1
|
Yang N, Zhang W, Ji C, Ge J, Zhang X, Li M, Wang M, Zhang T, He J, Zhu H. Metabolic alteration of circulating steroid hormones in women with gestational diabetes mellitus and the related risk factors. Front Endocrinol (Lausanne) 2023; 14:1196935. [PMID: 37396163 PMCID: PMC10310992 DOI: 10.3389/fendo.2023.1196935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Background Abnormally changed steroid hormones during pregnancy are closely related to the pathological process of gestational diabetes mellitus (GDM). Our aim was to systematically profile the metabolic alteration of circulating steroid hormones in GDM women and screen for risk factors. Methods This study was a case-control study with data measured from 40 GDM women and 70 healthy pregnant women during their 24-28 gestational weeks. 36 kinds of steroid hormones, including 3 kinds of corticosteroids, 2 kinds of progestins, 5 kinds of androgens and 26 kinds of downstream estrogens in serum were systematically measured using a combined sensitive UPLC-MS/MS method. The flux of different metabolic pathways of steroid hormones was analyzed. Logistic regression and ROC curve model analyses were performed to identify potential steroid markers closely associated with GDM development. Results Serum corticosteroids, progestins and almost all the estrogen metabolites via 16-pathway from parent estrogens were higher in GDM women compared with healthy controls. Most of the estrogen metabolites via 4-pathway and more than half of the metabolites via 2-pathway were not significantly different. 16α-hydroxyestrone (16OHE1), estrone-glucuronide/sulfate (E1-G/S) and the ratio of total 2-pathway estrogens to total estrogens were screened as three indicators closely related to the risk of GDM development. The adjusted odds ratios of GDM for the highest quartile compared with the lowest were 72.22 (95% CI 11.27-462.71, P trend <0.001) for 16OHE1 and 6.28 (95% CI 1.74-22.71, P trend <0.05) for E1-G/S. The ratio of 2-pathway estrogens to total estrogens was negatively associated with the risk of GDM. Conclusion The whole metabolic flux from cholesterol to downstream steroid hormones increased in GDM condition. The most significant changes were observed in the 16-pathway metabolism of estrogens, rather than the 2- or 4-pathway or other types of steroid hormones. 16OHE1 may be a strong marker associated with the risk for GDM.
Collapse
Affiliation(s)
- Na Yang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wei Zhang
- Nanjing Qlife Medical Technology Co., Ltd, Nanjing, Jiangsu, China
| | - Cheng Ji
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jiajia Ge
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xiaoli Zhang
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Meijuan Li
- Nanjing Qlife Medical Technology Co., Ltd, Nanjing, Jiangsu, China
| | - Min Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Tianqi Zhang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jun He
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Huaijun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
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: 1.0] [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
|
3
|
Wasniewski T, Woclawek-Potocka I. Altered expression of lysophosphatidic acid receptors, in association with the synthesis of estrogens and androgens in type 1 endometrial cancer biology. Gynecol Endocrinol 2018; 34:422-427. [PMID: 29182028 DOI: 10.1080/09513590.2017.1409707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
To establish association between two main lysophosphatidic acid (LPA) receptors (LPAR2 and LPAR1) with the synthesis of estrogens and androgens in type-1 endometrial carcinoma (EC), we evaluated correlation of LPARs expression with expression of steroid 5 alpha-reductase 2 - aromatase (SRD5A2), or cytochrome P450 family 19 subfamily A member 1-5α-reductase (CYP19A1) in EC. Moreover, we aimed to investigate SRD5A2 and CYP19A1 expression in type 1 endometrial cancer and normal endometrium with its correlation to selected clinicopathological features. The studied cancerous samples showed higher CYP19A1 and SRD5A2 expression comparing to normal endometria. We also documented positive correlations between LPAR1 and LPAR2 with responsible for proliferation SRD5A2 in EC tissue which suggests that intratumoral estrogen metabolism and synthesis are pivotal in endometrial carcinoma progression, with the involvement of LPA in this process. However, positive correlation between CYP19A1 and LPAR1 accounts for supporting role of LPA acting via LPAR1 in intratumoral DHT concentration and the ethiology of endometrial cancer progression. Also, owing to the highest expression of LPARs, CYP19A1 and SRD5A2 as well as their association with depth of myoinvasion and FIGO stage LPAR2 and LPAR1 seem to be the efficient candidate prognostic markers in the individual, targeted therapies for EC.
Collapse
Affiliation(s)
- Tomasz Wasniewski
- a Department of Gynecology and Obstetrics, Faculty of Medical Sciences , University of Warmia and Masuria , Olsztyn , Poland
| | - Izabela Woclawek-Potocka
- b Department of Gamete and Embryo Biology , Institute of Animal Reproduction and Food Research, Polish Academy of Sciences , Olsztyn , Poland
| |
Collapse
|
4
|
Audet-Delage Y, Villeneuve L, Grégoire J, Plante M, Guillemette C. Identification of Metabolomic Biomarkers for Endometrial Cancer and Its Recurrence after Surgery in Postmenopausal Women. Front Endocrinol (Lausanne) 2018; 9:87. [PMID: 29593653 PMCID: PMC5857535 DOI: 10.3389/fendo.2018.00087] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/23/2018] [Indexed: 11/24/2022] Open
Abstract
Endometrial cancer (EC) is the most frequent gynecological cancer in developed countries. Most EC occurs after menopause and is diagnosed as endometrioid (type I) carcinomas, which exhibit a favorable prognosis. In contrast, non-endometrioid (type II) carcinomas such as serous tumors have a poor prognosis. Our goal was to identify novel blood-based markers associated with EC subtypes and recurrence after surgery in postmenopausal women. Using mass spectrometry-based untargeted metabolomics, we examined preoperative serum metabolites among control women (n = 18) and those with non-recurrent (NR) and recurrent (R) cases of type I endometrioid (n = 24) and type II serous (n = 12) carcinomas. R and NR cases were similar with respect to pathological characteristics, body mass index, and age. A total of 1,592 compounds were analyzed including 14 different lipid classes. When we compared EC cases with controls, 137 metabolites were significantly different. A combination of spermine and isovalerate resulted in an age-adjusted area under the receiver-operating characteristic curve (AUCadj) of 0.914 (P < 0.001) for EC detection. The combination of 2-oleoylglycerol and TAG42:2-FA12:0 allowed the distinction of R cases from NR cases with an AUCadj of 0.901 (P < 0.001). Type I R cases were also characterized by much lower levels of bile acids and elevated concentrations of phosphorylated fibrinogen cleavage peptide, whereas type II R cases displayed higher levels of ceramides. The findings from our pilot study provide a detailed metabolomics study of EC and identify putative serum biomarkers for defining clinically relevant risk groups.
Collapse
Affiliation(s)
- Yannick Audet-Delage
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Lyne Villeneuve
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Jean Grégoire
- Gynecologic Oncology Service, CHU de Québec, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Marie Plante
- Gynecologic Oncology Service, CHU de Québec, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Chantal Guillemette
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy, Laval University, Québec, QC, Canada
- Canada Research Chair in Pharmacogenomi, Laval University, Québec, QC, Canada
- *Correspondence: Chantal Guillemette,
| |
Collapse
|
5
|
Ito K, Miki Y, Suzuki T, McNamara KM, Sasano H. In situ androgen and estrogen biosynthesis in endometrial cancer: focus on androgen actions and intratumoral production. Endocr Relat Cancer 2016; 23:R323-35. [PMID: 27287451 DOI: 10.1530/erc-15-0470] [Citation(s) in RCA: 18] [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: 05/17/2016] [Accepted: 06/10/2016] [Indexed: 01/20/2023]
Abstract
In situ estrogen biosynthesis is considered to play pivotal roles in the development and progression of human endometrial carcinoma. However, the biological roles of androgen have remained virtually unknown. Various epidemiological studies have revealed that elevated serum androgen levels are generally associated with an increased risk of developing endometrial carcinoma; however, studies directly examining androgens in carcinoma tissues are relatively rare and reviews summarizing this information are scarce. Therefore, we summarized recent studies on androgens in endometrial carcinoma, especially focusing androgen actions and in situ androgen biosynthesis. Among the enzymes required for local biosynthesis of androgen, 17β-hydroxysteroid dehydrogenase type 5 (conversion from androstenedione to testosterone) and 5α-reductase (reduction of testosterone to dihydrotestosterone (DHT)) are the principal enzymes involved in the formation of biologically most potent androgen, DHT. Both enzymes and androgen receptor were expressed in endometrial carcinoma tissues, and in situ production of DHT has been reported to exist in endometrial carcinoma tissues. However, testosterone is not only a precursor of DHT production, but also a precursor of estradiol synthesis, as a substrate of the aromatase enzyme. Therefore, aromatase could be another key enzyme serving as a negative regulator for in situ production of DHT by reducing amounts of the precursor. In an in vitro study, DHT was reported to exert antiproliferative effects on endometrial carcinoma cells. Intracrine mechanisms of androgens, the downstream signals of AR, which are directly related to anticancer progression, and the clinical significance of DHT-AR pathway in the patients with endometrial carcinoma have, however, not been fully elucidated.
Collapse
Affiliation(s)
- Kiyoshi Ito
- Department of Disaster Obstetrics and GynecologyInternational Research Institute of Disaster Science (IRIDeS), Tohoku University, Sendai, Japan Department of Disaster Obstetrics and GynecologyTohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Miki
- Department of Disaster Obstetrics and GynecologyInternational Research Institute of Disaster Science (IRIDeS), Tohoku University, Sendai, Japan
| | - Takashi Suzuki
- Department of Pathology and HistotechnologyTohoku University Graduate School of Medicine, Sendai, Japan
| | - Keely May McNamara
- Department of PathologyTohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of PathologyTohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
6
|
El-Gamal MI, Semreen MH, Foster PA, Potter BVL. Design, synthesis, and biological evaluation of new arylamide derivatives possessing sulfonate or sulfamate moieties as steroid sulfatase enzyme inhibitors. Bioorg Med Chem 2016; 24:2762-7. [PMID: 27143133 PMCID: PMC4896991 DOI: 10.1016/j.bmc.2016.04.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 01/15/2023]
Abstract
A series of new arylamide derivatives possessing terminal sulfonate or sulfamate moieties was designed and synthesized. The target compounds were tested for in vitro inhibitory effects against the steroid sulfatase (STS) enzyme in a cell-free assay system. The free sulfamate derivative 1j was the most active. It inhibited the enzymatic activity by 72.0% and 55.7% at 20 μM and 10 μM, respectively. Compound 1j was further tested for STS inhibition in JEG-3 placental carcinoma cells with high STS enzyme activity. It inhibited 93.9% of the enzyme activity in JEG-3 placental carcinoma cells at 20 μM with an efficacy near to that of the well-established drug STX64 as reference. At 10 μM, 1j inhibited 86.1% of the STS activity of JEG-3. Its IC50 value against the STS enzyme in JEG-3 cells was 0.421 μM. Thus, 1j represents an attractive new non-steroidal lead for further optimization.
