Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Publication year range
1.
FASEB J ; 34(5): 6111-6128, 2020 05.
Article in English | MEDLINE | ID: mdl-32190925

ABSTRACT

Hydroxysteroid (17ß) dehydrogenase type 3 (HSD17B3) deficiency causes a disorder of sex development in humans, where affected males are born with female-appearing external genitalia, but are virilized during puberty. The hormonal disturbances observed in the Hsd17b3 knockout mice (HSD17B3KO), generated in the present study, mimic those found in patients with HSD17B3 mutations. Identical to affected humans, serum T in the adult HSD17B3KO mice was within the normal range, while a striking increase was detected in serum A-dione concentration. This resulted in a marked reduction of the serum T/A-dione ratio, a diagnostic hallmark for the patients with HSD17B3 deficiency. However, unlike humans, male HSD17B3KO mice were born with normally virilized phenotype, but presenting with delayed puberty. In contrast to the current belief, data from HSD17B3KO mice show that the circulating T largely originates from the testes, indicating a strong compensatory mechanism in the absence of HSD17B3. The lack of testicular malignancies in HSD17B3KO mice supports the view that testis tumors in human patients are due to associated cryptorchidism. The HSD17B3KO mice presented also with impaired Leydig cell maturation and signs of undermasculinization in adulthood. The identical hormonal disturbances between HSD17B3 deficient knockout mice and human patients make the current mouse model valuable for understanding the mechanism of the patient phenotypes, as well as endocrinopathies and compensatory steroidogenic mechanisms in HSD17B3 deficiency.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Gonadal Steroid Hormones/blood , Infertility, Male/pathology , Leydig Cells/pathology , Mutation , 17-Hydroxysteroid Dehydrogenases/deficiency , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Female , Infertility, Male/etiology , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Am J Physiol Endocrinol Metab ; 319(3): E494-E508, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32691632

ABSTRACT

Hydroxysteroid 17ß dehydrogenase 12 (HSD17B12) is suggested to be involved in the elongation of very long chain fatty acids. Previously, we have shown a pivotal role for the enzyme during mouse development. In the present study we generated a conditional Hsd17b12 knockout (HSD17B12cKO) mouse model by breeding mice homozygous for a floxed Hsd17b12 allele with mice expressing the tamoxifen-inducible Cre recombinase at the ROSA26 locus. Gene inactivation was induced by administering tamoxifen to adult mice. The gene inactivation led to a 20% loss of body weight within 6 days, associated with drastic reduction in both white (83% males, 75% females) and brown (65% males, 60% females) fat, likely due to markedly reduced food and water intake. Furthermore, the knockout mice showed sickness behavior and signs of liver toxicity, specifically microvesicular hepatic steatosis and increased serum alanine aminotransferase (4.6-fold in males, 7.7-fold in females). The hepatic changes were more pronounced in females than males. Proinflammatory cytokines, such as interleukin-6 (IL-6), IL-17, and granulocyte colony-stimulating factor, were increased in the HSD17B12cKO mice indicating an inflammatory response. Serum lipidomics study showed an increase in the amount of dihydroceramides, despite the dramatic overall loss of lipids. In line with the proposed role for HSD17B12 in fatty acid elongation, we observed accumulation of ceramides, dihydroceramides, hexosylceramides, and lactosylceramides with shorter than 18-carbon fatty acid side chains in the serum. The results indicate that HSD17B12 is essential for proper lipid homeostasis and HSD17B12 deficiency rapidly results in fatal systemic inflammation and lipolysis in adult mice.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Homeostasis/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Behavior, Animal , Body Weight/genetics , Cytokines/metabolism , Fatty Acids/metabolism , Feeding Behavior , Female , Homeostasis/genetics , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Lipidomics , Liver Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Characteristics , Tamoxifen/pharmacology
3.
Horm Metab Res ; 44(13): 949-56, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22864907

