Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 304
Filter
1.
J Steroid Biochem Mol Biol ; 73(5): 203-10, 2000.
Article in English | MEDLINE | ID: mdl-11070349

ABSTRACT

17beta-Hydroxysteroid dehydrogenase type 1 (17HSD type 1) catalyzes the reduction of estrone (E(1)) to biologically more active estradiol (E(2)). In the present study, the effect of activin, inhibin, and follistatin on 17HSD activity and 17HSD type 1 expression in cultured, unluteinized rat granulosa cells was examined. Furthermore, the effects of these hormones on 17HSD type 1 expression were compared with the expression of P450 aromatase (P450arom). Rat granulosa cells were pre-incubated in serum-free media for 3 days, followed by a 2-day treatment with activin, inhibin, follistatin and 8-Br-cAMP. Activin in increasing concentrations appeared to effect a dose-dependent increase in 17HSD activity. In addition, increasing concentrations of activin also increased 17HSD type 1 mRNA expression. Addition of 8-Br-cAMP at concentrations of 0.25 and 1.5 mmol/l together with activin significantly augmented the stimulatory effects of activin alone in the cultured cells. Neither inhibin, nor follistatin, either alone or in combination with 8-Br-cAMP, had any notable effects on 17HSD activity and 17HSD type 1 expression. Preincubation of activin with increasing concentrations of follistatin significantly diminished the stimulatory effect of activin. In the presence of follistatin, activin did not significantly increase the 8-Br-cAMP-induced 17HSD activity and 17HSD type 1 expression. The culturing of granulosa cells in the presence or the absence of inhibin or follistatin with or without 8-Br-cAMP did not alter the effect of these peptides on P450arom expression in rat granulosa cells as judged by Northern blot analysis of total RNA. However, cAMP-induced P450arom expression was enhanced by activin treatment, except when follistatin was present. This is in line with the suggested role of follistatin as an activin-binding protein, which limits the bioavailability of activin to its membrane receptors. Thus, the results support the notion of a paracrine/autocrine role of activin in follicular steroidogenesis of growing follicles.


Subject(s)
Estradiol Dehydrogenases/genetics , Gene Expression Regulation, Enzymologic/drug effects , Granulosa Cells/enzymology , Inhibins/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Activins , Animals , Aromatase/genetics , Cells, Cultured , Culture Media, Serum-Free , Female , Follistatin , Glycoproteins/pharmacology , Humans , Kinetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology
2.
Breast Cancer Res Treat ; 57(2): 175-82, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10598044

ABSTRACT

17Beta-hydroxysteroid dehydrogenase activity represents a group of several isoenzymes (17HSDs) that catalyze the interconversion between highly active 17beta-hydroxy- and low activity 17-ketosteroids and thereby regulate the biological activity of sex steroids. The present study was carried out to characterize the expression of 17HSD isoenzymes in human mammary epithelial cells and breast tissue. In normal breast tissues 17HSD types 1 and 2 mRNAs were both evenly expressed in glandular epithelium. In two human mammary epithelial cell lines, mRNAs for 17HSD types 1, 2 and 4 were detected. In enzyme activity measurements only oxidative 17HSD activity, corresponding to either type 2 or type 4 enzyme, was present. The role of 17HSD type 4 in estrogen metabolism was further investigated, using several cell lines originating from various tissues. No correlation between the presence of 17HSD type 4 mRNA and 17HSD activity in different cultured cell lines was detected. Instead, oxidative 17HSD activity appeared in cell lines where 17HSD type 2 was expressed and reductive 17HSD activity was present in cells expressing 17HSD type 1. These data strongly suggest that in mammary epithelial cell lines the oxidative activity is due to type 2 17HSD and that oxidation of 17beta-hydroxysteroids is not the primary activity of the 17HSD type 4 enzyme.