Collapse
Affiliation(s)
- Mohammed I El-Gamal
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Medicinal Chemistry, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt.
| | - Mohammad H Semreen
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Paul A Foster
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2HT, United Kingdom.
| | - Barry V L Potter
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| |
Collapse
|
7
|
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: 1.8] [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
|
8
|
Rižner TL. The Important Roles of Steroid Sulfatase and Sulfotransferases in Gynecological Diseases. Front Pharmacol 2016; 7:30. [PMID: 26924986 PMCID: PMC4757672 DOI: 10.3389/fphar.2016.00030] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/03/2016] [Indexed: 01/08/2023] Open
Abstract
Gynecological diseases such as endometriosis, adenomyosis and uterine fibroids, and gynecological cancers including endometrial cancer and ovarian cancer, affect a large proportion of women. These diseases are estrogen dependent, and their progression often depends on local estrogen formation. In peripheral tissues, estrogens can be formed from the inactive precursors dehydroepiandrosterone sulfate and estrone sulfate. Sulfatase and sulfotransferases have pivotal roles in these processes, where sulfatase hydrolyzes estrone sulfate to estrone, and dehydroepiandrosterone sulfate to dehydroepiandrosterone, and sulfotransferases catalyze the reverse reactions. Further activation of estrone to the most potent estrogen, estradiol, is catalyzed by 17-ketosteroid reductases, while estradiol can also be formed from dehydroepiandrosterone by the sequential actions of 3β-hydroxysteroid dehydrogenase-Δ4-isomerase, aromatase, and 17-ketosteroid reductase. This review introduces the sulfatase and sulfotransferase enzymes, in terms of their structures and reaction mechanisms, and the regulation and different transcripts of their genes, together with the importance of their currently known single nucleotide polymorphisms. Data on expression of sulfatase and sulfotransferases in gynecological diseases are also reviewed. There are often unchanged mRNA and protein levels in diseased tissue, with higher sulfatase activities in cancerous endometrium, ovarian cancer cell lines, and adenomyosis. This can be indicative of a disturbed balance between the sulfatase and sulfotransferases enzymes, defining the potential for sulfatase as a drug target for treatment of gynecological diseases. Finally, clinical trials with sulfatase inhibitors are discussed, where two inhibitors have already concluded phase II trials, although so far with no convincing clinical outcomes for patients with endometrial cancer and endometriosis.
Collapse
Affiliation(s)
- Tea Lanišnik Rižner
- Faculty of Medicine, Institute of Biochemistry, University of Ljubljana Ljubljana, Slovenia
| |
Collapse
|
9
|
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: 3.9] [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
|
10
|
Sung CH, Im HJ, Park N, Kwon Y, Shin S, Ye DJ, Cho NH, Park YS, Choi HK, Kim D, Chun YJ. Induction of steroid sulfatase expression in PC-3 human prostate cancer cells by insulin-like growth factor II. Toxicol Lett 2013; 223:109-15. [PMID: 24055520 DOI: 10.1016/j.toxlet.2013.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 09/03/2013] [Accepted: 09/10/2013] [Indexed: 11/17/2022]
Abstract
Human steroid sulfatase (STS) plays an important role in regulating the formation of biologically active estrogens and may be a promising target for treating estrogen-mediated carcinogenesis. The molecular mechanism of STS gene expression, however, is still not clear. Growth factors are known to increase STS activity but the changes in STS expression have not been completely understood. To determine whether insulin-like growth factor (IGF)-II can induce STS gene expression, the effects of IGF-II on STS expression were studied in PC-3 human prostate cancer cells. RT-PCR and Western blot analysis showed that IGF-II treatment significantly increased the expression of STS mRNA and protein in concentration- and time-dependent manners. To understand the signaling pathway by which IGF-II induces STS gene expression, the effects of specific PI3-kinase/Akt and NF-κB inhibitors were determined. When the cells were treated with IGF-II and PI3-kinase/Akt inhibitors, such as LY294002, wortmannin, or Akt inhibitor IV, STS expression induced by IGF-II was significantly blocked. Moreover, we found that NF-κB inhibitors, such as MG-132, bortezomib, Bay 11-7082 or Nemo binding domain (NBD) binding peptide, also strongly prevented IGF-II from inducing STS gene expression. We assessed whether IGF-II activates STS promoter activity using transient transfection with a luciferase reporter. IGF-II significantly stimulated STS reporter activity. Furthermore, IGF-II induced expression of 17β-hydroxysteroid dehydrogenase (HSD) 1 and 3, whereas it reduced estrone sulfotransferase (EST) gene expression, causing enhanced estrone and β-estradiol production. Taken together, these results strongly suggest that IGF-II induces STS expression via a PI3-kinase/Akt-NF-κB signaling pathway in PC-3 cells and may induce estrogen production and estrogen-mediated carcinogenesis.
Collapse
Affiliation(s)
- Chul-Hoon Sung
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Suh BY, Jung JJ, Park N, Seong CH, Im HJ, Kwon Y, Kim D, Chun YJ. Induction of steroid sulfatase expression by tumor necrosis factor-α through phosphatidylinositol 3-kinase/Akt signaling pathway in PC-3 human prostate cancer cells. Exp Mol Med 2012; 43:646-52. [PMID: 21904110 DOI: 10.3858/emm.2011.43.11.073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Steroid sulfatase (STS) is responsible for the hydrolysis of aryl and alkyl steroid sulfates and has a pivotal role in regulating the formation of biologically active estrogens. STS may be considered a new promising drug target for treating estrogen-mediated carcinogenesis. However, the molecular mechanism of STS expression is not well-known. To investigate whether tumor necrosis factor (TNF)-α is able to regulate gene transcription of STS, we studied the effect of TNF-α on STS expression in PC-3 human prostate cancer cells. RT-PCR and Western blot analysis showed that TNF-α significantly induced the expression of STS mRNA and protein in a concentration- and time-dependent manner. Treatment with TNF-α resulted in a strong increase in the phosphorylation of Akt on Ser-473 and when cells were treated with phosphatidylinositol (PI) 3-kinase inhibitors such as LY294002 or wortmannin, or Akt inhibitor (Akt inhibitor IV), induction of STS mRNA expression by TNF-α was significantly prevented. Moreover, activation of Akt1 by expressing the constitutively active form of Akt1 increased STS expression whereas dominant-negative Akt suppressed TNF-α-mediated STS induction. We also found that TNF-α is able to increase STS mRNA expression in other human cancer cells such as LNCaP, MDA-MB-231, and MCF-7 as well as PC-3 cells. Taken together, our results strongly suggest that PI 3-kinase/Akt activation mediates induction of human STS gene expression by TNF-α in human cancer cells.
Collapse
Affiliation(s)
- Bo Young Suh
- College of Pharmacy, Chung-Ang University Seoul 156-756, Korea
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Synthesis and evaluation of analogues of estrone-3-O-sulfamate as potent steroid sulfatase inhibitors. Bioorg Med Chem 2012; 20:2506-19. [PMID: 22455789 DOI: 10.1016/j.bmc.2012.03.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 02/28/2012] [Accepted: 03/01/2012] [Indexed: 01/06/2023]
Abstract
Estrone sulfamate (EMATE) is a potent irreversible inhibitor of steroid sulfatase (STS). In order to further expand SAR, the compound was substituted at the 2- and/or 4-positions and its 17-carbonyl group was also removed. The following general order of potency against STS in two in vitro systems is observed for the derivatives: The 4-NO(2) > 2-halogens, 2-cyano > EMATE (unsubstituted)>17-deoxyEMATE > 2-NO(2) > 4-bromo>2-(2-propenyl), 2-n-propyl > 4-(2-propenyl), 4-n-propyl > 2,4-(2-propenyl)= 2,4-di-n-propyl. There is a clear advantage in potency to place an electron-withdrawing substituent on the A-ring with halogens preferred at the 2-position, but nitro at the 4-position. Substitution with 2-propenyl or n-propyl at the 2- and/or 4-position of EMATE, and also removal of the 17-carbonyl group are detrimental to potency. Three cyclic sulfamates designed are not STS inhibitors. This further confirms that a free or N-unsubstituted sulfamate group (H(2)NSO(2)O-) is a prerequisite for potent and irreversible inhibition of STS as shown by inhibitors like EMATE and Irosustat. The most potent derivative synthesized is 4-nitroEMATE (2), whose IC(50)s in placental microsomes and MCF-7 cells are respectively 0.8 nM and 0.01 nM.
Collapse
|
13
|
Woo LWL, Ganeshapillai D, Thomas MP, Sutcliffe OB, Malini B, Mahon MF, Purohit A, Potter BVL. Structure-activity relationship for the first-in-class clinical steroid sulfatase inhibitor Irosustat (STX64, BN83495). ChemMedChem 2011; 6:2019-34. [PMID: 21990014 PMCID: PMC3262147 DOI: 10.1002/cmdc.201100288] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 07/22/2011] [Indexed: 12/27/2022]
Abstract
Structure-activity relationship studies were conducted on Irosustat (STX64, BN83495), the first steroid sulfatase (STS) inhibitor to enter diverse clinical trials for patients with advanced hormone-dependent cancer. The size of its aliphatic ring was expanded; its sulfamate group was N,N-dimethylated, relocated to another position and flanked by an adjacent methoxy group; and series of quinolin-2(1H)-one and quinoline derivatives of Irosustat were explored. The STS inhibitory activities of the synthesised compounds were assessed in a preparation of JEG-3 cells. Stepwise enlargement of the aliphatic ring from 7 to 11 members increases potency, although a further increase in ring size is detrimental. The best STS inhibitors in vitro had IC50 values between 0.015 and 0.025 nM. Other modifications made to Irosustat were found to either abolish or significantly weaken its activity. An azomethine adduct of Irosustat with N,N-dimethylformamide (DMF) was isolated, and crystal structures of Irosustat and this adduct were determined. Docking studies were conducted to explore the potential interactions between compounds and the active site of STS, and suggest a sulfamoyl group transfer to formylglycine 75 during the inactivation mechanism.
