Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Chembiochem ; 18(1): 77-80, 2017 Jan 03.
Article En | MEDLINE | ID: mdl-27906500

17ß-Hydroxysteroid dehydrogenase (17ß-HSDcl) from the filamentous fungus Curvularia lunata (teleomorph Cochliobolus lunatus) catalyzes NADP(H)-dependent oxidoreductions of androgens and estrogens. Despite detailed biochemical and structural characterization of 17ß-HSDcl, its physiological function remains unknown. On the basis of amino acid sequence alignment, phylogenetic studies, and the recent identification of the physiological substrates of the homologous MdpC from Aspergillus nidulans and AflM from Aspergillus parasiticus, we propose an anthrahydroquinone as the physiological substrate of 17ß-HSDcl. This is also supported by our analysis of a secondary metabolite biosynthetic gene cluster in C. lunata m118, containing 17ß-HSDcl and ten other genes, including a polyketide synthase probably involved in emodin formation. Chemoenzymatic reduction of emodin by 17ß-HSDcl in the presence of sodium dithionite verified this hypothesis. On the basis of these results, the involvement of a 17ß-HSDcl in the biosynthesis of other anthrahydroquinone-derived natural products is proposed; hence, 17ß-HSDcl should be more appropriately referred to as a polyhydroxyanthracene reductase (PHAR).


17-Hydroxysteroid Dehydrogenases/metabolism , Anthraquinones/metabolism , Ascomycota/enzymology , 17-Hydroxysteroid Dehydrogenases/classification , 17-Hydroxysteroid Dehydrogenases/genetics , Anthraquinones/chemistry , Ascomycota/genetics , Biocatalysis , Emodin/chemistry , Emodin/metabolism , Multigene Family , Oxidation-Reduction , Phylogeny
2.
J Steroid Biochem Mol Biol ; 125(1-2): 66-82, 2011 May.
Article En | MEDLINE | ID: mdl-21193039

17ß-Hydroxysteroid dehydrogenases (17ß-HSDs) are oxidoreductases, which play a key role in estrogen and androgen steroid metabolism by catalyzing final steps of the steroid biosynthesis. Up to now, 14 different subtypes have been identified in mammals, which catalyze NAD(P)H or NAD(P)(+) dependent reductions/oxidations at the 17-position of the steroid. Depending on their reductive or oxidative activities, they modulate the intracellular concentration of inactive and active steroids. As the genomic mechanism of steroid action involves binding to a steroid nuclear receptor, 17ß-HSDs act like pre-receptor molecular switches. 17ß-HSDs are thus key enzymes implicated in the different functions of the reproductive tissues in both males and females. The crucial role of estrogens and androgens in the genesis and development of hormone dependent diseases is well recognized. Considering the pivotal role of 17ß-HSDs in steroid hormone modulation and their substrate specificity, these proteins are promising therapeutic targets for diseases like breast cancer, endometriosis, osteoporosis, and prostate cancer. The selective inhibition of the concerned enzymes might provide an effective treatment and a good alternative to the existing endocrine therapies. Herein, we give an overview of functional and structural aspects for the different 17ß-HSDs. We focus on steroidal and non-steroidal inhibitors recently published for each subtype and report on existing animal models for the different 17ß-HSDs and the respective diseases. Article from the Special issue on Targeted Inhibitors.


17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/metabolism , Enzyme Inhibitors/metabolism , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , 17-Hydroxysteroid Dehydrogenases/classification , Amino Acid Sequence , Androgens/chemistry , Androgens/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Catalytic Domain , Enzyme Inhibitors/chemistry , Estrogens/chemistry , Estrogens/metabolism , Female , Humans , Isoenzymes/classification , Male , Models, Molecular , Molecular Sequence Data , Molecular Structure , Phylogeny , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Protein Conformation , Sequence Alignment
4.
Mol Cell Endocrinol ; 301(1-2): 146-53, 2009 Mar 25.
Article En | MEDLINE | ID: mdl-18812208

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.


