1
|
Wang T, He K, Blaney L, Chung JS. 17β-Estradiol (E2) may be involved in the mode of crustacean female sex hormone (CFSH) action in the blue crab, Callinectes sapidus. Front Endocrinol (Lausanne) 2022; 13:962576. [PMID: 35957817 PMCID: PMC9358259 DOI: 10.3389/fendo.2022.962576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
17β-estradiol (E2) has been proved to control reproduction, sexual differentiation, and the development of the secondary sexual characteristics of vertebrate females. In decapod crustacean species, crustacean female sex hormone (CFSH), a protein hormone, is required for developing adult-specific ovigerous setae for embryo brooding and gonophores for mating at the blue crab Callinectes sapidus puberty molting. However, it is unclear that whether the mode of CFSH action involves a vertebrate-type sex steroid hormone in crustaceans. To this end, E2 levels were first measured using a competitive ELISA in the hemolymph and the potential CFSH target tissues from both prepuberty and adult females; the presence of E2 was further confirmed with a liquid chromatography tandem mass spectrometry method. Then, the cDNAs of the following genes known to be associated with vertebrate steroidogenic pathways were isolated: StAR-related lipid transfer protein 3 (StAR3); 3β-hydroxysteroid dehydrogenase (3βHSD); two isoforms of 17β-hydroxysteroid dehydrogenase 8 (17βHSD8); and, estradiol-related receptor (ERR). RT-PCR analysis revealed that these genes were widely distributed in the eyestalk ganglia, hepatopancreas, brain, ovary, spermathecae, ovigerous and plumose setae tissues of adult females. The 17βHSD8 transcripts were localized in the follicle cells, the periphery of the nuclear membrane of primary oocytes, and yolk granules of the vitellogenic oocytes using in situ hybridization, and the corresponding protein was detected in the follicle cells and ooplasm of primary oocytes using immunohistochemistry. Furthermore, the adult females injected with CFSH-dsRNA (n = 30 times) had E2 and StAR3 transcripts levels lower in the ovigerous and plumose setae, spermathecae than controls. These results suggested that the mode of CFSH action in C. sapidus might involve E2 in these adult-female-specific tissues.
Collapse
Affiliation(s)
- Tao Wang
- Department of Marine Biotechnology & Institute of Marine and Environmental Technology, University of Maryland Baltimore County, Baltimore, MD, United States
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental Science, Baltimore, MD, United States
| | - Ke He
- Department of Chemical, Biochemical, and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD, United States
| | - Lee Blaney
- Department of Chemical, Biochemical, and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD, United States
| | - J. Sook Chung
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental Science, Baltimore, MD, United States
| |
Collapse
|
2
|
Tchernof A, Mansour MF, Pelletier M, Boulet MM, Nadeau M, Luu-The V. Updated survey of the steroid-converting enzymes in human adipose tissues. J Steroid Biochem Mol Biol 2015; 147:56-69. [PMID: 25448733 DOI: 10.1016/j.jsbmb.2014.11.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 11/10/2014] [Accepted: 11/12/2014] [Indexed: 12/26/2022]
Abstract
Over the past decade, adipose tissues have been increasingly known for their endocrine properties, that is, their ability to secrete a number of adipocytokines that may exert local and/or systemic effects. In addition, adipose tissues have long been recognized as significant sites for steroid hormone transformation and action. We hereby provide an updated survey of the many steroid-converting enzymes that may be detected in human adipose tissues, their activities and potential roles. In addition to the now well-established role of aromatase and 11β-hydroxysteroid dehydrogenase (HSD) type 1, many enzymes have been reported in adipocyte cell lines, isolated mature cells and/or preadipocytes. These include 11β-HSD type 2, 17β-HSDs, 3β-HSD, 5α-reductases, sulfatases and glucuronosyltransferases. Some of these enzymes are postulated to bear relevance for adipose tissue physiology and perhaps for the pathophysiology of obesity. This elaborate set of steroid-converting enzymes in the cell types of adipose tissue deserves further scientific attention. Our work on 20α-HSD (AKR1C1), 3α-HSD type 3 (AKR1C2) and 17β-HSD type 5 (AKR1C3) allowed us to clarify the relevance of these enzymes for some aspects of adipose tissue function. For example, down-regulation of AKR1C2 expression in preadipocytes seems to potentiate the inhibitory action of dihydrotestosterone on adipogenesis in this model. Many additional studies are warranted to assess the impact of intra-adipose steroid hormone conversions on adipose tissue functions and chronic conditions such as obesity, diabetes and cancer.