Collapse
Affiliation(s)
- L W Lawrence Woo
- Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY (UK), Fax:(+44) 1225-386-114
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Ito K, Utsunomiya H, Niikura H, Yaegashi N, Sasano H. Inhibition of estrogen actions in human gynecological malignancies: new aspects of endocrine therapy for endometrial cancer and ovarian cancer. Mol Cell Endocrinol 2011; 340:161-7. [PMID: 21111773 DOI: 10.1016/j.mce.2010.11.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Revised: 11/10/2010] [Accepted: 11/10/2010] [Indexed: 10/18/2022]
Abstract
Endometrial carcinoma and possibly ovarian carcinoma are considered "estrogen-dependent tumors" in human gynecological malignancies. In endometrial carcinoma, the enzymes responsible for intratumoral estrogen metabolism and biosynthesis are different from those in human breast carcinoma, although both of them are considered "estrogen-dependent malignancies". Specific and effective endocrine treatment of endometrial carcinoma should be explored, although progestin agents have been widely used for a long time. Aromatase inhibitors, the most effective endocrine agents of breast carcinoma, retinoids, metabolites of vitamin A, and synthetic peroxisome proliferator-activated receptor (PPAR) gamma ligands, used for the treatment of insulin resistance in type II diabetes mellitus, may be the important candidates for possible endocrine treatment of endometrial carcinoma. In ovarian carcinoma, several clinical studies recently demonstrated that aromatase inhibitors had some therapeutic activity against recurrent ovarian carcinoma. However, at least at this juncture, further studies should be required to establish an aromatase inhibitor treatment as one form of endocrine therapy of ovarian carcinoma in future.
Collapse
Affiliation(s)
- Kiyoshi Ito
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan.
| | | | | | | | | |
Collapse
|
15
|
Woo LWL, Purohit A, Potter BVL. Development of steroid sulfatase inhibitors. Mol Cell Endocrinol 2011; 340:175-85. [PMID: 21238537 DOI: 10.1016/j.mce.2010.12.035] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 10/13/2010] [Accepted: 12/22/2010] [Indexed: 11/21/2022]
Abstract
Hydrolysis of biologically inactive steroid sulfates to unconjugated steroids by steroid sulfatase (STS) is strongly implicated in rendering estrogenic stimulation to hormone-dependent cancers such as those of the breast. Considerable progress has been made in the past two decades with regard to the discovery, design and development of STS inhibitors. We outline historical aspects of their development, cumulating in the discovery of the first clinical trial candidate STX64 (BN83495, Irosustat) and other sulfamate-based inhibitors. The development of reversible STS inhibitors and the design of dual inhibitors of both aromatase and STS is also discussed.
Collapse
Affiliation(s)
- L W Lawrence Woo
- Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | | | | |
Collapse
|
16
|
Steed HL, Chu QSC. Aromatase inhibition: a potential target for the management of recurrent or metastatic endometrial cancer by letrozole: more questions than answers? Expert Opin Investig Drugs 2011; 20:681-90. [PMID: 21413907 DOI: 10.1517/13543784.2011.566862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Endometrial cancer generally presents as early and resectable disease, but about 20% of patients present with either incurable or recurrent/metastatic disease. Patients with good performance status will be treated with hormonal agents, including progestins and tamoxifen, followed by cytotoxic chemotherapy. The options are restricted to hormonal agents for those with multiple comorbidities and older age. Therefore, there is a need to identify novel hormonal agents and other targeted therapeutics with improved therapeutic window in this setting. AREA COVERED Clinical trials of letrozole in localized and metastatic settings are reviewed. In the localized setting, limited by the small sample size, preliminary and conflicting clinical activities were observed. Despite the selection of Type I endometrial cancer, which is more estrogen-dependent for its growth, modest clinical activity was observed in the metastatic setting. Thus far, no biomarkers for efficacy have been identified. EXPERT OPINION Further understanding of the relevance of aromatase and estrogen receptor and their interplay with other growth pathways will be necessary to guide further development of letrozole. It is premature to declare letrozole a therapeutic option in recurrent/metastatic endometrial cancer.
Collapse
Affiliation(s)
- Helen L Steed
- Cross Cancer Institute, Department of Gynecological Oncology, Edmonton, AB, Canada
| | | |
Collapse
|
17
|
Pasqualini JR, Chetrite GS. Recent advances on the action of estrogens and progestogens in normal and pathological human endometrium. Horm Mol Biol Clin Investig 2010; 2:155-75. [DOI: 10.1515/hmbci.2010.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Accepted: 04/07/2010] [Indexed: 01/25/2023]
Abstract
AbstractHormonal control in the development of the normal endometrium is of the utmost importance. It is well established that the two main hormones involved in this process are estradiol and progesterone, which are also implicated in the pathological conditions concerning endometriosis and endometrial carcinoma. There are two types of endometrial carcinoma: type I which represents 80%–90% is hormone-dependent, whereas the remainder is type II and is hormone-independent. The endometrial tissue contains all the enzymatic systems in the formation and transformation of the various hormones, including aromatases, sulfatases, sulfotransferases, hydroxysteroid dehydrogenases, hydroxylases, and glucuronidases. It is interesting to note that increased sulfatase activity is correlated with severity of endometriosis. An increased sulfatase/sulfotransferase ratio represents a poor prognosis in patients with endometrial carcinoma. Treatment with hormone replacement therapy (estrogens+progestogens), as well as with tibolone, is most effective in protecting this tissue by climacteric alterations, owing to the significant decrease of ovarian hormones. In conclusion, enzymatic control can open appealing perspectives to protect this organ from possible pathological alterations.
Collapse
|
18
|
Gründker C, Günthert AR, Emons G. Hormonal heterogeneity of endometrial cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 630:166-88. [PMID: 18637491 DOI: 10.1007/978-0-387-78818-0_11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Endometrial cancer is the most common malignant tumor of the female genital tract in the developed world. Increasing evidence suggests that the majority of cases can be divided into two different types ofendometrial cancer based on clinico-pathological and molecular characteristics. Type I is associated with an endocrine milieu of estrogen predominance. These tumors are ofendometroid histology and develop from endometrial hyperplasia. They have good prognosis and are sensitive to endocrine treatment. Type II endometrial cancers are not associated with a history of unopposed estrogens and develop from the atrophic endometrium of elderly women. Mainly, they are of serous papillary or clear cell morphology, have a poor prognosis and do not react to endocrine treatment. Both types of endometrial cancer probably differ markedly with regard to the molecular mechanisms of transformation. The transition from normal endometrium to a malignant tumor is thought to involve a stepwise accumulation of alterations in cellular mechanisms leading to dysfunctional cell growth. This chapter reviews the current knowledge of the molecular mechanisms commonly associated with development of type I and type II endometrial cancer.
Collapse
Affiliation(s)
- Carsten Gründker
- Department of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| | | | | |
Collapse
|
19
|
Ito K, Utsunomiya H, Yaegashi N, Sasano H. Biological roles of estrogen and progesterone in human endometrial carcinoma--new developments in potential endocrine therapy for endometrial cancer. Endocr J 2007; 54:667-79. [PMID: 17785917 DOI: 10.1507/endocrj.kr-114] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Endometrial carcinoma is one of the most common female pelvic malignancies. It is well known that uterine endometrial cell proliferation is under the control of both estrogen and progesterone. In this review, results of the recent studies on the biosynthesis and action of estrogen and progestin in normal endometrium and its disorders will be summarized and the new aspects of hormonal therapies in the patients with endometrial carcinoma will be discussed including its future prospectives. We reported that the enzymes responsible for intratumoral estrogen metabolism and biosynthesis are markedly different between human breast and endometrial carcinoma, although both of them are considered "estrogen-dependent malignancies". In addition, the biological significance of Progesterone receptor (PR) isoforms is considered to differ between endometrial and breast carcinomas. Clinical data concerning Hormone replacement therapy (HRT) and estrogen-dependent cancer risk also support these findings. These basic and clinical findings help to understand the biology and provide the new knowledge for prevention, diagnosis and treatment of human endomerial carcinoma. Specific endocrine treatment of endometrial carcinoma should be explored in future, although aromatase inhibitors are the most effective endocrine treatments of estrogen-responsive breast carcinoma. Retinoid, metabolities of vitamin A, and synthetic peroxisome proliferator-activated receptor (PPAR) gamma ligands, which have been used for the treatment of insulin resistance in type II diabetes mellitus, may be the important candidates as drugs not only for prevention but also for possible endocrine treatment of endometrial carcinoma.
Collapse
MESH Headings
- Antineoplastic Agents, Hormonal/therapeutic use
- Aromatase/metabolism
- Aromatase/physiology
- Carcinoma, Endometrioid/drug therapy
- Carcinoma, Endometrioid/etiology
- Endometrial Neoplasms/drug therapy
- Endometrial Neoplasms/etiology
- Estrogen Replacement Therapy/adverse effects
- Estrogens/biosynthesis
- Estrogens/physiology
- Female
- Humans
- Hydroxysteroid Dehydrogenases/metabolism
- Models, Biological
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/etiology
- Progesterone/physiology
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/physiology
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/physiology
- Risk Factors
- Steryl-Sulfatase/metabolism
- Steryl-Sulfatase/physiology
- Sulfotransferases/metabolism
- Sulfotransferases/physiology
Collapse
Affiliation(s)
- Kiyoshi Ito
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | |
Collapse
|
20
|
Jackson T, Woo LWL, Trusselle MN, Chander SK, Purohit A, Reed MJ, Potter BVL. Dual aromatase-sulfatase inhibitors based on the anastrozole template: synthesis, in vitro SAR, molecular modelling and in vivo activity. Org Biomol Chem 2007; 5:2940-52. [PMID: 17728860 DOI: 10.1039/b707768h] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The synthesis and biological evaluation of a series of novel Dual Aromatase-Sulfatase Inhibitors (DASIs) are described. It is postulated that dual inhibition of the aromatase and steroid sulfatase enzymes, both responsible for the biosynthesis of oestrogens, will be beneficial in the treatment of hormone-dependent breast cancer. The compounds are based upon the Anastrozole aromatase inhibitor template which, while maintaining the haem ligating triazole moiety crucial for enzyme inhibition, was modified to include a phenol sulfamate ester motif, the pharmacophore for potent irreversible steroid sulfatase inhibition. Adaption of a synthetic route to Anastrozole was accomplished via selective radical bromination and substitution reactions to furnish a series of inhibitory aromatase pharmacophores. Linking these fragments to the phenol sulfamate ester moiety employed S(N)2, Heck and Mitsunobu reactions with phenolic precursors, from where the completed DASIs were achieved via sulfamoylation. In vitro, the lead compound, 11, had a high degree of potency against aromatase (IC(50) 3.5 nM), comparable with that of Anastrozole (IC(50) 1.5 nM) whereas, only moderate activity against steroid sulfatase was found. However, in vivo, 11 surprisingly exhibited potent dual inhibition. Compound 11 was modelled into the active site of a homology model of human aromatase and the X-ray crystal structure of steroid sulfatase.
