 22:327-49. [PMID: 22388879 DOI: 10.1007/s12110-011-9120-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Middle childhood, the period from 6 to 12 years of age, is defined socially by increasing autonomy and emotional regulation, somatically by the development of anatomical structures for subsistence, and endocrinologically by adrenarche, the adrenal production of dehydroepiandrosterone (DHEA). Here I suggest that DHEA plays a key role in the coordinated development of the brain and body beginning with middle childhood, via energetic allocation. I argue that with adrenarche, increasing levels of circulating DHEA act to down-regulate the release of glucose into circulation and hence limit the supply of glucose which is needed by the brain for synaptogenesis. Furthermore, I suggest the antioxidant properties of DHEA may be important in maintaining synaptic plasticity throughout middle childhood within slow-developing areas of the cortex, including the insula, thamalus, and anterior cingulate cortex. In addition, DHEA may play a role in the development of body odor as a reliable social signal of behavioral changes associated with middle childhood.
Collapse
|
36
|
Fujioka K, Kajita K, Wu Z, Hanamoto T, Ikeda T, Mori I, Okada H, Yamauchi M, Uno Y, Morita H, Nagano I, Takahashi Y, Ishizuka T. Dehydroepiandrosterone reduces preadipocyte proliferation via androgen receptor. Am J Physiol Endocrinol Metab 2012; 302:E694-704. [PMID: 22234368 DOI: 10.1152/ajpendo.00112.2011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Several studies have suggested that both testosterone and dehydroepiandrosterone (DHEA) have weight-reducing and antidiabetic effects, especially in rodent studies; however, the precise mechanism of their action remains unclear. Here, we investigated the effect of DHEA on cell growth in adipose tissue. The appearance of senescence-associated β-galactosidase in stromal vascular fraction (SVF) isolated from Otsuka Long-Evans Tokushima fatty rats, an animal model of inherent obese type 2 diabetes, was prevented by DHEA administration. Next, the effects of DHEA and testosterone were compared in vivo and in vitro to evaluate whether these hormones influence cell growth in adipose tissue. Both DHEA and testosterone reduced body weight and epididymal fat weight equivalently when administered for 4 wk. To assess the effect of DHEA and testosterone on cell growth in adipose tissue, 5-bromo-2'-deoxyuridine (BrdU) uptake by SVF was measured. Quantification analysis of BrdU uptake by examining DNA isolated from each SVF revealed that treatment with DHEA and testosterone reduced cell replication. These results indicated that DHEA- and testosterone-induced decreased adiposity was associated with reduced SVF growth. Incubation with DHEA and testosterone equally decreased BrdU uptake by 3T3-L1 preadipocytes. Pretreatment with the androgen receptor (AR) inhibitor flutamide, but not the estrogen receptor inhibitor fulvestrant, abolished these effects. Knockdown of AR with siRNA also inhibited DHEA-induced decreases in BrdU uptake. These results suggest that DHEA-induced growth suppression of preadipocytes is mediated via AR. Therefore, both DHEA and testosterone similarly decrease adipocyte growth possibly via a common mechanism.
Collapse
Affiliation(s)
- Kei Fujioka
- Dept. of General Internal Medicine, Gifu Univ. Graduate School of Medicine,Yanagido, Gifu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Tagawa N, Minamitani E, Yamaguchi Y, Kobayashi Y. Alternative mechanism for anti-obesity effect of dehydroepiandrosterone: possible contribution of 11β-hydroxysteroid dehydrogenase type 1 inhibition in rodent adipose tissue. Steroids 2011; 76:1546-53. [PMID: 21945397 DOI: 10.1016/j.steroids.2011.09.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 08/21/2011] [Accepted: 09/06/2011] [Indexed: 11/24/2022]
Abstract
Dehydroepiandrosterone (DHEA) has been suggested to have an anti-obesity effect; however, the mechanism underlying this effect remains unclear. The effect of DHEA on adipocytes opposes that of glucocorticoids, which potentiate adipogenesis. The key to the intracellular activation of glucocorticoids in adipocytes is 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which catalyses the production of active glucocorticoids (cortisol in humans and corticosterone in rodents) from an inactive 11-keto form (cortisone in humans and 11-dehydrocorticosterone in rodents). In humans and rodents, intracellular glucocorticoid reactivation is exaggerated in obese adipose tissue. Using differentiated 3T3-L1 adipocytes, we demonstrated that DHEA inhibited about 15.6% of 11β-HSD1 activity at a concentration of 1 μM within 10min. Inhibition was also observed in a cell-free system composed of microsomes prepared from rat adipose tissue and NADPH, a coenzyme of 11β-HSD1. A kinetic study revealed that DHEA acted as a non-competitive inhibitor of 11β-HSD1. Moreover, conversion from DHEA to estrogens was not observed by sensitive semi-micro HPLC equipped with electrochemical detector. These results indicate that the inhibition of 11β-HSD1 by DHEA depends on neither the transcriptional pathway nor the nonspecific manner. This is the first demonstration that the anti-obesity effect of DHEA is exerted by non-transcriptional inhibition of 11β-HSD1 in rodent adipocytes.