Collapse
Affiliation(s)
- André Tchernof
- Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Canada; École de Nutrition, Université Laval, Québec, Canada; Endocrinologe et Néphrologie, Centre Hospitalier Universitaire de Québec, Québec, Canada.
| | - Mohamed Fouad Mansour
- Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Canada; Endocrinologe et Néphrologie, Centre Hospitalier Universitaire de Québec, Québec, Canada
| | - Mélissa Pelletier
- Endocrinologe et Néphrologie, Centre Hospitalier Universitaire de Québec, Québec, Canada
| | - Marie-Michèle Boulet
- Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Canada; École de Nutrition, Université Laval, Québec, Canada
| | - Mélanie Nadeau
- Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Canada
| | - Van Luu-The
- Endocrinologe et Néphrologie, Centre Hospitalier Universitaire de Québec, Québec, Canada
| |
Collapse
|
3
|
Liu J, Zhang Z, Ma X, Liang S, Yang D. Characteristics of 17β-hydroxysteroid dehydrogenase 8 and its potential role in gonad of Zhikong scallop Chlamys farreri. J Steroid Biochem Mol Biol 2014; 141:77-86. [PMID: 24486454 DOI: 10.1016/j.jsbmb.2014.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/07/2014] [Accepted: 01/21/2014] [Indexed: 12/31/2022]
Abstract
17β-Hydroxysteroid dehydrogenases (17β-HSDs) are important enzymes catalyzing steroids biosynthesis and metabolism in vertebrates. Although studies indicate steroids play a potential role in reproduction of molluscs, little is known about the presence and function of 17β-HSDs in molluscs. In the present study, a full-length cDNA encoding 17β-HSD type 8 (17β-HSD8) was identified in the Zhikong scallop Chlamys farreri, which is 1104bp in length with an open reading frame of 759bp encoding a protein of 252 amino acids. Phylogenetic analysis revealed that the C. farreri 17β-HSD8 (Cf-17β-HSD8) belongs to the short chain dehydrogenase/reductase family (SDR) and shares high homology with other 17β-HSD8 homologues. Catalytic activity assay in vitro demonstrated that the refolded Cf-17β-HSD8 expressed in Escherichia coli could effectively convert estradiol-17β (E2) to estrone (E1), and weakly catalyze the conversion of testosterone (T) to androstenedione (A) in the presence of NAD(+). The Cf-17β-HSD8 mRNA was ubiquitously expressed in all tissues analyzed, including gonads. The expression levels of Cf-17β-HSD8 mRNA and protein increased with gametogenesis in both ovary and testis, and were significantly higher in testis than in ovary at growing stage and mature stage. Moreover, results of in situ hybridization and immunohistochemistry revealed that the mRNA and protein of Cf-17β-HSD8 were expressed in follicle cells and gametes at all stages except spermatozoa. Our findings suggest that Cf-17β-HSD8 may play an important role in regulating gametogenesis through modulating E2 levels in gonad of C. farreri.
Collapse
Affiliation(s)
- Jianguo Liu
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Zhifeng Zhang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China.
