Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
Acta Chim Slov ; 71(2): 256-263, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38919102

ABSTRACT

Breast cancer cell growth is often dependent on the presence of steroidal hormones. The 17ß-hydroxysteroid dehydrogenase type 1 isoform (17ßHSD1) catalyzes NADPH-dependent conversion of estrone to estradiol, a more potent estrogen, and represents potential drug target for breast cancer treatment.  To provide active enzyme for inhibitor screening, 17ßHSD1 is usually expressed in insect or mammalian cells, or isolated from human placenta. In the present study we describe a simple protocol for expression and purification of active human 17ßHSD1 from BL21(DE3) Escherichia coli cells. Soluble human 17ßHSD1 was expressed using a pET28a(+)-based plasmid, which encodes a hexahistidine tag fused to the N-terminus of the protein, and purified by nickel affinity chromatography. The enzyme activity of purified 17ßHSD1 was verified by three methods: thin-layer chromatography, an alkali assay and a spectroscopic assay. These non-radioactive enzyme assays require only standard laboratory equipment, and can be used for screening compounds that modulate 17ßHSD1 activity.


Subject(s)
17-Hydroxysteroid Dehydrogenases , Escherichia coli , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , 17-Hydroxysteroid Dehydrogenases/isolation & purification , 17-Hydroxysteroid Dehydrogenases/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/chemistry , Chromatography, Affinity , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/biosynthesis
2.
J Steroid Biochem Mol Biol ; 240: 106510, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38508472

ABSTRACT

The objective of this study was to examine the effect of 11 organochlorine pesticides on human and rat 17ß-Hydroxysteroid dehydrogenase 1 (17ß-HSD1) in human placental and rat ovarian microsome and on estradiol production in BeWo cells. The results showed that the IC50 values for endosulfan, fenhexamid, chlordecone, and rhothane on human 17ß-HSD1 were 21.37, 73.25, 92.80, and 117.69 µM. Kinetic analysis revealed that endosulfan acts as a competitive inhibitor, fenhexamid as a mixed/competitive inhibitor, chlordecone and rhothane as a mixed/uncompetitive inhibitor. In BeWo cells, all insecticides except endosulfan significantly decreased estradiol production at 100 µM. For rats, the IC50 values for dimethomorph, fenhexamid, and chlordecone were 11.98, 36.92, and 109.14 µM. Dimethomorph acts as a mixed inhibitor, while fenhexamid acts as a mixed/competitive inhibitor. Docking analysis revealed that endosulfan and fenhexamid bind to the steroid-binding site of human 17ß-HSD1. On the other hand, chlordecone and rhothane binds to a different site other than the steroid and NADPH-binding site. Dimethomorph binds to the steroid/NADPH binding site, and fenhexamid binds to the steroid binding site of rat 17ß-HSD1. Bivariate correlation analysis showed a positive correlation between IC50 values and LogP for human 17ß-HSD1, while a slight negative correlation was observed between IC50 values and the number of HBA. ADMET analysis provided insights into the toxicokinetics and toxicity of organochlorine pesticides. In conclusion, this study identified the inhibitory effects of 3-4 organochlorine pesticides and binding mechanisms on human and rat 17ß-HSD1, as well as their impact on hormone production.


Subject(s)
Hydrocarbons, Chlorinated , Molecular Docking Simulation , Pesticides , Animals , Humans , Rats , Hydrocarbons, Chlorinated/chemistry , Hydrocarbons, Chlorinated/pharmacology , Structure-Activity Relationship , Female , Pesticides/chemistry , Pesticides/metabolism , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/metabolism , 17-Hydroxysteroid Dehydrogenases/chemistry , Pregnancy , Placenta/metabolism , Estradiol/metabolism , Estradiol/chemistry , Insecticides/chemistry , Insecticides/pharmacology
3.
Ecotoxicol Environ Saf ; 273: 116173, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38452703