Collapse
Affiliation(s)
- Noriko Tagawa
- Department of Medical Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan.
| | | | | | | |
Collapse
|
38
|
Borcherding DC, Hugo ER, Idelman G, De Silva A, Richtand NW, Loftus J, Ben-Jonathan N. Dopamine receptors in human adipocytes: expression and functions. PLoS One 2011; 6:e25537. [PMID: 21966540 PMCID: PMC3180449 DOI: 10.1371/journal.pone.0025537] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 09/06/2011] [Indexed: 12/28/2022] Open
Abstract
Introduction Dopamine (DA) binds to five receptors (DAR), classified by their ability to increase (D1R-like) or decrease (D2R-like) cAMP. In humans, most DA circulates as dopamine sulfate (DA-S), which can be de-conjugated to bioactive DA by arylsulfatase A (ARSA). The objective was to examine expression of DAR and ARSA in human adipose tissue and determine whether DA regulates prolactin (PRL) and adipokine expression and release. Methods DAR were analyzed by RT-PCR and Western blotting in explants, primary adipocytes and two human adipocyte cell lines, LS14 and SW872. ARSA expression and activity were determined by qPCR and enzymatic assay. PRL expression and release were determined by luciferase reporter and Nb2 bioassay. Analysis of cAMP, cGMP, leptin, adiponectin and interleukin 6 (IL-6) was done by ELISA. Activation of MAPK and PI3 kinase/Akt was determined by Western blotting. Results DAR are variably expressed at the mRNA and protein levels in adipose tissue and adipocytes during adipogenesis. ARSA activity in adipocyte increases after differentiation. DA at nM concentrations suppresses cAMP, stimulates cGMP, and activates MAPK in adipocytes. Acting via D2R-like receptors, DA and DA-S inhibit PRL gene expression and release. Acting via D1R/D5R receptors, DA suppresses leptin and stimulates adiponectin and IL-6 release. Conclusions This is the first report that human adipocytes express functional DAR and ARSA, suggesting a regulatory role for peripheral DA in adipose functions. We speculate that the propensity of some DAR-activating antipsychotics to increase weight and alter metabolic homeostasis is due, in part, to their direct action on adipose tissue.
Collapse
Affiliation(s)
- Dana C. Borcherding
- Department of Cancer and Cell Biology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Eric R. Hugo
- Department of Cancer and Cell Biology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Gila Idelman
- Department of Cancer and Cell Biology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Anuradha De Silva
- Department of Cancer and Cell Biology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Nathan W. Richtand
- Department of Cancer and Cell Biology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Jean Loftus
- The Christ Hospital, Cincinnati, Ohio, United States of America
| | - Nira Ben-Jonathan
- Department of Cancer and Cell Biology, University of Cincinnati, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
39
|
Purohit A, Woo LWL, Potter BVL. Steroid sulfatase: a pivotal player in estrogen synthesis and metabolism. Mol Cell Endocrinol 2011; 340:154-60. [PMID: 21693170 DOI: 10.1016/j.mce.2011.06.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 06/14/2011] [Accepted: 06/15/2011] [Indexed: 02/07/2023]
Abstract
Steroid sulfatase plays a pivotal role in regulating the formation of biologically active steroids from inactive steroid sulfates. It is responsible for the hydrolysis of estrone sulfate and dehydroepiandrosterone sulfate to estrone and dehydroepiandrosterone, respectively, both of which can be subsequently reduced to steroids with estrogenic properties (i.e. estradiol and androstenediol) that can stimulate the growth of tumors in hormone-responsive tissues of the breast, endometrium and prostate. Hence, the action of steroid sulfatase is implicated in physiological processes and pathological conditions. It has been five years since our group last reviewed the important role of this enzyme in steroid synthesis and the progress made in the development of potent inhibitors of this important enzyme target. This timely review therefore concentrates on recent advances in steroid sulfatase research, and summarises the findings of clinical trials with Irosustat (BN83495), the only steroid sulfatase inhibitor that is being trialed in postmenopausal women with breast or endometrial cancer.