| | - Xiaoshi Ma
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Shaoshuai Liang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Dandan Yang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| |
Collapse
|
4
|
Ayan D, Maltais R, Roy J, Poirier D. A new nonestrogenic steroidal inhibitor of 17β-hydroxysteroid dehydrogenase type I blocks the estrogen-dependent breast cancer tumor growth induced by estrone. Mol Cancer Ther 2012; 11:2096-104. [PMID: 22914440 DOI: 10.1158/1535-7163.mct-12-0299] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) converts estrone (E1) into estradiol (E2) and is expressed in many steroidogenic tissues and breast cancer cell lines. Because the potent estrogen E2 stimulates the growth and development of hormone-dependent diseases, inhibition of the final step of E2 synthesis is considered a promising strategy for the treatment of breast cancer. On the basis of our previous study identifying 16β-(m-carbamoylbenzyl)-E2 (CC-156) as a lead compound for the inhibition of 17β-HSD1, we conducted a number of structural modifications to reduce its undesired residual estrogenic activity. The steroid derivative PBRM [3-(2-bromoethyl)-16β-(m-carbamoylbenzyl)-17β-hydroxy-1,3,5(10)-estratriene] emerged as a potent inhibitor of 17β-HSD1 with an IC(50) value of 68 nmol/L for the transformation of E1 into E2. When tested in the estrogen-sensitive breast cancer cell line T-47D and in mice, PBRM showed no estrogenic activity in the range of concentrations tested. Furthermore, with the purpose of evaluating the bioavailability of PBRM and CC-156 injected subcutaneously (2.3 mg/kg), we measured their plasmatic concentrations as a function of time, calculated the area under the curve (AUC(0-12h)) and showed a significant improvement for PBRM (772 ng*h/mL) compared with CC-156 (445 ng*h/mL). We next tested the in vivo efficiency of PBRM on the T-47D xenograft tumor model in female ovariectomized athymic nude mice. After a treatment with PBRM, tumor sizes in mice stimulated with exogenous E1 were completely reduced at the control group level (without E1 treatment). As a conclusion, PBRM is a promising nonestrogenic inhibitor of 17β-HSD1 for the treatment of estrogen-dependent diseases such as breast cancer.
Collapse
Affiliation(s)
- Diana Ayan
- Laboratory of Medicinal Chemistry, CHUQ (CHUL)-Research Center, 2705 Laurier Boulevard, Quebec G1V 4G2, Canada
| | | | | | | |
Collapse
|
5
|
Ahlem CN, Auci DL, Nicoletti F, Pieters R, Kennedy MR, Page TM, Reading CL, Enioutina EY, Frincke JM. Pharmacology and immune modulating properties of 5-androstene-3β,7β,17β-triol, a DHEA metabolite in the human metabolome. J Steroid Biochem Mol Biol 2011; 126:87-94. [PMID: 21570467 DOI: 10.1016/j.jsbmb.2011.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 04/23/2011] [Accepted: 04/24/2011] [Indexed: 10/18/2022]
Abstract
Androst-5-ene-3β,7β,17β-triol (βAET) is an anti-inflammatory metabolite of DHEA that is found naturally in humans, but in rodents only after exogenous DHEA administration. Unlike DHEA, C-7-oxidized DHEA metabolites cannot be metabolized into potent androgens or estrogens, and are not peroxisome proliferators in rodents. The objective of our current studies was to characterize the pharmacology of βAET to enable clinical trials in humans. The pharmacology of βAET was characterized by pharmacokinetics, drug metabolism, nuclear hormone receptor interactions, androgenicity, estrogenicity, and systemic toxicity studies. βAET's acute anti-inflammatory activity and immune modulating characteristics were measured in vitro in RAW264.7 cells and in vivo in murine models with parenteral administration. βAET was rapidly metabolized and cleared from circulation in mice and monkeys. βAET was weakly androgenic and estrogenic in immature rodents, but not bound by androgen, estrogen, progesterone, or glucocorticoid nuclear hormone receptors. βAET did not induce peroxisome proliferation, nor was it systemically toxic or trophic for sex hormone responsive tissues in mature rats and monkeys. βAET significantly attenuated acute inflammation both in vitro and in vivo, augmented immune responses in adult mice, and reversed immune senescence in aged mice. βAET may contribute to the anti-inflammatory activity in rodents attributed to DHEA. Unlike DHEA, βAET's anti-inflammatory activity cannot be ascribed to activation of PPARs, androgen, or estrogen nuclear hormone receptors. Exogenous βAET is unlikely to produce untoward toxicity or hormonal perturbations in humans.