Subject(s)
17-Hydroxysteroid Dehydrogenases/analysis , Breast/enzymology , Epithelial Cells/enzymology , 17-Hydroxysteroid Dehydrogenases/genetics , Adult , Blotting, Northern , Cell Line , Female , Gene Expression Regulation, Enzymologic , Humans , In Situ Hybridization , Middle Aged , RNA, Messenger/isolation & purification
3.
Endocrinology ; 140(8): 3478-87, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10433203

ABSTRACT

The function of the gene encoding human 17beta-hydroxysteroid dehydrogenase (17HSD) type 1, the hHSD17B1 gene, is regulated by a cell-specific enhancer at position -662 to -392. The adjacent hHSD17BP1 gene, whose function is not known, contains an analogous region in its 5'-flanking region. The identity between the hHSD17B1 enhancer and the hHSD17BP1 equivalent is as high as 98%, i.e. they differ by only five nucleotides. Results from reporter gene analyses showed that the hHSD17BP1 analog, a pseudoenhancer, has only 10% the activity of the hHSD17B1 enhancer. Furthermore, the results indicate that the reduced function of the pseudoenhancer is a consequence of the presence of G and A at positions -480 and -486, whereas the hHSD17B1 enhancer contains -480C and -486G. In addition, three protected areas were localized to regions -495/-485 (FP1), -544/-528 (FP2), and -589/-571 (FP3) in deoxyribonuclease I footprinting analysis of the hHSD17B1 enhancer. Replacement of the footprinted regions with a nonsense sequence demonstrated that the FP2 region is the most critical for enhancer activity. Mutations of FP2 or a short palindromic region within it led to almost complete abolishment of enhancer activity. We have identified several subelements that are essential for appropriate function of the hHSD17B1 enhancer. The results also show that the hHSD17B1 and hHSD17BP1 genes operate differently despite the high homology between them.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Enhancer Elements, Genetic , 17-Hydroxysteroid Dehydrogenases/biosynthesis , Base Sequence , Breast Neoplasms , Chloramphenicol O-Acetyltransferase/genetics , Choriocarcinoma , Female , Genes, Reporter , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation, Missense , Plasmids , Pregnancy , RNA, Messenger/genetics , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Uterine Neoplasms
4.
J Steroid Biochem Mol Biol ; 69(1-6): 431-9, 1999.
Article in English | MEDLINE | ID: mdl-10419022

ABSTRACT

Two 17beta-hydroxysteroid dehydrogenases (17HSDs), type 1 and type 7, are enzymes of estradiol biosynthesis, in addition to which rodent type 1 enzymes are also able to catalyze androgens. Both of the 17HSDs are abundantly expressed in ovaries, the type 1 enzyme in granulosa cells and type 7 in luteinized cells. The expression of 17HSD7, which has also been described as a prolactin receptor-associated protein (PRAP), is particularly up-regulated in corpus luteum during the second half of rodent pregnancy. A moderate or slight signal for mouse 17HSD7/PRAP mRNA has also been demonstrated in samples of placenta and mammary gland, for example. Human, but not rodent, 17HSD1 is expressed in placenta, breast epithelium and endometrium in addition to ovaries. A cell-specific enhancer, silencer and promoter in the hHSD17B1 gene participate in the regulation of type 1 enzyme expression. The enhancer consists of several subunits, including a retinoic acid response element, the silencer has a binding motif for GATA factors, and the proximal promoter contains adjacent and competing AP-2 and Sp binding sites.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Estradiol/biosynthesis , Isoenzymes/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary , Gene Expression Regulation, Enzymologic , Humans , Molecular Sequence Data
5.
Endocrinology ; 140(7): 3334-41, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385431

ABSTRACT

Human 17beta-hydroxysteroid dehydrogenase (17HSD) type 2 is a widely distributed enzyme that primarily converts the highly active 17beta-hydroxysteroids to their inactive keto forms. In the present study, full-length human 17HSD type 2 was localized in the endoplasmic reticulum using a double immunofluorescence labeling technique. As a consequence of its strong membrane interaction, full-length human 17HSD type 2 could not be solubilized as a biologically active form in vitro. However, by deleting the first 29 amino acids from the N-terminus, we were able to purify a catalytically active enzyme from the cytosolic fraction of Sf9 insect cells. Biochemical and catalytic properties of the purified truncated human 17HSD type 2 protein confirm its suitability for structure-function analyses of the enzyme. Both intact and truncated 17HSD type 2 enzymes efficiently catalyzed the oxidation of estradiol, testosterone, dihydrotestosterone, androstenediol, and 20alpha-dihydroprogesterone. The oxidation of estradiol brought about by human 17HSD type 2 was effectively inhibited by several other steroidal compounds, such as 2-hydroxyestradiol, 5beta-androstan-3alpha,17beta-diol, 5alpha-androstan-3alpha,17beta-diol, and 5alpha-androstan-3beta,17beta-diol. The broad substrate specificity of human 17HSD type 2 together with its predominant oxidative activity and intracellular location, as observed in this study, indicate the physiological role of the enzyme to be primarily an inactivator of highly active 17beta-hydroxysteroids.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Endoplasmic Reticulum/enzymology , Isoenzymes/genetics , Isoenzymes/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Base Sequence/genetics , Catalysis , Humans , Immunohistochemistry , Intracellular Membranes/enzymology , Molecular Sequence Data , Tissue Distribution/physiology
7.
Mol Endocrinol ; 12(7): 1048-59, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9658408