ABSTRACT

17ßHSD enzymes catalyze the stereospecific oxidation/reduction at carbon 17ß of androgens and estrogens, and are important players in intracrine sex hormone synthesis. The biological relevance of 17ßHSD14, first named retSDR3, is largely unknown. We generated and validated an antibody targeting the 17ßHSD14 antigen and used this for immunohistochemical evaluation of expression patterns in 33 healthy human tissues. Furthermore, sex steroid conversional activity in HSD17B14 overexpressing HEK293 and MCF10A cells was investigated by assessing interconversion products of estrone, estradiol, androstenedione, testosterone, and dehydroepiandrosterone. Immunohistochemical staining patterns of 17ßHSD14 with the enzyme being primarily expressed in glandular epithelial tissue reveal an enzyme with possible implications in the secretion or conversion of externally derived compounds. A role for 17ßHSD14 in sex steroid metabolism is supported by the finding that 17HSD14 oxidizes both estradiol and testosterone into less bioactive steroid metabolites estrone and androstenedione, respectively.


Subject(s)
17-Hydroxysteroid Dehydrogenases/analysis , 17-Hydroxysteroid Dehydrogenases/physiology , Antibody Specificity , HEK293 Cells , Humans , Immunohistochemistry
4.
Biochim Biophys Acta ; 1781(11-12): 694-702, 2008.
Article in English | MEDLINE | ID: mdl-18773970

ABSTRACT

Mice with inactivation of the D-specific multifunctional protein 2 (MFP2), a crucial enzyme of peroxisomal beta-oxidation, develop multiple pathologies in diverse tissues already starting in the postnatal period. Gene expression profiling performed on liver of 2-day-old pups revealed up-regulation of PPAR alpha responsive genes in knockout mice. Surprisingly, also genes involved in cholesterol biosynthesis were markedly induced. Real-time PCR confirmed the induction of PPAR alpha target genes and of HMGCR and SREBP2, both involved in cholesterol synthesis, in lactating and in adult MFP2 knockout mice. In accordance, the rate of cholesterol biosynthesis was significantly increased in liver of knockout mice but the hepatic cholesterol concentration was unaltered. In MFP2/PPAR alpha double knockout mice, up-regulations of SREBP2 and HMGCR were markedly attenuated. These data demonstrate a tight interrelationship between induction of PPAR alpha by endogenous ligands and up-regulation of genes of cholesterol biosynthesis through increased expression of SREBP2.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Disease Models, Animal , Enoyl-CoA Hydratase/physiology , Liver/metabolism , Multienzyme Complexes/physiology , PPAR alpha/biosynthesis , Sterol Regulatory Element Binding Protein 2/biosynthesis , Animals , Blotting, Western , Cells, Cultured , Cholesterol/biosynthesis , Gene Expression Profiling , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Lactation , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , PPAR alpha/genetics , Peroxisomal Multifunctional Protein-2 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 2/genetics , Up-Regulation
6.
J Endocrinol Invest ; 31(1): 85-91, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18296911

ABSTRACT

The 17beta-hydroxysteroid dehydrogenases (17betaHSD) gene family comprises different enzymes involved in the biosynthesis of active steroid hormones. The 17betaHSD type 3 (17betaHSD3) isoenzyme catalyzes the reductive conversion of the inactive C19-steroid, Delta4-androstenedione (Delta4- A), into the biologically active androgen, testosterone (T), in the Leydig cells of the testis. It is encoded by the 17beta-hydroxysteroid dehydrogenase type 3 (HSD17B3) gene, which maps to chromosome 9q22. Mutations in the HSD17B3 gene are associated with a rare form of 46,XY disorder of sex development referred to as 17betaHSD3 deficiency (or as 17-ketosteroid reductase deficiency), due to impaired testicular conversion of Delta4-A into T. 46,XY patients with 17betaHSD3 deficiency are usually classified as female at birth, raised as such, but develop secondary male features at puberty. Diagnosis, and consequently early treatment, is difficult because clinical signs from birth until puberty may be mild or absent. Biochemical diagnosis of 17betaHSD3 deficiency requires measurement of serum T/Delta4-A ratio after hCG stimulation test in pre-pubertal subjects, while baseline values seem to be informative in early infancy and adolescence. However, low basal T/Delta4-A ratio is not specific for 17betaHSD3 deficiency, being sometimes also found in patients with other defects in T synthesis or with Leydig cells hypoplasia. Mutational analysis of the 17HSDB3 gene is useful in confirming the clinical diagnosis of 17betaHSD3 deficiency. This review describes clinical findings, diagnosis, and molecular basis of this rare disease.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Disorders of Sex Development/diagnosis , Disorders of Sex Development/genetics , Endocrine System/physiology , 17-Hydroxysteroid Dehydrogenases/deficiency , 17-Hydroxysteroid Dehydrogenases/physiology , Amino Acid Sequence , Disorders of Sex Development/physiopathology , Disorders of Sex Development/therapy , Female , Gender Identity , Humans , Male , Molecular Diagnostic Techniques , Mutation , Testis/enzymology
7.
Anim Sci J ; 89(1): 158-166, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28877400