Collapse
|
6
|
|
7
|
Moeller G, Adamski J. Integrated view on 17beta-hydroxysteroid dehydrogenases. Mol Cell Endocrinol 2009; 301:7-19. [PMID: 19027824 DOI: 10.1016/j.mce.2008.10.040] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 10/27/2008] [Accepted: 10/27/2008] [Indexed: 10/21/2022]
Abstract
17beta-Hydroxysteroid dehydrogenases (17beta-HSDs) are important enzymes in steroid metabolism. Long known members of the protein family seemed to be well characterised concerning their role in the regulation of the biological potency of steroid hormones, but today more and more evidence points to pivotal contributions of these enzymes in a variety of other metabolic pathways. Therefore, studies on 17beta-HSDs develop towards metabolomic survey. Latest research results give new insights into the complex metabolic interconnectivity of the 17beta-HSDs. In this paper metabolic activities of 17beta-HSDs will be compared, their interplay with endogenous substrates summarised, and interlacing pathways depicted. Strategies on deciphering the physiological role of 17beta-HSDs and the genetic predisposition for associated diseases will be presented.
Collapse
Affiliation(s)
- Gabriele Moeller
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany.
| | | |
Collapse
|
8
|
Laplante Y, Rancourt C, Poirier D. Relative involvement of three 17beta-hydroxysteroid dehydrogenases (types 1, 7 and 12) in the formation of estradiol in various breast cancer cell lines using selective inhibitors. Mol Cell Endocrinol 2009; 301:146-53. [PMID: 18812208 DOI: 10.1016/j.mce.2008.08.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 08/27/2008] [Accepted: 08/27/2008] [Indexed: 11/25/2022]
Abstract
We investigated the relative involvement of three reductive 17beta-hydroxysteroid dehydrogenase (17beta-HSD) isoforms, namely types 1, 7 and 12, in the formation of potent estrogen estradiol (E2) in 10 human breast cancer cell lines (T-47D, MCF-7, ZR-75-1, CAMA-1, BT-20, BRC-17, BRC-31, BRC-32, BRC-36 and BRN-196) and also in 1 choriocarcinoma cell line (JEG-3) using selective inhibitors. In T-47D, BT-20 and JEG-3 cells, a 17beta-HSD1 inhibitor almost completely inhibited the formation of E2 at 1microM from 60nM of estrone (E1) (98%, 91% and 90%, respectively), whereas no significant inhibition of E2 formation was obtained using inhibitors of types 7 and 12. However, we obtained lower levels of inhibition (32%, 36% and 35% respectively using inhibitors of types 1, 7 and 12 at 10microM) in MCF-7 cells and even lower and variable levels of inhibition (15%, 23% and 18% respectively using inhibitors of types 1, 7 and 12 at 10microM) in ZR-75-1 cells. No inhibition of E2 formation was observed in CAMA-1 cells with a 17beta-HSD1 inhibitor at 1microM whereas inhibitors of types 7 and 12 inhibited 40% and 30% of E2 formation, respectively. In BRC and BRN cell lines, types 1, 7 and 12 17beta-HSDs were all involved in the formation of E2, but type 12 seemed to predominate. At 10microM, each inhibitor inhibited 10-50% of the formation of E2. Using MCF-7 and BRC-32 cell lines, a combination of the three inhibitors (3x10microM) does not fully inhibit the 17beta-HSD activity (65% and 75%). In addition to identify the relative importance of types 1, 7 and 12 17beta-HSDs in the formation of E2 in human breast cancer cell lines, our results show also a great variability between each cell line. In some cases the formation of E2 was completely inhibited, but this was not the result observed in other cell lines, suggesting the presence of another enzyme involved in the biosynthesis of E2.