17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/metabolism , Breast Neoplasms/enzymology , Enzyme Inhibitors/pharmacology , Estradiol/biosynthesis , 17-Hydroxysteroid Dehydrogenases/classification , Breast Neoplasms/genetics , Cell Line, Tumor , Estradiol/chemistry , Estrone/chemistry , Estrone/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity/drug effects
5.
Mol Cell Endocrinol ; 301(1-2): 259-65, 2009 Mar 25.
Article En | MEDLINE | ID: mdl-18775469

17beta-Hydroxysteroid dehydrogenase type 3 (17beta-HSD3) is expressed at high levels in the testes and seminal vesicles but has also been shown to be present in prostate tissue, suggesting its potential involvement in both gonadal and non-gonadal testosterone biosynthesis. The role of 17beta-HSD3 in testosterone biosynthesis makes this enzyme an attractive molecular target for small molecule inhibitors for the treatment of prostate cancer. Here we report the design of selective inhibitors of 17beta-HSD3 as potential anti-cancer agents. Due to 17beta-HSD3 being a membrane-bound protein a crystal structure is not yet available. A homology model of 17beta-HSD3 has been built to aid structure-based drug design. This model has been used with docking studies to identify a series of lead compounds that may give an insight as to how inhibitors interact with the active site. Compound 1 was identified as a potent selective inhibitor of 17beta-HSD3 with an IC(50)=700nM resulting in the discovery of a novel lead series for further optimisation. Using our homology model as a tool for inhibitor design compound 5 was discovered as a novel potent and selective inhibitor of 17beta-HSD3 with an IC(50) approximately 200nM.


17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/pharmacology , 17-Hydroxysteroid Dehydrogenases/classification , Azepines/chemical synthesis , Azepines/chemistry , Azepines/pharmacology , Catalytic Domain , Cell Line , Enzyme Inhibitors/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Piperidines/chemical synthesis , Piperidines/chemistry , Piperidines/pharmacology , Structural Homology, Protein
6.
Mol Cell Endocrinol ; 301(1-2): 251-8, 2009 Mar 25.
Article En | MEDLINE | ID: mdl-18786604

17beta-Hydroxysteroid dehydrogenases (17beta-HSDs) are responsible for the pre-receptor reduction/oxidation of steroids at the 17-position into active/inactive hormones, and the 15 known enzymes vary in their substrate specificity, localisation, and directional activity. 17beta-HSD Type 3 (17beta-HSD3) has been seen to be over-expressed in prostate cancer, and catalyses the reduction of androstenedione (Adione) to testosterone (T), which stimulates prostate tumour growth. Specific inhibitors of 17beta-HSD3 may have a role in the treatment of hormone-dependent prostate cancer and benign prostate hyperplasia, and also have potential as male anti-fertility agents. A 293-EBNA-based cell line with stable expression of transfected human 17beta-HSD3 was created and used to develop a whole cell radiometric TLC-based assay to assess the 17beta-HSD3 inhibitory potency of a series of compounds. STX2171 and STX2624 (IC(50) values in the 200-450nM range) were two of several active inhibitors identified. In similar TLC-based assays these compounds were found to be inactive against 17beta-HSD1 and 17beta-HSD2, indicating selectivity. A novel proof of concept model was developed to study the efficacy of the compounds in vitro using the androgen receptor positive hormone-dependent prostate cancer cell line, LNCaPwt, and its derivative, LNCaP[17beta-HSD3], transfected and selected for stable expression of 17beta-HSD3. The proliferation of the parental cell line was most efficiently stimulated by 5alpha-dihydrotestosterone (DHT), but the LNCaP[17beta-HSD3] cells were equally stimulated by Adione, indicating that 17beta-HSD3 efficiently converts Adione to T in this model. Adione-stimulated proliferation of LNCaP[17beta-HSD3] cells was inhibited in the presence of either STX2171 or STX2624. The compounds alone neither stimulated proliferation of the cells nor caused significant cell death, indicating that they are non-androgenic with low cytotoxicity. STX2171 inhibited Adione-stimulated growth of xenografts established from LNCaPwt cells in castrated mice in vivo. In conclusion, a primary screening assay and proof of concept model have been developed to study the efficacy of 17beta-HSD3 inhibitory compounds, which may have a role in the treatment of hormone-dependent cancer. Active compounds are selective for 17beta-HSD3 over 17beta-HSD1 and 17beta-HSD2, non-androgenic with low toxicity, and efficacious in both an in vitro proof of concept model and in an in vivo tumour model.