Collapse
Affiliation(s)
- Atul Purohit
- Oncology Drug Discovery Group, Section of Investigative Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, UK.
| | | | | |
Collapse
|
40
|
Suzuki T, Miki Y, Nakamura Y, Ito K, Sasano H. Steroid sulfatase and estrogen sulfotransferase in human carcinomas. Mol Cell Endocrinol 2011; 340:148-53. [PMID: 21073915 DOI: 10.1016/j.mce.2010.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2009] [Revised: 10/06/2010] [Accepted: 11/01/2010] [Indexed: 01/23/2023]
Abstract
Estrogens are closely involved in the development of hormone-dependent carcinomas. Estrone is locally produced from circulating inactive estrone sulfate by steroid sulfatase (STS), while estrone is inversely inactivated into estrone sulfate by estrogen sulfotransferase (EST). Recent studies suggested importance of this STS pathway in various human carcinomas. Therefore, in this review, we summarized recent results of STS and EST in several estrogen-dependent carcinomas. STS and EST expressions were detected in the breast and endometrial carcinomas, and activation of STS pathway due to increment in STS and/or decrement in EST expressions plays important role in their estrogen-dependent growth. STS expression was also reported in the ovarian and prostate carcinomas. STS/EST status was associated with intratumoral estrogen level in the colon carcinoma, and STS-negative/EST-positive colon carcinoma patients had longer survival. Therefore, STS pathway and estrogen actions may play an important role in the development of these carcinomas, and further investigations are required.
Collapse
Affiliation(s)
- Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University, Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | | | | | | | | |
Collapse
|
41
|
Hengstler JG, Foth H, Gebel T, Kramer PJ, Lilienblum W, Schweinfurth H, Völkel W, Wollin KM, Gundert-Remy U. Critical evaluation of key evidence on the human health hazards of exposure to bisphenol A. Crit Rev Toxicol 2011; 41:263-91. [PMID: 21438738 PMCID: PMC3135059 DOI: 10.3109/10408444.2011.558487] [Citation(s) in RCA: 225] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 01/19/2011] [Accepted: 01/25/2011] [Indexed: 01/08/2023]
Abstract
Despite the fact that more than 5000 safety-related studies have been published on bisphenol A (BPA), there seems to be no resolution of the apparently deadlocked controversy as to whether exposure of the general population to BPA causes adverse effects due to its estrogenicity. Therefore, the Advisory Committee of the German Society of Toxicology reviewed the background and cutting-edge topics of this BPA controversy. The current tolerable daily intake value (TDI) of 0.05 mg/kg body weight [bw]/day, derived by the European Food Safety Authority (EFSA), is mainly based on body weight changes in two- and three-generation studies in mice and rats. Recently, these studies and the derivation of the TDI have been criticized. After having carefully considered all arguments, the Committee had to conclude that the criticism was scientifically not justified; moreover, recently published additional data further support the reliability of the two- and three-generation studies demonstrating a lack of estrogen-dependent effects at and below doses on which the current TDI is based. A frequently discussed topic is whether doses below 5 mg/kg bw/day may cause adverse health effects in laboratory animals. Meanwhile, it has become clear that positive results from some explorative studies have not been confirmed in subsequent studies with higher numbers of animals or a priori defined hypotheses. Particularly relevant are some recent studies with negative outcomes that addressed effects of BPA on the brain, behavior, and the prostate in rodents for extrapolation to the human situation. The Committee came to the conclusion that rodent data can well be used as a basis for human risk evaluation. Currently published conjectures that rats are insensitive to estrogens compared to humans can be refuted. Data from toxicokinetics studies show that the half-life of BPA in adult human subjects is less than 2 hours and BPA is completely recovered in urine as BPA-conjugates. Tissue deconjugation of BPA-glucuronide and -sulfate may occur. Because of the extremely low quantities, it is only of minor relevance for BPA toxicity. Biomonitoring studies have been used to estimate human BPA exposure and show that the daily intake of BPA is far below the TDI for the general population. Further topics addressed in this article include reasons why some studies on BPA are not reproducible; the relevance of oral versus non-oral exposure routes; the degree to which newborns are at higher systemic BPA exposure; increased BPA exposure by infusions in intensive care units; mechanisms of action other than estrogen receptor activation; and the current regulatory status in Europe, as well as in the USA, Canada, Japan, New Zealand, and Australia. Overall, the Committee concluded that the current TDI for BPA is adequately justified and that the available evidence indicates that BPA exposure represents no noteworthy risk to the health of the human population, including newborns and babies.