ABSTRACT

17 beta-Hydroxysteroid dehydrogenases/17-ketosteroid reductases (17HSDs) modulate the biological activity of certain estrogens and androgens by catalyzing reductase or dehydrogenase reactions between 17-keto- and 17 beta-hydroxysteroids. In the present study, we demonstrate expression cloning of a novel type of 17HSD, chronologically named 17HSD type 7, from the HC11 cell line derived from mouse mammary gland. The cloned cDNA, 1.7 kb in size, encodes a protein of 334 amino acids with a calculated molecular mass of 37,317 Da. The primary structure contains segments characteristic of enzymes belonging to the short-chain dehydrogenase/reductase superfamily. Strikingly, mouse 17HSD type 7 (m17HSD7) shows 89% identity with a recently cloned rat protein called PRL receptor-associated protein (PRAP). The function of PRAP has not yet been demonstrated. The enzymatic characteristics of m17HSD7 and RT-PCR-cloned rat PRAP (rPRAP) were analyzed in cultured HEK-293 cells, where both of the enzymes efficiently catalyzed conversion of estrone (E1) to estradiol (E2). With other substrates tested no detectable 17HSD or 20 alpha-hydroxysteroid dehydrogenase activities were found. Kinetic parameters for m17HSD7 further indicate that E1 is a preferred substrate for this enzyme. Relative catalytic efficiencies (Vmax/K(m) values) for E1 and E2 are 244 and 48, respectively. As it is the case with rPRAP, m17HSD7 is most abundantly expressed in the ovaries of pregnant animals. Further studies show that the rat enzyme is primarily expressed in the middle and second half of pregnancy, in parallel with E2 secretion from the corpus luteum. The mRNA for m17HSD7 is also apparent in the placenta, and a slight signal for m17HSD7 is found in the ovaries of adult nonpregnant mice, in the mammary gland, liver, kidney, and testis. Altogether, because of their similar primary structures, enzymatic characteristics, and the tissue distribution of m17HSD7 and rPRAP, we suggest that rPRAP is rat 17HSD type 7. Furthermore, the results indicate that 17HSD7 is an enzyme of E2 biosynthesis, which is predominantly expressed in the corpus luteum of the pregnant animal.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Cloning, Molecular , Phosphoproteins , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA, Complementary/chemistry , Female , Gene Expression , Mammary Glands, Animal/enzymology , Mice , Molecular Sequence Data , Molecular Weight , Organ Specificity , Ovary/enzymology , Pregnancy , RNA, Messenger/analysis , Rats
8.
J Clin Endocrinol Metab ; 83(4): 1190-3, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9543139

ABSTRACT

Our purpose was to assess 17beta-hydroxysteroid dehydrogenase (17HSD) type 1 protein expression in normal breast tissue during the menstrual cycle and hormonal contraception. We analyzed 17HSD type 1 protein expression by immunohistochemistry during the regular menstrual cycle (n = 12) and hormonal contraception (n = 7) in women undergoing reduction mammoplasty. 17HSD type 1 protein was detected in normal breast epithelial cells throughout the menstrual cycle and in all women using hormonal contraception. Mean 17HSD type 1 staining intensity was higher in alveolar epithelial cells in women using hormonal contraception (2.14) than in untreated women (1.25; P < 0.04). For ducts, this difference approached significance (2.29 vs. 1.41; P = 0.06). There was a negative correlation between serum estradiol (E2) levels and 17HSD type 1 protein expression for both alveolar (r(s) = -0.68; P = 0.004) and ductal (r(s) = -0.75; P = 0.002) breast epithelial cells. Enhanced 17HSD type 1 protein expression might increase the conversion to E2 in normal breast tissue during hormonal contraception. The negative correlation between serum E2 levels and 17HSD type 1 suggests this enzyme to be one of the regulatory mechanisms of intratissue E2 concentration in normal breast tissue.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Breast/enzymology , Contraceptives, Oral, Hormonal/therapeutic use , Mammaplasty , Menstrual Cycle/physiology , Adult , Estradiol/blood , Female , Gonadal Steroid Hormones/blood , Humans , Linear Models , Middle Aged , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Reference Values , Steroids/blood
9.
J Clin Endocrinol Metab ; 83(4): 1319-24, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9543162