Collapse
Affiliation(s)
- Yannick Laplante
- Oncology and Molecular Endocrinology Laboratory (Medicinal Chemistry Division), CHUQ-CHUL Research Center and Université Laval, 2705 Laurier Boulevard, Québec, Québec G1V 4G2, Canada
| | | | | |
Collapse
|
9
|
Peychal SEM, Bilger A, Pitot HC, Drinkwater NR. Predominant modifier of extreme liver cancer susceptibility in C57BR/cdJ female mice localized to 6 Mb on chromosome 17. Carcinogenesis 2009; 30:879-85. [PMID: 19255062 PMCID: PMC2675651 DOI: 10.1093/carcin/bgp054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sex hormones influence the susceptibility of inbred mice to liver cancer. C57BR/cdJ (BR) females are extremely susceptible to spontaneous and chemically induced liver tumors, in part due to a lack of protection against hepatocarcinogenesis normally offered by ovarian hormones. BR males are also moderately susceptible, and the susceptibility of both sexes of BR mice to liver tumors induced with N,N-diethylnitrosamine relative to the resistant C57BL/6J (B6) strain is caused by two loci designated Hcf1 and Hcf2 (hepatocarcinogenesis in females) located on chromosomes 17 and 1, respectively. The Hcf1 locus on chromosome 17 is the predominant modifier of liver cancer in BR mice. To validate the existence of this locus and investigate its potential interaction with Hcf2, congenic mice for each region were generated. Homozygosity for the B6.BR(D17Mit164-D17Mit2) region resulted in a 4-fold increase in liver tumor multiplicity in females and a 4.5-fold increase in males compared with B6 controls. A series of 16 recombinants covering the entire congenic region was developed to further narrow the area containing Hcf1. Susceptible heterozygous recombinants demonstrated a 3- to 7-fold effect in females and a 1.5- to 2-fold effect in males compared with B6 siblings. The effect in susceptible lines completely recapitulated the susceptibility of heterozygous full-length chromosome 17 congenics and furthermore narrowed the location of the Hcf1 locus to a single region of the chromosome from 30.05 to 35.83 Mb.
Collapse
Affiliation(s)
- Stephanie E-M Peychal
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, 1400 University Avenue, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
10
|
Rotinen M, Celay J, Alonso MM, Arrazola A, Encio I, Villar J. Estradiol induces type 8 17beta-hydroxysteroid dehydrogenase expression: crosstalk between estrogen receptor alpha and C/EBPbeta. J Endocrinol 2009; 200:85-92. [PMID: 18852215 DOI: 10.1677/joe-08-0134] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hydroxysteroid (17-beta) dehydrogenase (HSD17B) are the enzymes responsible for the reversible interconversion of 17-hydroxy and 17-keto steroids. The human and mouse type 8 17beta-HSD (HSD17B8) selectively catalyze the conversion of estradiol (E2) to estrone (E1). We previously described thatHSD17B8 is transcriptionally regulated by C/EBPbeta, and that C/EBPbeta is bound to CCAAT boxes located at -5 and -46 of the transcription start site in basal conditions in HepG2 cells. Furthermore, ectopic expression of C/EBPbeta transactivated the HSD17B8 promoter activity. Here, we show that HSD17B8 expression is up-regulated in response to E2 in the estrogen receptor alpha (ERalpha) positive MCF-7 cells. Results showed that this induction is mediated by ERalpha because i) E2 did not induce HSD17B8 expression in ERalphanegative HepG2 cells, ii) ectopic expression of ERalpha restored E2-induced HSD17B8 expression, and iii) this induction was blocked by the anti-ER ICI 182,780. Additional experiments showed that no estrogen response element was necessary for this regulation. However, the CCAAT boxes located at the HSD17B8 proximal promoter were required for E2-induced transcription. Furthermore, co-immunoprecipitation studies revealed tethering of ERalphatoC/EBPbeta in response to E2 in cells expressing ERalpha. Additionally, chromatin immunoprecipitation assays demonstrated that, in response to E2, ERalpha is recruited to the CCAAT boxes in which C/EBPbeta is already bound. Taken together, our results reveal that ERalpha is involved in the transcriptional regulation of HSD17B8 gene in response to E2 through its interaction with C/EBPbeta.