17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Drug Evaluation, Preclinical , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Hormones/pharmacology , Prostatic Neoplasms/enzymology , 17-Hydroxysteroid Dehydrogenases/classification , Animals , Antineoplastic Agents/analysis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Enzyme Inhibitors/chemistry , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Transfection , Xenograft Model Antitumor Assays
7.
J Mol Endocrinol ; 35(2): 305-16, 2005 Oct.
Article En | MEDLINE | ID: mdl-16216911

Formation and inactivation of testosterone is performed by various members of the 17beta-hydroxysteroid dehydrogenase (17beta-HSD) family. The main player in testosterone formation is considered to be 17beta-HSD type 3, which catalyzes the reduction of androstenedione to testosterone with high efficiency and is almost exclusively expressed in testis. So far, only the mammalian homologs have been characterized but nothing is known about the role of 17beta-HSD type 3 in other vertebrates. In this study, we describe the identification and characterization of the zebrafish homolog. We found zebrafish 17beta-HSD type 3 to be expressed in embryogenesis from sphere to 84 h post-fertilization. Expression was also detected in various tissues of both male and female adults, but displayed sexual dimorphism. Interestingly, expression was not highest in male testis but in male liver. In female adults, strongest expression was observed in ovaries. At the subcellular level, both human and zebrafish 17beta-HSD type 3 localize to the endoplasmic reticulum. The zebrafish enzyme in vitro effectively catalyzed the conversion of androstenedione to testosterone by use of NADPH as cofactor. Among further tested androgens epiandrosterone and dehydroepiandrosterone were accepted as substrates and reduced at C-17 by the human and the zebrafish enzyme. Androsterone and androstanedione though, were only substrates of human 17beta-HSD type 3, not the zebrafish enzyme. Furthermore, we found that both enzymes can reduce 11-ketoandrostenedione as well as 11beta-hydroxyandrostenedione at C-17 to the respective testosterone forms. Our results suggest that 17beta-HSD type 3 might play slightly different roles in zebrafish compared with human although testosterone itself is likely to have similar functions in both organisms.


17-Hydroxysteroid Dehydrogenases/metabolism , Androgens/metabolism , Isoenzymes/metabolism , Zebrafish/metabolism , 17-Hydroxysteroid Dehydrogenases/classification , 17-Hydroxysteroid Dehydrogenases/genetics , Adult , Amino Acid Sequence , Androgens/chemistry , Animals , Endoplasmic Reticulum/enzymology , Female , Humans , Isoenzymes/classification , Isoenzymes/genetics , Liver/enzymology , Male , Mice , Molecular Sequence Data , Molecular Structure , NADP/metabolism , Phylogeny , Rats , Sequence Alignment , Sex Characteristics , Testis/enzymology , Zebrafish/embryology
8.
J Steroid Biochem Mol Biol ; 93(2-5): 277-83, 2005 Feb.
Article En | MEDLINE | ID: mdl-15860271