ABSTRACT

According to the current hypothesis, 17beta-hydroxysteroid dehydrogenases (17HSDs) regulate the extent of estrogen influence in the endometrium by converting estradiol (E2) locally into a biologically less active sex steroid, estrone (E1), and vice versa. Recently, we have shown that both 17HSD type 1 and type 2 are expressed in the human endometrium, and in the present work, using in situ hybridization, we show that 17HSD type 2 is localized in the glandular epithelial cells as previously shown for the type 1 enzyme, but in contrast to type 1, the expression of type 2 is highest at the end of the cycle. Hence, we hypothesize that the differential expression of the two 17HSD enzymes, with opposite activities in same cell types, could modulate intracellular E2 concentrations during the end of the luteal phase of the menstrual cycle. We further analyzed the expression of 17HSD type 1 and type 2 mRNAs in term human placenta. Expression of 17HSD type 1 mRNA was detected in the syncytiotrophoblasts, and signals for type 2 mRNA were found inside the villi, corresponding to cytotrophoblasts. The expression of 17HSD type 2 in the placenta may serve to maintain the presence of inactive sex steroids and attenuate the formation of biologically potent androgens and estrogens.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Endometrium/enzymology , Labor, Obstetric/physiology , Menstrual Cycle/physiology , Placenta/enzymology , RNA, Messenger/biosynthesis , Cell Division/physiology , Epithelial Cells/metabolism , Female , Humans , In Situ Hybridization , Intestine, Small/enzymology , Pregnancy
10.
Gene ; 208(2): 229-38, 1998 Feb 27.
Article in English | MEDLINE | ID: mdl-9524272

ABSTRACT

In the present study, the gene encoding rat 17 beta-hydroxysteroid dehydrogenase type 1 (rHSD17B1 gene) was cloned and characterized. Like the analogous human gene (hHSD17B1), rHSD17B1 contains six exons and five introns spanning approximately 2.2 kb. The identity between the exons and introns of the two genes ranges from 58% to 82% and 42% to 57%, respectively. In contrast to hHSD17B1, rHSD17B1 is not duplicated. The cap site for rHSD17B1 was localized to position -41 upstream of the ATG translation initiation codon. Sequence comparison of the first 200 bp upstream of the cap site showed 72% identity between the human and rat HSD17B1 genes, including a conserved GC-rich area. Further upstream, no significant identity between the two genes was observed and several, cis-acting elements known to modulate the expression of hHSD17B1 are not conserved in the rat gene. Rat HSD17B1 unlike hHSD17B1 with two cap sites, possesses two polyadenylation signals, thus resulting in two mRNAs.


Subject(s)
17-Hydroxysteroid Dehydrogenases/biosynthesis , 17-Hydroxysteroid Dehydrogenases/genetics , Rats/genetics , Transcription, Genetic , Animals , Base Sequence , Exons , Humans , Introns , Molecular Sequence Data , RNA, Messenger/biosynthesis , Regulatory Sequences, Nucleic Acid , Sequence Alignment , Sequence Homology, Nucleic Acid
11.
Proc Soc Exp Biol Med ; 217(3): 310-6, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9492340

