1
|
Adekunle YA, Samuel BB, Oluyemi WM, Adewumi AT, Mosebi S, Nahar L, Fatokun AA, Sarker SD. Oleanolic acid purified from the stem bark of Olax subscorpioidea Oliv. inhibits the function and catalysis of human 17 β-hydroxysteroid dehydrogenase 1. J Biomol Struct Dyn 2024:1-14. [PMID: 39485270 DOI: 10.1080/07391102.2024.2423173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 06/04/2024] [Indexed: 11/03/2024]
Abstract
Cancer is a leading cause of global death. Medicinal plants have gained increasing attention in cancer drug discovery. In this study, the stem bark extract of Olax subscorpioidea, which is used in ethnomedicine to treat cancer, was subjected to phytochemical investigation leading to the isolation of oleanolic acid (OA). The structure was elucidated by 1-dimensional and 2-dimensional nuclear magnetic resonance spectroscopic (NMR) data, and by comparing its data with previously reported data. Molecular docking was used to investigate the interactions of OA with nine selected cancer-related protein targets. OA docked well with human 17β-hydroxysteroid dehydrogenase type-1 (17βHSD1), caspase-3, and epidermal growth factor receptor tyrosine kinase (binding affinities: -9.8, -9.3, and -9.1 kcal/mol, respectively). OA is a triterpenoid compound with structural similarity to steroids. This similarity with the substrates of 17βHSD1 gives the inhibitor candidate an excellent opportunity to bind to 17βHSD1. The structural and functional dynamics of OA-17βHSD1 were investigated by molecular dynamics simulations at 240 ns. Molecular mechanics/Poisson-Boltzmann surface area (MMPBSA) studies showed that OA had a binding free energy that is comparable with that of vincristine (-52.76, and -63.56 kcal/mol, respectively). The average C-α root mean square of deviation (RMSD) value of OA (1.69 Å) compared with the unbound protein (2.01 Å) indicated its high stability at the protein's active site. The binding energy and stability at the active site of 17βHSD1 recorded in this study indicate that OA exhibited profound inhibitory potential. OA could be a good scaffold for developing new anti-breast cancer drugs.
Collapse
Affiliation(s)
- Yemi A Adekunle
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Babatunde B Samuel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Wande M Oluyemi
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Adeniyi T Adewumi
- Department of Life and Consumer Sciences, University of South Africa, Florida, South Africa
| | - Salerwe Mosebi
- Department of Life and Consumer Sciences, University of South Africa, Florida, South Africa
| | - Lutfun Nahar
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czech Republic
| | - Amos A Fatokun
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Satyajit D Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
2
|
Wen C, Chen H, Tang Y, Lin H, Xu C, Ying Y, Zhu Y, Miao X, Ge RS, Chen C, Chen S. Per- and polyfluoroalkyl substances inhibit human and rat 17β-hydroxysteroid dehydrogenase 1: Quantitative structure-activity relationship and molecular docking analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116173. [PMID: 38452703 DOI: 10.1016/j.ecoenv.2024.116173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/09/2024]
Abstract
Per- and polyfluoroalkyl (PFAS) substances are enduring industrial materials. 17β-Hydroxysteroid dehydrogenase isoform 1 (17β-HSD1) is an estrogen metabolizing enzyme, which transforms estrone into estradiol in human placenta and rat ovary. Whether PFAS inhibit 17β-HSD1 and what the structure-activity relationship (SAR) remains unexplored. We screened 18 PFAS for inhibiting human and rat 17β-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 17β-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 17β-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 17β-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 17β-HSD1 was more sensitive than rat enzyme. PFAS inhibited human and rat 17β-HSD1 in a mixed mode. Docking analysis revealed that they bind to the NADPH and steroid binding site of both 17β-HSD1 enzymes. The 3D quantitative SAR (3D-QSAR) showed that hydrophobic region, hydrogen bond acceptor and donor are key factors in binding to 17β-HSD1 active sites. In conclusion, PFAS exhibit inhibitory effects on human and rat 17β-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.
Collapse
Affiliation(s)
- Chao Wen
- Department of Neonatal Paediatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Huan Chen
- Department of Emergency, the Dingli Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yunbing Tang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Hang Lin
- Department of Anesthesiology and Perioperative Medicine, the Second Affiliated Hospital and Yuying Children's Hospital, Key Laboratory of Pediatric Anesthesiology, Ministry of Education; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Congcong Xu
- Department of Neonatal Paediatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yingfen Ying
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yang Zhu
- Key Laboratory of Structural Malformations in Children of Zhejiang Province and Key Laboratory of Male Health and Environment of Wenzhou, Zhejiang Province 325000, China
| | - Xinjun Miao
- Department of Emergency, the Dingli Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Ren-Shan Ge
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Department of Anesthesiology and Perioperative Medicine, the Second Affiliated Hospital and Yuying Children's Hospital, Key Laboratory of Pediatric Anesthesiology, Ministry of Education; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province and Key Laboratory of Male Health and Environment of Wenzhou, Zhejiang Province 325000, China.
| | - Chao Chen
- Department of Neonatal Paediatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | - Shangqin Chen
- Department of Neonatal Paediatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
3
|
Guo L, Malara D, Battaglia P, Waiho K, Davis DA, Deng Y, Shen Z, Rao K. Turnovers of Sex-Determining Mutation in the Golden Pompano and Related Species Provide Insights into Microevolution of Undifferentiated Sex Chromosome. Genome Biol Evol 2024; 16:evae037. [PMID: 38408866 PMCID: PMC10919887 DOI: 10.1093/gbe/evae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 02/28/2024] Open
Abstract
The suppression of recombination is considered a hallmark of sex chromosome evolution. However, previous research has identified undifferentiated sex chromosomes and sex determination by single SNP in the greater amberjack (Seriola dumerili). We observed the same phenomena in the golden pompano (Trachinotus ovatus) of the same family Carangidae and discovered a different sex-determining SNP within the same gene Hsd17b1. We propose an evolutionary model elucidating the turnover of sex-determining mutations by highlighting the contrasting dynamics between purifying selection, responsible for maintaining W-linked Hsd17b1, and neutral evolution, which drives Z-linked Hsd17b1. Additionally, sporadic loss-of-function mutations in W-linked Hsd17b1 contribute to the conversion of W chromosomes into Z chromosomes. This model was directly supported by simulations, closely related species, and indirectly by zebrafish mutants. These findings shed new light on the early stages of sex chromosome evolution.
Collapse
Affiliation(s)
- Liang Guo
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan Province 410081, China
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong Province 510300, China
| | - Danilo Malara
- Stazione Zoologica Anton Dohrn, Integrated Marine Ecology Department, CRIMAC, Calabria Marine Centre, Amendolara 87071, Italy
| | - Pietro Battaglia
- Integrated Marine Ecology Department, Stazione Zoologica Anton Dohrn, Sicily Marine Centre, Messina 98168, Italy
| | - Khor Waiho
- Higher Institution Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu 21300, Malaysia
- Centre for Chemical Biology, Universiti Sains Malaysia, Minden 11900, Malaysia
- Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand
| | - D Allen Davis
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36830, USA
| | - Yu Deng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan Province 410081, China
| | - Zhongyuan Shen
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan Province 410081, China
| | - Ke Rao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan Province 410081, China
| |
Collapse
|
4
|
Ibrahim AM, Hamed MT, EL-Khadragy MF, Morad MY. Molluscicidal activity of sodium hypochlorite against Biomphlaria alexandrina snails: Immunological and hepato-endocrine alterations with in silico docking study. Parasite Epidemiol Control 2023; 23:e00331. [PMID: 38148891 PMCID: PMC10750182 DOI: 10.1016/j.parepi.2023.e00331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/18/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023] Open
Abstract
Schistosomiasis is a tropical disease that widely neglected. Schistosoma mansoni reproduce asexually within the freshwater snail, Biomphlaria alexandrina. Sodium hypochlorite (NaOCl) is a widely used disinfectant, so its effect against gainst B. alexandrina snails was evaluated. The present results showed that NaOCl has a molluscicidal activity against adult B. alexandrina snails at LC50 1.25 ppm. Hemocytes displayed varied morphological forms after being exposed to the LC10 and LC25 concentrations of NaOCl in B. alexandrina snails, and the phagocytic index of B. alexandrina snail's hemocytes significantly increased. The phagocytic potency of exposed hemocytes to charcoal showed ruptured plasma membrane, engulfed particles, vacuolation in the cytoplasm and degeneration of nuclei. When B. alexandrina snails were treated with sublethal concentrations of NaOCl, transaminases (AST & ALT), alkaline and acid phosphatase activities were significantly increased. In contrast, the total protein, albumin concentrations, Testosterone (T) and 17β Estradiol (E) showed a significant decrease (p ≤ 0.05) as compared to the control groups. The molecular docking interaction showed high efficiency for the ligand, NaOCl against the receptor binding sites of the acid phosphatase, alanine aminotransferase, estrogen and testosterone. The present results showed that NaOCl could be used as an effective molluscicide against B. alexandrina snails but more attention should be paid to investigate the side effects on the non-target organisms living in the freshwater environment.
Collapse
Affiliation(s)
- Amina M. Ibrahim
- Medical Malacology Department, Theodor Bilharz Research Institute, Giza 12411, Egypt
| | - Mohamed T. Hamed
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt
| | - Manal F. EL-Khadragy
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mostafa Y. Morad
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt
| |
Collapse
|
5
|
Chen S, Wang S, Zheng J, Lu H, Chen H, Tang Y, Wang N, Zhu Y, Wang Y, Duan P, Ge RS. Bisphenol analogues inhibit human and rat 17β-hydroxysteroid dehydrogenase 1: 3D-quantitative structure-activity relationship (3D-QSAR) and in silico docking analysis. Food Chem Toxicol 2023; 181:114052. [PMID: 37758047 DOI: 10.1016/j.fct.2023.114052] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
Bisphenols, estrogenic endocrine-disrupting chemicals, disrupt at least one of three endocrine pathways (estrogen, androgen, and thyroid). 17β-Hydroxysteroid dehydrogenase 1 (17β-HSD1) is a steroidogenic enzyme that catalyzes the activation of estradiol from estrone in human placenta and rat ovary. However, whether bisphenols inhibit 17β-HSD1 and the mode of action remains unclear. This study we screened 17 bisphenols for inhibiting human 17β-HSD1 in placental microsomes and rat 17β-HSD1 in ovarian microsomes and determined 3D-quantitative structure-activity relationship (3D-QSAR) and mode of action. We observed some bisphenols with substituents were found to significantly inhibit both human and rat 17β-HSD1 with the most potent inhibition on human enzyme by bisphenol H (IC50 = 0.90 μM) when compared to bisphenol A (IC50 = 113.38 μM). Rat enzyme was less sensitive to the inhibition of bisphenols than human enzyme with bisphenol H (IC50 = 32.94 μM) for rat enzyme. We observed an inverse correlation between IC50 and hydrophobicity (expressed as Log P). Docking analysis showed that they bound steroid-binding site of 17β-HSD1. The 3D-QSAR models demonstrated that hydrophobic region, hydrophobic aromatic, ring aromatic, and hydrogen bond acceptor are key factors for the inhibition of steroid synthesis activity of 17β-HSD1.
