scispace - formally typeset
Search or ask a question

Showing papers on "Hydroxysteroid dehydrogenase published in 1997"


Journal ArticleDOI
TL;DR: Estrogen levels in the gonads of marine bivalves, the Pacific oyster Crassostrea gigas and scallop Patinopecten yessoensis were determined by high performance liquid chromatography (HPLC) and it was concluded that estrogens can be synthesized in the Gonad, that their levels vary with the reproductive cycle, and that they have a role in the development of gametes.
Abstract: Estrogen levels in the gonads of marine bivalves, the Pacific oyster Crassostrea gigas and scallop Patinopecten yessoensis were determined by high performance liquid chromatography (HPLC) using an electrochemical detector. Estrone (E 1 ), estradiol-17 β (E 2 ), and a small amount of estriol (E 3 ) were identified in the ovary, while only E 2 was found in the testis. The level of E 2 in the ovary was consistently higher than E 1 and it increased with sexual maturation. These results indicate that E 2 may play a role in the reproductive events of the oyster and scallop. In vitro experiments demonstrated the presence of 17 β -hydroxysteroid dehydrogenase (17 β -HSD) in the ovaries of both bivalves. The activity of 17 β -HSD in the ovary was lower in the postspawning stage than in the early differentiating stage. The evidence for the presence of aromatase activity in the scallop ovary was obtained by 3 H-water assay. The immunoreactivity against 3 β -hydroxysteroid dehydrogenase (3 β -HSD), P450 aromatase and E 2 was detected in the cells along the outside of germinal acini of the scallop ovary. It is concluded that estrogens can be synthesized in the gonad, that their levels vary with the reproductive cycle, and that they have a role in the development of gametes.

157 citations


Journal ArticleDOI
TL;DR: This review summarises the current knowledge of type-1 11β-hydroxysteroid dehydrogenase and discusses possible substrate/inhibitor interactions that might impair either the physiological function of glucocorticoids or the detoxification of non-steroid carbonyl compounds.
Abstract: Carbonyl reduction is a significant step in the biotransformation leading to the elimination, of endogenous and exogenous aldehydes, ketones and quinones. This reaction is mediated by members of the aldoketo reductase and short-chain dehydrogenase/reductase (SDR) superfamilies. The essential role of these enzymes in protecting organisms from damage by the accumulation of toxic carbonyl compounds is generally accepted, although their physiological roles are not always clear. Recently, the SDR enzyme 11β-hydroxysteroid dehydrogenase-1 has been identified to perform an important role in the detoxification of non-steroidal carbonyl compounds, in addition to metabolising its physiological glucocorticoid substrates. This review summarises the current knowledge of type-1 11β-hydroxysteroid dehydrogenase and discusses possible substrate/inhibitor interactions. They might impair either the physiological function of glucocorticoids or the detoxification of non-steroid carbonyl compounds.

105 citations


Journal ArticleDOI
TL;DR: This report provides the first evidence that despite the absence of the PR in the rat corpus luteum, progesterone exerts an intraluteal action despite the lack of progestersone receptors (PR) and whether progester one acts through the glucocorticoid receptor (GR) to enhance its own levels by down-regulating the expression of 20α-hydroxysteroid dehydrogenase (20α-HSD).
Abstract: In this study, we investigated whether progesterone exerts an intraluteal action despite the lack of progesterone receptors (PR) in the rat corpus luteum and whether progesterone acts through the glucocorticoid receptor (GR) to enhance its own levels by down-regulating the expression of 20α-hydroxysteroid dehydrogenase (20α-HSD). We first established that the corpus luteum constitutively expresses the GR throughout pregnancy and after parturition. We also generated a temperature sensitive SV-40 transformed luteal cell line (GG-CL) that expresses the GR and 20α-HSD but lacks the PR. Treatment with different doses of either progesterone or dexamethasone caused a dose-related decrease in 20α-HSD mRNA in both cultured corpora lutea and in the luteal cell line. RU486, a PR/GR antagonist, completely blocked both the progesterone and the dexamethasone mediated inhibition of 20α-HSD expression in GG-CL cells. In summary, this report provides the first evidence that despite the absence of the PR in the rat corpus ...