Experimental data suggest that sex steroids have a role in the development of breast and prostate cancers. The biological activity of sex steroid hormones in target tissues is regulated by several enzymes, including 17beta-hydroxysteroid dehydrogenases (17HSD). Changes in the expression patterns of these enzymes may significantly modulate the intracellular steroid content and play a pathophysiological role in malignant transformation. To further clarify the role of 17HSDs in breast cancer, we analyzed the mRNA expressions of the 17HSD type 1, 2, and 5 enzymes in 794 breast carcinoma specimens. Both 17HSD type 1 and 2 mRNAs were detected in normal breast tissue from premenopausal women but not in specimens from postmenopausal women. Of the breast cancer specimens, 16% showed signals for 17HSD type 1 mRNA, 25% for type 2, and 65% for type 5. No association between the 17HSD type 1, 2, and 5 expressions was detected. The patients with tumors expressing 17HSD type 1 mRNA or protein had significantly shorter overall and disease-free survival than the other patients. The expression of 17HSD type 5 was significantly higher in breast tumor specimens than in normal tissue. The group with 17HSD type 5 overexpression had a worse prognosis than the other patients. Cox multivariate analyses showed that 17HSD type 1 mRNA, tumor size, and ERalpha had independent prognostic significance. Using an LNCaP prostate cancer cell line, we developed a cell model to study the progression of prostate cancer. In this model, androgen-sensitive LNCaP cells are transformed in culture conditions into more aggressive, androgen-independent cells. The model was used to study androgen and estrogen metabolism during the transformation process. Our results indicate that substantial changes in androgen and estrogen metabolism occur in the cells during the process. A remarkable decrease in oxidative 17HSD activity was seen, whereas reductive activity seemed to increase. Since local steroid metabolism controls the bioavailability of active steroid hormones of target tissues, the variations in steroid-metabolizing enzymes during cancer progression may be crucial in the regulation of the growth and function of organs.


17-Hydroxysteroid Dehydrogenases/metabolism , Cell Transformation, Neoplastic/metabolism , 17-Hydroxysteroid Dehydrogenases/classification , 17-Hydroxysteroid Dehydrogenases/genetics , Androgens/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/etiology , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Estrogens/metabolism , Female , Humans , In Situ Hybridization , Male , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/etiology , Neoplasms, Hormone-Dependent/genetics , Oxidation-Reduction , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/etiology , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
9.
Int J Obes Relat Metab Disord ; 26(2): 165-75, 2002 Feb.
Article En | MEDLINE | ID: mdl-11850747

OBJECTIVES: To investigate (1) whether type 3 17beta-hydroxysteroid dehydrogenase (17beta-HSD), the enzyme which catalyzes the conversion of androstenedione to testosterone in the testis, is co-expressed with P450aromatase in the preadipocytes of women, and (2) whether the relative expression of type 3 17beta-HSD and aromatase varies in subcutaneous abdominal vs intra-abdominal adipose tissue of women. SUBJECTS: Subcutaneous abdominal and intra-abdominal adipose tissue was obtained from women undergoing elective abdominal surgery (age 22-78 y, body mass index (BMI) 22.4-52.9 kg/m(2)). MEASUREMENTS: Expression of type 3 17beta-HSD in adipose cell fractions was determined using RT-PCR. Preadipocyte steroidogenesis was investigated in primary cultures using androstenedione as substrate. Messenger RNA levels for type 3 17beta-HSD and aromatase were measured in adipose tissue from the subcutaneous abdominal and intra-abdominal depots using a quantitative multiplex competitive RT-PCR assay. RESULTS: Type 3 17beta-HSD is co-expressed with aromatase in the abdominal preadipocytes of women. Cultured preadipocytes from both subcutaneous abdominal (n=5) and intra-abdominal (n=5) sites converted androstenedione to testosterone, and there was minimal conversion of androstenedione to estrone. Consistent with this, the levels of type 3 17beta-HSD mRNA were significantly higher than aromatase mRNA at both sites (P<0.05; n=8 subcutaneous abdominal, n=12 intra-abdominal adipose tissue). The ratio of levels of 17beta-HSD mRNA to aromatase mRNA in intra-abdominal adipose tissue was positively correlated with BMI (n=11, r=0.61, P<0.05) and waist circumference (n=10, r=0.65, P<0.05). The converse was found in subcutaneous abdominal adipose tissue. CONCLUSION: The intra-abdominal adipose tissue of women may be substantially androgenic, increasingly so with increasing obesity, particularly central obesity. While androgen production by this adipose tissue deposit may not contribute to circulating testosterone levels due to hepatic clearance, it may have hitherto unrecognised local effects in the intra-abdominal adipose tissue and also on the liver via the hepatic portal system. These studies suggest a mechanism linking central obesity with insulin resistance and dyslipidaemia.