Collapse
Affiliation(s)
- Sailing Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Shaowei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jingyi Zheng
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Han Lu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Huiqian Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yunbing Tang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Nan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yang Zhu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yiyan Wang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Ping Duan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Ren-Shan Ge
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province and Key Laboratory of Male Health and Environment of Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
6
|
Li T, Song X, Stephen P, Yin H, Lin SX. New insights into the substrate inhibition of human 17β-hydroxysteroid dehydrogenase type 1. J Steroid Biochem Mol Biol 2023; 228:106246. [PMID: 36634828 DOI: 10.1016/j.jsbmb.2023.106246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 12/25/2022] [Accepted: 01/09/2023] [Indexed: 01/11/2023]
Abstract
Human type 1 17β-hydroxysteroid dehydrogenase (17β-HSD1),a member of the short-chain dehydrogenase/reductase family, catalyzes the last step in the bioactivation of the most potent estrogen estradiol with high specificity and is thus involved in estrogen-dependent diseases. As an oxidoreductase, 17β-HSD1 can utilize both triphosphate and diphosphate cofactors in reaction at the molecular level, but more specific with triphosphate cofactor. The NADPH is much higher than NADP+ in living cells leading to preliminary reduction action. The enzyme also showed substrate-induced inhibition unprecedented in other members of 17β-HSDs. Our previous study elucidated the structural mechanism of substrate inhibition is due to the reversely bound estrone (E1) in the substrate-binding pocket of the enzyme resulting in a dead-end complex. However, the effect of the cofactor preference on the substrate inhibition of the enzyme is not yet clear. In the present study, we solved the ternary crystal structures of 17β-HSD1 in complex with E1 and cofactor analog NAD+ . Combined with molecular dynamics simulation using the enzyme with NADH/NADPH and different oriented E1 (normally oriented, E1N; reversely oriented, E1R), such ternary structure provides a complete picture of enzyme-substrate-cofactor interactions. The results reveal that different cofactors and substrate binding mode affect the allosteric effect between the two subunits of the enzyme. And the results from MD simulations confirmed that His221 plays a key role in the formation of dead-end complex in NADPH complex, and the absence of stable interaction between His221 and E1R in the NADH complex should be the main reason for its lack of substrate inhibition.
Collapse
Affiliation(s)
- Tang Li
- Dalian Engineering Research Center for Carbohydrate Agricultural Preparations, Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; CHU de Québec Research Center and Department of Molecular Medicine, Laval University, Québec, QC, Canada.
| | - Xiaohui Song
- Dalian Engineering Research Center for Carbohydrate Agricultural Preparations, Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Preyesh Stephen
- CHU de Québec Research Center and Department of Molecular Medicine, Laval University, Québec, QC, Canada
| | - Heng Yin
- Dalian Engineering Research Center for Carbohydrate Agricultural Preparations, Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Sheng-Xiang Lin
- CHU de Québec Research Center and Department of Molecular Medicine, Laval University, Québec, QC, Canada.
| |
Collapse
|
7
|
Ngueta AD, Roy J, Maltais R, Poirier D. Chemical Synthesis and Biological Evaluation of 3-Substituted Estrone/Estradiol Derivatives as 17β-Hydroxysteroid Dehydrogenase Type 1 Inhibitors Acting via a Reverse Orientation of the Natural Substrate Estrone. Molecules 2023; 28:molecules28020632. [PMID: 36677690 PMCID: PMC9862175 DOI: 10.3390/molecules28020632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Estradiol (E2) plays an important role in the progression of diseases such as breast cancer and endometriosis. Inhibition of 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1), the enzyme that catalyzes the last step in the biosynthesis of the estrogenic hormone E2, therefore constitutes an interesting approach for the treatment of these two estrogen-dependent diseases. In order to obtain new inhibitors of 17β-HSD1, the impact of a m-carbamoylphenyloxy group at position three of an estrane nucleus was evaluated by preparing three derivatives of estrone (E1) and E2 using a microwave-assisted synthesis of diaryl ethers. Their inhibitory activity was addressed on two cell lines (T-47D and Z-12) representative of breast cancer and endometriosis, respectively, but unlike T-47D cells, Z-12 cells were not found suitable for testing potential 17β-HSD1 inhibitors. Thus, the addition of the m-carbamoylphenyl group at C3 of E1 (compound 5) did not increase the inhibition of E1 to E2 transformation by 17β-HSD1 present in T-47D cells (IC50 = 0.31 and 0.21 μM for 5 and E1, respectively), and this negative effect was more obvious for E2 derivatives 6 and 10 (IC50 = 1.2 and 1.3 μM, respectively). Molecular docking allowed us to identify key interactions with 17β-HSD1 and to highlight these new inhibitors' actions through an opposite orientation than natural enzyme substrate E1's classical one. Furthermore, molecular modeling experiments explain the better inhibitory activity of E1-ether derivative 5, as opposed to the E2-ether derivatives 6 and 10. Finally, when tested on T-47D and Z-12 cells, compounds 5, 6 and 10 did not stimulate the proliferation of these two estrogen-dependent cell lines. In fact, they reduced it.
Collapse
Affiliation(s)
- Adrien Djiemeny Ngueta
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec Research Center—Université Laval, Quebec, QC G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec Research Center—Université Laval, Quebec, QC G1V 4G2, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec Research Center—Université Laval, Quebec, QC G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec Research Center—Université Laval, Quebec, QC G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec, QC G1V 0A6, Canada
- Correspondence:
| |
Collapse
|
8
|
Lou D, Liu X, Tan J. An Overview of 7α- and 7β-Hydroxysteroid Dehydrogenases: Structure, Specificity and Practical Application. Protein Pept Lett 2021; 28:1206-1219. [PMID: 34397319 DOI: 10.2174/0929866528666210816114032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/27/2021] [Accepted: 06/17/2021] [Indexed: 11/22/2022]
Abstract
7α-Hydroxysteroid dehydrogenase and 7β-hydroxysteroid dehydrogenase are key enzymes involved in bile acid metabolism. They catalyze the epimerization of a hydroxyl group through 7-keto bile acid intermediates. Basic research of the two enzymes has focused on exploring new enzymes and the structure-function relationship. The application research focused on the in vitro biosynthesis of bile acid drugs and the exploration and improvement of their catalytic ability based on molecular engineering. This article summarized the primary and advanced structural characteristics, specificities, biochemical properties, and applications of the two enzymes. The emphasis is also given to obtaining of novel 7α-hydroxysteroid dehydrogenase and 7β-hydroxysteroid dehydrogenase that are thermally stable and active in the presence of organic solvents, high substrate concentration, and extreme pH values. To achieve these goals, enzyme redesigning based on protein engineering and genomics may be the most useful approaches.
Collapse
Affiliation(s)
- Deshuai Lou
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China
| | - Xi Liu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China
| |
Collapse
|
9
|
Sager CP, Weber S, Negri M, Banachowicz P, Möller G, Adamski J, Hartmann RW, Marchais-Oberwinkler S. Homology modeling meets site-directed mutagenesis: An ideal combination to elucidate the topology of 17β-HSD2. J Steroid Biochem Mol Biol 2021; 206:105790. [PMID: 33246154 DOI: 10.1016/j.jsbmb.2020.105790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/03/2020] [Accepted: 11/18/2020] [Indexed: 11/17/2022]
Abstract
17β-Hydroxysteroid dehydrogenase type 2 (17β-HSD2) catalyzes the conversion of highly active estrogens and androgens into their less active forms using NAD+ as cofactor. Substrate and cofactor specificities of 17β-HSD2 have been reported and potent 17β-HSD2 inhibitors have been discovered in a ligand-based approach. However, the molecular basis and the amino acids involved in the enzymatic functionality are poorly understood, as no crystal structure of the membrane-associated 17β-HSD2 exists. The functional properties of only few amino acids are known. The lack of topological information impedes structure-based drug design studies and limits the design of biochemical experiments. The aim of this work was the determination of the 17β-HSD2 topology. For this, the first homology model of 17β-HSD2 in complex with NAD+ and 17β-estradiol was built, using a multi-fragment "patchwork" approach. To confirm the quality of the model, fifteen selected amino acids were exchanged one by one using site directed mutagenesis. The mutants' functional behavior demonstrated that the generated model was of very good quality and allowed the identification of several key amino acids involved in either ligand or internal structure stabilization. The final model is an optimal basis for further experiments like, for example, lead optimization.