Collapse
Affiliation(s)
- J G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), University of Dortmund, Dortmund, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wang F, Koskela A, Hämäläinen E, Turpeinen U, Savolainen-Peltonen H, Mikkola TS, Vihma V, Adlercreutz H, Tikkanen MJ. Quantitative determination of dehydroepiandrosterone fatty acyl esters in human female adipose tissue and serum using mass spectrometric methods. J Steroid Biochem Mol Biol 2011; 124:93-8. [PMID: 21316450 DOI: 10.1016/j.jsbmb.2011.01.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/26/2011] [Accepted: 01/27/2011] [Indexed: 10/18/2022]
Abstract
Dehydroepiandrosterone-fatty acyl esters (DHEA-FAE) are naturally occurring water-insoluble metabolites of DHEA, which are transported in plasma exclusively by lipoproteins. To find out whether DHEA, like estradiol, might be stored in adipose tissue in FAE form, we set up a mass spectrometric method to quantify DHEA-FAE and free DHEA in human adipose tissue and serum. The method consists of chromatographic purification steps and final determination of hydrolyzed DHEA-FAE and free DHEA, which was carried out by gas chromatography-mass spectrometry (GC-MS) or liquid chromatography-tandem mass spectrometry (LC-MS/MS). Our results showed that no detectable amounts of DHEA-FAE could be found in adipose tissue although 32-178 pmol/g of free DHEA were determined by GC-MS and LC-MS/MS. The DHEA-FAE concentrations in serum quantified by GC-MS were 1.4±0.7 pmol/ml in premenopausal women (n=7), and 0.9±0.4 pmol/ml in postmenopausal women (n=5). Correspondingly, the free DHEA concentrations were 15.2±6.3 pmol/ml and 6.8±3.0 pmol/ml. In addition, the mean proportions of DHEA-FAE of total DHEA (DHEA-FAE+free DHEA) in serum were 8.6% and 11.2% in pre- and postmenopausal women, respectively. Serum DHEA-FAE concentration was below quantification limit for LC-MS/MS (signal-to-noise ratio, S/N=10), while free DHEA concentrations varied between 5.8 and 23.2 pmol/ml. In conclusion, the proportion of DHEA-FAE of total DHEA in serum was approximately 9%. However, in contrast to our previous findings for estradiol fatty acid esters in adipose tissue which constituted about 80% of total estradiol (esterified+free), the proportion of DHEA-FAE of total DHEA was below 5%. Four to ten times higher concentrations of free DHEA were quantified in adipose tissue compared to those in serum.