ABSTRACT

Per- and polyfluoroalkyl (PFAS) substances are enduring industrial materials. 17ß-Hydroxysteroid dehydrogenase isoform 1 (17ß-HSD1) is an estrogen metabolizing enzyme, which transforms estrone into estradiol in human placenta and rat ovary. Whether PFAS inhibit 17ß-HSD1 and what the structure-activity relationship (SAR) remains unexplored. We screened 18 PFAS for inhibiting human and rat 17ß-HSD1 in microsomes and studied their SAR and mode of action(MOA). Of the 11 perfluorocarboxylic acids (PFCAs), C8-C14 PFCAs at a concentration of 100 µM substantially inhibited human 17ß-HSD1, with order of C11 (half-maximal inhibition concentration, IC50, 8.94 µM) > C10 (10.52 µM) > C12 (14.90 µM) > C13 (30.97 µM) > C9 (43.20 µM) > C14 (44.83 µM) > C8 (73.38 µM) > others. Of the 7 per- and poly-fluorosulfonic acids (PFSAs), the potency was C8S (IC50, 14.93 µM) > C7S (80.70 µM) > C6S (177.80 µM) > others. Of the PFCAs, C8-C14 PFCAs at 100 µM markedly reduced rat 17ß-HSD1 activity, with order of C11 (IC50, 9.11 µM) > C12 (14.30 µM) > C10 (18.24 µM) > C13 (25.61 µM) > C9 (67.96 µM) > C8 (204.39 µM) > others. Of the PFSAs, the potency was C8S (IC50, 37.19 µM) > C7S (49.38 µM) > others. In contrast to PFOS (C6S), the partially fluorinated compound 6:2 FTS with an equivalent number of carbon atoms demonstrated no inhibition of human and rat 17ß-HSD1 activity at a concentration of 100 µM. The inhibition of human and rat enzymes by PFAS followed a V-shaped trend from C4 to C14, with a nadir at C11. Moreover, human 17ß-HSD1 was more sensitive than rat enzyme. PFAS inhibited human and rat 17ß-HSD1 in a mixed mode. Docking analysis revealed that they bind to the NADPH and steroid binding site of both 17ß-HSD1 enzymes. The 3D quantitative SAR (3D-QSAR) showed that hydrophobic region, hydrogen bond acceptor and donor are key factors in binding to 17ß-HSD1 active sites. In conclusion, PFAS exhibit inhibitory effects on human and rat 17ß-HSD1 depending on factors such as carbon chain length, degree of fluorination, and the presence of carboxylic acid or sulfonic acid groups, with a notable V-shaped shift observed at C11.


Subject(s)
Fluorocarbons , Quantitative Structure-Activity Relationship , Pregnancy , Female , Humans , Animals , Rats , Molecular Docking Simulation , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/metabolism , Estrone , Carbon , Fluorocarbons/toxicity
4.
Nat Commun ; 14(1): 5158, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37620305

ABSTRACT

Hydroxysteroid 17-beta-dehydrogenase 13 (HSD17B13) is a hepatic lipid droplet-associated enzyme that is upregulated in patients with non-alcoholic fatty liver disease. Recently, there have been several reports that predicted loss of function variants in HSD17B13 protect against the progression of steatosis to non-alcoholic steatohepatitis with fibrosis and hepatocellular carcinoma. Here we report crystal structures of full length HSD17B13 in complex with its NAD+ cofactor, and with lipid/detergent molecules and small molecule inhibitors from two distinct series in the ligand binding pocket. These structures provide insights into a mechanism for lipid droplet-associated proteins anchoring to membranes as well as a basis for HSD17B13 variants disrupting function. Two series of inhibitors interact with the active site residues and the bound cofactor similarly, yet they occupy different paths leading to the active site. These structures provide ideas for structure-based design of inhibitors that may be used in the treatment of liver disease.


Subject(s)
17-Hydroxysteroid Dehydrogenases , Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Lipid Droplet Associated Proteins , Lipids , 17-Hydroxysteroid Dehydrogenases/chemistry
5.
J Steroid Biochem Mol Biol ; 225: 106191, 2023 01.
Article in English | MEDLINE | ID: mdl-36154887

ABSTRACT

17ß-Hydroxysteroid dehydrogenase type 3 (17ß-HSD3) deficiency is rarely reported in Chinese patients with 46, XY disorders of sexual development (DSD). Seven subjects with 17ß-HSD3 deficiency were identified from 206 Chinese 46, XY DSD patients using targeted next-generation sequencing (NGS). Serum AD and T levels were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS). In silico and functional studies were performed to evaluate the enzymatic activity impairment of HSD17B3 variants. A minigene assay was performed in an exonic splicing variant. Our results showed that four novel and five reported HSD17B3 variants were identified in 7 unrelated patients. The patients showed cryptic presentation during childhood and classical virilization after puberty with T/AD ratio< 0.4. A heterozygous large deletion from the 5'UTR to exon 1 was identified in a patient with a monoallelic variant of p.N130S. Although predicted to be 'likely pathogenic', only p. S232P and p. S160F drastically reduced the enzymatic activity of 17ß-HSD3. A previously reported 'missense' variant c 0.277 G>A (p. E93K) was revealed to have no impact on enzyme activity but resulted in aberrant splicing of exon 3 and was reclassified as an exonic splicing variant. In our study, one nonsense, one exonic splicing, one deletion, one large deletion and five missense variants were detected in patients with 17ß-HSD3 deficiency, expanding the clinical and molecular profile of this disorder. In silico analysis should be cautiously interpreted when the heredity pattern and functional study are inconsistent.