Collapse
Affiliation(s)
- Christoph P Sager
- Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg 6, 35037 Marburg, Germany
| | - Susanne Weber
- Helmholtz Zentrum München, Research Unit Molecular Endocrinology and Metabolism, 85764 Neuherberg, Germany
| | - Matthias Negri
- Helmholtz Institute for Pharmaceutical Research Saarland, Campus E8.1, 66123 Saarbrücken, Germany
| | - Pauline Banachowicz
- Helmholtz Zentrum München, Research Unit Molecular Endocrinology and Metabolism, 85764 Neuherberg, Germany
| | - Gabriele Möller
- Helmholtz Zentrum München, Research Unit Molecular Endocrinology and Metabolism, 85764 Neuherberg, Germany
| | - Jerzy Adamski
- Helmholtz Zentrum München, Research Unit Molecular Endocrinology and Metabolism, 85764 Neuherberg, Germany; Lehrstuhl für Experimentelle Genetik, Technische Universität München, 85356 Freising-Weihenstephan, Germany; German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Rolf W Hartmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Campus E8.1, 66123 Saarbrücken, Germany; Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123 Saarbrücken, Germany
| | - Sandrine Marchais-Oberwinkler
- Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg 6, 35037 Marburg, Germany; Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123 Saarbrücken, Germany.
| |
Collapse
|
10
|
Zhang CY, Calvo EL, Yang CQ, Liu J, Sang XY, Lin SX. Transcriptome of 17β-hydroxysteroid dehydrogenase type 2 plays both hormone-dependent and hormone-independent roles in MCF-7 breast cancer cells. J Steroid Biochem Mol Biol 2019; 195:105471. [PMID: 31513846 DOI: 10.1016/j.jsbmb.2019.105471] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/06/2019] [Accepted: 09/07/2019] [Indexed: 12/11/2022]
Abstract
Breast cancer is a major cause of cancer-related death for women in western countries. 17β-Hydroxysteroid dehydrogenases (17β-HSDs) play important roles in the last step of sex-hormone activation and the first step of sex-hormone inactivation. 17β-HSD2 is responsible for oxidizing the sex hormones. We used microarray technology to analyze the effect of 17β-HSD2 on the MCF-7 cell transcript profile after knocking down 17β-HSD2. Five hundred forty-two genes were regulated 1.5-fold or higher after treatment with 17β-HSD2 siRNA. Knocking down 17β-HSD2 interrupted nucleosome assembly. Pathway-Act-Network analysis showed that the MAPK and apoptosis signaling pathways were most regulated. In the gene-gene interaction network analysis, UGT2B15, which is involved in hormone metabolism, was the most regulated core gene. FOS, GREB1, and CXCL12 were the most regulated genes, and CXCL12 was related to tumor migration. Following 17β-HSD2 knock-down, the cell viability decreased to 75.9%. The S-phase percentage decreased by 19.4%, the Q2-phase percentage in cell apoptosis testing increased by 1.5 times, and cell migration decreased to 66.0%. These results were consistent with our gene chip analysis and indicated that 17β-HSD2 plays both hormone-dependent and hormone-independent enzymatic roles. In-depth investigations of this enzyme on the genomic level will help clarify its related molecular mechanisms.
Collapse
Affiliation(s)
- Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, PR China; Laboratory of Molecular Endocrinology and Oncology, Centre Hospitalier Universitaire de Québec Research Centre (CHUQ, CHUL) and Department of Molecular Medicine, Québec G1V 4G2, Canada
| | - Ezequiel-Luis Calvo
- Laboratory of Molecular Endocrinology and Oncology, Centre Hospitalier Universitaire de Québec Research Centre (CHUQ, CHUL) and Department of Molecular Medicine, Québec G1V 4G2, Canada
| | - Chang-Qing Yang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, PR China
| | - Jie Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, PR China
| | - Xiao-Ye Sang
- Laboratory of Molecular Endocrinology and Oncology, Centre Hospitalier Universitaire de Québec Research Centre (CHUQ, CHUL) and Department of Molecular Medicine, Québec G1V 4G2, Canada
| | - Sheng-Xiang Lin
- Laboratory of Molecular Endocrinology and Oncology, Centre Hospitalier Universitaire de Québec Research Centre (CHUQ, CHUL) and Department of Molecular Medicine, Québec G1V 4G2, Canada.
| |
Collapse
|
11
|
Crystallographic Studies of Steroid-Protein Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1135:27-45. [PMID: 31098809 DOI: 10.1007/978-3-030-14265-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Steroid molecules have a wide range of function in eukaryotes, including the control and maintenance of membranes, hormonal control of transcription, and intracellular signaling. X-ray crystallography has served as a successful tool for gaining understanding of the structural and mechanistic aspects of these functions by providing snapshots of steroids in complex with various types of proteins. These proteins include nuclear receptors activated by steroid hormones, several families of enzymes involved in steroid synthesis and metabolism, and proteins involved in signaling and trafficking pathways. Proteins found in some bacteria that bind and metabolize steroids have been investigated as well. A survey of the steroid-protein complexes that have been studied using crystallography and the insight learned from them is presented.
Collapse
|
12
|
Heinosalo T, Saarinen N, Poutanen M. Role of hydroxysteroid (17beta) dehydrogenase type 1 in reproductive tissues and hormone-dependent diseases. Mol Cell Endocrinol 2019; 489:9-31. [PMID: 30149044 DOI: 10.1016/j.mce.2018.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/14/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022]
Abstract
Abnormal synthesis and metabolism of sex steroids is involved in the pathogenesis of various human diseases, such as endometriosis and cancers arising from the breast and uterus. Steroid biosynthesis is a multistep enzymatic process proceeding from cholesterol to highly active sex steroids via different intermediates. Human Hydroxysteroid (17beta) dehydrogenase 1 (HSD17B1) enzyme shows a high capacity to produce the highly active estrogen, estradiol, from a precursor hormone, estrone. However, the enzyme may also play a role in other steps of the steroid biosynthesis pathway. In this article, we have reviewed the literature on HSD17B1, and summarize the role of the enzyme in hormone-dependent diseases in women as evidenced by preclinical studies.
Collapse
Affiliation(s)
- Taija Heinosalo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| | - Niina Saarinen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Matti Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland; Institute of Medicine, The Sahlgrenska Academy, Gothenburg University, 413 45, Gothenburg, Sweden
| |
Collapse
|
13
|
Tchernof A, Lin SX, Luu-The V, Simard J. The Passing of Dr. Fernand Labrie (1937-2019): The legacy of excellence in science. J Steroid Biochem Mol Biol 2019; 189:63-64. [PMID: 30779933 DOI: 10.1016/j.jsbmb.2019.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
14
|
Badran MJ, Bertoletti N, Keils A, Heine A, Klebe G, Marchais-Oberwinkler S. Mutational and structural studies uncover crucial amino acids determining activity and stability of 17β-HSD14. J Steroid Biochem Mol Biol 2019; 189:135-144. [PMID: 30836176 DOI: 10.1016/j.jsbmb.2019.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 01/31/2023]
Abstract
17β-Hydroxysteroid dehydrogenase type 14 (17β-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 17β-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.
Collapse
Affiliation(s)
- Mohammed J Badran
- Institute for Pharmaceutical Chemistry, Philipps University Marburg, 35032 Marburg, Germany
| | - Nicole Bertoletti
- Institute for Pharmaceutical Chemistry, Philipps University Marburg, 35032 Marburg, Germany
| | - Aline Keils
- Institute for Pharmaceutical Chemistry, Philipps University Marburg, 35032 Marburg, Germany
| | - Andreas Heine
- Institute for Pharmaceutical Chemistry, Philipps University Marburg, 35032 Marburg, Germany
| | - Gerhard Klebe
- Institute for Pharmaceutical Chemistry, Philipps University Marburg, 35032 Marburg, Germany
| | | |
Collapse
|
15
|
Li T, Stephen P, Zhu DW, Shi R, Lin SX. Crystal structures of human 17β-hydroxysteroid dehydrogenase type 1 complexed with estrone and NADP + reveal the mechanism of substrate inhibition. FEBS J 2019; 286:2155-2166. [PMID: 30768851 DOI: 10.1111/febs.14784] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/28/2019] [Accepted: 02/13/2019] [Indexed: 12/22/2022]
Abstract
Human 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) catalyses the last step in estrogen activation and is thus involved in estrogen-dependent diseases (EDDs). Unlike other 17β-HSD members, 17β-HSD1 undergoes a significant substrate-induced inhibition that we have previously reported. Here we solved the binary and ternary crystal structures of 17β-HSD1 in complex with estrone (E1) and cofactor analog NADP+ , demonstrating critical enzyme-substrate-cofactor interactions. These complexes revealed a reversely bound E1 in 17β-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 17β-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 17β-HSD1 based on the dead-end inhibition model for efficiently targeting EDDs. DATABASES: Coordinates and structure factors of 17β-HSD1-E1 and 17β-HSD1-E1-NADP+ have been deposited in the Protein Data Bank with accession code 6MNC and 6MNE respectively. ENZYMES: 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) EC 1.1.1.62.
Collapse
Affiliation(s)
- Tang Li
- Axe Molecular Endocrinology and Nephrology, CHU de Québec Research Center, Department of Molecular Medicine, Laval University, Québec, Canada
| | - Preyesh Stephen
- Axe Molecular Endocrinology and Nephrology, CHU de Québec Research Center, Department of Molecular Medicine, Laval University, Québec, Canada
| | - Dao-Wei Zhu
- Axe Molecular Endocrinology and Nephrology, CHU de Québec Research Center, Department of Molecular Medicine, Laval University, Québec, Canada
| | - Rong Shi
- Département de Biochimie, de Microbiologie et de Bio-Informatique, IBIS et PROTEO, Université Laval, Pavillon Charles-Eugène Marchand, Québec, Canada
| | - Sheng-Xiang Lin
- Axe Molecular Endocrinology and Nephrology, CHU de Québec Research Center, Department of Molecular Medicine, Laval University, Québec, Canada
| |
Collapse
|
16
|
Li F, Zhu Z, Xue M, He W, Zhang T, Feng L, Lin S. siRNA-based breast cancer therapy by suppressing 17β-hydroxysteroid dehydrogenase type 1 in an optimized xenograft cell and molecular biology model in vivo. Drug Des Devel Ther 2019; 13:757-766. [PMID: 30863015 PMCID: PMC6391152 DOI: 10.2147/dddt.s180836] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose Hormone-dependent breast cancer is the most common form of breast cancer, and inhibiting 17β-HSD1 can play an attractive role in decreasing estrogen and cancer cell proliferation. However, the majority of existing inhibitors have been developed from estrogens and inevitably possess residual estrogenicity. siRNA knockdown provides a highly specific way to block a targeted enzyme, being especially useful to avoid estrogenicity. Application of 17β-HSD1-siRNA in vivo is limited by the establishment of an animal model, as well as the potential nuclease activity in vivo. We tried to reveal the in vivo potential of 17β-HSD1-siRNA-based breast cancer therapy. Materials and methods To establish a competent animal model, daily subcutaneous injection of an estrone micellar aqueous solution was adopted to provide the substrate for estradiol biosynthesis. The effects of three different doses of estrone (0.1, 0.5, and 2.5 µg/kg/day) on tumor growth in T47D-17β-HSD1-inoculated group were investigated and compared with the animals inoculated with wild type T47D cells. To solve in vivo delivery problem of siRNA, “17β-HSD1-siRNA/LPD”, a PEGylated and modified liposome–polycation–DNA nanoparticle containing 17β-HSD1-siRNA was prepared by the thin film hydration method and postinsertion technology. Finally, “17β-HSD1-siRNA/LPD” was tested in the optimized model. Tumor growth and 17β-HSD1 expression were assessed. Results Comparison with the untreated group revealed significant suppression of tumor growth in “17β-HSD1-siRNA/LPD”-treated group when HSD17B1 gene expression was knocked down. Conclusion These findings showed promising in vivo assessments of 17β-HSD1-siRNA candidates. This is the first report of an in vivo application of siRNA for steroid-converting enzymes in a nude mouse model.