ABSTRACT

Several flavonoids and isoflavonoids were found to inhibit 17beta-oxidoreduction of estrogens by the purified 17beta-HSOR type 1, or in cell lines expressing 17beta-HSOR type 1 enzyme (T-47D breast cancer cells) or type 2 (PC-3 prostate cancer cells). The structural demands for the inhibition of estrone (E1) reduction and estradiol (E2) oxidation catalyzed by 17beta-HSOR types 1 and 2, respectively, were not identical. Flavones, flavanones, and isoflavones hydroxylated at both the double ring (positions 5 and 7) and ring B (position 4') were the most potent inhibitors of E1 reduction in T-47D cells, and by the purified type 1 enzyme whereas flavones hydroxylated at positions 3, 5, and 7 of rings A and C, with or without a hydroxyl group in ring B, were capable of inhibiting E2 oxidation in PC-3 cells. Change to flavanone structure, or hydroxylation at position 3 of ring C of flavones, or methylation of the hydroxyl group at position 4' of ring B of flavones and isoflavones reduced or abolished their inhibitory activity on E1 reduction in T-47D cells. On the contrary, hydroxyl group at position 3 of flavones (flavonol structure) markedly increased the inhibition of E2 oxidation in PC-3 cells. Thus, changes in the number and location of hydroxyl groups may discriminate inhibition of E1 reduction and E2 oxidation. Some of the differences may be due to differences in pharmacokinetics of these compounds in T-47D and PC-3 cells. Inhibition of 17beta-HSORs could lead to an alteration in the availability of the highly active endogenous estrogen, but the effects of these compounds in vivo cannot be predicted on the basis of these results alone. Some of these compounds (isoflavones) are estrogenic per se, and they may replace endogenous estrogens, whereas flavones are only very weakly estrogenic or nonestrogenic. Regarding prevention or treatment of estrogen-related diseases, apigenin, coumestrol, and genistein raise special interest.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Breast Neoplasms/enzymology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Prostatic Neoplasms/enzymology , Estradiol/metabolism , Estrone/metabolism , Female , Humans , Male , Oxidation-Reduction , Structure-Activity Relationship , Tumor Cells, Cultured
12.
J Mol Endocrinol ; 20(1): 67-74, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9513083

ABSTRACT

17 beta-Hydroxysteroid dehydrogenase (17HSD) type 2 efficiently catalyzes the conversion of the high activity 17 beta-hydroxy forms of sex steroids into less potent 17-ketosteroids. In the present study in situ hybridization was utilized to analyze the cellular localization of 17HSD type 2 expression in adult male and female mice. The data indicate that 17HSD type 2 mRNA is expressed in several epithelial cell layers, including both absorptive and secretory epithelia as well as protective epithelium. In both males and females, strong expression of 17HSD type 2 was particularly detected in epithelial cells of the gastrointestinal and urinary tracts. The mRNA was expressed in the stratified squamous epithelium of the esophagus, and surface epithelial cells of the stomach, small intestine and colon. The hepatocytes of the liver and the thick limbs of the loops of Henle in the kidneys, as well as the epithelium of the urinary bladder, also showed strong expression of 17HSD type 2 mRNA in both male and female mice. In the genital tracts, low 17HSD type 2 expression was detected in the seminiferous tubules, the uterine epithelial cells and the surface epithelium of the ovary. Expression of the mRNA was also detected in the sebaceous glands of the skin. The results indicate that in both male and female mice, 17HSD type 2 is expressed mainly in the various epithelial cell types of the gastrointestinal and urinary tracts, and therefore suggest a role for the enzyme in steroid inactivation in a range of tissues and cell types not considered as classical sex steroid target tissues.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Digestive System/enzymology , Liver/enzymology , Urinary Tract/enzymology , Animals , Digestive System/cytology , Epithelial Cells/enzymology , Female , Liver/cytology , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , Skin/enzymology , Urinary Tract/cytology
13.
Mol Cell Endocrinol ; 134(1): 33-40, 1997 Oct 31.
Article in English | MEDLINE | ID: mdl-9406847