Collapse
Affiliation(s)
- Feng Wang
- Institute of Clinical Medicine, Department of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wright JL, Kwon EM, Ostrander EA, Montgomery RB, Lin DW, Vessella R, Stanford JL, Mostaghel EA. Expression of SLCO transport genes in castration-resistant prostate cancer and impact of genetic variation in SLCO1B3 and SLCO2B1 on prostate cancer outcomes. Cancer Epidemiol Biomarkers Prev 2011; 20:619-27. [PMID: 21266523 DOI: 10.1158/1055-9965.epi-10-1023] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Metastases from men with castration-resistant prostate cancer (CRPC) harbor increased tumoral androgens versus untreated prostate cancers. This may reflect steroid uptake by OATP (organic anion transporting polypeptide)/SLCO transporters. We evaluated SLCO gene expression in CRPC metastases and determined whether prostate cancer outcomes are associated with single nucleotide polymorphisms (SNP) in SLCO2B1 and SLCO1B3, transporters previously shown to mediate androgen uptake. METHODS Transcripts encoding eleven SLCO genes were analyzed in untreated prostate cancer and in metastatic CRPC tumors obtained by rapid autopsy. SNPs in SLCO2B1 and SLCO1B3 were genotyped in a population-based cohort of 1,309 Caucasian prostate cancer patients. Median survival follow-up was 7.0 years (0.77-16.4). The risk of prostate cancer recurrence/progression and prostate cancer-specific mortality (PCSM) was estimated with Cox proportional hazards analysis. RESULTS Six SLCO genes were highly expressed in CRPC metastases versus untreated prostate cancer, including SLCO1B3 (3.6-fold; P = 0.0517) and SLCO2B1 (5.5-fold; P = 0.0034). Carriers of the variant alleles SLCO2B1 SNP rs12422149 (HR: 1.99; 95% CI: 1.11-3.55) or SLCO1B3 SNP rs4149117 (HR: 1.76; 95% CI: 1.00-3.08) had an increased risk of PCSM. CONCLUSIONS CRPC metastases show increased expression of SLCO genes versus primary prostate cancer. Genetic variants of SLCO1B3 and SLCO2B1 are associated with PCSM. Expression and genetic variation of SLCO genes which alter androgen uptake may be important in prostate cancer outcomes. IMPACT OATP/SLCO genes may be potential biomarkers for assessing risk of PCSM. Expression and genetic variation in these genes may allow stratification of patients to more aggressive hormonal therapy or earlier incorporation of nonhormonal-based treatment strategies.
Collapse
|
44
|
Brookes KJ, Hawi Z, Park J, Scott S, Gill M, Kent L. Polymorphisms of the steroid sulfatase (STS) gene are associated with attention deficit hyperactivity disorder and influence brain tissue mRNA expression. Am J Med Genet B Neuropsychiatr Genet 2010; 153B:1417-24. [PMID: 20862695 PMCID: PMC3132592 DOI: 10.1002/ajmg.b.31120] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 08/03/2010] [Indexed: 11/09/2022]
Abstract
Previous studies in animals and humans have implicated the X-chromosome STS gene in the etiology of attentional difficulties and attention deficit hyperactivity disorder (ADHD). This family based association study has fine mapped a region of the STS gene across intron 1 and 2 previously associated with ADHD, in an extended sample of 450 ADHD probands and their parents. Significant association across this region is demonstrated individually with 7 of the 12 genotyped SNPs, as well as an allele specific haplotype of the 12 SNPs. The over transmitted risk allele of rs12861247 was also associated with reduced STS mRNA expression in normal human post-mortem frontal cortex brain tissue compared to the non-risk allele (P = 0.01). These results are consistent with the hypothesis arising from previous literature demonstrating that boys with deletions of the STS gene, and hence no STS protein are at a significantly increased risk of developing ADHD. Furthermore, this study has established the brain tissue transcript of STS, which except from adipose tissue, differs from that seen in all other tissues investigated. © 2010 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- KJ Brookes
- Bute Medical School, University of St AndrewsSt Andrews, Scotland, UK
| | - Z Hawi
- Department of Psychiatry, Trinity Centre for Health Sciences, St James's HospitalDublin, Ireland
| | - J Park
- Bute Medical School, University of St AndrewsSt Andrews, Scotland, UK
| | - S Scott
- Bute Medical School, University of St AndrewsSt Andrews, Scotland, UK
| | - M Gill
- Department of Psychiatry, Trinity Centre for Health Sciences, St James's HospitalDublin, Ireland
| | - L Kent
- Bute Medical School, University of St AndrewsSt Andrews, Scotland, UK,*Correspondence to: L. Kent, Bute Medical School, University of St Andrews, St Andrews, Scotland KY16 9TS, UK. E-mail:
| |
Collapse
|
45
|
Rice SPL, Zhang L, Grennan-Jones F, Agarwal N, Lewis MD, Rees DA, Ludgate M. Dehydroepiandrosterone (DHEA) treatment in vitro inhibits adipogenesis in human omental but not subcutaneous adipose tissue. Mol Cell Endocrinol 2010; 320:51-7. [PMID: 20176080 DOI: 10.1016/j.mce.2010.02.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 01/07/2010] [Accepted: 02/10/2010] [Indexed: 01/26/2023]
Abstract
Dehydroepiandrosterone (DHEA), a precursor sex steroid, circulates in sulphated form (DHEAS). Serum DHEAS concentrations are inversely correlated with metabolic syndrome components and in vivo/in vitro studies suggest a role in modulating adipose mass. To investigate further, we assessed the in vitro biological effect of DHEA in white (3T3-L1) and brown (PAZ6) preadipocyte cell lines and human primary preadipocytes. DHEA (from 10(-8)M) caused concentration-dependent proliferation inhibition of 3T3-L1 and PAZ6 preadipocytes. Cell cycle analysis demonstrated unaltered apoptosis but indicated blockade at G1/S or G2/M in 3T3-L1 and PAZ6, respectively. Preadipocyte cell-line adipogenesis was not affected. In human primary subcutaneous and omental preadipocytes, DHEA significantly inhibited proliferation from 10(-8)M. DHEA 10(-7)M had opposing effects on adipogenesis in the two fat depots. Subcutaneous preadipocyte differentiation was unaffected or increased whereas omental preadipocytes showed significantly reduced adipogenesis. We conclude that DHEA exerts fat depot-specific differences which modulate body composition by limiting omental fat production.