17-Hydroxysteroid Dehydrogenases/biosynthesis , Adipose Tissue/enzymology , Aromatase/biosynthesis , Obesity/enzymology , 17-Hydroxysteroid Dehydrogenases/classification , 17-Hydroxysteroid Dehydrogenases/genetics , Abdomen , Adipocytes , Adult , Aged , Aromatase/genetics , Body Mass Index , DNA Primers , Humans , Male , Middle Aged , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Skin
10.
Mol Biol Evol ; 18(12): 2154-61, 2001 Dec.
Article En | MEDLINE | ID: mdl-11719564

Short-chain alcohol dehydrogenases (SCAD) constitute a large and diverse family of ancient origin. Several of its members play an important role in human physiology and disease, especially in the metabolism of steroid substrates (e.g., prostaglandins, estrogens, androgens, and corticosteroids). Their involvement in common human disorders such as endocrine-related cancer, osteoporosis, and Alzheimer disease makes them an important candidate for drug targets. Recent phylogenetic analysis of SCAD is incomplete and does not allow any conclusions on very ancient divergences or on a functional characterization of novel proteins within this complex family. We have developed a 3D structure-based approach to establish the deep-branching pattern within the SCAD family. In this approach, pairwise superpositions of X-ray structures were used to calculate similarity scores as an input for a tree-building algorithm. The resulting phylogeny was validated by comparison with the results of sequence-based algorithms and biochemical data. It was possible to use the 3D data as a template for the reliable determination of the phylogenetic position of novel proteins as a first step toward functional predictions. We were able to discern new patterns in the phylogenetic relationships of the SCAD family, including a basal dichotomy of the 17beta-hydroxysteroid dehydrogenases (17beta-HSDs). These data provide an important contribution toward the development of type-specific inhibitors for 17beta-HSDs for the treatment and prevention of disease. Our structure-based phylogenetic approach can also be applied to increase the reliability of evolutionary reconstructions in other large protein families.


17-Hydroxysteroid Dehydrogenases/classification , Alcohol Dehydrogenase/classification , Alcohol Dehydrogenase/genetics , Phylogeny , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Crystallography, X-Ray , Databases, Protein , Humans , Likelihood Functions
11.
J Mol Endocrinol ; 25(1): 1-16, 2000 Aug.
Article En | MEDLINE | ID: mdl-10915214

In women and men, an important proportion of estrogens and androgens are synthesized locally at their site of action in peripheral target tissues. This new field of endocrinology has been called intracrinology. In postmenopausal women, 100% of active sex steroids are synthesized in peripheral target tissues from inactive steroid precursors while, in adult men, approximately 50% of androgens are made locally in intracrine target tissues. The last and key step in the formation of all estrogens and androgens is catalyzed by members of the family of 17beta-hydroxysteroid dehydrogenases (17 beta-HSDs) while different 17 beta-HSDs inactivate these steroids in the same cell where synthesis takes place. To date, seven human 17 beta-HSDs have been cloned, sequenced and characterized. The 17 beta-HSDs provide each cell with the means of precisely controlling the intracellular concentration of each sex steroid according to local needs.