Collapse
Affiliation(s)
- Mirja Rotinen
- Department of Health Sciences, Universidad Pública de Navarra, Pamplona, Spain
| | | | | | | | | | | |
Collapse
|
11
|
|
12
|
Abstract
It is well recognized that there are two androgens, namely testosterone (T) and dihydrotestosterone (DHT); T plays an important role in the testis and muscle, and DHT is crucial for the development, function and pathology of the prostate. It is generally thought that DHT is produced from the 5alpha-reduction of circulating T before being inactivated by 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD) that converts DHT into 5alpha-androstane-3alpha,17beta-diol (3alpha-diol). However, the presence of various steroidogenic enzymes in the prostate as well as the availability at high levels of various steroid precursors such as dehydroepiandrosterone sulphate (DHEAS), dehydroepiandrosterone (DHEA) and 4-androstenedione (4-dione) strongly suggest the existence of additional pathways involved in the biosynthesis and metabolism of DHT. Because steroidogenesis could be different in different species, data from the literature obtained from various human, dog, rat and mouse prostate tissues, as well as primary cells and prostatic cancer cell lines, provide a somewhat confusing picture. In the present chapter, we review the data in order to provide a clearer picture of the pathways involved in DHT biosynthesis and metabolism in the human prostate.
Collapse
Affiliation(s)
- Van Luu-The
- Laboratory of Molecular Endocrinology and Oncology, Laval University Hospital Research Center, Laval University, Québec, Canada.
| | | | | |
Collapse
|
13
|
Ohno S, Nishikawa K, Honda Y, Nakajin S. Expression in E. coli and tissue distribution of the human homologue of the mouse Ke 6 gene, 17beta-hydroxysteroid dehydrogenase type 8. Mol Cell Biochem 2007; 309:209-15. [PMID: 17978863 DOI: 10.1007/s11010-007-9637-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Accepted: 10/18/2007] [Indexed: 10/22/2022]
Abstract
Expression of the human Ke 6 gene, 17beta-hydroxysteroid dehydrogenase type 8, in E. coli and the substrate specificity of the expressed protein were examined. The tissue distribution of mRNA expression of the human Ke 6 gene was also studied using real-time PCR. Human Ke 6 gene was expressed as an enzymatically-active His-tag fusion protein, whose molecular weight was estimated to be 32.5 kDa by SDS-polyacrylamide gel electrophoresis. Expressed human Ke 6 gene effectively catalyzed the conversion of estradiol into estrone. Testosterone, 5alpha-dihydrotestosterone, and 5-androstene-3beta,17beta-diol were also catalyzed into the corresponding 17-ketosteroid at 2.4-5.9% that of estradiol oxidation. Furthermore, expressed enzyme catalyzed the reduction of estrone to estradiol, but the rate was a mere 2.3%. Human Ke 6 gene mRNA was expressed in the various tissues examined, such as brain, cerebellum, heart, lung, kidney, liver, small intestine, ovary, testis, adrenals, placenta, prostate, and stomach. Expression of human Ke 6 gene mRNA was especially abundant in prostate, placenta, and kidney. The levels in prostate and placenta were higher than that in kidney, where it is known to be expressed in large quantities.
Collapse
Affiliation(s)
- Shuji Ohno
- Department of Biochemistry, Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41 Ebara, Shinagawa, Tokyo 142-8501, Japan
| | | | | | | |
Collapse
|
14
|
Villar J, Celay J, Alonso MM, Rotinen M, de Miguel C, Migliaccio M, Encío I. Transcriptional regulation of the human type 8 17beta-hydroxysteroid dehydrogenase gene by C/EBPbeta. J Steroid Biochem Mol Biol 2007; 105:131-9. [PMID: 17583490 DOI: 10.1016/j.jsbmb.2006.12.106] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Accepted: 12/07/2006] [Indexed: 11/23/2022]
Abstract
17beta-Hydroxysteroid dehydrogenases (17beta-HSD) regulate the intracellular concentration of active sex steroid hormones in target tissues. To date, at least 14 different isozymes have been identified. The type 8 17beta-hydroxysteroid dehydrogenase (17beta-HSD8) selectively catalyzes the conversion of estradiol (E2) to estrone (E1). To map the promoter region and to investigate its regulation, we cloned and fused a 1600 bp DNA fragment upstream of the 17beta-HSD8 transcriptional start site to a luciferase reporter gene. After transient transfection in HepG2 cells, this fragment was shown to possess promoter activity. Deletion constructs of the 5' flanking region of the 17beta-HSD8 gene led to the identification of the minimal promoter region within the first 75 bp upstream of the transcriptional start site. This region included two CCAAT boxes and sequences closely resembling the consensus Sp1 and NF-kappaB motifs. Site directed mutagenesis revealed that the CCAAT boxes were essential for transcription in HepG2. EMSA, supershift and chromatin immunoprecipitation reflected that these sequences were binding sites for C/EBPbeta. Furthermore, promoter activity was increased by the co-transfection of a C/EBPbeta expression vector, and this transactivation was through both CCAAT boxes. Our studies indicate that C/EBPbeta is essential for the transcription of the 17beta-HSD8 gene in the liver.