Collapse
Affiliation(s)
- S P L Rice
- Centre for Endocrine and Diabetes Sciences, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | | | | | | | | | | | | |
Collapse
|
46
|
Dehydroepiandrosterone-Sulfate Modifies Human Fatty Acid Composition of Different Adipose Tissue Depots. Obes Surg 2010; 21:102-11. [DOI: 10.1007/s11695-009-0064-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 12/15/2009] [Indexed: 01/22/2023]
|
47
|
Nardi A, Pomari E, Zambon D, Belvedere P, Colombo L, Dalla Valle L. Transcriptional control of human steroid sulfatase. J Steroid Biochem Mol Biol 2009; 115:68-74. [PMID: 19429462 DOI: 10.1016/j.jsbmb.2009.02.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 02/19/2009] [Accepted: 02/24/2009] [Indexed: 10/21/2022]
Abstract
Steroid sulfatase (STS) is a membrane-bound microsomal enzyme that hydrolyzes various alkyl and aryl steroid sulfates, leading to the in situ formation of biologically active hormones. The entire human STS gene spans over approximately 200kbp of which the first 100kbp include the regulatory region, while the STS-coding region is located downstream. Previous studies indicated that STS expression, in different human tissues, could be regulated by at least six different promoters associated with alternative first exons. Here, we describe two new splicing patterns: the first, found in the prostatic cell line PC3, is based upon a partially coding new first exon (0d) that is spliced to a new second exon (1e). The second variant was found in the ovary and it is characterized by the novel splicing of the untranslated exon 0b to exon 0c, which is then spliced to the common exon 1b. We also report the results of a multiplex ligation-dependent probe amplification (RT-MLPA) analysis for the simultaneous detection, in qualitative and/or semi-quantitative terms, of the transcription patterns of STS in different tissues.
Collapse
|
48
|
Blouin K, Nadeau M, Mailloux J, Daris M, Lebel S, Luu-The V, Tchernof A. Pathways of adipose tissue androgen metabolism in women: depot differences and modulation by adipogenesis. Am J Physiol Endocrinol Metab 2009; 296:E244-55. [PMID: 18984855 DOI: 10.1152/ajpendo.00039.2008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The objective was to examine pathways of androgen metabolism in abdominal adipose tissue in women. Abdominal subcutaneous (SC) and omental (OM) adipose tissue samples were surgically obtained in women. Total RNA was isolated from whole adipose tissue samples and from primary preadipocyte cultures before and after induction of differentiation. Expression levels of several steroid-converting enzyme transcripts were examined by real-time RT-PCR. Androgen conversion rates were also measured. We found higher expression levels in SC compared with OM adipose tissue for type 1 3beta-hydroxysteroid dehydrogenase (3beta-HSD-1; P < 0.05), for aldo-keto reductase 1C3 (AKR1C3; P < 0.0001), for AKR1C2 (P < 0.0001), and for the androgen receptor (P < 0.0001). 17beta-HSD-2 mRNA levels were lower in SC adipose tissue (P < 0.05). Induction of adipocyte differentiation led to significantly increased expression levels in SC cultures for AKR1C3 (4.7-fold, P < 0.01), 11-cis-retinol dehydrogenase (6.9-fold, P < 0.02), AKR1C2 (5.6-fold, P < 0.004), P-450 aromatase (5.7-fold, P < 0.02), steroid sulfatase (3.1-fold, P < 0.02), estrogen receptor-beta (11.8-fold, P < 0.01), and the androgen receptor (4.0-fold, P < 0.0005). Generally similar but nonsignificant trends were obtained in OM cultures. DHT inactivation rates increased with differentiation, this effect being mediated by dexamethasone alone, through a glucocorticoid receptor-dependent mechanism. In conclusion, higher mRNA levels of enzymes synthesizing and inactivating androgens are found in differentiated adipocytes, consistent with higher androgen-processing rates in these cells. Glucocorticoid-induced androgen inactivation may locally modulate the exposure of adipose cells to active androgens.