ABSTRACT

The enzyme 3ß-hydroxysteroid dehydrogenase (3ß-HSD) plays an important role in androstenone metabolism in pig liver, and its defective expression is related to the development of boar taint. Early age castration is a common practice in many countries to avoid boar taint, yet whether and how castration affects porcine hepatic 3ß-HSD expression are still poorly understood. In this study, we aimed to compare the expression of 3ß-HSD between intact (boars) and castrated (barrows) male pigs, and to explore the potential factors regulating 3ß-HSD transcription. Compared to barrows, boars showed worse carcass quality. Boars had significantly higher levels of serum androstenone (P < 0.01), testosterone (P < 0.01) and hepatic cortisol (P < 0.05), which were contrary to significantly lower expression of 3ß-HSD messenger RNA (P < 0.01) and protein (P < 0.01) in the liver. Significant differences were detected for the hepatic expression of androgen receptor (AR) and CCAAT/enhancer binding protein ß (C/EBPß). Chromatin immunoprecipitation (ChIP) assay demonstrated reduced histone H3 acetylation (P < 0.05) but increased glucocorticoid receptor (GR) binding to 3ß-HSD gene promoter in boars (P < 0.05). These results indicate that GR binding to 3ß-HSD promoter is involved in the differential hepatic 3ß-HSD expression between boars and barrows.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Castration , Gene Expression , Receptors, Glucocorticoid/metabolism , Swine/genetics , Swine/metabolism , 17-Hydroxysteroid Dehydrogenases/physiology , Acetylation , Androsterone/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Histones/metabolism , Hydrocortisone/metabolism , Liver/metabolism , Male , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Testosterone/metabolism
8.
Biochim Biophys Acta ; 1761(9): 973-94, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16766224

ABSTRACT

The mammalian multifunctional protein-2 (MFP-2, also called multifunctional enzyme 2, D-bifunctional enzyme or 17-beta-estradiol dehydrogenase type IV) was identified by several groups about a decade ago. It plays a central role in peroxisomal beta-oxidation as it handles most, if not all, peroxisomal beta-oxidation substrates. Deficiency of this enzyme in man causes a severe developmental syndrome with abnormalities in several organs but in particular in the brain, leading to death within the first year of life. Accumulation of branched-long-chain fatty acids and very-long-chain fatty acids and a disturbed synthesis of bile acids were documented in these patients. A mouse model with MFP-2 deficiency only partly phenocopies the human disease. Although the expected metabolic abnormalities are present, no neurodevelopmental aberrations are observed. However, the survival of these mice into adulthood allowed to document the importance of this enzyme for the normal functioning of the brain, eyes and testis. In the present review, the identification and biochemical characteristics of MFP-2, and the consequences of MFP-2 dysfunction in humans and in mice will be discussed.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Enoyl-CoA Hydratase/physiology , Models, Molecular , Multienzyme Complexes/physiology , Peroxisomes/enzymology , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/deficiency , Abnormalities, Multiple/enzymology , Abnormalities, Multiple/pathology , Amino Acid Sequence , Animals , Brain/abnormalities , Brain/growth & development , Enoyl-CoA Hydratase/chemistry , Enoyl-CoA Hydratase/deficiency , Eye Abnormalities/enzymology , Eye Abnormalities/pathology , Fatty Acids/metabolism , Humans , Lipid Metabolism , Male , Mice , Mice, Knockout , Molecular Sequence Data , Multienzyme Complexes/chemistry , Multienzyme Complexes/deficiency , Organ Specificity , Peroxisomal Multifunctional Protein-2 , Testis/abnormalities , Testis/growth & development
9.
Reprod Biol Endocrinol ; 5: 30, 2007 Jul 10.
Article in English | MEDLINE | ID: mdl-17623101