Subject(s)
Disorder of Sex Development, 46,XY , Female , Humans , Disorder of Sex Development, 46,XY/genetics , Chromatography, Liquid , Tandem Mass Spectrometry , 17-Hydroxysteroid Dehydrogenases/chemistry , China
6.
Molecules ; 26(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34885749

ABSTRACT

17ß-Hydroxysteroid dehydrogenase type 3 (17ß-HSD3) is expressed at high levels in testes and seminal vesicles; it is also present in prostate tissue and involved in gonadal and non-gonadal testosterone biosynthesis. The enzyme is membrane-bound, and a crystal structure is not yet available. Selective aryl benzylamine-based inhibitors were designed and synthesised as potential agents for prostate cancer therapeutics through structure-based design, using a previously built homology model with docking studies. Potent, selective, low nanomolar IC50 17ß-HSD3 inhibitors were discovered using N-(2-([2-(4-chlorophenoxy)phenylamino]methyl)phenyl)acetamide (1). The most potent compounds have IC50 values of approximately 75 nM. Compound 29, N-[2-(1-Acetylpiperidin-4-ylamino)benzyl]-N-[2-(4-chlorophenoxy)phenyl]acetamide, has an IC50 of 76 nM, while compound 30, N-(2-(1-[2-(4-chlorophenoxy)-phenylamino]ethyl)phenyl)acetamide, has an IC50 of 74 nM. Racemic C-allyl derivative 26 (IC50 of 520 nM) was easily formed from 1 in good yield and, to determine binding directionality, its enantiomers were separated by chiral chromatography. Absolute configuration was determined using single crystal X-ray crystallography. Only the S-(+)-enantiomer (32) was active with an IC50 of 370 nM. Binding directionality was predictable through our in silico docking studies, giving confidence to our model. Importantly, all novel inhibitors are selective over the type 2 isozyme of 17ß-HSD2 and show <20% inhibition when tested at 10 µM. Lead compounds from this series are worthy of further optimisation and development as inhibitors of testosterone production by 17ß-HSD3 and as inhibitors of prostate cancer cell growth.


Subject(s)
17-Hydroxysteroid Dehydrogenases/chemistry , Benzylamines/chemistry , Prostatic Neoplasms/drug therapy , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/ultrastructure , Benzylamines/chemical synthesis , Benzylamines/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Male , Molecular Docking Simulation , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/pathology , Structure-Activity Relationship , Testosterone/biosynthesis
7.
Bioresour Technol ; 341: 125833, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34455250

ABSTRACT

Testosterone (TS) is a critical androgenic steroid that regulates human metabolism and maintains secondary sexual characteristics. The biotransformation from 4-androstene-3,17-done (4-AD) to TS is limited by the poor catalytic activity of 17ß-hydroxysteroid dehydrogenase type 3 (17ß-HSD3). Herein, we explored the structural characteristics and catalytic mechanism of 17ß-HSD3 and adopted the rational design strategy to improve its catalytic activity. Molecular docking and molecular dynamics simulations revealed the substrate-binding pocket and the binding mode of 4-AD to 17ß-HSD3. We located the pivotal residues and regulated their hydrophobicity and polarity. The obtained G186R/Y195W variant formed additional electrostatic interaction and hydrogen bond with 4-AD, increasing the binding affinity between the variant and 4-AD. Therefore, the G186R/Y195W variant produced 3.98 g/L of TS, which increased to 297%. The combination of structural and mechanism resolution drives the implementation of the rational design strategy, which provides guidance for bioproduction of TS catalyzed by 17ß-HSD3.