ABSTRACT

17beta-Hydroxysteroid dehydrogenase type 2 (17HSD type 2) catalyzes the inactivation of estradiol, testosterone and dihydrotestosterone into biologically less active 17-keto forms. Our recent Northern analysis indicated that the enzyme is expressed both in mouse placenta and fetus. The present data indicate that in the placenta the distribution of enzyme expression changes during pregnancy. In the choriovitelline placenta (day 8) 17HSD type 2 was expressed both in mural and polar giant cells. Later, on days 9-12.5, the mRNA was also detected in the junctional zone, and in late gestation (days 14.5-17.5), 17HSD type 2 mRNA was predominantly expressed only at the labyrinth region. In the fetus, 17HSD type 2 expression appears in the liver on day 11. At day 12 the expression was strongly increased in the liver, and at the same time moderate mRNA expression was also detected in the esophagus and intestine. In these tissues, high constitutive expression of 17HSD type 2 was then maintained throughout pregnancy. At later stages of development (days 15-16) the mRNA was, furthermore, detected in epithelial cells of the stomach, tongue, oropharynx and nasopharynx as well as in the kidney. We conclude that the expression pattern of 17HSD type 2 in the developing placenta and fetus suggests a role for the enzyme in maintaining a barrier to the transfer of active 17-hydroxy forms of sex steroids between the fetus and maternal circulation.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Fetus/metabolism , Gene Expression Regulation, Developmental/physiology , Placenta/metabolism , RNA, Messenger/metabolism , Animals , Epithelial Cells/metabolism , Female , Mice , Organ Specificity , Placenta/enzymology , Pregnancy , RNA, Messenger/genetics
14.
Endocrinology ; 138(8): 3417-25, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9231796

ABSTRACT

The 5'-flanking region from -78 to +9 in the HSD17B1 gene serves as a promoter, and an HSD17B1 silencer element is located in position -113 to -78. In the present studies, we have characterized three regulatory elements in the proximal 5'-flanking regions of the gene, using electrophoretic mobility shift assays and reporter gene analysis. First, nuclear factors recognized by antibodies against Sp1 and Sp3 were found to bind the Sp1 motif in the region from -52 to -43. Mutation of the Sp1-binding site decreased the promoter activity to 30% in JEG-3 cells and to 60% in JAR cells, suggesting that binding to the Sp1 motif has a substantial role in the complete functioning of the HSD17B1 promoter. Second, the binding of AP-2 to its motif in the region from -62 to -53 led to reduced binding of Sp1 and Sp3, and furthermore, mutation of the AP-2 element increased promoter activity to 260% in JEG-3 cells. The data thus implied that AP-2 can repress the function of the HSD17B1 promoter by preventing binding to the Sp1 motif. Finally, GATA factors, GATA-3 in particular, were demonstrated to bind their cognate sequence in the HSD17B1 silencer region, and mutations introduced into the GATA-binding site increased transcriptional activity to the level seen in constructs not containing the silencer element. Thus, GATA-3 seems to prevent transcription in the constructs, and hence, the GATA motif also may operate as a negative control element for HSD17B1 transcription.


Subject(s)
Choriocarcinoma/genetics , DNA-Binding Proteins/genetics , Hydroxysteroid Dehydrogenases/genetics , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , Sp1 Transcription Factor/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Base Sequence , Choriocarcinoma/pathology , Choriocarcinoma/physiopathology , DNA, Neoplasm/analysis , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , GATA2 Transcription Factor , GATA3 Transcription Factor , Gene Deletion , Genes, Reporter/genetics , Humans , Plasmids , Polymerase Chain Reaction , Transcription Factor AP-2 , Transcription, Genetic , Tumor Cells, Cultured
15.
Endocrinology ; 138(8): 3532-9, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9231808

ABSTRACT

Human 17beta-hydroxysteroid dehydrogenase (17-HSD) type 1 predominantly catalyzes the 17beta-reduction of estrone to estradiol. The present results, however, show that rat 17-HSD type 1 equally uses both estrone and androstenedione as substrates. Analyzing the activity of various rat/human chimeric enzymes indicated that the region between amino acids 148 and 268 is responsible for the difference in substrate specificity, which is in line with the structural data showing that the recognition end of the active site is primarily at residues 185-230. The enzymes are highly conserved between amino acids 148-191, and the data indicate that in this region Asn152HisAsp153Glu and Pro187Ala variations are most closely related to the differential steroid specificity. The structural analyses furthermore suggested that the presence of His instead of Asn at position 152 of the human enzyme might result in considerable rearrangement of the loop located close to the beta-face of the A- and B-rings of the bound substrate, and that the Pro187Ala variation could modify the flexible region involved in substrate recognition and access of the substrate to the active site. Altogether, our results indicate that the Asn152His and Pro187Ala variations, together with several amino acid variations at the recognition end of the catalytic cleft built by residues 190-230, alter the structure of the active site of rat 17-HSD type 1 to one more favorable to an androgenic substrate.