ABSTRACT

BACKGROUND: During human pregnancy, the placental villi produces high amounts of estradiol. This steroid is secreted by the syncytium, which is directly in contact with maternal blood. Estradiol has to cross placental foetal vessels to reach foetal circulation. The enzyme 17beta-hydroxysteroid dehydrogenase type 2 (17beta-HSD2) was detected in placental endothelial cells of foetal vessels inside the villi. This enzyme catalyzes the conversion of estradiol to estrone, and of testosterone to androstenedione. It was proposed that estradiol level into foetal circulation could be regulated by 17beta-HSD2. METHODS: We obtained placentas from 10 to 26 6/7 weeks of pregnancy from women undergoing voluntary termination of pregnancy, term placentas were collected after normal spontaneous vaginal deliveries. We quantified 17beta-HSD2 mRNA levels in mid-gestation and term human placenta by RT-QPCR. We produced a new anti-17beta-HSD2 antibody to study its spatio-temporal expression by immunohistochemistry. We also compared steroid levels (testosterone, estrone and estradiol) and 17beta-HSD2 mRNA and protein levels between term placenta and endometrium. RESULTS: High 17beta-HSD2 mRNA and protein levels were found in both mid-gestation and term placentas. However, we showed that 17beta-HSD2 mRNA levels increase by 2.27 fold between mid-gestation and term. This period coincides with a transitional phase in the development of the villous vasculature. In mid-gestation placenta, high levels of 17beta-HSD2 were found in mesenchymal villi and immature intermediate villi, more precisely in endothelial cells of the stromal channel. At term, high levels of 17beta-HSD2 were found in the numerous sinusoidal capillaries of terminal villi. 17beta-HSD2 mRNA and protein levels in term placentas were respectively 25.4 fold and 30 to 60 fold higher than in the endometrium. Steroid levels were also significantly higher in term placenta than in the endometrium. CONCLUSION: The spatial and temporal expression of 17beta-HSD2 in the placenta during pregnancy and the comparison of 17beta-HSD2 expression and steroid levels between placental villi and endometrium are compatible with a role in the modulation of active and inactive forms of estrogens. Our observations strongly support the hypothesis that 17beta-HSD2 acts as a barrier decreasing estradiol secretion rates in the foetal circulation.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Chorionic Villi/enzymology , Pregnancy Trimester, Second/genetics , Pregnancy Trimester, Third/genetics , RNA, Messenger/biosynthesis , 17-Hydroxysteroid Dehydrogenases/biosynthesis , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/physiology , Chorionic Villi/physiology , Estradiol Dehydrogenases , Female , Gene Expression Regulation, Enzymologic , Humans , Pregnancy , RNA, Messenger/analysis , RNA, Messenger/genetics
10.
J Steroid Biochem Mol Biol ; 104(3-5): 334-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17467981

ABSTRACT

Determining the functional aspects of a gene or protein is a difficult and time-consuming process. De novo analysis is surely the hardest and so it is often quite useful to start with a comparison to functionally or structurally related proteins. Although 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD 1) can hardly be called a new protein but rather the best characterized among the family of 17beta-HSDs some aspects of structure-function relationships remain unclear. We have sought new aspects of 17beta-HSD 1 function through a comparison with its closest homolog, a photoreceptor-associated retinol dehydrogenase (prRDH). Overall amino acid identity and size of the proteins are highly conserved, but major differences occur in the C-termini, where prRDH, but not 17beta-HSD 1, harbors motifs indicative of membrane localization. To gain insight into substrate discrimination by prRDH and 17beta-HSD 1, we constructed 3D-structure models of the corresponding zebrafish enzymes. Investigation of the substrate binding site revealed a few identical amino acids, and suggested a role for G143 in zebrafish 17beta-HSD 1 and M146 and M147 in the two zebrafish paralogs prRDH 1 and prRDH 2, respectively, in substrate specificity. Activity measurements of modified proteins in transiently transfected intact HEK 293 cells hint at a putative role of these amino acids in discrimination between steroid and retinoid substrates.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Alcohol Oxidoreductases/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , Alcohol Oxidoreductases/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Retinoids/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Steroids/metabolism , Substrate Specificity , Transfection , Zebrafish/genetics
11.
Mol Cell Endocrinol ; 248(1-2): 61-71, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16414178