Collapse
Affiliation(s)
- Toby Jackson
- Medicinal Chemistry, Department of Pharmacy and Pharmacology and Sterix Ltd, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | | | | | | | | | | | | |
Collapse
|
21
|
Woo LWL, Bubert C, Sutcliffe OB, Smith A, Chander SK, Mahon MF, Purohit A, Reed MJ, Potter BVL. Dual aromatase-steroid sulfatase inhibitors. J Med Chem 2007; 50:3540-60. [PMID: 17580845 DOI: 10.1021/jm061462b] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
By introducting the steroid sulfatase inhibitory pharmacophore into aromatase inhibitor 1 (YM511), two series of single agent dual aromatase-sulfatase inhibitors (DASIs) were generated. The best DASIs in vitro (JEG-3 cells) are 5, (IC50(aromatase) = 0.82 nM; IC50(sulfatase) = 39 nM), and 14, (IC50(aromatase) = 0.77 nM; IC50(sulfatase) = 590 nM). X-ray crystallography of 5, and docking studies of selected compounds into an aromatase homology model and the steroid sulfatase crystal structure are presented. Both 5 and 14 inhibit aromatase and sulfatase in PMSG pretreated adult female Wistar rats potently 3 h after a single oral 10 mg/kg dose. Almost complete dual inhibition is observed for 5 but the levels were reduced to 85% (aromatase) and 72% (sulfatase) after 24 h. DASI 5 did not inhibit aldosterone synthesis. The development of a potent and selective DASI should allow the therapeutic potential of dual aromatase-sulfatase inhibition in hormone-dependent breast cancer to be assessed.
Collapse
Affiliation(s)
- L W Lawrence Woo
- Medicinal Chemistry, Department of Pharmacy & Pharmacology and Sterix Limited, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ito K. Hormone replacement therapy and cancers: the biological roles of estrogen and progestin in tumorigenesis are different between the endometrium and breast. TOHOKU J EXP MED 2007; 212:1-12. [PMID: 17464097 DOI: 10.1620/tjem.212.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Hormone replacement therapy (HRT) has become available over the past few decades, but the risk of breast cancer with HRT remains controversial. The Women's Health Initiative Study has recently demonstrated that women receiving estrogen plus progestin (HRT) have an increased risk of invasive breast carcinoma, although women receiving estrogen alone (estrogen replacement therapy) exhibit no increased risk of breast carcinoma. By contrast, the risk of endometrial carcinoma increases with estrogen replacement therapy, while HRT reduces the risk of endometrial carcinoma. These clinical findings suggest that the biological roles of estrogen and progestin in tumorigenesis are certainly different between the endometrium and breast, although both are considered "estrogen-dependent tissues". In this review, I summarize the recent studies and indicate that the enzymes responsible for intratumoral estrogen metabolism and biosynthesis are markedly different between human breast and endometrial carcinomas. 17beta-hydroxysteroid dehydrogenases (17-HSDs) are enzymes estrogen replacement therapyinvolved in the formation of active sex steroids. Estrogens are interconverted by two enzymes, 17-HSD types 1 and 2. Type 1 converts estrone to estradiol, and type 2 catalyzes the reverse reaction. 17-HSD type 5 reduces androstenedione to testosterone. 17-HSD type 1 plays an important role in the regulation of high estradiol levels in breast carcinoma tissues, whereas 17-HSD types 2 and 5 appear to be essential for the maintenance of estradiol concentrations in endometrial carcinoma tissues. In addition, the biological significance of progesterone receptor isoforms differs between endometrial and breast carcinomas. These findings may provide new insights into the biology of "estrogen-dependent tissues".
Collapse
Affiliation(s)
- Kiyoshi Ito
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
23
|
Nakamura Y, Suzuki T, Fukuda T, Ito A, Endo M, Moriya T, Arai Y, Sasano H. Steroid sulfatase and estrogen sulfotransferase in human prostate cancer. Prostate 2006; 66:1005-12. [PMID: 16541422 DOI: 10.1002/pros.20426] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Estrogen sulfotransferase (EST) and steroid sulfatase (STS) are known to be involved in in situ estrogen production in estrogen dependent human cancer such as breast cancer, but unknown in prostate cancer. MATERIALS AND METHODS We first examined whether these enzymes above were expressed and actually involved in estrogen production and metabolism in prostate cancer cell lines (LNCaP, DU-145, and PC-3). We than examined the expression of EST and STS in human prostate cancer tissues obtained from surgery (n = 52) using immunohistochemistry. RESULTS mRNAs of both enzymes were detected in all prostate cancer cell lines examined, and the synthesis of estrone (E(1)) and estradiol (E(2)) was also confirmed in these cell lines. In addition, STS immunoreactivity was detected in 44 cases (85%) and EST in 39 cases (75%), respectively. CONCLUSIONS STS and EST are expressed and may be involved in local production and metabolism of estrogens in human prostate cancers.
Collapse
Affiliation(s)
- Yasuhiro Nakamura
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Ito K, Utsunomiya H, Suzuki T, Saitou S, Akahira JI, Okamura K, Yaegashi N, Sasano H. 17Beta-hydroxysteroid dehydrogenases in human endometrium and its disorders. Mol Cell Endocrinol 2006; 248:136-40. [PMID: 16406263 DOI: 10.1016/j.mce.2005.11.038] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In situ estrogen metabolism and synthesis have been considered to play a very important role in the development and progression of human endometrial carcinoma. 17Beta-hydroxysteroid dehydrogenases (17-HSDs) are enzymes involved in the formation of active sex steroids, including testosterone, estrone (E1) and estradiol (E2). Estrogens are interchanged by two enzymes, 17-HSD types 1 and 2, type 1 converts E1 to E2, and type 2 does reverse actions. 17-HSD type 5 catalyzes the reduction of androstenedione to testosterone. 17-HSD type 2 expression was decreased through normal endometrium, hyperplasia and carcinoma accordingly. There was a significant inverse correlation between intratumoral E2 concentration and the level of 17-HSD type 2 mRNA in endometrial carcinoma. 17-HSD type 5 expression was significantly increased through normal endometrium, hyperplasia and carcinoma accordingly. These results indicated that 17-HSD types 2 and 5 play an important role in the regulation of in situ estrogen production in endometrial carcinoma.
Collapse
Affiliation(s)
- Kiyoshi Ito
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kríz L, Bicíková M, Hill M, Hampl R. Steroid sulfatase and sulfuryl transferase activity in monkey brain tissue. Steroids 2005; 70:960-9. [PMID: 16157357 DOI: 10.1016/j.steroids.2005.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Revised: 06/29/2005] [Accepted: 07/06/2005] [Indexed: 10/25/2022]
Abstract
Dehydroepiandrosterone and its sulfated form are commonly known as modulators of gamma-aminobutyrate A and N-methyl-D-aspartate receptors. In spite of poor permeability of the blood-brain barrier for sulfated steroids, high concentrations of dehydroepiandrosterone and also its sulfate have been found in brain tissue. Physiological concentrations of these neuromodulators are maintained by two enzymes present in the blood and many peripheral tissues, including the brain, namely, steroid sulfatase and neurosteroid sulfuryl transferase (NSST). This prompted us to investigate activities of these enzymes in primate brain tissue. Rather low neurosteroid sulfuryl transferase activity was detectable in in vitro incubations of cytosol fractions from male and female Macaca mulatta brains, dissected to cerebral cortex, subcortex, and cerebellum. In male monkeys, the highest activity was found in the cerebellum followed by cortex and subcortex. On the other hand, in female monkeys, the highest activity was determined in the cortex followed by subcortex and cerebellum. Steroid sulfatase activity was determined in in vitro microsomal samples from each of the above-mentioned brain regions. Specific activities in female cerebral regions declined in the order: cerebellum, cortex, and subcortex. In male monkeys, no significant difference among the studied regions was observed. Using dehydroepiandrosterone sulfate as a substrate, the apparent kinetic characteristics of steroid sulfatase were determined as follows: K(M) 36.10 +/- 8.33 microM, V(max) 8.38 +/- 1.68 nmol/h/mg protein. These results will serve as a basis for further studies concerning the pathophysiology of human brain tumors.
Collapse
Affiliation(s)
- Lubomír Kríz
- Institute of Endocrinology, Národní 8, 116 94 Prague 1, Czech Republic
| | | | | | | |
Collapse
|
26
|
Ishibashi H, Suzuki T, Suzuki S, Moriya T, Kaneko C, Nakata T, Sunamori M, Handa M, Kondo T, Sasano H. Estrogen Inhibits Cell Proliferation throughIn situProduction in Human Thymoma. Clin Cancer Res 2005; 11:6495-504. [PMID: 16166425 DOI: 10.1158/1078-0432.ccr-04-2495] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We showed previously estrogen receptor (ER) alpha as an independent prognostic marker in human thymoma. Estrogen sulfotransferase (EST), steroid sulfatase (STS), 17beta-hydroxysteroid dehydrogenase (17beta-HSD), and aromatase are considered to play important roles in hormone metabolism of estrogen-dependent tumors. EXPERIMENTAL DESIGN We examined estrogen production using primary cultures of human thymoma epithelial cells (TEC), intratumoral estradiol (E(2)) concentrations, and status of these enzymes above using immunohistochemistry or semiquantitative reverse transcription-PCR. We then correlated these findings with clinicopathologic variables and/or clinical outcome in 132 patients. RESULTS E(2) inhibited cell proliferation via ERalpha in TEC, which synthesized estrone and E(2). Intratumoral E(2) concentrations were inversely correlated with EST, positively correlated with STS or 17beta-HSD type 1, and significantly higher in lower-grade or early-stage thymoma. EST status was positively correlated with tumor size, clinical stage, histologic differentiation, and Ki-67 labeling index and significantly associated with adverse clinical outcome and turned out to be a potent independent prognostic factor. STS and/or 17beta-HSD type 1 status was inversely correlated with Ki-67 labeling index and associated with lower histologic grade or early clinical stages. CONCLUSIONS E(2) inhibits proliferation of TEC through ERalpha, which suggests that E(2) may be effective in treatment of thymoma, especially inoperable tumor, possibly through suppressing its cell proliferation activity. EST status is a potent prognostic factor in thymoma through inactivating estrogens. In situ estrogen synthesis through intracrine mechanism therefore may play important roles in tumorigenesis and/or development of thymoma through regulation of cell proliferation in an intracrine manner.