Subject(s)
Hydroxysteroid Dehydrogenases/metabolism , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Amino Acids/analysis , Animals , Base Sequence , Cell Line , DNA Probes/analysis , DNA Probes/chemistry , DNA Probes/genetics , DNA, Complementary/analysis , DNA, Complementary/chemistry , DNA, Complementary/genetics , Estradiol/metabolism , Estrone/metabolism , Humans , Hydroxysteroid Dehydrogenases/chemistry , Hydroxysteroid Dehydrogenases/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Spodoptera/cytology , Substrate Specificity , Time Factors
16.
Cancer Res ; 57(16): 3356-9, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9269995

ABSTRACT

Several studies have indicated that in prostate cancer, frequent aberrations take place in several genomic regions. In the present study, we have analyzed allelic losses in chromosome 16 region q in 50 prostate cancer specimens of various histological grades. The most frequently deleted region was located at 16q23-16q24.2 between loci D16S504 and D16S422. The highest percentage of loss of heterozygosity (LOH) at 16q was also found within this area at loci HSD17B2 and D16S422 located at 16q24.1-q24.2. The LOH at 16q24.1-q24.2 was significantly associated with clinically aggressive behavior of the disease, metastatic disease, and higher tumor grade. Of the metastatic diseases, 83% showed LOH, whereas only 40% of the nonmetastatic diseases were found to show it. Similarly, LOH was found in 76% of the clinically aggressive diseases and in 33% of the nonaggressive diseases. The data suggest that a potentially important gene associated with prostate cancer progression is located close to 16q24.1-q24.2.


Subject(s)
Chromosomes, Human, Pair 16/genetics , Gene Deletion , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Chromosome Mapping , Humans , Male , Microsatellite Repeats , Neoplasm Metastasis/genetics
17.
Biochem J ; 325 ( Pt 1): 199-205, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9224647

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.


Subject(s)
17-Hydroxysteroid Dehydrogenases/biosynthesis , Blastocyst/enzymology , Gene Expression Regulation, Developmental , Isoenzymes/biosynthesis , Transcription, Genetic , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/genetics , Amino Acid Sequence , Animals , Base Sequence , Blastocyst/physiology , Cloning, Molecular , DNA, Complementary , Embryonic and Fetal Development , Female , Gene Expression Regulation, Enzymologic , Gestational Age , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Male , Mice , Molecular Sequence Data , Molecular Weight , Open Reading Frames , RNA, Messenger/biosynthesis , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Sex Characteristics , Substrate Specificity
18.
Endocrinology ; 138(7): 2886-92, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9202232

ABSTRACT

In the present study, we evaluated the expression and regulation of 17beta-hydroxysteroid dehydrogenase (17HSD) type 1 and type 2, cytochrome P450 aromatase (P450arom), and 20alpha-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 localization, expression, and regulation patterns for 17HSD type 1 and P450arom during the rat estrous cycle and pregnancy. Furthermore, compared with the two estradiol biosynthetic enzymes, a different expression pattern was detected for 20HSD messenger RNA. During the estrous cycle the enzyme was detected in the ovaries throughout the cycle, and in the ovaries of pregnant animals the enzyme showed an expression pattern the opposite of that observed for P450arom. Rat 17HSD type 2, not detected in the ovaries, was constitutively expressed in both female and male liver and small intestine in 21-day-old fetuses up to 6-week-old mature animals. Similarly, in these tissues the enzyme was constitutively expressed in normal cycling and pregnant animals, but it showed increasing expression in the placenta as pregnancy advanced. The relatively constitutive expression of the enzyme at all physiological stages of the animals suggests a general role for the enzyme in the inactivation of circulating sex steroids.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , 20-Hydroxysteroid Dehydrogenases/metabolism , Aging/metabolism , Aromatase/metabolism , Isoenzymes/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , 20-Hydroxysteroid Dehydrogenases/genetics , 20-alpha-Hydroxysteroid Dehydrogenase , Animals , Aromatase/genetics , Estradiol/metabolism , Estrone/metabolism , Female , Isoenzymes/genetics , Male , Ovary/enzymology , Placenta/enzymology , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
19.
Endocrinology ; 138(3): 898-904, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9048588