ABSTRACT

17Beta-hydroxysteroid dehydrogenases (17beta-HSDs) catalyze the NAD(P)(H) dependent oxidoreduction at C17 oxo/beta-hydroxyl groups of androgen and estrogen hormones. This reversible reaction constitutes an important pre-receptor control mechanism for nuclear receptor ligands, since the conversion "switches" between the 17beta-OH receptor ligands and their inactive 17-oxo metabolites. At present, 14 mammalian 17beta-HSDs are described, of which at least 11 exist within the human genome, encoded by different genes. The enzymes differ in their expression pattern, nucleotide cofactor preference, steroid substrate specificity and subcellular localization, and thus constitute a complex system ensuring cell-specific adaptation and regulation of sex steroid hormone levels. Broad and overlapping substrate specificities with enzymes involved in lipid metabolism suggest interactions of several 17beta-HSDs with other metabolic pathways. Several 17beta-HSDs enzymes constitute promising drug targets, of particular importance in cancer, metabolic diseases, neurodegeneration and possibly immunity.


Subject(s)
17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/physiology , 17-Hydroxysteroid Dehydrogenases/drug effects , Humans , Protein Conformation , Substrate Specificity
12.
Mol Cell Endocrinol ; 248(1-2): 141-8, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16406264

ABSTRACT

Sex steroid hormone signaling regulates the development, growth, and functioning of the breast and the prostate and plays a role in the development and progression of cancer in these organs. The intracellular concentration of active sex steroid hormones in target tissues is regulated by several enzymes, including 17beta-hydroxysteroid dehydrogenases (17HSDs). Changes in the expression patterns of these enzymes may play a pathophysiological role in malignant transformation. We recently analyzed the mRNA expressions of the 17HSD type 1, 2, and 5 enzymes in about 800 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. 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. Cox multivariate analyses showed that 17HSD type 1, tumor size, and estrogen receptor alpha (ERalpha) had independent prognostic significance. We developed, using a LNCaP prostate cancer cell line, a model to study the malignant transformation of prostate cancer and showed that androgen-sensitive LNCaP cells are transformed into neuroendocrine-like cells when cultured without androgens and, eventually into highly proliferating androgen-independent cells. We conducted Northern hybridizations and microarrays to analyze the gene expression during these processes. Substantial changes in the expressions of steroid metabolizing enzymes occurred during the transformation process. The variations in steroid-metabolizing enzymes during cancer progression may be crucial in the regulation of the growth and function of organs.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Breast Neoplasms/enzymology , Cell Proliferation , Gonadal Steroid Hormones/metabolism , Prostatic Neoplasms/enzymology , 17-Hydroxysteroid Dehydrogenases/analysis , 17-Hydroxysteroid Dehydrogenases/genetics , Androgens/metabolism , Breast Neoplasms/pathology , Female , Humans , Male , Prostate/enzymology , Prostate/metabolism
13.
Trends Endocrinol Metab ; 16(4): 167-75, 2005.
Article in English | MEDLINE | ID: mdl-15860413

ABSTRACT

Human 17beta-hydroxysteroid dehydrogenase type 10 (17beta-HSD10) is a mitochondrial enzyme encoded by the SCHAD gene, which escapes chromosome X inactivation. 17Beta-HSD10/SCHAD mutations cause a spectrum of clinical conditions, from mild mental retardation to progressive infantile neurodegeneration. 17Beta-HSD10/SCHAD is essential for the metabolism of isoleucine and branched-chain fatty acids. It can inactivate 17beta-estradiol and steroid modulators of GABA(A) receptors, and convert 5alpha-androstanediol into 5alpha-dihydrotestosterone (DHT). Certain malignant prostatic epithelial cells contain high levels of 17beta-HSD10, generating 5alpha-DHT in the absence of testosterone. 17Beta-HSD10 has an affinity for amyloid-beta peptide, and might be linked to the mitochondrial dysfunction seen in Alzheimer's disease. This versatile enzyme might provide a new drug target for neuronal excitability control and for intervention in Alzheimer's disease and certain cancers.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/etiology , Animals , Gonadal Steroid Hormones/metabolism , Humans , Isoleucine/metabolism , Neurotransmitter Agents/metabolism , Steroids/metabolism
14.
Biochem J ; 376(Pt 1): 49-60, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-12917011