87 citations


Journal ArticleDOI
Mika Mustonen1, Matti Poutanen1, V. Isomaa1, Pirkko Vihko1, Reijo Vihko1 
TL;DR: There is transient oestradiol production in the early days of embryonic development, after which inactivation of sex steroids predominates in the fetus and placenta.
Abstract: 17beta-Hydroxysteroid dehydrogenases (17HSDs) are responsible for the conversion of low-activity sex steroids to more potent forms, and vice versa. 17HSD activity is essential for the biosynthesis of sex steroids in the gonads, and it is also one of the key factors regulating the availability of active ligands for sex-steroid receptors in various extragonadal tissues. In this study, we have characterized mouse 17HSD type 2 cDNA, and analysed the relative expression of 17HSD types 1, 2, 3, 4 and 5 mRNAs in mouse embryos and adult male and female tissues. The cDNA characterized has a open reading frame of 1146 bp, and encodes a protein of 381 amino acids with a predicted molecular mass of 41837 kDa. Northern-blot analysis of adult mouse tissues revealed that, of the different 17HSDs, the type 2 enzyme is most abundantly expressed. High expression of the enzyme, which oxidizes both testosterone and oestradiol, in several large organs of both sexes indicates that it is the isoform having the most substantial role in the metabolism of sex steroids. Interestingly, four of the five 17HSD enzymes were also detected by Northern blots of whole mouse embryos, and each of the enzymes showed a unique pattern of expression. The oestradiol-synthesizing type 1 enzyme predominates in early days of development embryonic day 7, but after that the oxidative type 2 enzyme becomes the predominant form of all 17HSDs. The data therefore suggest that there is transient oestradiol production in the early days of embryonic development, after which inactivation of sex steroids predominates in the fetus and placenta.

64 citations


Journal ArticleDOI
TL;DR: This study describes the isolation, characterization, and tissue-specific expression of a sixth member of this gene family, 3β-HSD VI, which functions as an NAD+-dependent dehydrogenase/isomerase exhibiting very low Michaelis-Menten constant (Km) values for pregnenolone and dehydroepiandrosterone.
Abstract: The enzyme 3beta-hydroxysteroid dehydrogenase (3beta-HSD) is a key enzyme in the biosynthesis of steroid hormones. To date, this laboratory has isolated and characterized five distinct 3beta-HSD complementary DNAs (cDNAs) in the mouse (3beta-HSD I through V). These different forms are expressed in a tissue- and developmentally-specific manner and fall into two functionally distinct enzymes. 3beta-HSD I and III, and most likely II, function as dehydrogenase/isomerases, whereas 3beta-HSD IV and V function as 3-ketosteroid reductases. This study describes the isolation, characterization, and tissue-specific expression of a sixth member of this gene family, 3beta-HSD VI. This new isoform functions as an NAD+-dependent dehydrogenase/isomerase exhibiting very low Michaelis-Menten constant (Km) values for pregnenolone (approximately 0.035 microM) and dehydroepiandrosterone (approximately 0.12 microM). 3beta-HSD VI is the earliest isoform to be expressed during embryogenesis in cells of embryonic origin at 7 and 9.5 days postcoitum (pc), and is the major isoform expressed in uterine tissue at the time of implantation (4.5 days pc) and continues to be expressed in uterine tissue at 6.5, 7.5, and 9.5 days pc. 3beta-HSD VI is expressed in giant trophoblasts at 9.5 days pc and is expressed in the placenta through day 15.5 pc. In the adult mouse, 3beta-HSD VI appears to be the only isoform expressed in the skin and also is expressed in the testis, but to a lesser extent than 3beta-HSD I. Mouse 3beta-HSD VI cDNA is orthologous to human 3beta-HSD I cDNA. Human type I 3beta-HSD has been shown to be the only isoform expressed in the placenta and skin. The demonstration that mouse 3beta-HSD VI functions as a dehydrogenase/isomerase and is the predominant isoform expressed during the first half of pregnancy in uterine tissue and in embryonic cells suggests that this isoform may be involved in local production of progesterone, which is needed for successful implantation of the blastocyst and/or maintenance of early pregnancy.

59 citations


Journal ArticleDOI
TL;DR: The cell type-specific localization of 17 beta HSD type 2 is in accord with the proposed physiological role of the enzyme, namely to protect tissues, in this case the fetus, from bioactive estrogen and androgen.
Abstract: The enzymatic actions of the 17 beta-hydroxysteroid dehydrogenase (17 beta HSD) isozymes are crucial in steroid hormone metabolism/physiology. The type 1 isozyme catalyzes the conversion of the biologically inactive C18 steroid, estrone, to the active estrogen, 17 beta-estradiol, and the enzyme is predominantly expressed in the syncytiotrophoblast of the placenta and the granulosa cells of the ovary. 17 beta HSD type 2 is highly expressed in placenta, liver, and secretory endometrium and catalyzes the conversion of bioactive estrogens and androgens to biologically inactive 17-ketosteroid counterparts. The expression pattern of 17 beta HSD type 2 protein was determined in human term placenta and fetal liver by immunohistochemical analysis using monoclonal antibodies directed against distinct epitopes of the 17 beta HSD type 2 protein. In placenta, the protein was detected in the endothelial cells of fetal capillaries, but not in cytotrophoblasts or syncytiotrophoblast. There was dichotomous immunostaining seen among pairs of cotyledonary vessels and chorionic vessels. In the liver, on the other hand, staining was detected in the hepatocytes, but not in the cells lining blood vessels. We conclude that the cell type-specific localization of 17 beta HSD type 2 is in accord with the proposed physiological role of the enzyme, namely to protect tissues, in this case the fetus, from bioactive estrogen and androgen.