17-Hydroxysteroid Dehydrogenases/physiology , Endocrinology , 17-Hydroxysteroid Dehydrogenases/classification , 17-Hydroxysteroid Dehydrogenases/genetics , Adrenal Glands/physiology , Adult , Androgens/biosynthesis , Breast Neoplasms/physiopathology , Dehydroepiandrosterone/metabolism , Estrogens/biosynthesis , Female , Genitalia/physiology , Humans , Male , Middle Aged , Neoplasms, Hormone-Dependent/physiopathology
12.
J Mol Endocrinol ; 23(1): 1-11, 1999 Aug.
Article En | MEDLINE | ID: mdl-10431140

A number of enzymes possessing 17beta-hydroxysteroid dehydrogenase/17-ketosteroid reductase (17HSD/KSR) activities have been described and cloned, but their nomenclature needs specification. To clarify the present situation, descriptions of the eight cloned 17HSD/KSRs are given and guidelines for the classification of novel 17HSD/KSR enzymes are presented.


17-Hydroxysteroid Dehydrogenases/metabolism , Terminology as Topic , 17-Hydroxysteroid Dehydrogenases/classification , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Humans
13.
J Clin Endocrinol Metab ; 83(1): 187-94, 1998 Jan.
Article En | MEDLINE | ID: mdl-9435439

Human adipose tissue is known to have 17 beta-oxidoreductase activity, interconverting estrone (E1) and estradiol (E2), as well as androstenedione (A) and testosterone (T). We examined both the subcutaneous abdominal and intra-abdominal (visceral) adipose tissue of women for expression of types 1, 2, and 3 17 beta-hydroxysteroid dehydrogenase (17 beta-HSD) using ribonuclease (RNase) protection assay and RT-PCR/Southern blotting. Type 1 17 beta-HSD, which encodes the enzyme responsible for the conversion of E1 to E2 in the placenta and ovary, was expressed in the subcutaneous abdominal and intra-abdominal adipose tissue of women, but the messenger RNA transcripts were predominantly incompletely spliced and therefore unlikely to encode an active protein. A pseudogene for type 1 17 beta-HSD was also expressed in these tissues, but messenger RNA transcripts were again unspliced. Type 2 17 beta-HSD, which encodes an enzyme that can catalyze the conversion of T to A and E2 to E1, was expressed in both the subcutaneous abdominal and intra-abdominal adipose tissue of women. Type 3 17 beta-HSD was also expressed in adipose tissue from both sites studied. Type 3 17 beta-HSD encodes the enzyme that catalyzes the conversion of A to T in the testis and also converts E1 to E2. Together with aromatase, which is known to be expressed in adipose tissue, the expression of types 2 and 3 17 beta-HSD indicates that sex steroid production in the adipose tissue of women is a complex process. The association of visceral obesity with the development of insulin resistance and dyslipidaemia raises the question of the role of steroid production in adipose tissue in the pathogenesis of these disorders.


17-Hydroxysteroid Dehydrogenases/biosynthesis , Adipose Tissue/enzymology , Transcription, Genetic , 17-Hydroxysteroid Dehydrogenases/classification , 17-Hydroxysteroid Dehydrogenases/genetics , Abdomen , Adolescent , Adult , Aged , Blotting, Southern , Female , Humans , Middle Aged , Ovary/enzymology , Placenta/enzymology , Polymerase Chain Reaction , Pregnancy , Pseudogenes , RNA Splicing , RNA, Messenger/biosynthesis , Skin
14.
Mol Pharmacol ; 50(5): 1157-66, 1996 Nov.
Article En | MEDLINE | ID: mdl-8913347