ABSTRACT

17beta-hydroxysteroid dehydrogenases (17beta-HSDs) catalyse the conversion of 17beta-OH (-hydroxy)/17-oxo groups of steroids, and are essential in mammalian hormone physiology. At present, eleven 17beta-HSD isoforms have been defined in mammals, with different tissue-expression and substrate-conversion patterns. We analysed 17beta-HSD type 10 (17beta-HSD10) from humans and Drosophila, the latter known to be essential in development. In addition to the known hydroxyacyl-CoA dehydrogenase, and 3alpha-OH and 17beta-OH activities with sex steroids, we here demonstrate novel activities of 17beta-HSD10. Both species variants oxidize the 20beta-OH and 21-OH groups in C21 steroids, and act as 7beta-OH dehydrogenases of ursodeoxycholic or isoursodeoxycholic acid (also known as 7beta-hydroxylithocholic acid or 7beta-hydroxyisolithocholic acid respectively). Additionally, the human orthologue oxidizes the 7alpha-OH of chenodeoxycholic acid (5beta-cholanic acid, 3alpha,7alpha-diol) and cholic acid (5beta-cholanic acid). These novel substrate specificities are explained by homology models based on the orthologous rat crystal structure, showing a wide hydrophobic cleft, capable of accommodating steroids in different orientations. These properties suggest that the human enzyme is involved in glucocorticoid and gestagen catabolism, and participates in bile acid isomerization. Confocal microscopy and electron microscopy studies reveal that the human form is localized to mitochondria, whereas Drosophila 17beta-HSD10 shows a cytosolic localization pattern, possibly due to an N-terminal sequence difference that in human 17beta-HSD10 constitutes a mitochondrial targeting signal, extending into the Rossmann-fold motif.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxyacyl CoA Dehydrogenases , Bile Acids and Salts/metabolism , Gonadal Steroid Hormones/metabolism , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/physiology , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Drosophila melanogaster/enzymology , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Isoenzymes/physiology , Kinetics , Mitochondria/chemistry , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Steroids/metabolism , Substrate Specificity
15.
Exp Hematol ; 27(3): 451-60, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10089907

ABSTRACT

Hormones such as 1 alpha, 25-dihydroxy vitamin D3 (D3), all-trans retinoic acid, and 9-cis retinoic acid stimulate differentiation of myeloid progenitor cells via their interaction with specific hormone receptors. However, the sensitivity of cells to these agents is not merely governed by the expression of their receptors and the availability of ligand to bind them. Recent studies from our group suggested that the actions of D3 and retinoids on myelopoiesis also are influenced by endogenous mechanisms involving other steroid hormones. In this study we examined the influence of local estrogen metabolism on the differentiation of HL60 cells and normal primitive myeloid progenitor cells. Quantitative thin-layer chromatography (TLC) analyses showed that HL60 and normal cells are able to generate estrone (E1) from estradiol (E2). Neither cell population generated significant amounts of E2 from E1. Reverse transcriptase polymerase chain reaction and Northern analyses confirmed that normal and leukemic myeloid progenitor cells expressed mRNA for the type I and IV isoforms of 17 beta-hydroxysteroid dehydrogenase. Conversion of E2 to E1 was upregulated within 24 hours when HL60 cells were treated with either all-trans retinoic acid or D3 at doses that induce their differentiation toward neutrophils or monocytes, respectively. Similarly, D3-induced monocyte differentiation of normal myeloid progenitor cells was associated with increased capacity to generate E1 from E2. When HL60 cells or normal myeloid progenitor cells were exposed to exogenous E1 they became more sensitive to the differentiation-inducing effects of D3. Data presented provide further evidence for the local modulation of myelopoiesis by intracrine mechanisms. In particular, our findings suggest that local metabolism of steroids by normal as well as leukemic myeloid cells influences their responsiveness to D3 and retinoids.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Estradiol/metabolism , Estrone/metabolism , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Protein Isoforms/physiology , 17-Hydroxysteroid Dehydrogenases/biosynthesis , 17-Hydroxysteroid Dehydrogenases/genetics , Aromatase/metabolism , Cell Differentiation/drug effects , Cholecalciferol/pharmacology , Enzyme Induction/drug effects , Fetal Blood/cytology , Gene Expression Regulation/drug effects , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells/drug effects , HL-60 Cells/enzymology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Humans , Monocytes/cytology , Neoplasm Proteins/metabolism , Neutrophils/cytology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptors, Estrogen/metabolism , Tretinoin/pharmacology
16.
Endocrinology ; 135(5): 1963-71, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7956918