Subject(s)
17-Hydroxysteroid Dehydrogenases/chemistry , Molecular Dynamics Simulation , Testosterone , Molecular Docking Simulation , Protein Engineering , Saccharomycetales , Testosterone/biosynthesis
8.
J Steroid Biochem Mol Biol ; 210: 105846, 2021 06.
Article in English | MEDLINE | ID: mdl-33609690

ABSTRACT

A new androsterone derivative bearing a 16ß-picolyl group (compound 5; FCO-586-119) was synthetized in four steps from the lead compound 1 (RM-532-105). We measured its inhibitory activity on 17ß-HSD3 using microsomal fraction of rat testes as well as transfected LNCaP[17ß-HSD3] cells. We then assessed its metabolic stability as well as its cytotoxic effect against a panel of cancer cell lines. The addition of a picolyl moiety at C-16 of RM-532-105 steroid core improves the 17ß-HSD3 inhibitory activity in the microsomal fraction of rat testes, but not in whole LNCaP[17ß-HSD3] cells. Interestingly, this structural modification enhances 3-fold the metabolic stability in conjunction with a significant cytotoxic effect against pancreatic, ovarian, breast, lung, and prostate cancer cells. Because the inhibitory activity data against 17ß-HSD3 suggested that both steroid derivatives are non-competitive inhibitors, we performed docking and molecular dynamics simulations using a homology model of this membrane-associated enzyme. The results of these simulations revealed that both RM-532-105 (1) and FCO-586-119 (5) can compete for the cofactor-binding site displaying better binding energy than NADP+.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Androsterone/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/metabolism , Androstanes/chemistry , Androsterone/analogs & derivatives , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Stability , Enzyme Inhibitors/chemical synthesis , Humans , Magnetic Resonance Spectroscopy , Male , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Rats, Sprague-Dawley , Sulfonamides/chemistry
9.
Biomolecules ; 10(9)2020 08 21.
Article in English | MEDLINE | ID: mdl-32825572

ABSTRACT

Progressive mitochondrial dysfunction due to the accumulation of amyloid beta (Aß) peptide within the mitochondrial matrix represents one of the key characteristics of Alzheimer's disease (AD) and appears already in its early stages. Inside the mitochondria, Aß interacts with a number of biomolecules, including cyclophilin D (cypD) and 17ß-hydroxysteroid dehydrogenase type 10 (17ß-HSD10), and affects their physiological functions. However, despite intensive ongoing research, the exact mechanisms through which Aß impairs mitochondrial functions remain to be explained. In this work, we studied the interactions of Aß with cypD and 17ß-HSD10 in vitro using the surface plasmon resonance (SPR) method and determined the kinetic parameters (association and dissociation rates) of these interactions. This is the first work which determines all these parameters under the same conditions, thus, enabling direct comparison of relative affinities of Aß to its mitochondrial binding partners. Moreover, we used the determined characteristics of the individual interactions to simulate the concurrent interactions of Aß with cypD and 17ß-HSD10 in different model situations associated with the progression of AD. This study not only advances the understanding of Aß-induced processes in mitochondria during AD, but it also provides a new perspective on research into complex multi-interaction biomolecular processes in general.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Mitochondrial Proteins/metabolism , Peptidyl-Prolyl Isomerase F/metabolism , 17-Hydroxysteroid Dehydrogenases/chemistry , Amyloid beta-Peptides/chemistry , Biosensing Techniques , Peptidyl-Prolyl Isomerase F/chemistry , Humans , Mitochondrial Proteins/chemistry , Surface Plasmon Resonance
10.
Mol Nutr Food Res ; 64(16): e2000289, 2020 08.
Article in English | MEDLINE | ID: mdl-32640069

ABSTRACT

SCOPE: Urolithin A and B are gut metabolites of ellagic acid and ellagitannins associated with many beneficial effects. Evidence in vitro pointed to their potential as estrogenic modulators. However, both molecular mechanisms and biological targets involved in such activity are still poorly characterized, preventing a comprehensive understanding of their bioactivity in living organisms. This study aimed at rationally identifying novel biological targets underlying the estrogenic-modulatory activity of urolithins. METHODS AND RESULTS: The work relies on an in silico/in vitro target fishing study coupling molecular modeling with biochemical and cell-based assays. Estrogen sulfotransferase and 17ß-hydroxysteroid dehydrogenase are identified as potentially subject to inhibition by the investigated urolithins. The inhibition of the latter undergoes experimental confirmation either in a cell-free or cell-based assay, validating computational outcomes. CONCLUSIONS: The work describes target fishing as an effective tool to identify unexpected targets of food bioactives detailing the interaction at a molecular level. Specifically, it described, for the first time, 17ß-hydroxysteroid dehydrogenase as a target of urolithins and highlighted the need of further investigations to widen the understanding of urolithins as estrogen modulators in living organisms.