Collapse
|
27
|
Wang LQ, James MO. Sulfotransferase 2A1 forms estradiol-17-sulfate and celecoxib switches the dominant product from estradiol-3-sulfate to estradiol-17-sulfate. J Steroid Biochem Mol Biol 2005; 96:367-74. [PMID: 16011896 DOI: 10.1016/j.jsbmb.2005.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Accepted: 05/09/2005] [Indexed: 11/24/2022]
Abstract
Using recombinant sulfotransferases (SULTs) expressed in E. coli, beta-estradiol (E2) sulfonation was examined to determine which SULT enzyme is responsible for producing E2-17-sulfate (E2-17-S). SULTs 1A1*1, 1A1*2, 1A3, 1E1 and 2A1 all sulfated E2 to varying extents. No activity was observed with SULT1B1. Among the SULTs studied, SULT2A1 produced primarily E2-3-sulfate (E2-3-S), but also some E2-17-S and trace amounts of E2 disulfate. SULT2A1 had a K(m) value of 1.52 microM for formation of E2-3-S and 2.95 microM for formation of E2-17-S. SULT2A1 had the highest V(max) of 493 pmol/min/mg protein for formation of E2-3-S, which was 8.8- and 47-fold higher than the maximal rates of formation of E2-17-S and E2 disulfate, respectively. SULT2A1 formed E2-3-S more efficiently. However, when celecoxib (0-160 microM) was included in the incubation with either SULT2A1 or human liver cytosol, sulfonation switched from E2-3-S to E2-17-S in a concentration-dependent manner. The ratio of E2-17-S/E2-3-S went up to 15 with SULT2A1, and was saturated at 1 with human liver cytosol. In both cases, more E2-17-S was formed, with the unreacted E2 remained unchanged, suggesting celecoxib probably bound to a separate effector site to cause a conformational change in SULT2A1, which favored production of E2-17-S. The ability of celecoxib to alter the position of sulfonation of E2 may in part explain its success in the experimental prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Li-Quan Wang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | | |
Collapse
|
28
|
Segawa T, Shozu M, Murakami K, Kasai T, Shinohara K, Nomura K, Ohno S, Inoue M. Aromatase Expression in Stromal Cells of Endometrioid Endometrial Cancer Correlates with Poor Survival. Clin Cancer Res 2005; 11:2188-94. [PMID: 15788666 DOI: 10.1158/1078-0432.ccr-04-1859] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE AND EXPERIMENTAL DESIGN To assess the prognostic significance of intratumoral aromatase in endometrioid endometrial cancer, sections from 55 patients with endometrial cancer were evaluated for expression of aromatase using immunohistochemistry, and the correlation between aromatase expression and clinicopathologic parameters were analyzed. RESULTS Immunohistochemical staining for aromatase was positive for 32 (58%), 20 (36%), and 19 (34%) patients in cancer epithelial cells, stromal cells, and myometrial cells around the flank invasion, respectively. In situ hybridization also detected aromatase mRNA in all three types of cells. RT-PCR analysis revealed that aromatase mRNA was 2.5 +/- 1.0 amol/mug total RNA (mean +/- SE; n = 7) in tumor tissue. Western blot analysis detected the expected aromatase protein size of 58 kDa in cancer tissues more abundantly than in cancer-free endometrium (n = 3). The immunoreactivity in stromal cells correlated positively with advanced surgical stage and poor survival. Survival analysis revealed that the immunoreactivity of stromal cells was a significant prognostic factor, independent of histologic grade, muscular invasion, and lymph node metastasis, but dependent on surgical stage. By contrast, the immunoreactivity of aromatase both in cancer epithelial cells and myometrial cells did not correlate with prognosis. CONCLUSIONS To the best of our knowledge, this is the first evidence associating intratumoral aromatase expression in stromal cells and poor survival in endometrioid endometrial cancer. This positive linkage indicates that local expression of aromatase plays a role in tumor progression through the formation of in situ estrogens. In situ expression of aromatase may offer a potential target for management of endometrial cancers.
Collapse
Affiliation(s)
- Tomoya Segawa
- Department of Obstetrics and Gynecology, Kanazawa University School of Medicine, Kanazawa 920-0934, Japan
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Pasqualini JR, Chetrite GS. Recent insight on the control of enzymes involved in estrogen formation and transformation in human breast cancer. J Steroid Biochem Mol Biol 2005; 93:221-36. [PMID: 15860265 DOI: 10.1016/j.jsbmb.2005.02.007] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The great majority of breast cancers are in their early stage hormone-dependent and it is well accepted that estradiol (E2) plays an important role in the genesis and evolution of this tumor. Human breast cancer tissues contain all the enzymes: estrone sulfatase, 17beta-hydroxysteroid dehydrogenase, aromatase involved in the last steps of E2 bioformation. Sulfotransferases which convert estrogens into the biologically inactive estrogen sulfates are also present in this tissue. Quantitative data show that the 'sulfatase pathway', which transforms estrogen sulfates into the bioactive unconjugated E2, is 100-500 times higher than the 'aromatase pathway', which converts androgens into estrogens. The treatment of breast cancer patients with anti-aromatases is largely developed with very positive results. However, the formation of E2 via the 'sulfatase pathway' is very important in the breast cancer tissue. In recent years it was found that antiestrogens (e.g. tamoxifen, 4-hydroxytamoxifen), various progestins (e.g. promegestone, nomegestrol acetate, medrogestone, dydrogesterone, norelgestromin), tibolone and its metabolites, as well as other steroidal (e.g. sulfamates) and non-steroidal compounds, are potent sulfatase inhibitors. In another series of studies, it was found that E2 itself has a strong anti-sulfatase action. This paradoxical effect of E2 adds a new biological response of this hormone and could be related to estrogen replacement therapy in which it was observed to have either no effect or to decrease breast cancer mortality in postmenopausal women. Interesting information is that high expression of steroid sulfatase mRNA predicts a poor prognosis in patients with +ER. These progestins, as well as tibolone, can also block the conversion of estrone to estradiol by the inhibition of the 17beta-hydroxysteroid dehydrogenase type I (17beta-HSD-1). High expressison of 17beta-HSD-1 can be an indicator of adverse prognosis in ER-positive patients. It was shown that nomegestrol acetate, medrogestone, promegestone or tibolone, could stimulate the sulfotransferase activity for the local production of estrogen sulfates. This is an important point in the physiopathology of this disease, as it is well known that estrogen sulfates are biologically inactive. A possible correlation between this stimulatory effect on sulfotransferase activity and breast cancer cell proliferation is presented. In agreement with all this information, we have proposed the concept of selective estrogen enzyme modulators (SEEM). In conclusion, the blockage in the formation of estradiol via sulfatase, or the stimulatory effect on sulfotransferase activity in combination with anti-aromatases can open interesting and new possibilities in clinical applications in breast cancer.
Collapse
MESH Headings
- 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors
- 17-Hydroxysteroid Dehydrogenases/genetics
- 17-Hydroxysteroid Dehydrogenases/metabolism
- Biotransformation
- Breast/enzymology
- Breast/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Enzyme Inhibitors/therapeutic use
- Estrogen Antagonists/therapeutic use
- Estrogens/metabolism
- Female
- Humans
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/enzymology
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Norpregnenes/therapeutic use
- Progesterone Congeners/therapeutic use
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Receptors, Estrogen/metabolism
- Sulfatases/antagonists & inhibitors
- Sulfatases/genetics
- Sulfatases/metabolism
Collapse
Affiliation(s)
- Jorge R Pasqualini
- Hormones and Cancer Research Unit, Institut de Puériculture, 26 Boulevard Brune, 75014 Paris, France.
| | | |
Collapse
|
30
|
Nakamura Y, Suzuki T, Sasano H. Estrogen actions and in situ synthesis in human vascular smooth muscle cells and their correlation with atherosclerosis. J Steroid Biochem Mol Biol 2005; 93:263-8. [PMID: 15860269 DOI: 10.1016/j.jsbmb.2004.12.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Various epidemiological studies have demonstrated a relatively low incidence of cardiovascular events in premenopausal women and its marked increment after menopause. In addition, estrogens have been postulated to exert direct anti-atherogenic effects via binding to estrogen receptors (ERs) in vascular smooth muscle cells (VSMCs). However, not all postmenopausal women develop atherosclerosis despite decreased levels of serum estrogen. Therefore, it is considered important to examine the status of estrogen metabolism in situ and of ER expression in the human cardiovascular system. Estrone sulfate (E1S) is a major circulating plasma estrogen that is converted into the biologically active estrogen, estrone (E1) by steroid sulfatase (STS). E1 is also sulfated and reverted into E1S by estrogen sulfotransferase (EST). These two enzymes have recently been shown to play important roles in the in situ estrogen actions of estrogen-dependent human tissues. STS and EST, however, have not been studied in detail in the human vascular system associated with atherosclerotic changes. Therefore, the relative abundance of STS- and EST-immunoreactive protein and mRNA expression in human aorta were evaluated using immunohistochemistry and reverse transcription followed by quantitative polymerase chain reaction in addition to enzyme activity. Furthermore, we evaluated the relative abundance of messenger RNA (mRNA) of both ER subtypes (ERalpha and ERbeta) in the human aorta using reverse transcription followed by quantitative polymerase chain reaction (RT-qPCR), as well as the immunoreactivity of both ERs in VSMCs of human atherosclerotic lesions. STS expression levels were found to be significantly higher in the VSMCs obtained from female aortas with mild atherosclerotic changes than in those with severe atherosclerotic changes and in male aortas regardless of atherosclerotic changes. EST expression levels in the VSMCs of these aortas, however, were significantly higher in female aortas with severe atherosclerotic changes and in male aortas than in female aortas with mild atherosclerotic changes. In addition, the number of ERalpha and/or ERbeta double positive cells in the neointima was higher in female aortas with a mild degree of atherosclerosis than in female aortas with severe atherosclerosis. They indicate that both abundance of these estrogen-metabolizing enzymes in female aorta and relative levels of ER in VSMCs of female neointima may be associated with the status of atherosclerotic changes.
Collapse
Affiliation(s)
- Yasuhiro Nakamura
- Department of Pathology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | |
Collapse
|
31
|
Maitoko K, Sasaki H. Gonadotropin-releasing hormone agonist inhibits estrone sulfatase expression of cystic endometriosis in the ovary. Fertil Steril 2004; 82:322-6. [PMID: 15302278 DOI: 10.1016/j.fertnstert.2003.12.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2003] [Revised: 12/18/2003] [Accepted: 12/18/2003] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To clarify the inhibitory effect of GnRH agonist on estrone (E(1)) sulfatase expression. DESIGN Retrospective immunohistochemical study. SETTING The Jikei University Hospital, Tokyo, Japan. PATIENT(S) Thirty-three women who had undergone cystectomy of the ovary or oophorectomy and were proved histopathologically to have cystic endometriosis in the ovary. INTERVENTION(S) Fifteen of the 33 patients were treated with GnRH agonists monthly for 2-6 months before surgery. The other 18 patients did not receive any hormonal therapy. Tissue sections were immunostained with an anti-E(1) sulfatase monoclonal antibody (KM1049) originating from human placenta. MAIN OUTCOME MEASURE(S) Microscopic evaluation to assess the presence and localization of E(1) sulfatase and to describe any variations in its expression with or without treatment with GnRH agonist. RESULT(S) Immunostaining showed that E(1) sulfatase was localized only on the glandular epithelial cells of cystic endometriosis in the ovary. The immunostaining with anti-E(1) sulfatase proved that GnRH agonist inhibited E(1) sulfatase expression in the cystic endometriosis in the ovary. CONCLUSION(S) Gonadotropin-releasing hormone agonist inhibits E(1) sulfatase expression in cystic endometriosis in the ovary.