58 citations


Journal ArticleDOI
TL;DR: The absent expression of 11 beta-HSD2 in late gestation may facilitate glucocorticoid-dependent maturation of mouse fetal tissues, and the sexual-dimorphic expression in kidney and colon may reflect male-female differences in sodium homeostasis.

55 citations


Journal ArticleDOI
TL;DR: It is evident that compared with P450arom, 17HSD type 1 is more widely expressed in the follicles during the various maturational stages of folliculogenesis.
Abstract: In the present study, we evaluated the expression and regulation of 17β-hydroxysteroid dehydrogenase (17HSD) type 1 and type 2, cytochrome P450 aromatase (P450arom), and 20α-hydroxysteroid dehydrogenase (20HSD) in mature and pregnant rats. Immunohistochemical analysis of rat 17HSD type 1 showed that the enzyme is exclusively expressed in the granulosa cells of developing, healthy, primary, secondary, and tertiary follicles at all stages of the estrous cycle and pregnancy, and is not detected in the corpora lutea. The data showed that the amount of the enzyme expressed in the follicle increases as follicular maturation progresses and is highest in tertiary and Graafian follicles. However, Northern blot analysis of total RNA from whole ovaries showed a rather constitutive expression of the 17HSD type 1 enzyme. It is evident that compared with P450arom, 17HSD type 1 is more widely expressed in the follicles during the various maturational stages of folliculogenesis. Hence, the data indicate distinct localiza...

55 citations


Journal ArticleDOI
TL;DR: Tissue distribution of mRNA encoding both types 1 and 3 17betaHSD was studied using reverse transcription and the polymerase chain reaction (RT-PCR).

48 citations


Journal ArticleDOI
01 Feb 1997-Steroids
TL;DR: Evidence is provided supporting the suggestion that endogenous substances may regulate the glucocorticoid inactivating isoenzymes, 11β-HSD (hydroxysteroid dehydrogenase) 1 (liver) and 11 β- HSD2 (kidney), and the importance of the structural conformation of the A and B Rings in conferring specific inhibitory properties on these compounds is indicated.

33 citations


Journal ArticleDOI
TL;DR: The data suggests that leukocytes interact with the ovarian cells through cytokine secretion and/or cell–cell contact to increase the 11 β -HSD activity in human granulosa cells.

Journal ArticleDOI
TL;DR: It is hypothesize that there is at least one 3beta-hydroxysteroid dehydrogenase enzyme involved in the biosynthetic pathway of ecdysteroids, as this activity was specific of molting glands.

Journal ArticleDOI
TL;DR: The results emphasized the complexity of calcium contribution with the protein kinase A and C pathways in the regulation of P450scc, 3 β-HSD-1 and 17β-HSd-1 mRNA levels.

Journal ArticleDOI
TL;DR: Immunohistochemical and in-situ hybridization studies revealed that 17β-HSD type IV was preferentially expressed in the RPE cells, which may have a role of maintenance of the neural retina.

Journal ArticleDOI
TL;DR: The existence of 3beta-HSD in osteoblast-like cells indicates that these cells convert delta5 androgens into more biologically active delta4 3-keto steroids, suggesting that the osteobwin cells play an important role in facilitating hormonal action in bone tissue.
Abstract: This paper describes for the first time the presence of 3beta-hydroxysteroid dehydrogenase (3beta-HSD) activity in osteoblast-like cells and investigates its characteristics. 3beta-HSD activity was detected by the formation of androstenedione from [3H]dehydroepiandrosterone (DHEA) in whole cell assays of human osteoblast-like cells, HOS and MG-63. The radiolabeled product, androstenedione, was purified by thin-layer chromatography and identified by recrystallization on admixture with authentic androstenedione to show constant specific activities. The apparent Michaelis constant (Km) for DHEA in HOS was found to be 9.9 microM and that in MG-63 was 80.4 microM. The expression of the 3beta-HSD messenger ribonucleic acid in HOS and MG-63 was demonstrated through a reverse transcription-polymerase chain reaction. The PCR products were confirmed by Southern blot analysis. The existence of 3beta-HSD in osteoblast-like cells indicates that these cells convert delta5 androgens into more biologically active delta4 3-keto steroids. These results, together with the demonstration of other steroid converting enzyme systems, suggest that the osteoblast cells play an important role in facilitating hormonal action in bone tissue.