Subject(s)
Coumarins/pharmacology , Enzyme Inhibitors/pharmacology , Estradiol Dehydrogenases/antagonists & inhibitors , Proteins/metabolism , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/metabolism , Cell-Free System , Computer Simulation , Coumarins/chemistry , Coumarins/metabolism , Humans , Ligands , MCF-7 Cells , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Proteins/chemistry , Sulfotransferases/chemistry , Sulfotransferases/metabolism
11.
J Steroid Biochem Mol Biol ; 199: 105586, 2020 05.
Article in English | MEDLINE | ID: mdl-31926269

ABSTRACT

Recent studies have shown that an adrenal steroid 11ß-hydroxy-4-androstene-3,17-dione serves as the precursor to androgens, 11-ketotestosterone and 11-ketodihydrotestosterone (11KDHT). The biosynthetic pathways include the reduction of 3- and 17-keto groups of the androgen precursors 11-keto-C19-steroids, which has been reported to be mediated by three human enzymes; aldo-keto reductase (AKR)1C2, AKR1C3 and 17ß-hydroxysteroid dehydrogenase (HSD) type-3. To explore the contribution of the enzymes in the reductive metabolism, we kinetically compared the substrate specificity for 11-keto-C19-steroids among purified recombinant preparations of four AKRs (1C1, 1C2,1C3 and 1C4) and DHRS11, which shows 17ß-HSD activity. Although AKR1C1 did not reduce the 11-keto-C19-steroids, AKR1C3 and DHRS11 reduced 17-keto groups of 11-keto-4-androstene-3,17-dione, 11-keto-5α-androstane-3,17-dione (11K-Adione) and 11-ketoandrosterone with Km values of 5-28 µM. The 3-keto groups of 11KDHT and 11K-Adione were reduced by AKR1C4 (Km 1 µM) more efficiently than by AKR1C2 (Km 5 and 8 µM, respectively). GC/MS analysis of the products showed that DHRS11 acts as 17ß-HSD, and that AKR1C2 and AKR1C4 are predominantly 3α-HSDs, but formed a minor 3ß-metabolite from 11KDHT. Since DHRS11 was thus newly identified as 11-keto-C19-steroid reductase, we also investigated its substrate-binding mode by molecular docking and site-directed mutagenesis of Thr163 and Val200, and found the following structural features: 1). There is a space that accommodates the 11-keto group of the 11-keto-C19-steroids in the substrate-binding site. 2) Val200 is a critical determinant for exhibiting the strict 17ß-HSD activity of the enzyme, because the Val200Leu mutation resulted in both significant impairment of the 17ß-HSD activity and emergence of 3ß-HSD activity towards 5α-androstanes including 11KDHT.


Subject(s)
17-Hydroxysteroid Dehydrogenases/chemistry , 20-Hydroxysteroid Dehydrogenases/chemistry , Aldo-Keto Reductases/chemistry , Steroids/biosynthesis , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , 20-Hydroxysteroid Dehydrogenases/genetics , 20-Hydroxysteroid Dehydrogenases/metabolism , Aldo-Keto Reductase Family 1 Member C3/chemistry , Aldo-Keto Reductase Family 1 Member C3/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Aldo-Keto Reductases/genetics , Aldo-Keto Reductases/metabolism , Androgens/biosynthesis , Androgens/chemistry , Biosynthetic Pathways/genetics , Humans , Molecular Docking Simulation , Oxidoreductases/chemistry , Oxidoreductases/genetics , Oxidoreductases/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Steroids/chemistry , Substrate Specificity , Testosterone/analogs & derivatives , Testosterone/metabolism
12.
J Steroid Biochem Mol Biol ; 199: 105597, 2020 05.
Article in English | MEDLINE | ID: mdl-31958634