Collapse
Affiliation(s)
- Joaquín Villar
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, Avda. Barañain s/n, 31008 Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
15
|
Bolduc C, Yoshioka M, St-Amand J. Transcriptomic characterization of the long-term dihydrotestosterone effects in adipose tissue. Obesity (Silver Spring) 2007; 15:1107-32. [PMID: 17495187 DOI: 10.1038/oby.2007.623] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To study the long-term transcriptomic effects of dihydrotestosterone (DHT) in adipose tissue. Fat distribution is regulated by sexual hormones. It is still unclear if androgens are promoting or reducing intra-abdominal fat accumulation. RESEARCH METHODS AND PROCEDURES Retroperitoneal adipose tissue were isolated from each group of gonadectomized (GDX) C57BL6 male mice treated with vehicle or DHT for 21 days. Serial analysis of gene expression (SAGE) was performed to generate approximately 150,000 SAGE tags from each sample. RESULTS Among the numerous genes regulated by DHT, transcripts involved in glycolysis, such as aldolase 1 A isoform and pyruvate kinase muscle as well as lipogenic transcripts, such as malic enzyme supernatant and ELOVL family member 6 elongation of long chain fatty acids were down-regulated by androgen supplementation. In contrast, transcripts involved in lipolysis and fatty acid oxidation, such as carboxylesterase 3, acetyl-coenzyme A acyltransferase 1, 3-ketoacyl-CoA thiolase B and enoyl-coenzyme A hydratase/3-hydroxyacyl coenzyme A dehydrogenase were up-regulated by DHT. Pro-apoptotic transcripts such as cell death-inducing DFFA-like effector c, BCL2/adenovirus E1B 19 kDa-interacting protein 1 NIP3 and -interacting protein 3-like were up-regulated by DHT, whereas transcripts involved in promotion of cell cycle such as cyclin D2 were down-regulated by DHT. DISCUSSION These results suggest that chronic androgen treatment may help to improve metabolic profile by regulating various critical pathways involved in adipose tissue physiology. In addition, several genes associated with a healthier metabolic profile, such as adiponectin and CD36 antigen, were up-regulated by 21 days of DHT treatment.