Collapse
Affiliation(s)
- Fang Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, and Shanghai Engineer and Technology Research Center of Reproductive Health Drug and Devices, Shanghai 200032, China,
| | - ZhiHan Zhu
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, and Shanghai Engineer and Technology Research Center of Reproductive Health Drug and Devices, Shanghai 200032, China,
| | - Man Xue
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, and Shanghai Engineer and Technology Research Center of Reproductive Health Drug and Devices, Shanghai 200032, China,
| | - WanHong He
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, and Shanghai Engineer and Technology Research Center of Reproductive Health Drug and Devices, Shanghai 200032, China,
| | - Ting Zhang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, and Shanghai Engineer and Technology Research Center of Reproductive Health Drug and Devices, Shanghai 200032, China,
| | - LingLin Feng
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, and Shanghai Engineer and Technology Research Center of Reproductive Health Drug and Devices, Shanghai 200032, China,
| | - ShengXiang Lin
- Axe Molecular Endocrinology and Nephrology, CHU Research Center and Department of Molecular Medicine, Laval University, Québec, G1V 4G2, QC, Canada,
| |
Collapse
|
17
|
Li T, Maltais R, Poirier D, Lin SX. Combined Biophysical Chemistry Reveals a New Covalent Inhibitor with a Low-Reactivity Alkyl Halide. J Phys Chem Lett 2018; 9:5275-5280. [PMID: 30148957 DOI: 10.1021/acs.jpclett.8b02225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) plays a pivotal role in the progression of estrogen-related diseases because of its involvement in the biosynthesis of estradiol (E2), constituting a valuable therapeutic target for endocrine treatment. In the present study, we successfully cocrystallized the enzyme with the reversible inhibitor 2-methoxy-16β-( m-carbamoylbenzyl)-E2 (2-MeO-CC-156) as well as the enzyme with the irreversible inhibitor 3-(2-bromoethyl)-16β-( m-carbamoylbenzyl)-17β-hydroxy-1,3,5(10)-estratriene (PBRM). The structures of ternary complexes of 17β-HSD1-2-MeO-CC-156-NADP+ and 17β-HSD1-PBRM-NADP+ comparatively show the formation of a covalent bond between His221 and the bromoethyl side chain of the inhibitor in the PBRM structure. A dynamic process including beneficial molecular interactions that favor the specific binding of a low-reactivity inhibitor and subsequent N-alkylation event through the participation of His221 in the enzyme catalytic site clearly demonstrates the covalent bond formation. This finding opens the door to a new design of alkyl halide-based specific covalent inhibitors as potential therapeutic agents for different enzymes, contributing to the development of highly efficient inhibitors.
Collapse
Affiliation(s)
- Tang Li
- CHU de Québec - Research Center , 2705 Boulevard Laurier , Québec , QC G1V 4G2 , Canada
- Faculty of Medicine , Université Laval , Québec , QC G1V 0A6 , Canada
| | - René Maltais
- CHU de Québec - Research Center , 2705 Boulevard Laurier , Québec , QC G1V 4G2 , Canada
| | - Donald Poirier
- CHU de Québec - Research Center , 2705 Boulevard Laurier , Québec , QC G1V 4G2 , Canada
- Faculty of Medicine , Université Laval , Québec , QC G1V 0A6 , Canada
| | - Sheng-Xiang Lin
- CHU de Québec - Research Center , 2705 Boulevard Laurier , Québec , QC G1V 4G2 , Canada
- Faculty of Medicine , Université Laval , Québec , QC G1V 0A6 , Canada
| |
Collapse
|
18
|
Li T, Zhu D, Labrie F, Lin S. Crystal Structures of Human 17<i>β</i>-Hydroxysteroid Dehydrogenase Type 1 Complexed with the Dual-Site Inhibitor EM-139. Health (London) 2018. [DOI: 10.4236/health.2018.108081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
19
|
Clinical, genetic, and structural basis of apparent mineralocorticoid excess due to 11β-hydroxysteroid dehydrogenase type 2 deficiency. Proc Natl Acad Sci U S A 2017; 114:E11248-E11256. [PMID: 29229831 DOI: 10.1073/pnas.1716621115] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mutations in 11β-hydroxysteroid dehydrogenase type 2 gene (HSD11B2) cause an extraordinarily rare autosomal recessive disorder, apparent mineralocorticoid excess (AME). AME is a form of low renin hypertension that is potentially fatal if untreated. Mutations in the HSD11B2 gene result either in severe AME or a milder phenotype (type 2 AME). To date, ∼40 causative mutations have been identified. As part of the International Consortium for Rare Steroid Disorders, we have diagnosed and followed the largest single worldwide cohort of 36 AME patients. Here, we present the genotype and clinical phenotype of these patients, prominently from consanguineous marriages in the Middle East, who display profound hypertension and hypokalemic alkalosis. To correlate mutations with phenotypic severity, we constructed a computational model of the HSD11B2 protein. Having used a similar strategy for the in silico evaluation of 150 mutations of CYP21A2, the disease-causing gene in congenital adrenal hyperplasia, we now provide a full structural explanation for the clinical severity of AME resulting from each known HSD11B2 missense mutation. We find that mutations that allow the formation of an inactive dimer, alter substrate/coenzyme binding, or impair structural stability of HSD11B2 yield severe AME. In contrast, mutations that cause an indirect disruption of substrate binding or mildly alter intramolecular interactions result in type 2 AME. A simple in silico evaluation of novel missense mutations could help predict the often-diverse phenotypes of an extremely rare monogenic disorder.
Collapse
|
20
|
Mendez-Arroyo J, d’Aquino AI, Chinen AB, Manraj YD, Mirkin CA. Reversible and Selective Encapsulation of Dextromethorphan and β-Estradiol Using an Asymmetric Molecular Capsule Assembled via the Weak-Link Approach. J Am Chem Soc 2017; 139:1368-1371. [DOI: 10.1021/jacs.6b10027] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Jose Mendez-Arroyo
- Department of Chemistry and
International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Andrea I. d’Aquino
- Department of Chemistry and
International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Alyssa B. Chinen
- Department of Chemistry and
International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Yashin D. Manraj
- Department of Chemistry and
International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A. Mirkin
- Department of Chemistry and
International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
21
|
Lin SX, Shi R, Hu XJ, Penning TM. Current physico-biochemistry in steroid research and status of structural biology for steroid-converting enzymes. J Steroid Biochem Mol Biol 2016; 161:1-4. [PMID: 27196263 PMCID: PMC5278676 DOI: 10.1016/j.jsbmb.2016.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- S X Lin
- Axe of Endocrinology and Nephrology, CHU research center and Faculty of Medicine, Laval University; Quebec, Canada.
| | - R Shi
- Département de Biochimie, de Microbiologie et de Bio-Informatique, IBIS et PROTEO, Université Laval, Pavillon Charles-Eugène Marchand, Québec City, Canada
| | - X J Hu
- School of Life Sciences, Fudan University, Shanghai 200438, PR China
| | - T M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems, Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, United States
| |
Collapse
|
22
|
Current knowledge of the multifunctional 17β-hydroxysteroid dehydrogenase type 1 (HSD17B1). Gene 2016; 588:54-61. [PMID: 27102893 DOI: 10.1016/j.gene.2016.04.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 02/10/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023]
Abstract
At the late 1940s, 17β-HSD1 was discovered as the first member of the 17β-HSD family with its gene cloned. The three-dimensional structure of human 17β-HSD1 is the first example of any human steroid converting enzyme. The human enzyme's structure and biological function have thus been studied extensively in the last two decades. In humans, the enzyme is expressed in placenta, ovary, endometrium and breast. The high activity of estrogen activation provides the basis of 17β-HSD1's implication in estrogen-dependent diseases, such as breast cancer, endometriosis and non-small cell lung carcinomas. Its dual function in estrogen activation and androgen inactivation has been revealed in molecular and breast cancer cell levels, significantly stimulating the proliferation of such cells. The enzyme's overexpression in breast cancer was demonstrated by clinical samples. Inhibition of human 17β-HSD1 led to xenograft tumor shrinkage. Unfortunately, through decades of studies, there is still no drug using the enzyme's inhibitors available. This is due to the difficulty to get rid of the estrogenic activity of its inhibitors, which are mostly estrogen analogues. New non-steroid inhibitors for the enzyme provide new hope for non-estrogenic inhibitors of the enzyme.
Collapse
|
23
|
Bacsa I, Jójárt R, Schneider G, Wölfling J, Maróti P, Herman BE, Szécsi M, Mernyák E. Synthesis of A-ring halogenated 13α-estrone derivatives as potential 17β-HSD1 inhibitors. Steroids 2015; 104:230-6. [PMID: 26476182 DOI: 10.1016/j.steroids.2015.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/01/2015] [Accepted: 10/09/2015] [Indexed: 12/17/2022]
Abstract
13α-Estrone and its 3-methyl or benzyl ether were halogenated in ring A with N-bromo- or N-iodosuccinimide or 1,3-dibromo-5,5-dimethylhydantoin as electrophile triggers. The chemo- and regioselectivities of the reactions depended greatly on the nature of the substituent on C-3. Bromination of the ethers led to 2- and 4-regioisomers. Bis-halogenation occurred only in the case of the phenolic derivative. Iodination and bromination resulted in similar products, except that the 3-benzyl ether could not be iodinated under the applied conditions. The potential inhibitory action of the new halogenated 13α-estrones on human 17β-hydroxysteroid dehydrogenase 1 activity was investigated via in vitro radiosubstrate incubation. Some compounds proved to be effective inhibitors, with IC50 values in the submicromolar range.
Collapse
Affiliation(s)
- Ildikó Bacsa
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary
| | - Rebeka Jójárt
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary
| | - Gyula Schneider
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary
| | - János Wölfling
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary
| | - Péter Maróti
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary
| | - Bianka Edina Herman
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720 Szeged, Hungary
| | - Mihály Szécsi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720 Szeged, Hungary.