ABSTRACT

Sex steroid hormones play important roles in fish sex differentiation, gonadal development and secondary sexual characteristics. Olive flounder Paralichthys olivaceus is a valuable commercial marine fish species and has marked sexual dimorphism. However, the mechanisms of action of sex hormones in flounder sex are still unclear. In this study, a total of ten Hsd17b family genes, including Hsd17b3, -4, -7, -8, -9, -10, -12a, -12b, -14 and -15, were identified in the flounder, which encoded critical enzymes acting on sex steroid synthesis and metabolism. Hsd17b genes were distributed on eight chromosomes. Hsd17b12a and -12b were located on chromosomes 19 and 7, respectively. It was speculated that these two genes were just highly similar rather than different transcripts derived from the same gene. According to the results of domain and motif analyses, they all belonged to the SDR superfamily and contained conserved Hsd17b motifs TGxxxGxG, PGxxxT, NNAG and YxxxK. Analysis of amino acid sequences predicted that Hsd17b1, -4, -7, -12a and -14 were hydrophilic proteins. The stability of Hsd17b1, -3 and -12b proteins was predicted to be low. The various Hsd17b family genes differed in tissue expression pattern, and Hsd17b10, -12a and -12b were highly expressed in the flounder ovary. Moreover, throughout gonadal development, Hsd17b3 was highly expressed in the testis, and Hsd17b1, -12a and -12b were highly expressed in the ovary, suggesting that they might play an important role in testosterone synthesis in the testis or estrogen synthesis in the ovary. Activities of Hsd17b3 at stages I-V were all significantly higher in the testis than in the ovary (P < 0.05, P < 0.01). Transfection analysis in HEK293T cells showed that Hsd17b1 and -3 were located in both the cytoplasm and nucleus. Additionally, after challenging fish with tamoxifen, Hsd17b3 expression level in the testis decreased significantly (P < 0.01), and in the ovary no significant change was observed. Moreover, the expression of Hsd17b1 in the ovary was significantly upregulated after injection with flutamide (P < 0.05). These findings introduce the characteristics of the flounder Hsd17b in subfamily, which contribute to our understanding of the regulation of sex steroid hormone synthesis in fish gonadal development.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Fish Proteins/genetics , Flounder/genetics , Gonadal Steroid Hormones/genetics , 17-Hydroxysteroid Dehydrogenases/chemistry , Amino Acid Sequence/genetics , Animals , Female , Gene Expression Regulation, Developmental/genetics , Gonadal Steroid Hormones/biosynthesis , Gonads/growth & development , Gonads/metabolism , Male , Multigene Family/genetics , Ovary/growth & development , Ovary/metabolism , Sex Characteristics , Testis/growth & development , Testis/metabolism
13.
Int J Mol Sci ; 21(24)2020 Dec 20.
Article in English | MEDLINE | ID: mdl-33419257

ABSTRACT

In early stages of Alzheimer's disease (AD), amyloid beta (Aß) accumulates in the mitochondrial matrix and interacts with mitochondrial proteins, such as cyclophilin D (cypD) and 17ß-hydroxysteroid dehydrogenase 10 (17ß-HSD10). Multiple processes associated with AD such as increased production or oligomerization of Aß affect these interactions and disbalance the equilibrium between the biomolecules, which contributes to mitochondrial dysfunction. Here, we investigate the effect of the ionic environment on the interactions of Aß (Aß1-40, Aß1-42) with cypD and 17ß-HSD10 using a surface plasmon resonance (SPR) biosensor. We show that changes in concentrations of K+ and Mg2+ significantly affect the interactions and may increase the binding efficiency between the biomolecules by up to 35% and 65% for the interactions with Aß1-40 and Aß1-42, respectively, in comparison with the physiological state. We also demonstrate that while the binding of Aß1-40 to cypD and 17ß-HSD10 takes place preferentially around the physiological concentrations of ions, decreased concentrations of K+ and increased concentrations of Mg2+ promote the interaction of both mitochondrial proteins with Aß1-42. These results suggest that the ionic environment represents an important factor that should be considered in the investigation of biomolecular interactions taking place in the mitochondrial matrix under physiological as well as AD-associated conditions.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/chemistry , Biosensing Techniques/methods , Surface Plasmon Resonance/methods , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/genetics , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Peptidyl-Prolyl Isomerase F/chemistry , Peptidyl-Prolyl Isomerase F/genetics , Humans , Ions/chemistry , Mitochondria/chemistry , Mitochondrial Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/genetics
14.
Molecules ; 24(15)2019 Jul 29.
Article in English | MEDLINE | ID: mdl-31362457