Collapse
Affiliation(s)
- Kazuhiro Maitoko
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nihonbashi, Minato-ku, Tokyo 105-8461, Japan
| | | |
Collapse
|
32
|
Abstract
There is substantial evidence that mammary cancer tissue contains all the enzymes responsible for the local biosynthesis of estradiol (E2) from circulating precursors. Two principal pathways are implicated in the final steps of E2 formation in breast cancer tissue: the 'aromatase pathway' that transforms androgens into estrogens and the 'sulfatase pathway' that converts estrone sulfate (E1S) into estrone (E1) via estrone sulfatase. The final step is the conversion of weak E1 to potent biologically active E2 via reductive 17beta-hydroxysteroid dehydrogenase type 1 activity. It is also well established that steroid sulfotransferases, which convert estrogens into their sulfates, are present in breast cancer tissues. One of the possible means of blocking E2 effects in breast cancer is to use anti-estrogens, which act by binding to the estrogen receptor (ER). Another option is to block E2 using anti-enzymes (anti-sulfatase, anti-aromatase, or anti-17beta-hydroxysteroid dehydrogenase (17beta-HSD). Various progestins (e.g. promegestone, nomegestrol acetate, medrogestone, 17-deacetyl norgestimate, dydrogesterone and its 20-dihydro derivative), as well as tibolone and its metabolites, have been shown to inhibit estrone sulfatase and 17beta-hydroxysteroid dehydrogenase. Some progestins and tibolone can also stimulate sulfotransferase activity. These various progestins may therefore provide a new option for the treatment of breast cancer.
Collapse
Affiliation(s)
- J R Pasqualini
- Hormones and Cancer Research Unit, Institut de Puériculture, 26 Blvd. Brune, Paris 75014, France.
| |
Collapse
|
33
|
Nakamura Y, Miki Y, Suzuki T, Nakata T, Darnel AD, Moriya T, Tazawa C, Saito H, Ishibashi T, Takahashi S, Yamada S, Sasano H. Steroid sulfatase and estrogen sulfotransferase in the atherosclerotic human aorta. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:1329-39. [PMID: 14507642 PMCID: PMC1868299 DOI: 10.1016/s0002-9440(10)63492-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/18/2003] [Indexed: 11/19/2022]
Abstract
Various epidemiological studies have demonstrated a relatively low incidence of cardiovascular events in premenopausal women and its marked increment after menopause. In addition, estrogens have been postulated to exert direct anti-atherogenic effects via binding to estrogen receptors in vascular smooth muscle cells (VSMCs). However, not all postmenopausal women develop atherosclerosis despite decreased levels of serum estrogen. Therefore, we believe it is important to examine the status of estrogen metabolism in situ in the human cardiovascular system. Estrone sulfate (E1S) is a major circulating plasma estrogen that is converted into the biologically active estrogen, estrone (E1) by steroid sulfatase (STS). E1 is also sulfated and reverted into E1S by estrogen sulfotransferase (EST). These two enzymes have recently been shown to play important roles in the in situ estrogen actions of estrogen-dependent human tissues and various sex steroid-dependent tumors. STS and EST, however, have not been studied in detail in the human vascular system associated with atherosclerotic changes. In the present study, we evaluated the relative abundance of STS- and EST-immunoreactive protein and mRNA expression in human aorta using immunohistochemistry and reverse transcription followed by quantitative polymerase chain reaction in addition to enzyme activity. STS expression levels were found to be significantly higher in the VSMCs obtained from female aortas with mild atherosclerotic changes than in those with severe atherosclerotic changes and in male aortas regardless of atherosclerotic changes. EST expression levels in the VSMCs of these aortas, however, were significantly higher in female aortas with severe atherosclerotic changes and in male aortas than in female aortas with mild atherosclerotic changes. We believe it is important to examine factors regulating the expression and activity of these estrogen-metabolizing enzymes in the human aorta. Various cytokines have been proposed to function as regulators of these enzymes in other tissues. In the present study, we studied the effects of interleukin (IL)-1beta, known to be produced in human atherosclerotic lesions, on the expression of these enzymes using cultured human VSMCs originally obtained from a female patient. IL-1beta markedly inhibited the expression of STS mRNA and enzyme activity, but stimulated the expression of EST mRNA and enzyme activity. In addition, IL-1beta also reduced E2 production from E1S and E1 in VSMCs. Results from the present study seem to suggest that the expression levels of both STS and EST mRNA and activity may be significantly associated with the degree of atherosclerotic changes in the female aorta, which may be related to cytokines produced in situ, such as IL-1beta, in human atherosclerotic lesions.
Collapse
MESH Headings
- Adult
- Aged
- Aorta/drug effects
- Aorta/enzymology
- Aortic Diseases/enzymology
- Arteriosclerosis/enzymology
- Arylsulfatases/genetics
- Arylsulfatases/metabolism
- Cells, Cultured
- Computer Systems
- Female
- Humans
- Immunohistochemistry/methods
- In Vitro Techniques
- Interleukin-1/pharmacology
- Male
- Middle Aged
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Staining and Labeling
- Steryl-Sulfatase
- Sulfotransferases/genetics
- Sulfotransferases/metabolism
Collapse
Affiliation(s)
- Yasuhiro Nakamura
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Woo LWL, Sutcliffe OB, Bubert C, Grasso A, Chander SK, Purohit A, Reed MJ, Potter BVL. First dual aromatase-steroid sulfatase inhibitors. J Med Chem 2003; 46:3193-6. [PMID: 12852749 DOI: 10.1021/jm034033b] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Aromatase inhibitors in clinical use block the biosynthesis of estrogens. Hydrolysis of estrone 3-sulfate by steroid sulfatase is an important additional source of tumor estrogen, and blockade of both enzymes should provide a more effective endocrine therapy. Sulfamoylated derivatives of the aromatase inhibitor YM511 inhibited sulfatase and aromatase in JEG-3 cells with respective IC(50) values of 20-227 and 0.82-100 nM (cf. letrozole, 0.89 nM). One dual inhibitor was potent against both enzymes in vivo, validating the concept.
Collapse
Affiliation(s)
- L W Lawrence Woo
- Medicinal Chemistry, Department of Pharmacy and Pharmacology and Sterix Ltd., University of Bath, Claverton Down, Bath BA2 7AY, UK
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Berstein LM, Tchernobrovkina AE, Gamajunova VB, Kovalevskij AJ, Vasilyev DA, Chepik OF, Turkevitch EA, Tsyrlina EV, Maximov SJ, Ashrafian LA, Thijssen JHH. Tumor estrogen content and clinico-morphological and endocrine features of endometrial cancer. J Cancer Res Clin Oncol 2003; 129:245-9. [PMID: 12695909 DOI: 10.1007/s00432-003-0427-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2002] [Accepted: 02/19/2003] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To compare estrogen concentrations in endometrial cancer tissue with those in macroscopically normal endometrium and with certain morphological characteristics of the tumor and endocrine parameters in patients. METHODS The estradiol content was evaluated by radioimmunoassay after homogenization and extraction in 78 adenocarcinomas (61 from postmenopausal patients). RESULTS Higher concentrations of estradiol in tumor tissue samples than in macroscopically normal endometrium were found in patients of both reproductive and postmenopausal age. This difference was the same in patients with either endometrial carcinoma type I or type II. No association between tumor steroid receptor levels, estradiol concentrations in blood serum, and timing of menopause with intratumoral estradiol contents was discovered. Estradiol concentrations in tumor tissues correlated positively with the clinical stage of disease and rate of tumor invasion (in patients with peripheric/lower type of fat topography), and negatively with tumor differentiation stage (in patients with central/upper type of fat topography) and the percentage of intact double-stranded DNA in normal endometrium. CONCLUSIONS Tumor estrogen content in endometrial cancer has clinical significance that is modified in the presence of certain endocrine characteristics related to insulin resistance. The role of local estrogen production (aromatase activity) in this setting deserves special study.
Collapse
Affiliation(s)
- L M Berstein
- Lab. Oncoendocrinology, N.N. Petrov Research Institute of Oncology, Pesochny-2, 197758, St. Petersburg, Russia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Miki Y, Nakata T, Suzuki T, Darnel AD, Moriya T, Kaneko C, Hidaka K, Shiotsu Y, Kusaka H, Sasano H. Systemic distribution of steroid sulfatase and estrogen sulfotransferase in human adult and fetal tissues. J Clin Endocrinol Metab 2002; 87:5760-8. [PMID: 12466383 DOI: 10.1210/jc.2002-020670] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Estrogens play a key role in various target tissues. Enzymes involved in the biosynthesis and metabolism of these sex steroids also regulate estrogenic actions in these tissues. Estrone sulfate (E1S) is a major circulating plasma estrogen that is converted into the biologically active estrogen, estrone (E1), by steroid sulfatase (STS). E1 is also sulfated and reverted into E1S by estrogen sulfotransferase (EST). These two enzymes have recently been shown to play important roles in the in situ estrogen actions of various sex steroid-dependent human tumors. However, the distribution of STS and EST in normal adult and fetal human tissues remains largely unknown. Therefore, in this study, in addition to examining the tissue distribution of both STS and EST mRNA in human adult and fetal tissues using RT followed by quantitative PCR, we studied the activity of these enzymes using (3)H-labeled E1/E1S as substrates in the homogenates of various human adult tissues. We also examined the localization of STS and EST protein in human adult and fetal tissues using immunohistochemistry, and that of EST mRNA in the adult kidney using laser dissection microscopy and PCR. STS mRNA, enzyme activity, and immunoreactivity were either absent or detected at very low levels in all adult and fetal tissues examined in this study. EST mRNA expression, however, was detected in all of the tissues examined, except for adult spleen and pancreas. EST enzyme activities were consistent with those of mRNA expression in the great majority of the tissues examined. Marked EST immunoreactivity was detected in hepatocytes, adrenal gland (adult, zona fasciculate to the reticularis; fetus, fetal zone), and epithelial cells of the gastrointestinal tract, smooth muscle cells of the tunica media in aorta, Leydig cells of the testis, and syncytiotrophoblast of the placenta. Patterns of EST immunolocalization were similar between adult and fetal human tissues, but EST immunoreactivity was detected in the urinary tubules of adult kidney, whereas in the fetal kidney, it was localized in the interstitial cells surrounding the urinary tubules. In the adult kidney, the presence of EST mRNA was also confirmed in the cells of urinary tubules using laser dissection microscopy and RT-PCR. Although the number of human tissues available for examination in this study was limited, our results suggest that between the enzymes involved in estrogen activation or inactivation, EST and not STS is the more widely expressed enzyme in various peripheral tissues in humans. We speculate that EST may play an important role in protecting peripheral tissues from possible excessive estrogenic effects.