Collapse
Affiliation(s)
- Karine Blouin
- Molecular Endocrinology and Oncology Research Ctr., Laval University Medical Research Ctr., 2705 Laurier Blvd. (T3-67 Québec, QC, Canada G1V 4G2
| | | | | | | | | | | | | |
Collapse
|
49
|
Dalla Valle L, Toffolo V, Nardi A, Fiore C, Armanini D, Belvedere P, Colombo L. The expression of the human steroid sulfatase-encoding gene is driven by alternative first exons. J Steroid Biochem Mol Biol 2007; 107:22-9. [PMID: 17601726 DOI: 10.1016/j.jsbmb.2007.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We have analyzed steroid sulfatase (STS) gene transcription in 10 human tissues: ovary, adrenal cortex, uterus, thyroid, liver, pancreas, colon, mammary gland, dermal papilla of the hair follicle, and peripheral mononuclear leukocytes. Overall, six different promoters were found to drive STS expression, giving rise to transcripts with unique first exons that were labeled 0a, 0b, 0c, 1a, 1c, and 1d, of which the last two and 0c are newly reported. All of them, except exon 1d, vary in length owing to the occurrence of multiple transcriptional start sites. While placental exon 1a is partially coding, the other five first exons are all untranslated. Three of these (0a, 0b, and 0c) are spliced to the common partially coding exon 1b, whereas the other two (1c and 1d) are spliced to the coding exon 2, which occurs in all transcripts. Whatever the ATG actually used, the differences are restricted to the signal peptide which is post-transcriptionally cleaved. Transcripts with exons 0a and 0b have the broadest tissue distribution, occurring, in 6 out of the 12 tissues so far investigated, while the other first exons are restricted to one or two tissues. The proximal promoter of each first exon was devoid of TATA box or initiator element and lacked consensus elements for transcription factors related to steroidogenesis, suggesting that regulatory sequences are probably placed at greater distance. In conclusion, the regulation of STS transcription appears to be more complex than previously thought, suggesting that this enzyme plays a substantial role in intercellular integration.
Collapse
Affiliation(s)
- Luisa Dalla Valle
- Comparative Endocrinology Laboratory, Department of Biology, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy.
| | | | | | | | | | | | | |
Collapse
|
50
|
Zaichuk T, Ivancic D, Scholtens D, Schiller C, Khan SA. Tissue-specific transcripts of human steroid sulfatase are under control of estrogen signaling pathways in breast carcinoma. J Steroid Biochem Mol Biol 2007; 105:76-84. [PMID: 17596930 DOI: 10.1016/j.jsbmb.2006.12.101] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Accepted: 12/18/2006] [Indexed: 11/30/2022]
Abstract
Steroid sulfatase (STS) increases the pool of precursors of biologically active steroids, thereby playing an important role in breast cancer development. Mechanisms that control STS expression remain poorly understood. In present study we investigated alterations in the 5' region of STS gene to gain insight into the mechanism(s) that regulates its expression in mammary epithelial cells. We found that at least four alternatively spliced transcripts of STS gene can be produced from at least four different leader exons. Distinct expression patterns of the STS variants were observed in human tissues. Expression profiles of estrogen receptor alpha (ERalpha)-positive and ERalpha-negative breast carcinomas showed that these two categories of tumors and their adjacent benign tissues display remarkably different expression of STS isoforms. Coexpression of STS isoforms with ER isotypes suggests their cell-type specific coregulation. In addition, we identified ERalpha as essential regulator of STS transcription and provide evidence of direct estradiol-dependent binding of ERalpha to multiple STS cis-regulatory regions in vivo. Our results indicate that STS isoforms are under control of estrogen signaling pathways and their differential expression may play a significant role in breast cancer biology.
Collapse
Affiliation(s)
- Tetiana Zaichuk
- Department of Surgery, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA.
| | | | | | | | | |
Collapse
|