ABSTRACT

: It has long been established that mitochondrial dysfunction in Alzheimer's disease (AD) patients can trigger pathological changes in cell metabolism by altering metabolic enzymes such as the mitochondrial 17ß-hydroxysteroid dehydrogenase type 10 (17ß-HSD10), also known as amyloid-binding alcohol dehydrogenase (ABAD). We and others have shown that frentizole and riluzole derivatives can inhibit 17ß-HSD10 and that this inhibition is beneficial and holds therapeutic merit for the treatment of AD. Here we evaluate several novel series based on benzothiazolylurea scaffold evaluating key structural and activity relationships required for the inhibition of 17ß-HSD10. Results show that the most promising of these compounds have markedly increased potency on our previously published inhibitors, with the most promising exhibiting advantageous features like low cytotoxicity and target engagement in living cells.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/chemistry , Benzothiazoles/chemistry , Urea/chemistry , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Cell Line , Dose-Response Relationship, Drug , Drug Design , Humans , Mitochondria/metabolism , Molecular Structure , Structure-Activity Relationship
15.
J Med Chem ; 62(15): 7070-7088, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31268309

ABSTRACT

Decreasing the intratumoral androgen biosynthesis by using an inhibitor of 17ß-hydroxysteroid dehydrogenase type 3 (17ß-HSD3) is a strategy to treat prostate cancer. The androsterone (ADT) derivative 1 (RM-532-105) has shown strong inhibitory activity on 17ß-HSD3, but needs to be improved. Herein, we describe the chemical synthesis and characterization of two series of analogues to address the impact of A- and D-ring modifications on 17ß-HSD3 inhibitory activity, androgenic effect, and metabolic stability. Structure-activity relationships were generated by adding different groups at C16/C17 (D-ring diversification) or replacing the ADT backbone by a nor-androstane or an estrane backbone (A-ring diversification). D-ring derivatives were less potent inhibitors than lead compound 1, whereas steroidal backbone (A-ring) change led to identifying promising novel estrane derivatives. This culminated with potent 17ß-HSD3 inhibitors 23, 27, 31, and 33 (IC50 = 0.10, 0.02, 0.13, and 0.17 µM, respectively), which did not stimulate LAPC-4 cell proliferation and displayed higher plasma concentration in mice than lead compound 1.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/chemistry , Androsterone/analogs & derivatives , Androsterone/pharmacology , Androsterone/therapeutic use , Animals , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
16.
Curr Biol ; 29(11): 1901-1909.e8, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31130458

ABSTRACT

Vertebrate sex development consists largely of two processes: "sex determination," the initial bifurcation of sexual identity, and "sex differentiation," which subsequently facilitates maleness or femaleness according to the sex determination signal. Steroid hormones promote multiple types of sexual dimorphism in eutherian mammals and avians [1-3], in which they are indispensable for proper sex differentiation. By contrast, in many poikilothermic vertebrates, steroid hormones have been proposed to be key players in sex determination as well as sex differentiation [4-8]. This hypothesis was introduced more than 50 years ago but has never been rigorously tested due to difficulties in discriminating the roles of steroids in sex determination and differentiation. We found that a missense SNP in the gene encoding the steroidogenic enzyme 17ß-hydroxysteroid dehydrogenase 1 (Hsd17b1) was perfectly associated with ZZ/ZW sex determination in Seriola fishes. Biochemical analyses revealed that a glutamate residue present specifically in Z-type HSD17B1 attenuated interconversion between 17-keto and 17ß-hydroxy steroids relative to the allelic product from the W chromosome, which harbors glycine at that position, by disrupting the hydrogen bond network between the steroid and the enzyme's catalytic residues. Hsd17b1 mRNA is constitutively expressed in undifferentiated and differentiating gonads of both genotypic sexes, whereas W-type mRNA is expressed only in genotypic females. Meanwhile, Cyp19a1 is predominantly expressed in differentiating ovary. We conclude that the combination of Hsd17b1 alleles determines sex by modulating endogenous estrogen levels in Seriola species. These findings strongly support the long-standing hypothesis on steroids in sex determination.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Fish Proteins/genetics , Fishes/genetics , Polymorphism, Single Nucleotide , Sex Differentiation/genetics , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/metabolism , Amino Acid Sequence , Animals , Fish Proteins/metabolism , Fishes/growth & development , Phenotype , Phylogeny , Sequence Alignment/veterinary , Sex Determination Processes/genetics
18.
Mol Cell Endocrinol ; 489: 98-106, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30864548