To better understand the molecular mechanisms of the pleiotropic responses induced by exposure to peroxisome proliferator chemicals (PPCs), we conducted a systematic search for genes whose mRNA levels are modulated by the PPC WY-14,643 (WY) in rat liver. The sequence of one up-regulated cDNA (2480 bp) was predicted to encode a protein of 735 aa with 82% identity to the porcine 17 beta-hydroxysteroid dehydrogenase type IV (HSD IV). Like the porcine enzyme, the rat HSD IV contains' a region homologous to yeast hydratase-dehydrogenase-epimerases and to sterol carrier proteins, indicating that the rat HSD IV has broad substrate specificity and contributes to cholesterol metabolism. The rat HSD IV was regulated by diverse PPCs via two distinct mechanisms. Induction of HSD IV and acyl-CoA oxidase (ACO) proteins in rat liver at different treatment times and concentrations of gemfibrozil and di-n-butyl phthalate were almost identical, indicating that HSD IV mRNA induction involves the peroxisome proliferator-activated receptor alpha, a regulator of ACO. In contrast, HSD IV protein levels were only weakly induced by WY, a strong inducer of ACO protein, even though the levels of HSD IV and ACO mRNA were strongly stimulated by WY and gemfibrozil. Thus, HSD IV protein levels were uniquely regulated pretranslationally by WY via a novel mechanism. Increased conversion of estradiol to the less-active estrone by HSD IV induction may explain how phthalate exposure leads to decreases in serum estradiol levels and suppression of ovulation.


17-Hydroxysteroid Dehydrogenases/genetics , Anticholesteremic Agents/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Isoenzymes/genetics , Microbodies/physiology , Pyrimidines/pharmacology , 17-Hydroxysteroid Dehydrogenases/biosynthesis , 17-Hydroxysteroid Dehydrogenases/classification , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , DNA, Complementary/metabolism , Enzyme Induction/drug effects , Estradiol/metabolism , Gene Expression Regulation, Enzymologic/physiology , Isoenzymes/biosynthesis , Liver/drug effects , Liver/enzymology , Liver/physiology , Male , Microbodies/drug effects , Microbodies/enzymology , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/drug effects , Transcription Factors/physiology , Up-Regulation/drug effects
15.
Bioessays ; 18(1): 63-70, 1996 Jan.
Article En | MEDLINE | ID: mdl-8593166

11beta-hydroxysteroid dehydrogenases regulate glucocorticoid concentrations and 17beta-hydroxysteroid dehydrogenases regulate estrogen and androgen concentrations in mammals. Phylogenetic analysis of the sequences from two 11beta-hydroxysteroid dehydrogenases and four mammalian 17beta-hydroxysteroid dehydrogenases indicates unusual evolution in these enzymes. Type 1 11beta- and 17beta-hydroxysteroid dehydrogenases are on the same branch; Type 2 enzymes cluster on another branch with beta-hydroxybutyrate dehydrogenase,11-cis-retinol dehydrogenase and retinol dehydrogenase; Type 3 17beta-hydroxysteroid dehydrogenase is on a third branch; while the pig dehydrogenase clusters with a yeast multifunctional enzyme on a fourth branch. Pig 17beta-hydroxysteroid dehydrogenase appears to have evolved independently from the other three 17beta-hydroxysteroid dehydrogenases; in which case, the evolution of 17beta-hydroxysteroid dehydrogenase activity is an example of functional convergence. The phylogeny also suggests that independent evolution of specificity toward C11 substituents on glucocorticoids and C17 substituents on androgens and estrogens has occurred in Types 1 and 2 11beta- and 17beta-hydroxysteroid dehydrogenases.