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary.
| |
Collapse
|
24
|
Abdelsamie AS, Bey E, Gargano EM, van Koppen CJ, Empting M, Frotscher M. Towards the evaluation in an animal disease model: Fluorinated 17β-HSD1 inhibitors showing strong activity towards both the human and the rat enzyme. Eur J Med Chem 2015; 103:56-68. [DOI: 10.1016/j.ejmech.2015.08.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 08/07/2015] [Accepted: 08/13/2015] [Indexed: 01/22/2023]
|
25
|
Zhang CY, Wang WQ, Chen J, Lin SX. Reductive 17beta-hydroxysteroid dehydrogenases which synthesize estradiol and inactivate dihydrotestosterone constitute major and concerted players in ER+ breast cancer cells. J Steroid Biochem Mol Biol 2015; 150:24-34. [PMID: 25257817 DOI: 10.1016/j.jsbmb.2014.09.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 09/02/2014] [Accepted: 09/21/2014] [Indexed: 11/26/2022]
Abstract
The reductive 17β-hydroxysteroid dehydrogenases which catalyze the last step in estrogen activation for estrogen dependent breast cancer cells were studied. Their biological function and the effects of their knockdown for cancer cell proliferation were demonstrated. The multidisciplinary study involves enzyme catalysis, sex-hormone and cell cycle regulation, as well as cell proliferation in breast cancer cells. Reductive 17β-HSD1, -7 and -12 were studied in the main breast cancer epithelial cells MCF-7 and T47D. Modification of estradiol and 5α-dihydrotestosterone concentrations was monitored by ELISA assay while corresponding cell viability measured by MTT assay. Cell cycle was determined by flow cytometry. Dual activity of estradiol activation and 5α-dihydrotestosterone reduction by 17β-HSD1 and -7 was critical for breast cancer cell (T47D and MCF-7) viability. Cell viability was decreased by 35.8% ± 1.6% in T47D cells after simultaneously knocking down 17β-HSD1 and -7. MCF-7 cell viability was decreased by 29.3% ± 4.2% using a combination of siRNAs and inhibitors. By knocking down 17β-HSD7, we have provided the first demonstration of the significant role of this enzyme in the stimulation of breast cancer cell viability as a result of its high activity on androgen reduction with positive feedback on estradiol production. A further decrease in cell viability was not observed with additional knockdown of 17β-HSD12 after 17β-HSD1 and 7. Breast cancer cell cycle progression was impeded to enter the S phase from G0-G1 after knocking down 17β-HSD1 and -7. In summary, this is the first demonstration that the dual activity in estrone activation and 5α-dihydrotestosterone reduction are the functional basis of reductive 17β-HSDs in breast cancer cells. 17β-HSD1 and -7 are principal reductive 17β-HSDs and major players in the viability of estrogen-dependent breast cancer cells. Combined targeting of these enzymes may be potential for molecular therapy of such cancer.
Collapse
Affiliation(s)
- Chen-Yan Zhang
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, Québec City, Québec G1V4G2, Canada; Key Laboratory for Space Bioscience and Biotechnology, Faculty of Life Sciences, Northwestern Polytechnic University, Xi'an, Shaanxi, China
| | - Wei-Qi Wang
- Shanghai Engineer and technology Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Jiong Chen
- Shanghai Engineer and technology Research Center of Reproductive Health Drug and Devices, Shanghai, China
| | - Sheng-Xiang Lin
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, Québec City, Québec G1V4G2, Canada; Shanghai Engineer and technology Research Center of Reproductive Health Drug and Devices, Shanghai, China.
| |
Collapse
|
26
|
Järvensivu P, Saloniemi-Heinonen T, Awosanya M, Koskimies P, Saarinen N, Poutanen M. HSD17B1 expression enhances estrogen signaling stimulated by the low active estrone, evidenced by an estrogen responsive element-driven reporter gene in vivo. Chem Biol Interact 2015; 234:126-34. [DOI: 10.1016/j.cbi.2015.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/19/2014] [Accepted: 01/07/2015] [Indexed: 01/13/2023]
|
27
|
Miralinaghi P, Schmitt C, Hartmann RW, Frotscher M, Engel M. 6-Hydroxybenzothiophene Ketones: Potent Inhibitors of 17β-Hydroxysteroid Dehydrogenase Type 1 (17β-HSD1) Owing to Favorable Molecule Geometry and Conformational Preorganization. ChemMedChem 2014; 9:2294-308. [DOI: 10.1002/cmdc.201402050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Indexed: 01/20/2023]
|
28
|
Cysteine-10 on 17 β -Hydroxysteroid Dehydrogenase 1 Has Stabilizing Interactions in the Cofactor Binding Region and Renders Sensitivity to Sulfhydryl Modifying Chemicals. Int J Cell Biol 2013; 2013:769536. [PMID: 24348564 PMCID: PMC3855964 DOI: 10.1155/2013/769536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/15/2013] [Indexed: 11/29/2022] Open
Abstract
17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) catalyzes the conversion of estrone to the potent estrogen estradiol. 17β-HSD1 is highly expressed in breast and ovary tissues and represents a prognostic marker for the tumor progression and survival of patients with breast cancer and other estrogen-dependent tumors. Therefore, the enzyme is considered a promising drug target against estrogen-dependent cancers. For the development of novel inhibitors, an improved understanding of the structure-function relationships is essential. In the present study, we examined the role of a cysteine residue, Cys10, in the Rossmann-fold NADPH binding region, for 17β-HSD1 function and tested the sensitivity towards sulfhydryl modifying chemicals. 3D structure modeling revealed important interactions of Cys10 with residues involved in the stabilization of amino acids of the NADPH binding pocket. Analysis of enzyme activity revealed that 17β-HSD1 was irreversibly inhibited by the sulfhydryl modifying agents N-ethylmaleimide (NEM) and dithiocarbamates. Preincubation with increasing concentrations of NADPH protected 17β-HSD1 from inhibition by these chemicals. Cys10Ser mutant 17β-HSD1 was partially protected from inhibition by NEM and dithiocarbamates, emphasizing the importance of Cys10 in the cofactor binding region. Substitution of Cys10 with serine resulted in a decreased protein half-life, without significantly altering kinetic properties. Despite the fact that Cys10 on 17β-HSD1 seems to have limited potential as a target for new enzyme inhibitors, the present study provides new insight into the structure-function relationships of this enzyme.
Collapse
|
29
|
Thomas MP, Potter BVL. The structural biology of oestrogen metabolism. J Steroid Biochem Mol Biol 2013; 137:27-49. [PMID: 23291110 PMCID: PMC3866684 DOI: 10.1016/j.jsbmb.2012.12.014] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 12/10/2012] [Accepted: 12/12/2012] [Indexed: 02/07/2023]
Abstract
Many enzymes catalyse reactions that have an oestrogen as a substrate and/or a product. The reactions catalysed include aromatisation, oxidation, reduction, sulfonation, desulfonation, hydroxylation and methoxylation. The enzymes that catalyse these reactions must all recognise and bind oestrogen but, despite this, they have diverse structures. This review looks at each of these enzymes in turn, describing the structure and discussing the mechanism of the catalysed reaction. Since oestrogen has a role in many disease states inhibition of the enzymes of oestrogen metabolism may have an impact on the state or progression of the disease and inhibitors of these enzymes are briefly discussed. This article is part of a Special Issue entitled 'CSR 2013'.
Collapse
Key Words
- 17β-HSD
- 17β-Hydroxysteroid dehydrogenase
- 17β-hydroxysteroid dehydrogenase
- 3,5-dinitrocatechol
- 3-(((8R,9S,13S,14S,16R,17S)-3,17-dihydroxy-13-methyl-7,8,9,11,12,13,14,15,16,17-decahydro-6H-cyclopenta[a]phenanthren-16-yl)methyl)benzamide
- 3′-phosphoadenosine-5′-phosphate
- 3′-phosphoadenosine-5′-phosphosulfate
- Aromatase
- COMT
- DHEA(S)
- DHETNA
- DNC
- E1(S)
- E2(S)
- E2B
- E3
- E4
- ER
- FAD/FMN
- FG
- HFG(S)
- NADP(+)
- NADPH
- O5′-[9-(3,17β-dihydroxy-1,3,5(10)-estratrien-16β-yl)-nonanoyl]adenosine
- Oestrogen
- PAP
- PAPS
- Protein structure
- Reaction mechanism
- S-adenosyl methionine
- SAM
- SDR
- Sulfatase
- Sulfotransferase
- catechol-O-methyl transferase
- dehydroepiandrosterone (sulfate)
- estetrol
- estradiol (sulfate)
- estriol
- estrogen receptor
- estrone (sulfate)
- flavin adenine dinucleotide/flavin mononucleotide
- formylglycine
- hydroxyformylglycine (sulfate)
- mb-COMT
- membrane-bound COMT
- nicotinamide adenine dinucleotide phosphate (oxidised)
- nicotinamide adenine dinucleotide phosphate (reduced)
- s-COMT
- short-chain dehydrogenase/reductase
- soluble COMT
Collapse
Affiliation(s)
- Mark P Thomas
- Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| | | |
Collapse
|
30
|
Farhane S, Fournier MA, Poirier D. Chemical synthesis, characterisation and biological evaluation of lactonic-estradiol derivatives as inhibitors of 17β-hydroxysteroid dehydrogenase type 1. J Steroid Biochem Mol Biol 2013; 137:322-31. [PMID: 23685015 DOI: 10.1016/j.jsbmb.2013.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/28/2013] [Accepted: 05/01/2013] [Indexed: 11/17/2022]
Abstract
To control estradiol (E2) formation, we are interested in synthesizing inhibitors of 17β-hydroxyteroid dehydrogenase type 1 (17β-HSD1). Since the results of docking experiments have shown that E2-lactone derivatives substituted in position 19 or 20 (E-ring) could generate interactions with the active site of the enzyme, we carried out their chemical synthesis. After having prepared the 16β,17β-γ-lactone-E2 in four steps starting from estrone (E1), we introduced the molecular diversity by adding a hydroxymethyl, a methylcarboxylate, a carboxy or an allyl group. The allyl derivative was used as a key intermediate to generate a hydroxyethyl side chain in α or β position. Two lactols were also obtained from two hydroxyalkyl lactones. Enzymatic assays revealed that lactone and lactol derivatives weakly inhibited 17β-HSD1 in homogenized HEK-293 cells overexpressing 17β-HSD1 (34-60% at 1 μM) and in intact T-47D cells expressing 17β-HSD1 (10-40% at 10 μM). This article is part of a Special Issue entitled "Synthesis and biological testing of steroid derivatives as inhibitors".