ABSTRACT

Ovarian estradiol (E2) production is regulated by complex interaction of different hormones, such as gonadotropins, steroids, and growth factors. Despite the key role of 17 beta-hydroxysteroid dehydrogenase (17HSD) in E2 biosynthesis, little is known about its regulation in the ovary. Recently, we have characterized the structure of rat 17HSD type 1 and demonstrated that its expression is regulated by gonadotropins and diethylstilbestrol (DES) in rat ovary in vivo. In the present study, the hormonal regulation of 17HSD type 1, and the expression of cytochrome P450 aromatase were examined in parallel in cultured granulosa cells obtained from DES-primed immature rats. Under these conditions, the cells show high expression of 17HSD type 1. Both the enzyme activity and 17HSD type 1 messenger RNA expression in these cells decreased over 2 days of culture in serum-free medium. However, recombinant FSH (recFSH) partially prevented the decreases in enzyme activity and messenger RNA expression in a dose-dependent manner. This effect appears to be mediated by a cAMP-dependent pathway. In contrast to recFSH, neither estrogens (DES or E2) nor androgens (testosterone or dihydrotestosterone) alone affected expression of the enzyme in the cultured cells. However, both estrogens and androgens clearly enhanced the effect of recFSH on 17HSD type 1 expression and 17HSD activity in a dose-dependent manner. Among the growth factors, epidermal growth factor (EGF) has previously been shown to decrease the expression of cytochrome P450 aromatase and E2 biosynthesis in granulosa cells. In the present study, we found that treatment with EGF caused a marked decrease in the effect of recFSH on 17HSD type 1 expression and 17HSD activity. The fact that 17HSD type 1 expression and 17HSD activity always behaved in parallel suggests that 17HSD type 1 is the major 17HSD enzyme involved in estradiol biosynthesis in rat granulosa cells. In conclusion, these data indicate that expression of 17HSD type 1 in rat granulosa cells is under multihormonal regulation. The enzyme is regulated by FSH, via cAMP, and the effect of FSH is modulated by estrogens, androgens, and EGF.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Androgens/pharmacology , Epidermal Growth Factor/pharmacology , Estrogens/pharmacology , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/cytology , Granulosa Cells/enzymology , 17-Hydroxysteroid Dehydrogenases/analysis , Animals , Blotting, Northern , Cells, Cultured , Dose-Response Relationship, Drug , Estradiol/metabolism , Female , Gene Expression Regulation, Enzymologic , Granulosa Cells/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology
17.
J Clin Endocrinol Metab ; 89(11): 5661-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15531526