17-Hydroxysteroid Dehydrogenases/genetics , Alcohol Oxidoreductases/genetics , Evolution, Molecular , Hydroxysteroid Dehydrogenases/genetics , Mammals/genetics , 11-beta-Hydroxysteroid Dehydrogenases , 17-Hydroxysteroid Dehydrogenases/classification , Alcohol Oxidoreductases/classification , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cattle , Cytochrome P450 Family 2 , Escherichia coli/enzymology , Escherichia coli/genetics , Fatty Acids/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Hydroxybutyrate Dehydrogenase/genetics , Hydroxysteroid Dehydrogenases/classification , Mammals/metabolism , Multigene Family , Phylogeny , Rats , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Species Specificity , Steroids/metabolism , Swine/genetics , Swine/metabolism , Vitamin A/metabolism
16.
Endocrinology ; 135(6): 2629-34, 1994 Dec.
Article En | MEDLINE | ID: mdl-7988452

17 beta-Hydroxysteroid dehydrogenase type 1 (17HSD type 1) is a steroidogenic enzyme that catalyzes the reversible interconversion of estrone and estradiol. In this study, we investigated the roles of epidermal growth factor (EGF) and tumor growth factor-alpha (TGF alpha) in the regulation of 17HSD type 1 gene expression and catalytic activity in cultured JAR, JEG-3, and BeWo choriocarcinoma cells. EGF and TGF alpha increased 17HSD type 1 protein concentrations in JAR and JEG-3 cells, as measured by time-resolved immunofluorometric assay, and 17HSD catalytic activity, as determined by production of estradiol from estrone. These increases were accompanied by parallel increases in concentrations of the 1.3-kilobase messenger RNA coding for 17HSD type 1 in these cells. EGF receptor tyrosine kinase activity inhibitors, tyrphostins, inhibited EGF action in JEG-3 cells, indicating that tyrosine kinase activity is needed for stimulation of the 17HSD type 1 gene. Treatment with 8-bromo-cAMP or phorbol 12-myristate 13-acetate increased the amount of 17HSD type 1 protein. Furthermore, phorbol 12-myristate 13-acetate potentiated the stimulatory effect of EGF. These results suggest that EGF and/or TGF alpha may play an important role in 17HSD type 1 regulation and, consequently, in estrogen production in the human placenta.


17-Hydroxysteroid Dehydrogenases/metabolism , Choriocarcinoma/enzymology , Epidermal Growth Factor/pharmacology , Transforming Growth Factor alpha/pharmacology , Tyrphostins , 17-Hydroxysteroid Dehydrogenases/classification , 17-Hydroxysteroid Dehydrogenases/genetics , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Catechols/pharmacology , Choriocarcinoma/pathology , Humans , Nitriles/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
17.
Genomics ; 23(1): 250-2, 1994 Sep 01.
Article En | MEDLINE | ID: mdl-7829082

EDH17B2, the gene encoding 17 beta-hydroxysteroid dehydrogenase type 1, has been suggested as a candidate for the familial breast cancer gene, BRCA1, located on 17q12-q21. We analyzed the promoter region of EDH17B2 in DNA from 20 control individuals and 40 patients with familial breast cancer. Two frequent (designated vI and vIII) and two rare (vII and vIV) nucleotide variations were present in both the breast cancer patients and the controls, except the alteration vII, which was found only in one patient. Although the data do not support the identification of EDH17B2 as the BRCA1 gene, it is of interest that point mutation vIV (A-->C) was located in the putative TATA box of the EDH17B2 gene. Reporter gene analyses showed that the mutation vIV decreases EDH17B2 promoter activity by an average of 45% in in vitro assays, suggesting that nucleotide A at position -27 is significant for efficient transcription.


17-Hydroxysteroid Dehydrogenases/genetics , Breast Neoplasms/genetics , Chromosomes, Human, Pair 17 , Isoenzymes/genetics , Neoplasm Proteins , Neoplastic Syndromes, Hereditary/genetics , Point Mutation , TATA Box , Transcription Factors , 17-Hydroxysteroid Dehydrogenases/classification , BRCA1 Protein , Base Sequence , Breast Neoplasms/enzymology , DNA Mutational Analysis , DNA, Neoplasm/genetics , Genes , Genes, Reporter , Genetic Linkage , Genetic Testing , Humans , Molecular Sequence Data , Polymorphism, Single-Stranded Conformational , Transcription, Genetic
...