Collapse
Affiliation(s)
- Siham Farhane
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL) - Research Center and Laval University, Québec (Québec), G1V 4G2, Canada
| | | | | |
Collapse
|
31
|
Saloniemi T, Jokela H, Strauss L, Pakarinen P, Poutanen M. The diversity of sex steroid action: novel functions of hydroxysteroid (17β) dehydrogenases as revealed by genetically modified mouse models. J Endocrinol 2012; 212:27-40. [PMID: 22045753 DOI: 10.1530/joe-11-0315] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Disturbed action of sex steroid hormones, i.e. androgens and estrogens, is involved in the pathogenesis of various severe diseases in humans. Interestingly, recent studies have provided data further supporting the hypothesis that the circulating hormone concentrations do not explain all physiological and pathological processes observed in hormone-dependent tissues, while the intratissue sex steroid concentrations are determined by the expression of steroid metabolising enzymes in the neighbouring cells (paracrine action) and/or by target cells themselves (intracrine action). This local sex steroid production is also a valuable treatment option for developing novel therapies against hormonal diseases. Hydroxysteroid (17β) dehydrogenases (HSD17Bs) compose a family of 14 enzymes that catalyse the conversion between the low-active 17-keto steroids and the highly active 17β-hydroxy steroids. The enzymes frequently expressed in sex steroid target tissues are, thus, potential drug targets in order to lower the local sex steroid concentrations. The present review summarises the recent data obtained for the role of HSD17B1, HSD17B2, HSD17B7 and HSD17B12 enzymes in various metabolic pathways and their physiological and pathophysiological roles as revealed by the recently generated genetically modified mouse models. Our data, together with that provided by others, show that, in addition to having a role in sex steroid metabolism, several of these HSD17B enzymes possess key roles in other metabolic processes: for example, HD17B7 is essential for cholesterol biosynthesis and HSD17B12 is involved in elongation of fatty acids. Additional studies in vitro and in vivo are to be carried out in order to fully define the metabolic role of the HSD17B enzymes and to evaluate their value as drug targets.
Collapse
Affiliation(s)
- Taija Saloniemi
- Department of Physiology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20014 Turku, Finland
| | | | | | | | | |
Collapse
|
32
|
Klein T, Henn C, Negri M, Frotscher M. Structural basis for species specific inhibition of 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1): computational study and biological validation. PLoS One 2011; 6:e22990. [PMID: 21857977 PMCID: PMC3153478 DOI: 10.1371/journal.pone.0022990] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 07/07/2011] [Indexed: 11/19/2022] Open
Abstract
17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) catalyzes the reduction of estrone to estradiol, which is the most potent estrogen in humans. Inhibition of 17β-HSD1 and thereby reducing the intracellular estradiol concentration is thus a promising approach for the treatment of estrogen dependent diseases. In the past, several steroidal and non-steroidal inhibitors of 17β-HSD1 have been described but so far there is no cocrystal structure of the latter in complex with 17β-HSD1. However, a distinct knowledge of active site topologies and protein-ligand interactions is a prerequisite for structure-based drug design and optimization. An elegant strategy to enhance this knowledge is to compare inhibition values obtained for one compound toward ortholog proteins from various species, which are highly conserved in sequence and differ only in few residues. In this study the inhibitory potencies of selected members of different non-steroidal inhibitor classes toward marmoset 17β-HSD1 were determined and the data were compared with the values obtained for the human enzyme. A species specific inhibition profile was observed in the class of the (hydroxyphenyl)naphthols. Using a combination of computational methods, including homology modelling, molecular docking, MD simulation, and binding energy calculation, a reasonable model of the three-dimensional structure of marmoset 17β-HSD1 was developed and inhibition data were rationalized on the structural basis. In marmoset 17β-HSD1, residues 190 to 196 form a small α-helix, which induces conformational changes compared to the human enzyme. The docking poses suggest these conformational changes as determinants for species specificity and energy decomposition analysis highlighted the outstanding role of Asn152 as interaction partner for inhibitor binding. In summary, this strategy of comparing the biological activities of inhibitors toward highly conserved ortholog proteins might be an alternative to laborious x-ray or site-directed mutagenesis experiments in certain cases. Additionally, it facilitates inhibitor design and optimization by offering new information on protein-ligand interactions.
Collapse
Affiliation(s)
- Tobias Klein
- Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, Germany
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Claudia Henn
- Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, Germany
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Matthias Negri
- Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, Germany
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Martin Frotscher
- Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, Germany
| |
Collapse
|
33
|
Hong Y, Chen S. Aromatase, estrone sulfatase, and 17β-hydroxysteroid dehydrogenase: structure-function studies and inhibitor development. Mol Cell Endocrinol 2011; 340:120-6. [PMID: 20888390 PMCID: PMC3035767 DOI: 10.1016/j.mce.2010.09.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 09/15/2010] [Accepted: 09/18/2010] [Indexed: 11/23/2022]
Abstract
Aromatase, estrone sulfatase, and 17β-hydroxysteroid dehydrogenase type 1 are involved in the key steps of 17β-estradiol biosynthesis. Structure-function studies of aromatase, estrone sulfatase and 17β-hydroxysteroid dehydrogenase type 1 are important to evaluate the molecular basis of the interaction between these enzymes and their inhibitors. Selective and potent inhibitors of the three enzymes have been developed as antiproliferative agents in hormone-dependent breast carcinoma. New treatment strategies for hormone-dependent breast cancer are discussed.
Collapse
Affiliation(s)
- Yanyan Hong
- Division of Tumor Cell Biology, Beckman Research Institute of the City of Hope, 1450 E. Duarte Road, Duarte, CA 91010, United States
| | | |
Collapse
|
34
|
Poirier D. Contribution to the development of inhibitors of 17β-hydroxysteroid dehydrogenase types 1 and 7: key tools for studying and treating estrogen-dependent diseases. J Steroid Biochem Mol Biol 2011; 125:83-94. [PMID: 21182944 DOI: 10.1016/j.jsbmb.2010.12.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 11/17/2010] [Accepted: 12/13/2010] [Indexed: 02/06/2023]
Abstract
17β-Hydroxysteroid dehydrogenases (17β-HSDs) belong to a group of key enzymes involved in the biosynthesis of steroidal hormones by catalyzing the reduction of 17-ketosteroids or the oxidation of 17β-hydroxysteroids. From three members known in the early nineties, the 17β-HSD functional family has grown to 15 members over the last 20 years. This growing number of 17β-HSD isoforms questioned the importance of each member, especially in their implication in estrogen- and androgen-dependent diseases, such as breast and prostate cancers. One of the strategies used to address the physiological importance of 17β-HSDs is to use potent and selective inhibitors. Furthermore, enzyme inhibitors could also be of therapeutic interest by reducing the level of estradiol (E2). Focusing on estrogens, we targeted 17β-HSD types 1 and 7, two enzymes able to transform the weak estrogen estrone (E1) into the potent estrogen E2. The present review article gives a description of different classes of inhibitors of 17β-HSD1 (C6-derivatives of E2, C16-derivatives of E2 as alkylating and dual action compounds, E2-adenosine hybrids, E2-simplified adenosine hybrids, and C16-derivatives of E1 or E2) and of inhibitors of 17β-HSD7, all these inhibitors developed in our laboratory. The chemical structures and inhibitory activity of these steroidal inhibitors, their potential as therapeutic agents, and their use as tools to elucidate the role of these enzymes in particular biological systems will be discussed. Article from the Special issue on Targeted Inhibitors.
Collapse
Affiliation(s)
- Donald Poirier
- Laval University (Faculty of Medicine) and CHUQ (CHUL)-Research Center (Laboratory of Medicinal Chemistry, Endocrinology and Genomic Unit), 2705 Laurier Boulevard, Quebec (Quebec) G1V 4G2, Canada.
| |
Collapse
|
35
|
Marchais-Oberwinkler S, Henn C, Möller G, Klein T, Negri M, Oster A, Spadaro A, Werth R, Wetzel M, Xu K, Frotscher M, Hartmann RW, Adamski J. 17β-Hydroxysteroid dehydrogenases (17β-HSDs) as therapeutic targets: protein structures, functions, and recent progress in inhibitor development. J Steroid Biochem Mol Biol 2011; 125:66-82. [PMID: 21193039 DOI: 10.1016/j.jsbmb.2010.12.013] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 12/03/2010] [Accepted: 12/20/2010] [Indexed: 01/18/2023]
Abstract
17β-Hydroxysteroid dehydrogenases (17β-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, 17β-HSDs act like pre-receptor molecular switches. 17β-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 17β-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 17β-HSDs. We focus on steroidal and non-steroidal inhibitors recently published for each subtype and report on existing animal models for the different 17β-HSDs and the respective diseases. Article from the Special issue on Targeted Inhibitors.
Collapse
|
36
|
Biochemical and biological evaluation of novel potent coumarin inhibitor of 17β-HSD type 1. Chem Biol Interact 2011; 191:60-5. [DOI: 10.1016/j.cbi.2011.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 12/23/2010] [Accepted: 01/04/2011] [Indexed: 01/23/2023]
|
37
|
Kratschmar DV, Vuorinen A, Da Cunha T, Wolber G, Classen-Houben D, Doblhoff O, Schuster D, Odermatt A. Characterization of activity and binding mode of glycyrrhetinic acid derivatives inhibiting 11β-hydroxysteroid dehydrogenase type 2. J Steroid Biochem Mol Biol 2011; 125:129-42. [PMID: 21236343 DOI: 10.1016/j.jsbmb.2010.12.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Revised: 12/24/2010] [Accepted: 12/31/2010] [Indexed: 12/21/2022]
Abstract
Modulation of intracellular glucocorticoid availability is considered as a promising strategy to treat glucocorticoid-dependent diseases. 18β-Glycyrrhetinic acid (GA), the biologically active triterpenoid metabolite of glycyrrhizin, which is contained in the roots and rhizomes of licorice (Glycyrrhiza spp.), represents a well-known but non-selective inhibitor of 11β-hydroxysteroid dehydrogenases (11β-HSDs). However, to assess the physiological functions of the respective enzymes and for potential therapeutic applications selective inhibitors are needed. In the present study, we applied bioassays and 3D-structure modeling to characterize nine 11β-HSD1 and fifteen 11β-HSD2 inhibiting GA derivatives. Comparison of the GA derivatives in assays using cell lysates revealed that modifications at the 3-hydroxyl and/or the carboxyl led to highly selective and potent 11β-HSD2 inhibitors. The data generated significantly extends our knowledge on structure-activity relationship of GA derivatives as 11β-HSD inhibitors. Using recombinant enzymes we found also potent inhibition of mouse 11β-HSD2, despite significant species-specific differences. The selected GA derivatives potently inhibited 11β-HSD2 in intact SW-620 colon cancer cells, although the rank order of inhibitory potential differed from that obtained in cell lysates. The biological activity of compounds was further demonstrated in glucocorticoid receptor (GR) transactivation assays in cells coexpressing GR and 11β-HSD1 or 11β-HSD2. 3D-structure modeling provides an explanation for the differences in the selectivity and activity of the GA derivatives investigated. The most potent and selective 11β-HSD2 inhibitors should prove useful as mechanistic tools for further anti-inflammatory and anti-cancer in vitro and in vivo studies. Article from the Special issue on Targeted Inhibitors.