ABSTRACT

The 17ß-hydroxysteroid dehydrogenases (17ß-HSDs) comprise enzymes initially identified by their ability to interconvert active and inactive forms of sex steroids, a vital process for the tissue-specific control of estrogen and androgen balance. However, most 17ß-HSDs have now been shown to accept substrates other than sex steroids, including bile acids, retinoids and fatty acids, thereby playing unanticipated roles in cell physiology. This functional divergence is often reflected by their different subcellular localization, with 17ß-HSDs found in the cytosol, peroxisome, mitochondria, endoplasmic reticulum and in lipid droplets. Moreover, a subset of 17ß-HSDs are integral membrane proteins, with their specific topology dictating the cellular compartment in which they exert their enzymatic activity. Here, we summarize the present knowledge on the subcellular localization and membrane topology of the 17ß-HSD enzymes and discuss the correlation with their biological functions.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Cell Membrane/chemistry , 17-Hydroxysteroid Dehydrogenases/chemistry , Animals , Catalytic Domain , Humans , Lipid Droplets/metabolism , Subcellular Fractions
19.
J Steroid Biochem Mol Biol ; 189: 135-144, 2019 05.
Article in English | MEDLINE | ID: mdl-30836176

ABSTRACT

17ß-Hydroxysteroid dehydrogenase type 14 (17ß-HSD14) catalyzes the conversion of highly active estrogens and androgens into their less active oxidized forms in presence of NAD+ as cofactor. The crystal structure of 17ß-HSD14 has been determined, however, the role of individual amino acids likely involved in the enzymatic function remains poorly understood. Objective of this study was to further characterize the enzyme by site-directed mutagenesis considering five amino acids next to the catalytic center. The tools used for the characterization of the enzyme variants are X-ray crystallography and enzyme kinetics. Lys158 was confirmed to belong to the catalytic triad. Tyr253', located on the C-terminal loop of the adjacent monomer, enters into the active site of the neighboring monomer and interacts with the catalytic Tyr154. Therefore, Tyr253' helps to tie the two monomers together. Cys255, located at the interface between both monomers, can form a disulfide bridge with the Cys255' from the adjacent monomer. In contrast to the contact provided by Tyr253, the latter interaction is not crucial for dimer formation. His93 and Gln148 are located at the rim of the substrate binding pocket. His93 does not interact directly with the ligand in the active site. However, it influences the turnover of the enzyme. The Gln148 restricts in size the access tunnel of the substrate to the binding pocket.


Subject(s)
17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/genetics , Amino Acid Substitution , Crystallography, X-Ray , Enzyme Stability , Humans , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Protein Multimerization
20.
FEBS J ; 286(11): 2155-2166, 2019 06.
Article in English | MEDLINE | ID: mdl-30768851

ABSTRACT

Human 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1) catalyses the last step in estrogen activation and is thus involved in estrogen-dependent diseases (EDDs). Unlike other 17ß-HSD members, 17ß-HSD1 undergoes a significant substrate-induced inhibition that we have previously reported. Here we solved the binary and ternary crystal structures of 17ß-HSD1 in complex with estrone (E1) and cofactor analog NADP+ , demonstrating critical enzyme-substrate-cofactor interactions. These complexes revealed a reversely bound E1 in 17ß-HSD1 that provides the basis of the substrate inhibition, never demonstrated in estradiol complexes. Structural analysis showed that His221 is the key residue responsible for the reorganization and stabilization of the reversely bound E1, leading to the formation of a dead-end complex, which exists widely in NADP(H)-preferred enzymes for the regulation of their enzymatic activity. Further, a new inhibitor is proposed that may inhibit 17ß-HSD1 through the formation of a dead-end complex. This finding indicates a simple mechanism of enzyme regulation in the physiological background and introduces a pioneer inhibitor of 17ß-HSD1 based on the dead-end inhibition model for efficiently targeting EDDs. DATABASES: Coordinates and structure factors of 17ß-HSD1-E1 and 17ß-HSD1-E1-NADP+ have been deposited in the Protein Data Bank with accession code 6MNC and 6MNE respectively. ENZYMES: 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1) EC 1.1.1.62.


Subject(s)
17-Hydroxysteroid Dehydrogenases/chemistry , Estrone/chemistry , NADP/chemistry , Protein Conformation , 17-Hydroxysteroid Dehydrogenases/genetics , Amino Acid Sequence/genetics , Binding Sites/genetics , Catalysis , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Estrogens/chemistry , Estrogens/genetics , Humans , Models, Molecular , Oxidation-Reduction , Protein Binding/genetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...