Journal ArticleDOI
C H Blomquist1, B S Leung, C Beaudoin, D Poirier, Yves Tremblay 
TL;DR: It appears that within A431 cells 17-HSD type 2 is regulated at the level of catalysis to function almost exclusively as a dehydrogenase and that its role as a reductase in testosterone formation by either the delta 4 or delta 5 pathway is limited.
Abstract: There is growing evidence that various isoforms of 17 beta-hydroxysteroid dehydrogenase (17-HSD) are regulated at the level of catalysis in intact cells. A number of investigators have proposed that the NAD(P)/NAD(P)H ratio may control the direction of reaction. In a previous study, we obtained evidence that A431 cells, derived from an epidermoid carcinoma of the vulva, are enriched in 17-HSD type 2, a membrane-bound isoform reactive with C18 and C19 17 beta-hydroxysteroids and 17-ketosteroids. The present investigation was undertaken to confirm the presence of 17-HSD type 2 in A431 cells and to assess intracellular regulation of 17-HSD at the level of catalysis by comparing the activity of homogenates and microsomes with that of cell monolayers. Northern blot analysis confirmed the presence of 17-HSD type 2 mRNA. Exposure of cells to epidermal growth factor resulted in an increase in type 2 mRNA and, for microsomes, increases in maximum velocity (Vmax) with no change in Michaelis constant (Km) for testosterone and androstenedione, resulting in equivalent increases in the Vmax/Km ratio consistent with the presence of a single enzyme. Initial velocity data and inhibition patterns were consistent with a highly ordered reaction sequence in vitro in which testosterone and androstenedione bind only to either an enzyme-NAD or an enzyme-NADH complex respectively. Microsomal dehydrogenase activity with testosterone was 2- to 3-fold higher than reductase activity with androstenedione. In contrast, although cell monolayers rapidly converted testosterone to androstenedione, reductase activity with androstenedione or dehydroepiandrosterone (DHEA) was barely detectable. lactate but not glucose, pyruvate or isocitrate stimulated the conversion of androstenedione to testosterone by monolayers, suggesting that cytoplasmic NADH may be the cofactor for 17-HSD type 2 reductase activity with androstenedione. However, exposure to lactate did not result in a significant change in the NAD/NADH ratio of cell monolayers. It appears that within A431 cells 17-HSD type 2 is regulated at the level of catalysis to function almost exclusively as a dehydrogenase. These findings give further support to the concept that 17-HSD type 2 functions in vivo principally as a dehydrogenase and that its role as a reductase in testosterone formation by either the delta 4 or delta 5 pathway is limited.

Journal ArticleDOI
TL;DR: The results showed that STZ26 is a biologically active inhibitor for H SD1 activity in the luteal tissue and suggested that the depletion of progesterone levels toward the end of pseudopregnancy is not solely due to the elevation of HSD1 activity.
Abstract: 20alpha-Hydroxysteroid dehydrogenase (20alpha-HSD, E.C.1.1.1.149) in rat luteal tissue, which catalyzes conversion of progesterone to a biologically inactive steroid, 20alpha-hydroxypregn-4-ene-3-one (20alpha-OHP), suppresses progesterone secretion into the circulation. An increase in 20alpha-HSD activity in luteal tissue in rats is a prerequisite for functional corpus luteum regression. This study was undertaken to find a steroid inhibitor for ovarian cytosolic 20alpha-HSD activity among derivatives based on progesterone structure. A derivative designated as STZ26 (D-homo-16-oxa-4-androstene-3,16alpha-dione) was found to inhibit potently 20alpha-HSD activity in cultured luteal cells. Ovarian 20alpha-HSD activity consists of two isoforms (HSD1 and HSD2). Kinetic analyses of STZ26 for HSD1 and HSD2 showed that the compound suppressed only HSD1 activity by competitive inhibition. Pseudopregnant rats were treated with STZ26 from 13 to 19 days after cervical stimulation. Either an elevation of peripheral 20alpha-OHP levels or a concomitant depletion of peripheral progesterone levels at the end of pseudopregnancy was considerably inhibited in treated animals, although not completely. The results showed that STZ26 is a biologically active inhibitor for HSD1 activity in the luteal tissue and suggested that the depletion of progesterone levels toward the end of pseudopregnancy is not solely due to the elevation of HSD1 activity.