Collapse
Affiliation(s)
- Denise V Kratschmar
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Starčević Š, Božnar P, Turk S, Gobec S, Rižner TL. Design and synthesis of substrate mimetics based on an indole scaffold: potential inhibitors of 17β-HSD type 1. Horm Mol Biol Clin Investig 2011; 6:201-9. [PMID: 25961256 DOI: 10.1515/hmbci.2011.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Accepted: 02/07/2011] [Indexed: 11/15/2022]
Abstract
BACKGROUND Human 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) acts at a pre-receptor level. It catalyzes NADPH-dependent reduction of the weak estrogen estrone into the most potent estrogen estradiol, which exerts its proliferative effects via estrogen receptors. Overexpression of 17β-HSD1 in estrogen-responsive tissues is related to the development of hormone-dependent diseases, such as breast cancer and endometriosis. 17β-HSD1 thus represents an attractive target for development of new drugs. METHODS We designed and synthesized a series of 3-, 5- and 6-phenyl indole derivatives as mimetics of the steroid substrate estrone. All of these compounds were evaluated for inhibition of recombinant human 17β-HSD1 from Escherichia coli, at concentrations of 0.6 μM and 6.0 μM. RESULTS Among 14 indole derivatives, compound 9 was an initial hit inhibitor of 17β-HSD1, with moderate inhibition (64% at 6 μM). Molecular docking into the crystal structure of 17β-HSD1 (1A27) revealed that this 5-phenyl indole derivative binds to 17β-HSD1 similarly to co-crystalized E2. Compound 9 forms two H-bonds with 17β-HSD1: one between the indole nitrogen and His222, and the second between the phenolic OH group and catalytic Tyr155. CONCLUSIONS The indole scaffold is one of the possible starting points for the design of substrate mimetics of the steroid substrate estrone. Our study shows that these 6- and, especially, 5-phenol indole derivatives can act as moderate inhibitors of 17β-HSD1. Based on inhibition assays and docking simulations, we can infer further improvements of the 5-phenol indole derivatives that might result in better inhibition profiles.
Collapse
|
39
|
Oster A, Klein T, Henn C, Werth R, Marchais‐Oberwinkler S, Frotscher M, Hartmann RW. Bicyclic Substituted Hydroxyphenylmethanone Type Inhibitors of 17 β‐Hydroxysteroid Dehydrogenase Type 1 (17 β‐HSD1): The Role of the Bicyclic Moiety. ChemMedChem 2011; 6:476-87. [DOI: 10.1002/cmdc.201000457] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 01/01/2011] [Indexed: 11/11/2022]
Affiliation(s)
- Alexander Oster
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Tobias Klein
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Claudia Henn
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Ruth Werth
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Sandrine Marchais‐Oberwinkler
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Martin Frotscher
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Rolf W. Hartmann
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| |
Collapse
|
40
|
Marchais-Oberwinkler S, Wetzel M, Ziegler E, Kruchten P, Werth R, Henn C, Hartmann RW, Frotscher M. New Drug-Like Hydroxyphenylnaphthol Steroidomimetics As Potent and Selective 17β-Hydroxysteroid Dehydrogenase Type 1 Inhibitors for the Treatment of Estrogen-Dependent Diseases. J Med Chem 2010; 54:534-47. [DOI: 10.1021/jm1009082] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - Marie Wetzel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2 3, D-66123 Saarbrücken, Germany
| | - Erika Ziegler
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2 3, D-66123 Saarbrücken, Germany
| | - Patricia Kruchten
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2 3, D-66123 Saarbrücken, Germany
| | - Ruth Werth
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2 3, D-66123 Saarbrücken, Germany
| | - Claudia Henn
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2 3, D-66123 Saarbrücken, Germany
| | - Rolf W. Hartmann
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2 3, D-66123 Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Campus C2 3, D-66123 Saarbrücken, Germany
| | - Martin Frotscher
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2 3, D-66123 Saarbrücken, Germany
| |
Collapse
|
41
|
Starčević Š, Brožič P, Turk S, Cesar J, Lanišnik Rižner T, Gobec S. Synthesis and Biological Evaluation of (6- and 7-Phenyl) Coumarin Derivatives as Selective Nonsteroidal Inhibitors of 17β-Hydroxysteroid Dehydrogenase Type 1. J Med Chem 2010; 54:248-61. [PMID: 21138273 DOI: 10.1021/jm101104z] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Štefan Starčević
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Petra Brožič
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Samo Turk
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Jožko Cesar
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
42
|
Negri M, Recanatini M, Hartmann RW. Insights in 17beta-HSD1 enzyme kinetics and ligand binding by dynamic motion investigation. PLoS One 2010; 5:e12026. [PMID: 20706575 PMCID: PMC2919385 DOI: 10.1371/journal.pone.0012026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 07/06/2010] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Bisubstrate enzymes, such as 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1), exist in solution as an ensemble of conformations. 17beta-HSD1 catalyzes the last step of the biosynthesis of estradiol and, thus, it is a potentially attractive target for breast cancer treatment. METHODOLOGY/PRINCIPAL FINDINGS To elucidate the conformational transitions of its catalytic cycle, a structural analysis of all available crystal structures was performed and representative conformations were assigned to each step of the putative kinetic mechanism. To cover most of the conformational space, all-atom molecular dynamic simulations were performed using the four crystallographic structures best describing apoform, opened, occluded and closed state of 17beta-HSD1 as starting structures. With three of them, binary and ternary complexes were built with NADPH and NADPH-estrone, respectively, while two were investigated as apoform. Free energy calculations were performed in order to judge more accurately which of the MD complexes describes a specific kinetic step. CONCLUSIONS/SIGNIFICANCE Remarkably, the analysis of the eight long range trajectories resulting from this multi-trajectory/-complex approach revealed an essential role played by the backbone and side chain motions, especially of the betaF alphaG'-loop, in cofactor and substrate binding. Thus, a selected-fit mechanism is suggested for 17beta-HSD1, where ligand-binding induced concerted motions of the FG-segment and the C-terminal part guide the enzyme along its preferred catalytic pathway. Overall, we could assign different enzyme conformations to the five steps of the random bi-bi kinetic cycle of 17beta-HSD1 and we could postulate a preferred pathway for it. This study lays the basis for more-targeted biochemical studies on 17beta-HSD1, as well as for the design of specific inhibitors of this enzyme. Moreover, it provides a useful guideline for other enzymes, also characterized by a rigid core and a flexible region directing their catalysis.
Collapse
Affiliation(s)
- Matthias Negri
- Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany
| | - Maurizio Recanatini
- Department of Pharmaceutical Sciences, University of Bologna, Bologna, Italy
| | - Rolf W. Hartmann
- Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany
| |
Collapse
|
43
|
Manning JR, Bailey MA, Soares DC, Dunbar DR, Mullins JJ. In silico structure-function analysis of pathological variation in the HSD11B2 gene sequence. Physiol Genomics 2010; 42:319-30. [PMID: 20571110 DOI: 10.1152/physiolgenomics.00053.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
11beta-Hydroxysteroid dehydrogenase type 2 (11betaHSD2) is a short-chain dehydrogenase/reductase (SDR) responsible for inactivating cortisol and preventing its binding to the mineralocorticoid receptor (MR). Nonfunctional mutations in HSD11B2, the gene encoding 11betaHSD2, cause the hypertensive syndrome of apparent mineralocorticoid excess (AME). Like other such Mendelian disorders, AME is rare but has nevertheless helped to illuminate principles fundamental to the regulation of blood pressure. Furthermore, polymorphisms in HSD11B2 have been associated with salt sensitivity, a major risk factor for cardiovascular mortality. It is therefore highly likely that sequence variation in HSD11B2, having subtle functional ramifications, will affect blood pressure in the wider population. In this study, a three-dimensional homology model of 11betaHSD2 was created and used to hypothesize the functional consequences in terms of protein structure of published mutations in HSD11B2. This approach underscored the strong genotype-phenotype correlation of AME: severe forms of the disease, associated with little in vivo enzyme activity, arise from mutations occurring in invariant alignment positions. These were predicted to exert gross structural changes in the protein. In contrast, those mutations causing a mild clinical phenotype were in less conserved regions of the protein that were predicted to be relatively more tolerant to substitution. Finally, a number of pathogenic mutations are shown to be associated with regions predicted to participate in dimer formation, and in protein stabilization, which may therefore suggest molecular mechanisms of disease.
Collapse
Affiliation(s)
- Jonathan R Manning
- Centre for Cardiovascular Science, Queen's Medical Research Institute, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | | | |
Collapse
|
44
|
Möller G, Husen B, Kowalik D, Hirvelä L, Plewczynski D, Rychlewski L, Messinger J, Thole H, Adamski J. Species used for drug testing reveal different inhibition susceptibility for 17beta-hydroxysteroid dehydrogenase type 1. PLoS One 2010; 5:e10969. [PMID: 20544026 PMCID: PMC2882332 DOI: 10.1371/journal.pone.0010969] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Accepted: 05/10/2010] [Indexed: 01/27/2023] Open
Abstract
Steroid-related cancers can be treated by inhibitors of steroid metabolism. In searching for new inhibitors of human 17beta-hydroxysteroid dehydrogenase type 1 (17β-HSD 1) for the treatment of breast cancer or endometriosis, novel substances based on 15-substituted estrone were validated. We checked the specificity for different 17β-HSD types and species. Compounds were tested for specificity in vitro not only towards recombinant human 17β-HSD types 1, 2, 4, 5 and 7 but also against 17β-HSD 1 of several other species including marmoset, pig, mouse, and rat. The latter are used in the processes of pharmacophore screening. We present the quantification of inhibitor preferences between human and animal models. Profound differences in the susceptibility to inhibition of steroid conversion among all 17β-HSDs analyzed were observed. Especially, the rodent 17β-HSDs 1 were significantly less sensitive to inhibition compared to the human ortholog, while the most similar inhibition pattern to the human 17β-HSD 1 was obtained with the marmoset enzyme. Molecular docking experiments predicted estrone as the most potent inhibitor. The best performing compound in enzymatic assays was also highly ranked by docking scoring for the human enzyme. However, species-specific prediction of inhibitor performance by molecular docking was not possible. We show that experiments with good candidate compounds would out-select them in the rodent model during preclinical optimization steps. Potentially active human-relevant drugs, therefore, would no longer be further developed. Activity and efficacy screens in heterologous species systems must be evaluated with caution.