Collapse
Affiliation(s)
- Carl Bolduc
- Functional Genomics Laboratory, Molecular Endocrinology and Oncology Research Center, Laval University Medical Center, 2705 Boulevard Laurier, Quebec City, Quebec, G1V 4G2 Canada
| | | | | |
Collapse
|
16
|
Expression and localization of estrogenic type 12 17beta-hydroxysteroid dehydrogenase in the cynomolgus monkey. BMC BIOCHEMISTRY 2007; 8:2. [PMID: 17280614 PMCID: PMC1800897 DOI: 10.1186/1471-2091-8-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2006] [Accepted: 02/05/2007] [Indexed: 11/10/2022]
Abstract
BACKGROUND We have recently discovered that human type 12 17beta-HSD (h17beta-HSD12), a homolog of type 3 17beta-HSD, is a new estrogen-specific 17beta-hydroxysteroid dehydrogenase involved in the production of estradiol (E2). To further characterize this estradiol-producing enzyme, we have isolated the corresponding cDNA in the cynomolgus monkey (Macaca fascicularis), characterized its enzymatic activities and performed cellular localization using in situ hybridization. RESULTS Using HEK-293 cells stably expressing Macaca fascicularis type 12 17beta-HSD (mf17beta-HSD12), we have found that the mf17beta-HSD12 catalyzes efficiently and selectively the transformation of El into E2, in analogy with the h17beta-HSD12. We have also quantified the mf17beta-HSD12 mRNA expression levels in a series of Macaca fascicularis tissues using Quantitative RealTime PCR. The Macaca fascicularis 17beta-HSD12 mRNA is widely expressed with the highest levels tissues found in the cerebellum, spleen and adrenal with moderate level observed in all the other examined, namely the testis, ovary, cerebral cortex, liver, heart, prostate, mammary gland, myometrium, endometrium, skin, muscle and pancreas. To gain knowledge about the cellular localization of the mf17beta-HSD12 mRNA expression, we performed in situ hybridization using a 35S-labeled cRNA probe. Strong labeling was observed in epithelial cells and stromal cells of the mammary gland. In the uterus, the labeling is detected in epithelial cells and stromal cells of the endometrium. CONCLUSION These results strongly suggest that the Macaca fascicularis 17beta-HSD12 is an essential partner of aromatase in the biosynthesis of estradiol (E2). It strongly suggests that in the estradiol biosynthesis pathway, the step of 17-ketoreduction comes after the step of the aromatization (the aromatization of 4-androstendione to estrone followed by the conversion of estrone into estradiol by estrogen specific l7beta-HSDs) which is in contrast with the hypothesis suggesting that 4-androstenedione is converted to testosterone followed by the aromatization of testosterone.
Collapse
|
17
|
Abstract
For many decades, androgens have dominated endocrine research in hair growth control. Androgen metabolism and the androgen receptor currently are the key targets for systemic, pharmacological hair growth control in clinical medicine. However, it has long been known that estrogens also profoundly alter hair follicle growth and cycling by binding to locally expressed high-affinity estrogen receptors (ERs). Besides altering the transcription of genes with estrogen-responsive elements, 17beta-estradiol (E2) also modifies androgen metabolism within distinct subunits of the pilosebaceous unit (i.e., hair follicle and sebaceous gland). The latter displays prominent aromatase activity, the key enzyme for androgen conversion to E2, and is both an estrogen source and target. Here, we chart the recent renaissance of estrogen research in hair research; explain why the hair follicle offers an ideal, clinically relevant test system for studying the role of sex steroids, their receptors, and interactions in neuroectodermal-mesodermal interaction systems in general; and illustrate how it can be exploited to identify novel functions and signaling cross talks of ER-mediated signaling. Emphasizing the long-underestimated complexity and species-, gender-, and site-dependence of E2-induced biological effects on the hair follicle, we explore targets for pharmacological intervention in clinically relevant hair cycle manipulation, ranging from androgenetic alopecia and hirsutism via telogen effluvium to chemotherapy-induced alopecia. While defining major open questions, unsolved clinical challenges, and particularly promising research avenues in this area, we argue that the time has come to pay estrogen-mediated signaling the full attention it deserves in future endocrinological therapy of common hair growth disorders.
Collapse
Affiliation(s)
- Ulrich Ohnemus
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Lübeck, University of Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany
| | | | | | | | | |
Collapse
|
18
|
Abstract
17Beta-hydroxysteroid dehydrogenases (17beta-HSDs) belong to the family of short chain dehydrogenases/reductases (SDRs) and aldoketo-reductases (AKRs). Some of the enzymes were discovered and named due to their enzymatic activity on steroid substrates or according to their sequence homology to other 17beta-HSDs. During characterisation of these enzymes it turned out that their substrate specificity is broader than first expected and key functions of some 17beta-HSDs in vivo are probably not in steroid metabolism but in basic metabolic pathways. The issue of such multifunctionality is the topic of this review.
Collapse
Affiliation(s)
- Gabriele Moeller
- GSF-National Research Center of Environment and Health, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany.
| | | |
Collapse
|