Collapse
Affiliation(s)
- Yasuhiro Miki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Immunohistologic Localization of Estrone Sulfatase in Uterine Endometrium and Adenomyosis. Obstet Gynecol 2001. [DOI: 10.1097/00006250-200111000-00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Woo LL, Purohit A, Malini B, Reed MJ, Potter BV. Potent active site-directed inhibition of steroid sulphatase by tricyclic coumarin-based sulphamates. CHEMISTRY & BIOLOGY 2000; 7:773-91. [PMID: 11033081 DOI: 10.1016/s1074-5521(00)00023-5] [Citation(s) in RCA: 137] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND There is now abundant evidence that inhibition of steroid sulphatase alone or in conjunction with inhibition of aromatase may enhance the response of postmenopausal patients with hormone-dependent breast cancer to this type of endocrine therapy. Additionally, sulphatase inhibition has been proposed to be of potential therapeutic benefit in the immune system and for neuro-degenerative diseases. After the finding that our first highly potent active site-directed steroid sulphatase inhibitor, oestrone-3-O-sulphamate (EMATE), was highly oestrogenic, we proposed non-steroidal coumarin sulphamates such as 4-methylcoumarin-7-O-sulphamate (COUMATE) as alternative non-steroidal steroid sulphatase inhibitors. In this work, we describe how tricyclic coumarin-based sulphamates have been developed which are even more potent than COUMATE, are non-oestrogenic and orally active. We also discuss potential mechanisms of action. RESULTS 4-Ethyl- (4), 4-(n-propyl)- (6), 3-ethyl-4-methyl- (8), 4-methyl-3-(n-propyl)coumarin-7-O-sulphamate (11); the tricyclic derivatives 665COUMATE (13), 666COUMATE (15), 667COUMATE (17), 668COUMATE (20) and the tricyclic oxepin sulphamate (22) were synthesised. In a placental microsome preparation, all of these analogues were found to be more active than COUMATE in the inhibition of oestrone sulphatase, with the most potent inhibitor being 667COUMATE which has an IC(50) of 8 nM, some 3-fold lower than that for EMATE (25 nM). In addition, 667COUMATE was also found to inhibit DHEA-sulphatase some 25-fold more potently than EMATE in a placental microsome preparation. Like EMATE, 667COUMATE acts in a time- and concentration-dependent manner, suggesting that it is an active site-directed inhibitor. However, in contrast to EMATE, 667COUMATE has the important advantage of not being oestrogenic. In addition, we propose several diverse mechanisms of action for this active site-directed steroid sulphatase inhibitor in the light of recent publications on the crystal structures of human arylsulphatases A and B and the catalytic site topology for the hydrolysis of a sulphate ester. CONCLUSIONS A highly potent non-steroidal, non-oestrogenic and irreversible steroid sulphatase inhibitor has been developed. Several mechanisms of action for an active site-directed steroid sulphatase inhibitor are proposed. With 667COUMATE now in pre-clinical development for clinical trial, this should allow the biological and/or clinical significance of steroid sulphatase inhibitors in the treatment of postmenopausal women with hormone-dependent breast cancer and other therapeutic indications to be fully evaluated.
Collapse
Affiliation(s)
- L L Woo
- Department of Pharmacy and Pharmacology and Sterix Ltd., University of Bath, UK
| | | | | | | | | |
Collapse
|
40
|
Chetrite GS, Cortes-Prieto J, Philippe JC, Wright F, Pasqualini JR. Comparison of estrogen concentrations, estrone sulfatase and aromatase activities in normal, and in cancerous, human breast tissues. J Steroid Biochem Mol Biol 2000; 72:23-7. [PMID: 10731634 DOI: 10.1016/s0960-0760(00)00040-6] [Citation(s) in RCA: 223] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In the present study, the concentrations of estrone (E(1)), estradiol (E(2)) and their sulfates (E(1)S and E(2)S), as well as the sulfatase and aromatase activities, were evaluated in post-menopausal patients with breast cancer. Comparative studies of the evaluation of these parameters were carried out in (a) tumor tissue, (b) areas surrounding the tumor, and (c) areas distant from the tumor (glandular tissue) which were considered as normal tissue. The levels (in pm/g; mean +/- SEM) were: for E(1) in the (a) area: 320+/-95; in (b): 232+/-86; and in (c): 203+/-71; for E(2) in the (a) area: 388+/-106; in (b): 224+/-48; and in (c): 172+/-80; for E(1)S in the (a) area: 454+/-110; in (b): 259+/-90; and in (c): 237+/-65; for E(2)S in the (a) area:318+/-67; in (b): 261+/-72; and in (c): 232+/-75, respectively. The values of E(1)S and E(2) were significantly higher in the tumor tissue than in the area considered as normal. In all the tissues studied, the sulfatase activity was much higher than aromatase (130-200). In addition, the sulfatase levels were significantly higher in the peripheral and in the tumor tissue than in the area considered as normal. The levels of aromatase were significantly higher in tumoral than in normal tissue. The present data extend the "intracrine concept" for breast cancer tumors. The physiopathology and clinical significance as promoter parameters in breast cancer is to be explored.
Collapse
Affiliation(s)
- G S Chetrite
- Hormones and Cancer Research Unit, 26 and 45 Blvd Brune, 75014, Paris, France
| | | | | | | | | |
Collapse
|
41
|
Saeki T, Takashima S, Sasaki H, Hanai N, Salomon DS. Localization of Estrone Sulfatase in Human Breast Carcinomas. Breast Cancer 1999; 6:331-337. [PMID: 11091739 DOI: 10.1007/bf02966449] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We generated anti-human E1-STS monoclonal antibodies to localize estrone sulfatase (E1-STS) in human breast carcinomas. In particular, we examined the MCF-7 clone E3, ZR-75-1, MDA-MB 231, and MDA-MB-468 breast cancer cell lines and 25 breast carcinomas by either immunohistochemistry or Western blotting analysis. Simultaneously, we analyzed histological data, estrogen receptor (ER) status, progesterone receptor (PgR) status and epidermal growth factor receptor (EGFR) in breast tissue. All were surgical specimens from female patients. Nine of 25 carcinomas were obtained from premenopausal women, and 16 carcinomas were obtained frompostmenopausal women. All cell lines demonstrated positive staining for E1-STS.Interestingly, fine granulated staining of E1-STS on the cell membrane was observed. In addition, Western blotting analysis detected a 65 kD protein with an E1-STS specific band in all breast cancer cell lines regardless of the presence orabsence of E2. Twenty-two of 25 (88.0%) carcinomas showed positive staining forE1-STS, whereas negative staining was observed in the interstitial tissue surrounding tumors. In the premenopausal patients, 8 of 10 carcinomas (80.0%) showed positive staining for E1-STS, whereas 14 of 15 carcinomas (93.3%) revealed positive staining in the postmenopausal patients. The frequency of E1-STS expression was relatively higher in postmenopausal patients than in premenopausal patients but not statistically significant. The intensity of immunostaining for E1-STS depended upon the size of the tumor (NS). There was no correlation between E1-STS expression and other parameters. This evidence suggests E1-STS expression may beinvolved in the development of breast cancer. Further studies are necessary to clarify the relationship between E1-STS expression and prognostic factors. Immunoreactive E1-STS may be localized in cancer cells but not in surrounding tissuesin breast cancer.
Collapse
Affiliation(s)
- T Saeki
- Department of Hematology and Oncology, National Cancer Hospital East, 6-5-1 Kashiwanoha, Kashiwa 277-8577, Japan
| | | | | | | | | |
Collapse
|
42
|
Misao R, Nakanishi Y, Fujimoto J, Tamaya T. Effects of danazol and progesterone on sex hormone-binding globulin mRNA expression in human endometrial cancer cell line Ishikawa. J Steroid Biochem Mol Biol 1997; 62:321-5. [PMID: 9408086 DOI: 10.1016/s0960-0760(97)00045-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To ascertain one of the biological effects of danazol and progesterone on the uterine endometrial cancer cell line, Ishikawa, we investigated the effects of these steroids on sex hormone-binding globulin (SHBG) mRNA expression by competitive reverse transcription-polymerase chain reaction-Southern blot analysis (RT-PCR-SBA). Estradiol-17beta (E2) in any concentration given did not exert any significant effect on the expression of SHBG mRNA. Danazol and progesterone significantly (P < 0.05) suppressed the expression of SHBG mRNA dose-dependently starting at a concentration of 10(-6) and 10(-8) M, respectively. Progesterone, in a low concentration (10[-10] M) with E2 (10[-8] M), significantly (P < 0.05) increased the expression of SHBG mRNA, but danazol did not. In contrast, danazol and progesterone in high concentrations (10[-6] to 10[-5] M) with E2 (10[-8] M) significantly (P < 0.05) suppressed its expression. The time course study showed the time-dependent decrease of SHBG mRNA level by danazol and progesterone (10[-6] M) with or without E2 (10[-8] M), except for a temporal increase by progesterone. These findings suggest that danazol and progesterone in a superphysiological milieu down-regulate the intracellular SHBG-related steroidal actions, and that progesterone in a physiological milieu with estrogen up-regulates it in a hormone-dependent cell line. A decrease of intracellular SHBG caused by high-dose danazol or progesterone might partly contribute to the abolition of the intracellular estrogen-dominant milieu, and be related to the inhibition of estrogen-dependent growth of some endometrial cancer cells.
Collapse
Affiliation(s)
- R Misao
- Department of Obstetrics and Gynecology, Gifu University School of Medicine, Japan
| | | | | | | |
Collapse
|
43
|
Park IH, Han BK, Jo DH. Distribution and characterization of neurosteroid sulfatase from the bovine brain. J Steroid Biochem Mol Biol 1997; 62:315-20. [PMID: 9408085 DOI: 10.1016/s0960-0760(97)00042-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We investigated the regional and subcellular distribution of neurosteroid sulfatase (NSS) in the bovine brain and its enzymatic properties by using dehydroepiandrosterone sulfate (DHEA-S) as a substrate. Bovine NSS was highly concentrated in the region of the midbrain and in the hypothalamus. The enzyme was found to be a microsomal enzyme. The optimal temperature of the enzyme was 50 degrees C, which was slightly lower than that of other steroid sulfatases. The optimal pH of bovine NSS was 7.4 with a second optimum at pH 4.0. The second optimal pH of 4.0 was the most characteristic property of bovine NSS. Employing DHEA-S as the substrate, apparent Km and Vmax values were 113 +/- 21 microM and 4.1 +/- 0.4 nmol/mg protein/h, respectively, whereas Km and Vmax values were found to be 1.6 +/- 0.2 M and 1.9 +/- 0.3 micromol/mg protein/h with p-nitrophenyl sulfate (NP-S) as the substrate. NSS has thus been shown to have a higher affinity for the steroid sulfate than the phenolic compound. When DHEA-S was used as the substrate, pregnenolone sulfate (Preg-S) was a competitive inhibitor with an apparent Ki value of 46 microM, and NP-S was a non-competitive inhibitor (apparent Ki=12 mM).