Collapse
Affiliation(s)
- Gabriele Möller
- Helmholtz Zentrum München, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany
| | - Bettina Husen
- Solvay Pharmaceuticals Research Laboratories, Hannover, Germany
| | - Dorota Kowalik
- Helmholtz Zentrum München, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany
| | | | - Dariusz Plewczynski
- Interdisciplinary Centre for Mathematical and Computational Modelling, Warsaw University, Warsaw, Poland
| | | | - Josef Messinger
- Solvay Pharmaceuticals Research Laboratories, Hannover, Germany
| | - Hubert Thole
- Solvay Pharmaceuticals Research Laboratories, Hannover, Germany
| | - Jerzy Adamski
- Helmholtz Zentrum München, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Freising-Weihenstephan, Germany
- * E-mail:
| |
Collapse
|
45
|
Aka JA, Mazumdar M, Chen CQ, Poirier D, Lin SX. 17beta-hydroxysteroid dehydrogenase type 1 stimulates breast cancer by dihydrotestosterone inactivation in addition to estradiol production. Mol Endocrinol 2010; 24:832-45. [PMID: 20172961 DOI: 10.1210/me.2009-0468] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The active estrogen estradiol (E2) stimulates breast cancer cell (BCC) growth, whereas the androgen dihydrotestosterone (DHT) has shown an antiproliferative effect. The principal product synthesized by the 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1) is E2, although we have demonstrated that the purified enzyme also inactivates DHT. However, the direct roles of 17beta-HSD1 in sex-hormone regulation and BCC proliferation have not been completely established. Here, we show that 17beta-HSD1 inhibition suppresses DHT catabolism by 19%, whereas knockdown of the gene expression increases the concentration of DHT by 41% in the T47D BCC line. The 17beta-HSD1/DHT complex crystal structure reveals that DHT binds in both normal and reverse modes, but the latter mode leading to O3 reduction is preferred with stronger interactions. Using RNA interference and an inhibitor of 17beta-HSD1, we demonstrate that 17beta-HSD1 expression is negatively correlated to DHT levels in BCC but positively correlated to estrone reduction, E2 levels, and cell proliferation. 17beta-HSD1 inhibition reduces DHT inactivation, increasing the antiproliferative effect by DHT in T47D cells after 8 d treatment. Thus, 17beta-HSD1 up-regulates BCC growth by a dual action on estradiol synthesis and DHT inactivation. We have further demonstrated that 17beta-HSD1 can enhance the E2-induced expression of the endogenous estrogen-responsive gene pS2, providing an important information regarding the modulation of the estrogen responsiveness by 17beta-HSD1 that may also contribute to BCC growth. These results strongly support the rationale for inhibiting 17beta-HSD1 in breast cancer therapy to eliminate estrogen activation via the sulfatase pathway while avoiding the deprivation of DHT.
Collapse
Affiliation(s)
- Juliette A Aka
- Research Center of the Laval University Hospital Center (CHUQ-CHUL) and Laval University, Québec, Canada
| | | | | | | | | |
Collapse
|
46
|
Binary and ternary crystal structure analyses of a novel inhibitor with 17β-HSD type 1: a lead compound for breast cancer therapy. Biochem J 2009; 424:357-66. [DOI: 10.1042/bj20091020] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Oestradiol is a well-characterized sex hormone that stimulates breast cancer and other oestrogen-related diseases. 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) catalyses the last step in the synthesis of oestradiol and androstenediol in breast tumour tissue. The enzyme's high expression and activity after simultaneous blockade of oestrogen receptors and inhibition of aromatase in the tumour shows the necessity for its inhibition as a requirement for breast cancer therapy. In the present paper, we report structures of the binary and ternary complexes of 17β-HSD1 with a new inhibitor E2B {3-[3′,17′β-dihydroxyestra-1′,3′,5′(10′)-trien-16′β-methyl]benzamide}, and the enzyme inhibition by the later. The IC50 value for E2B was determined to be 42 nM in T47D cells. Multiple interactions between E2B and the enzyme include hydrogen bonds and hydrophobic interactions, as well as π–π interactions. A kinetic study demonstrated that E2B inhibits the enzyme's reduction forming oestradiol from oestrone, with a Ki of 0.9±0.15 nM. Such strong inhibition is in agreement with its extensive interaction with the enzyme, suggesting its potential as a lead compound for breast cancer therapy. In fact, this possibility is enhanced by its capacity for cell penetration similar to natural steroids. Such inhibitors that block oestrogen synthesis to suppress the sulfatase pathway producing oestradiol can be used in adjuvant therapies with oestrogen receptor blockade, opening a new orientation of breast cancer treatment.
Collapse
|
47
|
Michiels PJA, Ludwig C, Stephan M, Fischer C, Möller G, Messinger J, van Dongen M, Thole H, Adamski J, Günther UL. Ligand-based NMR spectra demonstrate an additional phytoestrogen binding site for 17beta-hydroxysteroid dehydrogenase type 1. J Steroid Biochem Mol Biol 2009; 117:93-8. [PMID: 19631742 DOI: 10.1016/j.jsbmb.2009.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 07/13/2009] [Accepted: 07/15/2009] [Indexed: 01/21/2023]
Abstract
The enzyme 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1) has become an important drug target for breast cancer because it catalyzes the interconversion of estrone to the biologically more potent estradiol which also plays a crucial role in the etiology of breast cancer. Patients with an increased expression of the 17beta-HSD1 gene have a significantly worse outcome than patients without. Inhibitors for 17beta-HSD1 are therefore included in therapy development. Here we have studied binding of 17beta-HSD1 to substrates and a number of inhibitors using NMR spectroscopy. Ligand observed NMR spectra show a strong pH dependence for the phytoestrogens luteolin and apigenin but not for the natural ligands estradiol and estrone. Moreover, NMR competition experiments show that the phytoestrogens do not replace the estrogens despite their similar inhibition levels in the in vitro assay. These results strongly support an additional 17beta-HSD1 binding site for phytoestrogens which is neither the substrate nor the co-factor binding site. Docking experiments suggest the dimer interface as a possible location. An additional binding site for the phytoestrogens may open new opportunities for the design of inhibitors, not only for 17beta-HSD1, but also for other family members of the short chain dehydrogenases.
Collapse
Affiliation(s)
- Paul J A Michiels
- HWB-NMR, CR UK Institute of Cancer Sciences, University of Birmingham, Birmingham, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bey E, Marchais-Oberwinkler S, Negri M, Kruchten P, Oster A, Klein T, Spadaro A, Werth R, Frotscher M, Birk B, Hartmann RW. New Insights into the SAR and Binding Modes of Bis(hydroxyphenyl)thiophenes and -benzenes: Influence of Additional Substituents on 17β-Hydroxysteroid Dehydrogenase Type 1 (17β-HSD1) Inhibitory Activity and Selectivity. J Med Chem 2009; 52:6724-43. [DOI: 10.1021/jm901195w] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Emmanuel Bey
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| | | | - Matthias Negri
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| | - Patricia Kruchten
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| | - Alexander Oster
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| | - Tobias Klein
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| | - Alessandro Spadaro
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| | - Ruth Werth
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| | - Martin Frotscher
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| | - Barbara Birk
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| | - Rolf W. Hartmann
- Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 15 11 50, D-66041 Saarbrücken, Germany
| |
Collapse
|
49
|
Lilienkampf A, Karkola S, Alho-Richmond S, Koskimies P, Johansson N, Huhtinen K, Vihko K, Wähälä K. Synthesis and Biological Evaluation of 17β-Hydroxysteroid Dehydrogenase Type 1 (17β-HSD1) Inhibitors Based on a Thieno[2,3-d]pyrimidin-4(3H)-one Core. J Med Chem 2009; 52:6660-71. [DOI: 10.1021/jm900928k] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Annamaria Lilienkampf
- Laboratory of Organic Chemistry, Department of Chemistry, P.O. Box 55, FIN-00014 University of Helsinki, Finland
| | - Sampo Karkola
- Laboratory of Organic Chemistry, Department of Chemistry, P.O. Box 55, FIN-00014 University of Helsinki, Finland
| | - Sari Alho-Richmond
- Laboratory of Organic Chemistry, Department of Chemistry, P.O. Box 55, FIN-00014 University of Helsinki, Finland
| | - Pasi Koskimies
- Hormos Medical Ltd., PharmaCity, FIN-20520 Turku, Finland
| | - Nina Johansson
- Hormos Medical Ltd., PharmaCity, FIN-20520 Turku, Finland
| | - Kaisa Huhtinen
- Hormos Medical Ltd., PharmaCity, FIN-20520 Turku, Finland
| | - Kimmo Vihko
- Hormos Medical Ltd., PharmaCity, FIN-20520 Turku, Finland
| | - Kristiina Wähälä
- Laboratory of Organic Chemistry, Department of Chemistry, P.O. Box 55, FIN-00014 University of Helsinki, Finland
| |
Collapse
|
50
|
Bérubé M, Poirier D. Improved synthesis of EM-1745, preparation of its C17-ketone analogue and comparison of their inhibitory potency on 17β-hydroxysteroid dehydrogenase type 1. J Enzyme Inhib Med Chem 2009; 24:832-43. [DOI: 10.1080/14756360802399761] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Marie Bérubé
- Medicinal Chemistry Division, Oncology and Molecular Endocrinology Laboratory, CHUQ-CHUL Research Center, Québec, G1V 4G2, Canada
| | - Donald Poirier
- Medicinal Chemistry Division, Oncology and Molecular Endocrinology Laboratory, CHUQ-CHUL Research Center, Québec, G1V 4G2, Canada
| |
Collapse
|