ABSTRACT

Human 17 beta-hydroxysteroid dehydrogenase type 1 (17HSD type 1) primarily catalyzes the reduction of low activity estrone to high activity estradiol in ovarian granulosa cells and placental trophoblasts 17HSD type 1 is also present in certain peripheral tissues, such as breast tissue. In the present study we investigated the effects of retinoic acids (RAs) together with other stimuli known to modulate estradiol production and/or cell growth on expression of 17HSD type 1 in JEG-3 choriocarcinoma cells and estrogen-responsive T47D breast cancer cells. Treatment of cultured JEG-3 and T47D cells with all-trans-RA and 9-cis-RA increased reductive 17HSD activity and 17HSD type 1 messenger RNA expression severalfold in both cell lines. On the other hand, epidermal growth factor (EGF), Ca ionophore, the protein kinase C activator 12-O-tetradecanoylphorbol-13-acetate (TPA), and cAMP elevated 17HSD type 1 expression only in JEG-3 cells. Correspondingly, the effects of RAs were potentiated by EGF, TPA, and cAMP in JEG-3 cells, whereas no such phenomenon was observed in T47D cells. In JEG-3 cells, simultaneous administration of RAs with TPA and EGF maximally resulted in approximately 40- and 20-fold increases in 17HSD type 1 messenger RNA expression, respectively. The present data indicate that RAs may stimulate estradiol biosynthesis by regulating 17HSD type 1 expression in certain breast cancer and choriocarcinoma cells. The results suggest that interaction of multiple regulatory pathways is involved in maintaining high 17HSD type 1 expression in the placenta. In addition, regulation of 17HSD type 1 expression may be different in trophoblast cells from that in breast epithelial cells.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Breast Neoplasms/metabolism , Choriocarcinoma/metabolism , Epidermal Growth Factor/pharmacology , Isoenzymes/metabolism , Tretinoin/pharmacology , Uterine Neoplasms/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , Breast Neoplasms/pathology , Choriocarcinoma/pathology , Cyclic AMP/pharmacology , Drug Synergism , Female , Humans , Pregnancy , RNA, Messenger/metabolism , Stimulation, Chemical , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , Uterine Neoplasms/pathology
20.
Mol Endocrinol ; 11(1): 77-86, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8994190

ABSTRACT

Human 17 beta-hydroxysteroid dehydrogenase (17-HSD) type 1 catalyzes the conversion of the low activity estrogen, estrone, into highly active estradiol, both in the gonads and in target tissues. The present study was carried out to characterize the dimerization, microheterogeneity, and phosphorylation of human 17-HSD type 1 and to evaluate the current model of hydride transfer and substrate recognition of the enzyme, based on its x-ray structure. 17-HSD type 1 is a homodimer consisting of noncovalently bound subunits, and the data in the present study indicate an exceptionally strong association between the monomers [dissociation constant (Kd) < 5 pmol/monomers liter]. Furthermore, substitutions constructed at the hydrophobic dimer interface always resulted in inactive aggregates of the protein. The enzyme was shown to be phosphorylated by protein kinase A exclusively at Ser134 only in vitro. However, in contrast to previous suggestions, phosphorylation of Ser134 was shown to play no role in the activity or microheterogeneity of human 17-HSD type 1. The presence of microheterogeneity in the recombinant enzyme also indicates that it does not result from the frequent protein polymorphism previously found for the enzyme. In line with the x-ray structure and the proposed catalytic mechanism of the enzyme, our results indicate that Ser142, Tyr155, and Lys159 are all critical for hydride transfer in human 17-HSD type 1. In contrast, the proposed interaction between His221, Glu282, and the 3-OH group of the steroid at the substrate recognition helix could not be shown to exist. Neither of these residues plays a critical role in the catalytic action of the enzyme in cultured cells.


Subject(s)
Estradiol Dehydrogenases/chemistry , Protein Conformation , Binding Sites , Catalysis , Dimerization , Estradiol/metabolism , Estradiol Dehydrogenases/genetics , Estradiol Dehydrogenases/metabolism , Estrone/metabolism , Humans , Hydrogen Bonding , Isoelectric Focusing , Mutagenesis, Site-Directed , Phosphorylation , Protein Processing, Post-Translational , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Serine/physiology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...