Collapse
Affiliation(s)
- I H Park
- Department of Biotechnology, College of Engineering, Ajou University, Suwon, Republic of Korea (South)
| | | | | |
Collapse
|
44
|
Chetrite G, Pasqualini JR. Steroid sulphotransferase and 17beta-hydroxysteroid dehydrogenase activities in Ishikawa human endometrial adenocarcinoma cells. J Steroid Biochem Mol Biol 1997; 61:27-34. [PMID: 9328207 DOI: 10.1016/s0960-0760(97)00010-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The present studies concern sulphotransferase activities for estrogens and other steroids, and 17beta-hydroxysteroid dehydrogenase (17beta-HSD) activities for estrogens in Ishikawa endometrial adenocarcinoma cells. When physiological concentrations of various estrogens (estrone, estradiol, estriol) are incubated, most of the transformation product is the respective sulphate. The sulphotransferase activity is very rapid, and 2 h after incubation 70-95% are converted to the sulphated form. Sulphates are found exclusively in the culture medium, which suggests that as soon as the sulphate is biosynthesized it is secreted to the medium. Comparative data using neutral steroids (dehydroepiandrosterone, testosterone, and pregnenolone) show that sulphotransferase activity for these compounds is very limited. In another series of studies, 17beta-HSD activity was explored for the interconversion estrone estradiol. At low concentrations (5 x 10(-9)-5 x 10(-8) M), when estradiol (E2) is incubated, most of the unconjugated material remains as E2 in the cellular compartment, but at high concentrations (5 x 10(-7)-5 x 10(-6) M) a great proportion (70-80%) of the E2 is converted to estrone (E1). On the other hand, after incubation of E1 at all concentrations most remained as unchanged E1. It is suggested that, in Ishikawa cells, at very low concentrations of E1 or E2, sulphotransferases are predominant, but when this enzyme is saturated 17beta-HSD activity is orientated to the oxidative form.
Collapse
Affiliation(s)
- G Chetrite
- Steroid Hormone Research Unit, Paris, France
| | | |
Collapse
|
45
|
Anderson C, Freeman J, Lucas LH, Farley M, Dalhoumi H, Widlanski TS. Estrone sulfatase: probing structural requirements for substrate and inhibitor recognition. Biochemistry 1997; 36:2586-94. [PMID: 9054565 DOI: 10.1021/bi961536t] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The enzyme-catalyzed desulfation of steroids is a transformation that plays an important role in steroid biosynthesis. Conversion of steroid sulfates to unconjugated steroids may provide a source of steroids for processes such as steroid transport and the growth and proliferation of breast cancer. Steroid sulfatase catalyzes the hydrolysis of 3beta-hydroxysteroid sulfates. To identify structural features important in enzyme-inhibitor interaction, a variety of steroidal and non-steroidal phosphate esters were synthesized and tested as inhibitors of steroid sulfatase activity. We report that the basic structure for enzyme-inhibitor binding does not include the steroid nucleus. Furthermore, the hydrophobicity of the non-steroidal phosphates was determined to be an important factor for optimal inhibition. The monoanionic form of the phosphorylated compounds was found to be the inhibitory species. The best non-steroidal inhibitor of steroid sulfatase activity was n-lauroyl tryamine phosphate with a Ki of 3.6 microM and 520 nM at pH 7.5 and 7.0. The poorest non-steroidal based inhibitor of sulfatase activity was tetrahydronaphthyl phosphate with a Ki of 870 and 360 microM at pH 7.5 and 7.0.
Collapse
Affiliation(s)
- C Anderson
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, USA
| | | | | | | | | | | |
Collapse
|
46
|
Misao R, Nakanishi Y, Ichigo S, Hori M, Fujimoto J, Tamaya T. Expression of sex hormone-binding globulin mRNA in human endometrial cancers. J Steroid Biochem Mol Biol 1995; 52:517-22. [PMID: 7779755 DOI: 10.1016/0960-0760(95)00061-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
To more fully understand the role of sex hormone-binding globulin (SHBG) on the intracellular steroidal action in endometrial cancers, we investigated the expression of SHBG mRNA as the substitute of SHBG expression in human endometrial cancers. In the present study, the levels of SHBG mRNA were analyzed using competitive reverse transcription-polymerase chain reaction (RT-PCR)-Southern-blot analysis. The higher level of SHBG mRNA tended to be expressed in the normal secretory and late proliferative phase endometrium > early proliferative phase endometrium > well differentiated adenocarcinoma of the endometrium (G1) > moderately differentiated adenocarcinoma (G2) > poorly differentiated adenocarcinoma (G3), in the order shown. These studies indicate that endometrial cancer cells might synthesize intracellular SHBG to conserve their estrogen-dependent properties. Further, it indicates that endometrial cancer cell synthesis of SHBG mRNA is lost as these cells undergo de-differentiation.
Collapse
Affiliation(s)
- R Misao
- Department of Obstetrics and Gynecology, Gifu University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Chetrite G, Blumberg-Tick J, Pasqualini JR. Effect of Decapeptyl (a GnRH analogue) and of transforming growth factor-alpha (TGF-alpha), in the presence of heparin, on the sulfatase activity of human breast cancer cells. J Steroid Biochem Mol Biol 1995; 52:451-7. [PMID: 7748810 DOI: 10.1016/0960-0760(95)00004-j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The effects of the polypeptide Decapeptyl (a gonadotropin-releasing hormone (GnRH) agonist analogue) and of transforming growth factor-alpha (TGF-alpha), on estrone sulfate-sulfatase activities in the homogenates of various breast cancer cell lines were studied in the presence of heparin. In hormone-dependent MCF-7 breast cancer cells, Decapeptyl can inhibit sulfatase activity, and this effect is significantly augmented in the presence of heparin. In the other hormone-dependent T-47D breast cancer cell line, the decrease of sulfatase activity was only significant when Decapeptyl was associated with heparin. No significant effect on sulfatase activity elicited by heparin, Decapeptyl or a mixture of both was found in the hormone-independent MDA-MB-231 breast cancer cells. TGF-alpha stimulates sulfatase activity in the MDA-MB-231 cells but has no effect in the MCF-7 cells; in contrast, TGF-alpha combined with heparin provokes a decrease of the sulfatase activity in both cell lines. It is concluded that the sulfatase activity in some types of breast cancer cell can be inhibited by heparin combined with the polypeptides Decapeptyl or TGF-alpha.
Collapse
Affiliation(s)
- G Chetrite
- C.N.R.S. Steroid Hormone Research Unit, Foundation for Hormone Research, Paris, France
| | | | | |
Collapse
|
48
|
Evans TR, Rowlands MG, Sahota SS, Coombes RC. Lack of inhibition of placental estrone sulfatase and aromatase enzymes by vitamin D3 and its analogs. J Steroid Biochem Mol Biol 1994; 48:563-6. [PMID: 8180120 DOI: 10.1016/0960-0760(94)90209-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The aromatase and estrone sulfatase enzymes are important sources of biologically active estrogens in postmenopausal women with breast cancer. Promising initial results in the treatment of endocrine-responsive breast cancer have been exhibited by 1 alpha 25-dihydroxyvitamin D3 and the synthetic vitamin D analogues MC903 and EB1089. However, these compounds together with vitamin D3 and vitamin D3 sulfate did not inhibit the human placental aromatase enzyme when assayed up to 20 microns. Only vitamin D3 sulfate and 1 alpha 25-dihydroxyvitamin D inhibited the estrone sulfatase activity in human placental microsomes, albeit at high concentration (32 and 37% inhibition, respectively with 50 microns each inhibitor). It is unlikely that inhibition of aromatase or estrone sulfatase enzymes contribute to the inhibitory effect of this group of compounds on breast cancer cells in vivo.
Collapse
Affiliation(s)
- T R Evans
- Department of Medical Oncology, St George's Hospital Medical School, London, England
| | | | | | | |
Collapse
|
49
|
Evans TR, Rowlands MG, Law M, Coombes RC. Intratumoral oestrone sulphatase activity as a prognostic marker in human breast carcinoma. Br J Cancer 1994; 69:555-61. [PMID: 8123487 PMCID: PMC1968849 DOI: 10.1038/bjc.1994.101] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Oestrone sulphatase is an important source of local synthesis of biologically active oestrogens in human breast cancer. The oestrone sulphatase enzyme in the particulate fraction of human breast carcinoma was characterised. The Km was 8.91 microM, and the Vmax was 0.022 nmol min-1 mg-1. Oestrone sulphatase activity was detected in 93 of 104 human breast carcinoma samples (89%), and mean activity was 0.041 nmol min-1 mg-1 (range 0-0.399 nmol min-1 mg-1). There was no significant correlation between intratumoral oestrone sulphatase activity and oestrogen receptor status, or with any other prognostic factors. Intratumoral enzyme levels were not associated with time to recurrence or with overall survival time. It thus appears that, although a useful source of intratumoral oestrogens, oestrone sulphatase activity is not of prognostic significance in breast carcinoma.
Collapse
Affiliation(s)
- T R Evans
- Department of Medical Oncology, St George's Hospital Medical School, London, UK
| | | | | | | |
Collapse
|
50
|
Söderqvist G, Olsson H, Wilking N, von Schoultz B, Carlström K. Metabolism of estrone sulfate by normal breast tissue: influence of menopausal status and oral contraceptives. J Steroid Biochem Mol Biol 1994; 48:221-4. [PMID: 8142298 DOI: 10.1016/0960-0760(94)90148-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The metabolism of [3H]estrone sulfate ([3H]E1S) was studied in normal breast tissue from 10 premenopausal women without oral contraceptives (OC), in 12 OC users and in 9 untreated postmenopausal women. [3H]E1S was converted into estrone ([3H]E1) and estradiol-17 beta ([3H]E2) by tissue samples from all three groups of women, with only minor formation of other unconjugated compounds. The rate of [3H]E2 formation was significantly higher in premenopausal women without OC than in postmenopausal women. Among premenopausal women, OC users had a significantly lower rate of total hydrolysis and of [3H]E1 formation than non-users. The rate of total hydrolysis of [3H]E1S in normal breast tissue from all three groups of women was similar to that in muscle, but the rate of [3H]E2 formation was ten times higher. Both total hydrolysis rate and rate of [3H]E2 formation were significantly lower in normal breast tissue than in breast carcinoma and in normal and neoplastic endometrium. The specific ability of normal breast tissue to convert E1S into the terminal biologically active estrogen E2 may be important for estrogenic stimulation of the breast in subjects with low circulating E2 levels. The lower rate of E1 formation in OC users may reflect an inhibitory effect of the progestagen compound in such preparations.
Collapse
Affiliation(s)
- G Söderqvist
- Department of Obstetrics and Gynecology, Karolinska Institute, Karolinska Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|