ABSTRACT

Expression of 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) was compared between leiomyoma and myometrium. Cytosolic fractions from leiomyoma homogenate displayed 5-fold higher activity (estrone to estradiol), compared with surrounding myometrium (n = 6, P < 0.05), whereas microsomal fractions showed no difference. Oxidative activity (estradiol to estrone) did not differ between leiomyoma and myometrium. Levels of mRNA for 17beta-HSDs were then measured using real-time PCR techniques. Among the eight different types of 17beta-HSDs (types 1-5, 7, 8, and 10), type 1 was the only enzyme displaying differential expression between leiomyoma and myometrium. Mean concentration of type 1 17beta-HSD mRNA was 4-fold higher in leiomyoma than in surrounding myometrium (n = 20, P < 0.05). Type 1 transcript levels correlated significantly with reductive activity in individual samples (n = 6, P < 0.05). Northern blot analysis of leiomyoma and myometrium tissues detected 2.3- and 1.0-kb transcripts of type 1 enzyme, whereas the major 1.3-kb transcript for 17beta-HSD in placenta-derived JEG-3 cells was not detected. None of the factors increasing mRNA levels for type 1 enzyme in placenta increased mRNA levels in leiomyoma. These results indicate that leiomyoma tissues overexpress type 1 17beta-HSD, resulting in high conversion of estrone to estradiol. In situ expression of type 1 17beta-HSD may play a role in self-supported growth of leiomyoma cells.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Estradiol/biosynthesis , Leiomyoma/metabolism , Uterine Neoplasms/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , Adult , Estrone/metabolism , Female , Gene Expression Regulation, Enzymologic , Humans , Middle Aged , RNA, Messenger/analysis
18.
J Mol Endocrinol ; 25(1): 1-16, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10915214

ABSTRACT

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.


Subject(s)
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
19.
Mol Cell Endocrinol ; 171(1-2): 1-4, 2001 Jan 22.
Article in English | MEDLINE | ID: mdl-11165003

ABSTRACT

17beta-Hydroxysteroid dehydrogenases (17beta-HSD) are pivotal in controlling the biological potency of steroid hormones by catalyzing oxidation or reduction at position 17. Several 17beta-HSDs may as well metabolize further substrates including alcohols, bile acids, fatty acids and retinols. This review summarizes recent progress in the field of 17beta-HSD research provides an update of nomenclature.


Subject(s)
17-Hydroxysteroid Dehydrogenases , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/physiology , Animals , Humans , Oxidation-Reduction , Substrate Specificity , Terminology as Topic
20.
Mol Cell Endocrinol ; 223(1-2): 43-54, 2004 Aug 31.
Article in English | MEDLINE | ID: mdl-15279910

ABSTRACT

Granulosa cells of small follicles differentiate in vitro in serum-free medium, resulting in increased estradiol secretion and abundance of mRNA encoding cytochrome P450aromatase (P450arom). We tested the hypothesis that differentiation in vitro also involves increased expression of 3beta- and 17beta-hydroxysteroid dehydrogenases (HSD) in the absence of steroidogenic acute regulatory protein (StAR) expression, as has been observed in vivo. Granulosa cells from small (<6 mm diameter) follicles were cultured for up to 6 days, and mRNA levels quantified by Northern hybridization or RT-PCR. Estradiol and progesterone concentrations in medium increased with time in culture, as did mRNA encoding P450arom, 3beta- and 17beta-HSD but not P450scc. Both P450arom and 17beta-HSD were significantly correlated with estradiol accumulation in culture medium. Progesterone secretion was correlated with 3beta-HSD but not P450scc mRNA levels. StAR mRNA was detectable by RT-PCR, did not change with duration of culture and was not correlated with progesterone secretion. FSH significantly stimulated P450arom and 17beta-HSD mRNA levels. Cell origin (from the antral or the basal layer of the membrana granulosa) did not affect steroidogenesis. We conclude that under the present cell culture system granulosa cells do not luteinize, and show expression of key steroidogenic enzymes in patterns similar to those occurring in differentiating follicles in vivo. Further, the data suggest that 17beta-HSD may be as important as P450arom in regulating estradiol secretion, and that 3beta-HSD is more important than P450scc as a regulator of progesterone secretion in non-luteinizing granulosa cells.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Granulosa Cells/metabolism , Phosphoproteins/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , 17-Hydroxysteroid Dehydrogenases/physiology , Animals , Aromatase/genetics , Aromatase/physiology , Cattle , Cell Differentiation , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/physiology , Female , Gene Expression , Gonadal Steroid Hormones/metabolism , Granulosa Cells/enzymology , Phosphoproteins/metabolism , Phosphoproteins/physiology , RNA, Messenger/analysis , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL