1
|
Nsanzamahoro S, Bezuneh TT, Nan F, Wu Z, Iradukunda Y, Shen L, Li B, Yu WW. In-situ formed silicon nanoparticles as high-fidelity fluorometric probe for sulfatase activity assay. Talanta 2025; 295:128314. [PMID: 40378763 DOI: 10.1016/j.talanta.2025.128314] [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: 03/17/2025] [Revised: 04/28/2025] [Accepted: 05/12/2025] [Indexed: 05/19/2025]
Abstract
Analyte-triggered fluorometric nanosensors is desirable for establishing novel analytical tools with better sensing responses. In this study, we illustrated that pyrocatechol (PCT) easily reacted with (N-[3-(trimethoxysilyl) propyl]-ethylenediamine) (DAMO) to generate strong yellow fluorescent silicon nanoparticles (Si NPs). Based on this and the sulfatase-activated conversion of pyrocatechol sulfate (PCTS) into PCT, we designed a sensing approach using PCTS/DAMO encountered fluorescence turn-on for sulfatase activity assay. Through such simple and accessible sensing method with clear reaction mechanism, the designed probe revealed high detection performance with excellent sensitivity and selectivity towards sulfatase compared to the rest of enzymes. The sulfatase activity was measured in a linear range of 0.2-60 U/L, with a detection limit of 0.12 U/L. For real samples study, sulfatase activity was measured in fetal bovine serum and human serum albumin with successful recoveries. Furthermore, the designed sensing method was used for sulfatase inhibitor screening using estrone-3-O-sulfamate (EMATE) as a model.
Collapse
Affiliation(s)
- Stanislas Nsanzamahoro
- School of Chemistry and Chemical Engineering, Ministry of Education Key Laboratory of Special Functional Aggregated Materials, Shandong Key Laboratory of Advanced Organosilicon Materials and Technologies, Shandong University, Jinan, 250100, China; Shandong Provincial Key Laboratory for Science of Material Creation and Energy Conversion, Science Center for Material Creation and Energy Conversion, Shandong University, Qingdao, 266237, China
| | - Terefe Tafese Bezuneh
- School of Chemistry and Chemical Engineering, Ministry of Education Key Laboratory of Special Functional Aggregated Materials, Shandong Key Laboratory of Advanced Organosilicon Materials and Technologies, Shandong University, Jinan, 250100, China; Shandong Provincial Key Laboratory for Science of Material Creation and Energy Conversion, Science Center for Material Creation and Energy Conversion, Shandong University, Qingdao, 266237, China
| | - Fuchun Nan
- School of Chemistry and Chemical Engineering, Ministry of Education Key Laboratory of Special Functional Aggregated Materials, Shandong Key Laboratory of Advanced Organosilicon Materials and Technologies, Shandong University, Jinan, 250100, China; Shandong Provincial Key Laboratory for Science of Material Creation and Energy Conversion, Science Center for Material Creation and Energy Conversion, Shandong University, Qingdao, 266237, China
| | - Zilong Wu
- School of Chemistry and Chemical Engineering, Ministry of Education Key Laboratory of Special Functional Aggregated Materials, Shandong Key Laboratory of Advanced Organosilicon Materials and Technologies, Shandong University, Jinan, 250100, China; Shandong Provincial Key Laboratory for Science of Material Creation and Energy Conversion, Science Center for Material Creation and Energy Conversion, Shandong University, Qingdao, 266237, China
| | - Yves Iradukunda
- Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Lanbo Shen
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Bin Li
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China.
| | - William W Yu
- School of Chemistry and Chemical Engineering, Ministry of Education Key Laboratory of Special Functional Aggregated Materials, Shandong Key Laboratory of Advanced Organosilicon Materials and Technologies, Shandong University, Jinan, 250100, China; Shandong Provincial Key Laboratory for Science of Material Creation and Energy Conversion, Science Center for Material Creation and Energy Conversion, Shandong University, Qingdao, 266237, China.
| |
Collapse
|
2
|
Liu H, Zhang J, Liu L, Li W, Yang J, Wang P. A sensitive fluorescent nanoprobe for sulfatase detection and imaging in living cells and in vivo. Chem Commun (Camb) 2025; 61:4403-4406. [PMID: 39992205 DOI: 10.1039/d5cc00196j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
In this study, we developed a fluorescent probe TCF-SULF for sulfatase detection with satisfactory selectivity and sensitivity. After being encapsulated by the DSPE-PEG2000, the nanoprobe TCF-SULF NPs can be applied to imaging endogenous sulfatase in MCF-7 cells with low cytotoxicity. Importantly, TCF-SULF NPs were successfully used to monitor sulfatase in vivo.
Collapse
Affiliation(s)
- Huijia Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Jiaqi Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Li Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Wenqing Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Jing Yang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Peng Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
3
|
Zheng C, Cui M, Zhang Y, Liu L, Li W, Zhang J, Ji M, Chen W, Jiang W, Wang P, Zhang W. Universal sulfatase-based chemiluminescence biosensing platform: Validation via AFP detection in clinical blood samples. Biosens Bioelectron 2025; 267:116771. [PMID: 39265427 DOI: 10.1016/j.bios.2024.116771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Enzyme-catalyzed chemiluminescence has been widely used in the field of biomedicine, especially in the test kit for various biomarkers. However, the currently reported enzyme-catalyzed chemiluminescence systems suffered from the addition of oxidizing substances, short emission wavelength, and susceptibility to interference by autofluorescence. In this paper, a universal sulfatase-based chemiluminescence system with NIR was developed, in which the designed substrate QM-CF could be transformed into 1,2-dioxetane derivate in the presence of sulfatase and oxygen. This system exhibited long emission wavelengths and CL half-time, a high signal-noise ratio, and without other additives. Importantly, the sulfatase-based chemiluminescence enzyme-linked immunoassay platform was successfully constructed and could be generally applied to detect biomarkers. As a proof of concept, the sulfatase-labeled AFP antibody and substate QM-CF were conveniently suitable for commercial AFP test kits, leading to satisfactory detection results of AFP in clinical blood samples.
Collapse
Affiliation(s)
- Cheng Zheng
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China; Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China; Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610044, China
| | - Mengyuan Cui
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yingyu Zhang
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, the First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Li Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Wenqing Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiaqi Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Min Ji
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Wei Chen
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610044, China
| | - Wen Jiang
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210009, China.
| | - Peng Wang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China; Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China.
| | - Wancun Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.
| |
Collapse
|
4
|
He XB, Jia X, Zhao PQ, Fang Z, Qing FL. Photoredox-Catalysis Fluorosulfonyldifluoromethylation of Unactivated Alkenes and (Hetero)arenes with ICF 2SO 2F. Org Lett 2024; 26:6900-6904. [PMID: 39115249 DOI: 10.1021/acs.orglett.4c02538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The fluorosulfonyldifluoromethylation of unactivated alkenes and (hetero)arenes with iododifluoromethanesulfonyl fluoride (ICF2SO2F) under visible light photoredox catalysis was successfully developed. Key to the successful fluorosulfonyldifluoromethylation of aromatic compounds was the usage of AgOTf as an additive to promote the formation of the CF2SO2F radical. The protocol provided a straightforward way to introduce the interesting and useful CF2SO2F group on sp3 and sp2 carbons.
Collapse
Affiliation(s)
- Xu-Biao He
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Lu, Shanghai 200032, China
| | - Xin Jia
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Lu, Shanghai 200032, China
| | - Pin-Qiao Zhao
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Lu, Shanghai 200032, China
| | - Zeguo Fang
- New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, Hubei University of Technology, Wuhan 430068, China
| | - Feng-Ling Qing
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Lu, Shanghai 200032, China
| |
Collapse
|
5
|
Chen J, Peng Z, Ji M, Wang P. A novel fluorescent probe for rapid detection of sulfatase in vitro and in living cells. Bioorg Chem 2023; 138:106655. [PMID: 37300960 DOI: 10.1016/j.bioorg.2023.106655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/23/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
Sulfatase participates in a variety of physiological processes in organisms including hormone regulation, cell signaling, and bacterial pathogenesis. Current sulfatase fluorescent probes can be used to track sulfate esterase overexpression in cancer cells for diagnostic purposes and to understand the pathological activity of sulfate esterase. However, some sulfatase fluorescent probes based on the hydrolysis of the sulfate bond were easily disturbed by the catalytic activity of sulfatase. Herein, we developed the fluorescent probe BQM-NH2 for sulfatase detection, which was based on the quinoline-malononitrile. The probe BQM-NH2 showed a fast response to sulfatase within 1 min and satisfactory sensitivity with a calculated LOD of 1.73 U/L. Importantly, it was successfully used to monitor endogenous sulfate in tumor cells, indicating BQM-NH2 has the potential to monitor sulfatase under physiological and pathological conditions.
Collapse
Affiliation(s)
- Junqing Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China.
| | - Zihao Peng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Min Ji
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, PR China
| | - Peng Wang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
6
|
Xiang MH, Jiang ZY, Zhao WL, Zhang E, Xia L, Kong RM, Zhao Y, Kong W, Liu X, Qu F, Tan W. Activatable Near-Infrared Fluorescent and Photoacoustic Dual-Modal Probe for Highly Sensitive Imaging of Sulfatase In Vivo. ACS Sens 2023; 8:2021-2029. [PMID: 37167101 DOI: 10.1021/acssensors.3c00201] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Sulfatase is an important biomarker closely associated with various diseases. However, the state-of-the-art sulfatase probes are plagued with a short absorption/emission wavelength and limited sensitivity. Developing highly sensitive fluorescent probes for in vivo imaging of sulfatase remains a grand challenge. Herein, for the first time, an activatable near-infrared fluorescence/photoacoustic (NIRF/PA) dual-modal probe (Hcy-SA) for visualizing sulfatase activity in living cells and animals is developed. Hcy-SA is composed of a sulfate ester moiety as the recognition unit and a NIR fluorophore hemicyanine (Hcy-OH) as the NIRF/PA reporter. The designed probe exhibits a rapid response, excellent sensitivity, and high specificity for sulfatase detection in vitro. More importantly, cells and in vivo experiments confirm that Hcy-SA can be successfully applied for PA/NIRF dual-modal imaging of sulfatase activity in living sulfatase-overexpressed tumor cells and tumor-bearing animals. This probe can serve as a promising tool for sulfatase-related pathological research and cancer diagnosis.
Collapse
Affiliation(s)
- Mei-Hao Xiang
- College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, P. R. China
| | - Zhi-Yuan Jiang
- College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, P. R. China
| | - Wen-Long Zhao
- College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, P. R. China
| | - Ensheng Zhang
- College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, P. R. China
| | - Lian Xia
- College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, P. R. China
| | - Rong-Mei Kong
- College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, P. R. China
| | - Yan Zhao
- College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, P. R. China
| | - Weiheng Kong
- College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, P. R. China
| | - Xianjun Liu
- College of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, Hunan, P. R. China
| | - Fengli Qu
- College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, P. R. China
- Cancer Hospital of Zhejiang Province, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Weihong Tan
- Cancer Hospital of Zhejiang Province, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| |
Collapse
|
7
|
Poirier D. Description of Chemical Synthesis, Nuclear Magnetic Resonance Characterization and Biological Activity of Estrane-Based Inhibitors/Activators of Steroidogenesis. Molecules 2023; 28:molecules28083499. [PMID: 37110733 PMCID: PMC10143840 DOI: 10.3390/molecules28083499] [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: 03/10/2023] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Steroid hormones play a crucial role in several aspects of human life, and steroidogenesis is the process by which hormones are produced from cholesterol using several enzymes that work in concert to obtain the appropriate levels of each hormone at the right time. Unfortunately, many diseases, such as cancer, endometriosis, and osteoporosis as examples, are caused by an increase in the production of certain hormones. For these diseases, the use of an inhibitor to block the activity of an enzyme and, in doing so, the production of a key hormone is a proven therapeutic strategy whose development continues. This account-type article focuses on seven inhibitors (compounds 1-7) and an activator (compound 8) of six enzymes involved in steroidogenesis, namely steroid sulfatase, aldo-keto reductase 1C3, types 1, 2, 3, and 12 of the 17β-hydroxysteroid dehydrogenases. For these steroid derivatives, three topics will be addressed: (1) Their chemical synthesis from the same starting material, estrone, (2) their structural characterization using nuclear magnetic resonance, and (3) their in vitro or in vivo biological activities. These bioactive molecules constitute potential therapeutic or mechanistic tools that could be used to better understand the role of certain hormones in steroidogenesis.
Collapse
Affiliation(s)
- Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec Research Center-Université Laval, Québec, QC G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
8
|
Mohamed A, Salah M, Tahoun M, Hawner M, Abdelsamie AS, Frotscher M. Dual Targeting of Steroid Sulfatase and 17β-Hydroxysteroid Dehydrogenase Type 1 by a Novel Drug-Prodrug Approach: A Potential Therapeutic Option for the Treatment of Endometriosis. J Med Chem 2022; 65:11726-11744. [PMID: 35993890 DOI: 10.1021/acs.jmedchem.2c00589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A novel approach for the dual inhibition of steroid sulfatase (STS) and 17β-hydroxysteroid dehydrogenase type 1(17β HSD1) by a single drug was explored, starting from in-house 17β HSD1 inhibitors via masking their phenolic OH group with a sulfamate ester. The sulfamates were intentionally designed as drugs for the inhibition of STS and, at the same time, prodrugs for 17β-HSD1 inhibition ("drug-prodrug approach"). The most promising sulfamates 13, 16, 18-20, 22-24, 36, and 37 showed nanomolar IC50 values for STS inhibition in a cellular assay and their corresponding phenols displayed potent 17β-HSD1 inhibition in cell-free and cellular assays, high selectivity over 17β-HSD2, reasonable metabolic stability, and low estrogen receptor α affinity. A close relationship was found between the liberation of the phenolic compound by sulfamate hydrolysis and 17β-HSD1 inactivation. These results showed that the envisaged drug-prodrug concept was successfully implemented. The novel compounds constitute a promising class of therapeutics for the treatment of endometriosis and other estrogen-dependent diseases.
Collapse
Affiliation(s)
- Abdelrahman Mohamed
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany.,Pharmaceutical Organic Chemistry Department, Assiut University, Assiut 71526, Egypt
| | - Mohamed Salah
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts, Cairo 12451, Egypt
| | - Mariam Tahoun
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany
| | - Manuel Hawner
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany
| | - Ahmed S Abdelsamie
- Department of Chemistry of Natural and Microbial Products, Institute of Pharmaceutical and Drug Industries Research, National Research Centre, El-Buhouth St., Dokki, P.O. Box 12622 Cairo 12451, Egypt.,Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E81, Saarbrücken 66123, Germany
| | - Martin Frotscher
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany
| |
Collapse
|
9
|
Biernacki K, Ciupak O, Daśko M, Rachon J, Kozak W, Rak J, Kubiński K, Masłyk M, Martyna A, Śliwka-Kaszyńska M, Wietrzyk J, Świtalska M, Nocentini A, Supuran CT, Demkowicz S. Development of Sulfamoylated 4-(1-Phenyl-1 H-1,2,3-triazol-4-yl)phenol Derivatives as Potent Steroid Sulfatase Inhibitors for Efficient Treatment of Breast Cancer. J Med Chem 2022; 65:5044-5056. [PMID: 35235747 PMCID: PMC8958511 DOI: 10.1021/acs.jmedchem.1c02220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We present here the advances achieved in the development of new sulfamoylated 4-(1-phenyl-1H-1,2,3-triazol-4-yl)phenol derivatives as potent steroid sulfatase (STS) inhibitors for the treatment of breast cancer. Prompted by promising biological results and in silico analysis, the initial series of similar compounds were extended, appending a variety of m-substituents at the outer phenyl ring. The inhibition profiles of the newly synthesized compounds were evaluated using a radioisotope enzymatic assay and, together with the preceding reported derivatives, using a radioisotope assay in MCF-7 cells. The most active compound, 5l, demonstrated an extraordinary STS inhibitory potency in MCF-7 cells with an IC50 value improved 5-fold compared to that of the reference Irosustat (0.21 vs 1.06 nM). The five most potent compounds were assessed in vivo in a 67NR mouse mammary gland cancer model, with 4b measured to induce up to 51% tumor growth inhibition at 50 mg/kg with no evidence of side effects and toxicity.
Collapse
Affiliation(s)
- Karol Biernacki
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Olga Ciupak
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Janusz Rachon
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Witold Kozak
- Department of Physical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Janusz Rak
- Department of Physical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Konrad Kubiński
- Department of Molecular Biology, Faculty of Biotechnology and Environment Sciences, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708 Lublin, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Faculty of Biotechnology and Environment Sciences, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708 Lublin, Poland
| | - Aleksandra Martyna
- Department of Molecular Biology, Faculty of Biotechnology and Environment Sciences, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708 Lublin, Poland
| | - Magdalena Śliwka-Kaszyńska
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Rudolfa Weigla 12, 53-114 Wrocław, Poland
| | - Marta Świtalska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Rudolfa Weigla 12, 53-114 Wrocław, Poland
| | - Alessio Nocentini
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via U. Schiff 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Claudiu T Supuran
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via U. Schiff 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| |
Collapse
|
10
|
Herman BE, Gardi J, Julesz J, Tömböly C, Szánti-Pintér E, Fehér K, Skoda-Földes R, Szécsi M. Steroidal ferrocenes as potential enzyme inhibitors of the estrogen biosynthesis. Biol Futur 2021; 71:249-264. [PMID: 34554507 DOI: 10.1007/s42977-020-00023-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 06/04/2020] [Indexed: 01/13/2023]
Abstract
The potential inhibitory effect of diverse triazolyl-ferrocene steroids on key enzymes of the estrogen biosynthesis was investigated. Test compounds were synthesized via copper-catalyzed cycloaddition of steroidal azides and ferrocenyl-alkynes using our efficient methodology published previously. Inhibition of human aromatase, steroid sulfatase (STS) and 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) activities was investigated with in vitro radiosubstrate incubations. Some of the test compounds were found to be potent inhibitors of the STS. A compound bearing ferrocenyl side chain on the C-2 displayed a reversible inhibition, whereas C-16 and C-17 derivatives displayed competitive irreversible binding mechanism toward the enzyme. 17α-Triazolyl-ferrocene derivatives of 17β-estradiol exerted outstanding inhibitory effect and experiments demonstrated a key role of the ferrocenyl moiety in the enhanced binding affinity. Submicromolar IC50 and Ki parameters enroll these compounds to the group of the most effective STS inhibitors published so far. STS inhibitory potential of the steroidal ferrocenes may lead to the development of novel compounds able to suppress in situ biosynthesis of 17β-estradiol in target tissues.
Collapse
Affiliation(s)
- Bianka Edina Herman
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary
| | - János Gardi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary
| | - János Julesz
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary
| | - Csaba Tömböly
- Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvári körút 62, P. O. Box 521, Szeged, 6726, Hungary
| | - Eszter Szánti-Pintér
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Egyetem utca 10, P. O. Box 158, Veszprém, 8200, Hungary
| | - Klaudia Fehér
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Egyetem utca 10, P. O. Box 158, Veszprém, 8200, Hungary
| | - Rita Skoda-Földes
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Egyetem utca 10, P. O. Box 158, Veszprém, 8200, Hungary.
| | - Mihály Szécsi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary.
| |
Collapse
|
11
|
Huang L, Huang J, Nie H, Li Y, Song L, Wu F. Design, synthesis and biological evaluation of combretastatin A-4 sulfamate derivatives as potential anti-cancer agents. RSC Med Chem 2021; 12:1374-1380. [PMID: 34458740 PMCID: PMC8372205 DOI: 10.1039/d0md00372g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 05/17/2021] [Indexed: 01/20/2023] Open
Abstract
A series of combretastatin A-4 (CA-4) sulfamate derivatives were synthesized and their structure-activity relationship on tubulin, arylsulfatase and tumor cell antiproliferation inhibition was studied. Among them, compound 16a showed excellent potency as well as CA-4 under the same conditions against six tumor cells including HTC-116, HeLa, HepG2, MGC803, MKN45 and MCF-7 cells, respectively. Molecular docking revealed that several important hydrogen bond interactions were formed between the sulfamate group of 16a and the colchicine binding site of tubulin and steroid sulfatase respectively. Although compound 16a was less active than CA-4 in regard to its in vitro activity as an inhibitor of tubulin polymerization, it was effective as an inhibitor of arylsulfatase. This novel combretastatin A-4 sulfamate derivative has the potential to be developed as a dual inhibitor of tubulin polymerization and arylsulfatase for cancer therapy.
Collapse
Affiliation(s)
- Leilei Huang
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Jinwen Huang
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
- Shanghai Engineering Research Center of Green Fluoropharmaceutical Technology China
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry CAS China
| | - Hui Nie
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Yingzi Li
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Lixing Song
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Fanhong Wu
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
- Shanghai Engineering Research Center of Green Fluoropharmaceutical Technology China
| |
Collapse
|
12
|
Novel 1,2,3-Triazole Derivatives as Mimics of Steroidal System-Synthesis, Crystal Structures Determination, Hirshfeld Surfaces Analysis and Molecular Docking. Molecules 2021; 26:molecules26134059. [PMID: 34279404 PMCID: PMC8271369 DOI: 10.3390/molecules26134059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 11/17/2022] Open
Abstract
Herein, we present the synthesis and crystal structures determination of five 4-(1-phenyl-1H-1,2,3-triazol-4-yl)phenol derivatives containing halogen atoms, 6a-e, which may be used as an excellent mimic of steroids in the drug development process. Good quality crystals obtained for all of the synthesized compounds allowed the analysis of their molecular structures. Subsequently, the determined crystal structures were used to calculate the Hirshfeld surfaces for each of the synthesized compounds. Furthermore, results of our docking studies indicated that synthesized derivatives are able to bind effectively to the active sites of selected enzymes and receptors involved in the hormone biosynthesis and signaling pathways, analogously to the native steroids.
Collapse
|
13
|
Ciupak O, Daśko M, Biernacki K, Rachon J, Masłyk M, Kubiński K, Martyna A, Demkowicz S. New potent steroid sulphatase inhibitors based on 6-(1-phenyl-1 H-1,2,3-triazol-4-yl)naphthalen-2-yl sulphamate derivatives. J Enzyme Inhib Med Chem 2021; 36:238-247. [PMID: 33322953 PMCID: PMC7744152 DOI: 10.1080/14756366.2020.1858820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In the present work, we report a new class of potent steroid sulphatase (STS) inhibitors based on 6-(1-phenyl-1H-1,2,3-triazol-4-yl)naphthalen-2-yl sulphamate derivatives. Within the set of new STS inhibitors, 6-(1-(1,2,3-trifluorophenyl)-1H-1,2,3-triazol-4-yl)naphthalen-2-yl sulphamate 3L demonstrated the highest activity in the enzymatic assay inhibiting the STS activity to 7.98% at 0.5 µM concentration. Furthermore, to verify whether the obtained STS inhibitors are able to pass through the cellular membrane effectively, cell line experiments have been carried out. We found that the lowest STS activities were measured in the presence of compound 3L (remaining STS activity of 5.22%, 27.48% and 99.0% at 100, 10 and 1 nM concentrations, respectively). The measured STS activities for Irosustat (used as a reference) were 5.72%, 12.93% and 16.83% in the same concentration range. Moreover, a determined IC50 value of 15.97 nM for 3L showed that this compound is a very promising candidate for further preclinical investigations.
Collapse
Affiliation(s)
- Olga Ciupak
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Karol Biernacki
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Janusz Rachon
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Konrad Kubiński
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Aleksandra Martyna
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| |
Collapse
|
14
|
Anbar HS, Isa Z, Elounais JJ, Jameel MA, Zib JH, Samer AM, Jawad AF, El-Gamal MI. Steroid sulfatase inhibitors: the current landscape. Expert Opin Ther Pat 2021; 31:453-472. [PMID: 33783295 DOI: 10.1080/13543776.2021.1910237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Steroid sulfatase (STS) enzyme is responsible for transforming the inactive sulfate metabolites of steroid sex hormones into the active free steroids. Both the deficiency and the over-expression of STS are associated with the pathophysiology of certain diseases. This article provides the readership with a comprehensive review about STS enzyme and its recently reported inhibitors.Areas covered: In the present article, we reviewed the structure, location, and substrates of STS enzyme, physiological functions of STS, and disease states related to over-expression or deficiency of STS enzyme. STS inhibitors reported during the last five years (2016-present) have been reviewed as well.Expert opinion: Irosustat is the most successful STS inhibitor drug candidate so far. It is currently under investigation in clinical trials for treatment of estrogen-dependent breast cancer. Non-steroidal sulfamate is the most favorable scaffold for STS inhibitor design. They can be beneficial for the treatment of hormone-dependent cancers and neurodegenerative disorders without significant estrogenic side effects. Moreover, dual-acting molecules (inhibitors of STS + another synergistic mechanism) can be therapeutically efficient.
Collapse
Affiliation(s)
- Hanan S Anbar
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Zahraa Isa
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Jana J Elounais
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Mariam A Jameel
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Joudi H Zib
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Aya M Samer
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Aya F Jawad
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Mohammed I El-Gamal
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medicinal Chemistry, Faculty of Pharmacy, University of Mansoura, Mansoura, Egypt
| |
Collapse
|
15
|
A Targeted-Covalent Inhibitor of 17β-HSD1 Blocks Two Estrogen-Biosynthesis Pathways: In Vitro (Metabolism) and In Vivo (Xenograft) Studies in T-47D Breast Cancer Models. Cancers (Basel) 2021; 13:cancers13081841. [PMID: 33924352 PMCID: PMC8069897 DOI: 10.3390/cancers13081841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary 17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) is responsible for the production of estrogens estradiol (E2) and 5-androsten-3β,17β-diol (5-diol). This enzyme is therefore a target of choice for the treatment of estrogen-dependent diseases such as breast cancer and endometriosis, by blocking estrogen biosynthesis. After we developed the first irreversible and non-estrogenic 17β-HSD1 inhibitor, a molecule named PBRM, our goal was to demonstrate its therapeutic potential. PBRM was able to block the formation of E2 and 5-diol in T-47D human breast cancer cells. When given orally to mice, PBRM was also able to block the tumor growth without any observed toxic effects. Thanks to its irreversible type of inhibition, PBRM retained its anti-tumor growth effect, even after reducing its frequency of administration to only once a week, a clear advantage over reversible inhibitors. These results strongly support the use of PBRM as a new approach in the treatment of breast cancer. Abstract 17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) plays an important role in estrogen-dependent breast tumor growth. In addition to being involved in the production of estradiol (E2), the most potent estrogen in women, 17β-HSD1 is also responsible for the production of 5-androsten-3β,17β-diol (5-diol), a weaker estrogen than E2, but whose importance increases after menopause. 17β-HSD1 is therefore a target of choice for the treatment of estrogen-dependent diseases such as breast cancer and endometriosis. After we developed the first targeted-covalent (irreversible) and non-estrogenic inhibitor of 17β-HSD1, a molecule named PBRM, our goal was to demonstrate its therapeutic potential. Enzymatic assays demonstrated that estrone (E1) and dehydroepiandrosterone (DHEA) were transformed into E2 and 5-diol in T-47D human breast cancer cells, and that PBRM was able to block these transformations. Thereafter, we tested PBRM in a mouse tumor model (cell-derived T-47D xenografts). After treatment of ovariectomized (OVX) mice receiving E1 or DHEA, PBRM given orally was able to reduce the tumor growth at the control (OVX) level without any observed toxic effects. Thanks to its irreversible type of inhibition, PBRM retained its anti-tumor growth effect, even after reducing its frequency of administration to only once a week, a clear advantage over reversible inhibitors.
Collapse
|
16
|
Daśko M, Demkowicz S, Biernacki K, Ciupak O, Kozak W, Masłyk M, Rachon J. Recent progress in the development of steroid sulphatase inhibitors - examples of the novel and most promising compounds from the last decade. J Enzyme Inhib Med Chem 2020; 35:1163-1184. [PMID: 32363947 PMCID: PMC7241464 DOI: 10.1080/14756366.2020.1758692] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 01/08/2023] Open
Abstract
The purpose of this review article is to provide an overview of recent achievements in the synthesis of novel steroid sulphatase (STS) inhibitors. STS is a crucial enzyme in the biosynthesis of active hormones (including oestrogens and androgens) and, therefore, represents an extremely attractive molecular target for the development of hormone-dependent cancer therapies. The inhibition of STS may effectively reduce the availability of active hormones for cancer cells, causing a positive therapeutic effect. Herein, we report examples of novel STS inhibitors based on steroidal and nonsteroidal cores that contain various functional groups (e.g. sulphamate and phosphorus moieties) and halogen atoms, which may potentially be used in therapies for hormone-dependent cancers. The presented work also includes examples of multitargeting agents with STS inhibitory activities. Furthermore, the fundamental discoveries in the development of the most promising drug candidates exhibiting STS inhibitory activities are highlighted.
Collapse
Affiliation(s)
- Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Karol Biernacki
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Olga Ciupak
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Witold Kozak
- Department of Physical Chemistry, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Faculty of Biotechnology and Environment Sciences, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Janusz Rachon
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| |
Collapse
|
17
|
Daśko M, Demkowicz S, Rachon J, Biernacki K, Aszyk J, Kozak W, Masłyk M, Kubiński K. New potent STS inhibitors based on fluorinated 4-(1-phenyl-1 H-[1,2,3]triazol-4-yl)-phenyl sulfamates. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2020; 22:1037-1044. [PMID: 31773975 DOI: 10.1080/10286020.2019.1680642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/08/2019] [Indexed: 06/10/2023]
Abstract
A series of fluorinated analogs based on the frameworks of 4-(1-phenyl-1H-[1,2,3]triazol-4-yl)-phenyl sulfamates have been synthesized as steroid sulfatase (STS) inhibitors. The design of chemical structures of new potential STS inhibitors was supported by molecular docking techniques to identify potential interactions between inhibitors and amino acid residues located in the STS active site. The STS inhibitory potency was evaluated on STS isolated from human placenta. We found that compounds substituted with fluorine atom at the meta position demonstrated the highest inhibitory effects in enzymatic STS assay. The most active analog 12e - inhibited STS enzyme with the IC50 value of 36 nM.
Collapse
Affiliation(s)
- Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, Gdansk 80-233, Poland
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, Gdansk 80-233, Poland
| | - Janusz Rachon
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, Gdansk 80-233, Poland
| | - Karol Biernacki
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, Gdansk 80-233, Poland
| | - Justyna Aszyk
- Department of Analytical Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, Gdansk 80-233, Poland
| | - Witold Kozak
- Department of Physical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk 80-308, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Faculty of Biotechnology and Environment Sciences, the John Paul II Catholic University of Lublin, Konstantynów 1i, Lublin 20-708, Poland
| | - Konrad Kubiński
- Department of Molecular Biology, Faculty of Biotechnology and Environment Sciences, the John Paul II Catholic University of Lublin, Konstantynów 1i, Lublin 20-708, Poland
| |
Collapse
|
18
|
Maltais R, Ngueta Djiemeny A, Roy J, Barbeau X, Lambert JP, Poirier D. Design and synthesis of dansyl-labeled inhibitors of steroid sulfatase for optical imaging. Bioorg Med Chem 2020; 28:115368. [PMID: 32122754 DOI: 10.1016/j.bmc.2020.115368] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/28/2020] [Accepted: 02/03/2020] [Indexed: 02/06/2023]
Abstract
Steroid sulfatase (STS) is an important enzyme regulating the conversion of sulfated steroids into their active hydroxylated forms. Notably, the inhibition of STS has been shown to decrease the levels of active estrogens and was translated into clinical trials for the treatment of breast cancer. Based on quantitative structure-activity relationship (QSAR) and molecular modeling studies, we herein report the design of fluorescent inhibitors of STS by adding a dansyl group on an estrane scaffold. Synthesis of 17α-dansylaminomethyl-estradiol (7) and its sulfamoylated analog 8 were achieved from estrone in 5 and 6 steps, respectively. Inhibition assays on HEK-293 cells expressing exogenous STS revealed a high level of inhibition for compound 7 (IC50 = 69 nM), a value close to the QSAR model prediction (IC50 = 46 nM). As an irreversible inhibitor, sulfamate 8 led to an even more potent inhibition in the low nanomolar value (IC50 = 2.1 nM). In addition, we show that the potent STS inhibitor 8 can be employed as an optical imaging tool to investigate intracellular enzyme sub-localization as well as inhibitory behavior. As a result, confocal microscopy analysis confirmed good penetration of the STS fluorescent inhibitor 8 in cells and its localization in the endoplasmic reticulum where STS is localized.
Collapse
Affiliation(s)
- René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada
| | - Adrien Ngueta Djiemeny
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada
| | - Xavier Barbeau
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada
| | - Jean-Philippe Lambert
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
19
|
Hng Y, Lin MH, Lin TS, Liu IC, Lin IC, Lu YL, Chang CN, Chiu PF, Tsai KC, Chen MJ, Liang PH. Design and synthesis of 3-benzylaminocoumarin-7-O-sulfamate derivatives as steroid sulfatase inhibitors. Bioorg Chem 2020; 96:103618. [PMID: 32059152 DOI: 10.1016/j.bioorg.2020.103618] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 01/06/2023]
Abstract
Steroid sulfatase (STS) is a sulfatase enzyme that catalyzes the conversion of sulfated steroid precursors to free steroid. The inhibition of STS could abate estrogenic steroids that stimulate the proliferation and development of breast cancer, and therefore STS is a potential target for adjuvant endocrine therapy. In this study, a series of 3-benzylaminocoumarin-7-O-sulfamate derivatives targeting STS were designed and synthesized. Structure-relationship activities (SAR) analysis revealed that attachment of a benzylamino group at the 3-position of coumarin improved inhibitory activity. Compound 3j was found to have the highest inhibition activity against human placenta isolated STS (IC50 0.13 μM) and MCF-7 cell lines (IC50 1.35 µM). Kinetic studies found compound 3j to be an irreversible inhibitor of STS, with KI and kinact value of 86.9 nM and 158.7 min-1, respectively.
Collapse
Affiliation(s)
- Yue Hng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Mei-Hsiang Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Tzung-Sheng Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - I-Chen Liu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - I-Chun Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yeh-Lin Lu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan; School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Chiao-Nien Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Pei-Fang Chiu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan
| | - Mei-Jou Chen
- Department of Obstetrics and Gynecology and Livia Shangyu Wan Scholar, National Taiwan University Hospital, National Taiwan University, College of Medicine, Taipei 100, Taiwan
| | - Pi-Hui Liang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Genomics Research Center, Academia Sinica, Taipei 128, Taiwan.
| |
Collapse
|
20
|
Synthesis and in vitro evaluation of piperazinyl-ureido sulfamates as steroid sulfatase inhibitors. Eur J Med Chem 2019; 182:111614. [PMID: 31422224 DOI: 10.1016/j.ejmech.2019.111614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/26/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Two new piperazinyl-ureido single ring aryl sulfamate-based inhibitor series were designed against the emerging oncology drug target steroid sulfatase (STS), for which there are existing potent steroidal and non-steroidal agents in clinical trials. 4-(Piperazinocarbonyl)aminosulfamates (5-31) were obtained by reacting 4-hydroxyarylamines with phenylchloroformate, subsequent sulfamoylation of the resulting hydroxyarylcarbamates and coupling of the product with 1-substituted piperazines. Pyrimidinyl-piperazinourea sulfamates (35-42) were synthesized by pyrimidine ring closure of 4-Boc-piperazine-1-carboxamidine with 3-(dimethylamino)propenones, deprotection and coupling with the sulfamoylated building block. Target ureidosulfamates 5-31 and 35-42 were evaluated both as STS inhibitors in vitro using a lysate of JEG-3 human placenta choriocarcinoma cell line and in a whole cell assay. SAR conclusions were drawn from both series. In series 35-42 the best inhibitory activity is related to the presence of a benzofuryl on the pyrimidine ring. In series 5-31 the best inhibitory activity was shown by the ureas bearing 4-chlorophenyl, 3,4-dichlorophenyl groups or aliphatic chains at the piperazino 4-nitrogen displaying IC50 in the 33-94 nM concentration range. Final optimization to the low nanomolar level was achieved through substitution of the arylsulfamate ring with halogens. Four halogenated arylsulfamates of high potency were achieved and two of these 19 and 20 had IC50 values of 5.1 and 8.8 nM respectively and are attractive for potential in vivo evaluation and further development. We demonstrate the optimization of this new series to low nanomolar potency, employing fluorine substitution, providing potent membrane permeant inhibitors with further development potential indicating piperazinyl-ureido aryl sulfamate derivatives as an attractive new class of STS inhibitors.
Collapse
|
21
|
Daśko M, Demkowicz S, Biernacki K, Harrous A, Rachon J, Kozak W, Martyna A, Masłyk M, Kubiński K, Boguszewska‐Czubara A. Novel steroid sulfatase inhibitors based on
N
‐thiophosphorylated 3‐(4‐aminophenyl)‐coumarin‐7‐O‐sulfamates. Drug Dev Res 2019; 80:857-866. [DOI: 10.1002/ddr.21569] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/31/2019] [Accepted: 06/29/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of ChemistryGdansk University of Technology Gdansk Poland
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Faculty of ChemistryGdansk University of Technology Gdansk Poland
| | - Karol Biernacki
- Department of Organic Chemistry, Faculty of ChemistryGdansk University of Technology Gdansk Poland
| | - Amira Harrous
- Department of Organic Chemistry, Faculty of ChemistryGdansk University of Technology Gdansk Poland
| | - Janusz Rachon
- Department of Organic Chemistry, Faculty of ChemistryGdansk University of Technology Gdansk Poland
| | - Witold Kozak
- Department of Physical Chemistry, Faculty of ChemistryUniversity of Gdansk Gdansk Poland
| | - Aleksandra Martyna
- Department of Molecular Biology, Faculty of Biotechnology and Environment SciencesThe John Paul II Catholic University of Lublin Lublin Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Faculty of Biotechnology and Environment SciencesThe John Paul II Catholic University of Lublin Lublin Poland
| | - Konrad Kubiński
- Department of Molecular Biology, Faculty of Biotechnology and Environment SciencesThe John Paul II Catholic University of Lublin Lublin Poland
| | | |
Collapse
|
22
|
Sulfamates in drug design and discovery: Pre-clinical and clinical investigations. Eur J Med Chem 2019; 179:257-271. [PMID: 31255926 DOI: 10.1016/j.ejmech.2019.06.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022]
Abstract
In the present article, we reviewed the sulfamate-containing compounds reported as bioactive molecules. The possible molecular targets of sulfamate derivatives include steroid sulfatase enzyme, carbonic anhydrases, acyl transferase, and others. Sulfamate derivatives can help treat hormone-dependent tumors including breast, prostate, and endometrial cancers, Binge eating disorder, migraine, glaucoma, weight loss, and epilepsy. Sulfamate derivatives can act also as calcium sensing receptor agonists and can aid in osteoporosis. Furthermore, acyl sulfamate derivatives can act as antibacterial agents against Gram-positive bacteria. A recent study revealed a new side effect of topiramate, a sulfamate-containing compound, which is sialolithiasis. The structural and biological characteristics of the reviewed compounds are presented in detail.
Collapse
|
23
|
Kim Y, Lee J, Jung J, Kim SG. Chiral Brønsted acid-catalyzed Friedel–Crafts reaction of 3-indolylsulfamidates with indoles: Synthesis of enantioenriched bisindolylmethane sulfamates. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
24
|
Saha T, Makar S, Swetha R, Gutti G, Singh SK. Estrogen signaling: An emanating therapeutic target for breast cancer treatment. Eur J Med Chem 2019; 177:116-143. [PMID: 31129450 DOI: 10.1016/j.ejmech.2019.05.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/15/2022]
Abstract
Breast cancer, a most common malignancy in women, was known to be associated with steroid hormone estrogen. The discovery of estrogen receptor (ER) gave us not only a powerful predictive and prognostic marker, but also an efficient target for the treatment of hormone-dependent breast cancer with various estrogen ligands. ER consists of two subtypes i.e. ERα and ERβ, that are mostly G-protein-coupled receptors and activated by estrogen, specially 17β-estradiol. The activation is followed by translocation into the nucleus and binding with DNA to modulate activities of different genes. ERs can manage synthesis of RNA through genomic actions without directly binding to DNA. Receptors are tethered by protein-protein interactions to a transcription factor complex to communicate with DNA. Estrogens also exhibit nongenomic actions, a characteristic feature of steroid hormones, which are so rapid to be considered by the activation of RNA and translation. These are habitually related to stimulation of different protein kinase cascades. Majority of post-menopausal breast cancer is estrogen dependent, mostly potent biological estrogen (E2) for continuous growth and proliferation. Estrogen helps in regulating the differentiation and proliferation of normal breast epithelial cells. In this review we have investigated the important role of ER in development and progression of breast cancer, which is complicated by receptor's interaction with co-regulatory proteins, cross-talk with other signal transduction pathways and development of treatment strategies viz. selective estrogen receptor modulators (SERMs), selective estrogen receptor down regulators (SERDs), aromatase and sulphatase inhibitors.
Collapse
Affiliation(s)
- Tanmay Saha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Subhajit Makar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Rayala Swetha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Gopichand Gutti
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Sushil K Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India.
| |
Collapse
|
25
|
New organotin(IV) chlorides derived from N-(2-hydroxyphenyl)aryloxy sulfamates. Synthesis, characterization and DSC investigation. J CHEM SCI 2019. [DOI: 10.1007/s12039-018-1586-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
蒋 国, 刘 亚, 赵 婉, 王 道, 董 淑, 童 旭. [Effect of gap junction modulation on antitumor effects of adriamycin in estrogen receptor-positive breast cancer cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:780-786. [PMID: 33168517 PMCID: PMC6765543 DOI: 10.3969/j.issn.1673-4254.2018.07.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To observe the effect of functional modulation of gap junctions (GJ) on the antitumor effect of adriamycin in breast cancer cells positive for estrogen receptor (ER). METHODS The inhibitory effect of 0 to 24.0 μmol/L adriamycin on the surviving fraction of ER-positive human breast cancer MCF-7 cells and ER-negative MDA-MB-231 cells was assessed with MTT assay; Western blotting and immunofluorescence assay were used to detect the expressions of Cx43 total protein and membrane protein in the cells. A parachute assay was used to evaluate the function of the GJ in MCF-7 cells. The cytotoxic effect of adriamycin was observed in the cells treated with retinoic acid (RA) for enhancing GJ function, in cells treated with oleamide and 18-α- glycyrrhizic acid (18-α-ga) for inhibiting GJ function, and also in cells transfected with Cx43siRNA for Cx43 knockdown. RESULTS ER-positive MCF-7 cells expressed a significantly higher level of Cx43 with stronger GJ function than ER-negative MDA- MB-231 cells. Adriamycin significantly inhibited the proliferation of MCF-7 cells (P < 0.01), and RA treatment further increased the cytotoxicity of adriamycin (P < 0.01) while oleamide and 18-α-GA obviously attenuated the cytotoxicity of adriamycin (P < 0.01). In the cells with Cx43 knockdown, the expressions of total Cx43 protein and Cx43 on the membrane were significantly reduced, the function of GJ was attenuated, and the cytotoxicity of adriamycin was significantly decreased (P < 0.01). CONCLUSIONS ER-positive breast cancer cells have stronger Cx43 expressions and GJ function than the ERnegative cells. The cytotoxicity of adriamycin against the breast cancer cells can be strengthened by enhancing GJ function and attenuated by inhibiting GJ function. Cx43 silencing inhibits the function of GJ to lower the cytotoxicity of adriamycin in human breast cancer MCF-7 cells.
Collapse
Affiliation(s)
- 国君 蒋
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 亚明 刘
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 婉晨 赵
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 道鑫 王
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 淑英 董
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| | - 旭辉 童
- />蚌埠医学院药学院药理教研室,安徽 蚌埠 233030Pharmacology department of Bengbu Medical college, Bengbu 233030, China
| |
Collapse
|
27
|
Quest for steroidomimetics: Amino acids derived steroidal and nonsteroidal architectures. Eur J Med Chem 2017; 133:139-151. [DOI: 10.1016/j.ejmech.2017.03.054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 11/18/2022]
|
28
|
Kahveci B, Yılmaz F, Menteşe E, Ülker S. Design, Synthesis, and Biological Evaluation of Coumarin-Triazole Hybrid Molecules as Potential Antitumor and Pancreatic Lipase Agents. Arch Pharm (Weinheim) 2017; 350. [PMID: 28543820 DOI: 10.1002/ardp.201600369] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/07/2017] [Accepted: 04/17/2017] [Indexed: 12/25/2022]
Abstract
The design, synthesis, and investigation of antitumor and anti-lipase activities of some coumarin-triazole hybrid molecules are reported. The synthesis of these hybrid molecules was performed under microwave irradiation and conventional heating procedures. The newly synthesized hybrid molecules were investigated as inhibitors against four tumor cell lines (BT20 human breast carcinoma, SK-Mel 128 melanoma, DU-145 prostate carcinoma, and A549 lung carcinoma) and porcine pancreatic lipase (PPL). Most of these compounds showed notable antitumor activities against the tested tumor cell lines, and compounds 8i and 8l showed the best anti-lipase activity of 99.30 ± 0.56% and 99.85 ± 1.21%, respectively, at a concentration of 10 μM.
Collapse
Affiliation(s)
- Bahittin Kahveci
- Faculty of Health Sciences, Department of Nutrition and Diethetics, Karadeniz Technical University, Trabzon, Turkey
| | - Fatih Yılmaz
- Faculty of Arts and Sciences, Department of Chemistry, Recep Tayyip Erdogan University, Rize, Turkey
| | - Emre Menteşe
- Faculty of Arts and Sciences, Department of Chemistry, Recep Tayyip Erdogan University, Rize, Turkey
| | - Serdar Ülker
- Faculty of Arts and Sciences, Department of Biology, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
29
|
Daśko M, Rachon J, Masłyk M, Kubiński K, Demkowicz S. Synthesis and biological evaluation ofN-acylated tyramine sulfamates containing C-F bonds as steroid sulfatase inhibitors. Chem Biol Drug Des 2017; 90:156-161. [DOI: 10.1111/cbdd.12931] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 11/30/2016] [Accepted: 12/13/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Mateusz Daśko
- Department of Organic Chemistry; Faculty of Chemistry; Gdansk University of Technology; Gdansk Poland
| | - Janusz Rachon
- Department of Organic Chemistry; Faculty of Chemistry; Gdansk University of Technology; Gdansk Poland
| | - Maciej Masłyk
- Department of Molecular Biology; Faculty of Biotechnology and Environmental Sciences; The John Paul II Catholic University of Lublin; Lublin Poland
| | - Konrad Kubiński
- Department of Molecular Biology; Faculty of Biotechnology and Environmental Sciences; The John Paul II Catholic University of Lublin; Lublin Poland
| | - Sebastian Demkowicz
- Department of Organic Chemistry; Faculty of Chemistry; Gdansk University of Technology; Gdansk Poland
| |
Collapse
|
30
|
Garbacz WG, Jiang M, Xie W. Sex-Dependent Role of Estrogen Sulfotransferase and Steroid Sulfatase in Metabolic Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:455-469. [PMID: 29224107 DOI: 10.1007/978-3-319-70178-3_21] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sulfonation and desulfation are two opposing processes that represent an important layer of regulation of estrogenic activity via ligand supplies. Enzymatic activities of families of enzymes, known as sulfotransferases and sulfatases, lead to structural and functional changes of the steroids, thyroids, xenobiotics, and neurotransmitters. Estrogen sulfotransferase (EST) and steroid sulfatase (STS) represent negative and positive regulation of the estrogen activity, respectively. This is because EST-mediated sulfation deactivates estrogens, whereas STS-mediated desulfation converts the inactive estrogen sulfates to active estrogens. In addition to the known functions of estrogens, EST and STS in reproductive processes, regulation of estrogens and other signal molecules especially at the local tissue levels has gained increased attention in the context of metabolic disease in recent years. EST expression is detectable in the subcutaneous adipose tissue in both obese women and men, and the expression of EST is markedly induced in the livers of rodent models of obesity and type 2 diabetes. STS was found to be upregulated in patients with chronic inflammatory liver diseases. Interestingly, the tissue distribution and the transcriptional regulation of EST and STS exhibit obvious sex and species specificity. EST ablation produces completely opposite metabolic phenotype in female and male obese mice. Adipogenesis is also differentially regulated by EST in murine and human adipocytes. This chapter focuses on the recent progress in our understanding of the expression and regulation EST and STS in the context of metabolic homeostasis.
Collapse
Affiliation(s)
- Wojciech G Garbacz
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mengxi Jiang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
31
|
Ouellet C, Maltais R, Ouellet É, Barbeau X, Lagüe P, Poirier D. Discovery of a sulfamate-based steroid sulfatase inhibitor with intrinsic selective estrogen receptor modulator properties. Eur J Med Chem 2016; 119:169-82. [PMID: 27155470 DOI: 10.1016/j.ejmech.2016.04.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/15/2016] [Accepted: 04/16/2016] [Indexed: 01/27/2023]
Abstract
Steroid sulfatase (STS), the enzyme which converts inactive sulfated steroid precursors into active hormones, is a promising therapeutic target for the treatment of estrogen-sensitive breast cancer. We report herein the synthesis and in vitro study of dual-action STS inhibitors with selective estrogen-receptor modulator (SERM) effects. A library of tetrahydroisoquinoline-N-substituted derivatives (phenolic compounds) was synthesized by solid-phase chemistry and tested on estrogen-sensitive breast cancer T-47D cells. Three phenolic compounds devoid of estrogenic activity and toxicity emerged from this screening. Their sulfamate analogs were then synthesized, tested in STS-transfected HEK-293 cells, and found to be potent inhibitors of the enzyme (IC50 of 3.9, 8.9, and 16.6 nM). When tested in T-47D cells they showed no estrogenic activity and produced a moderate antiestrogenic activity. The compounds were further tested on osteoblast-like Saos-2 cells and found to significantly stimulate their proliferation as well as their alkaline phosphatase activity, thus suggesting a SERM activity. These results are supported by molecular docking experiments.
Collapse
Affiliation(s)
- Charles Ouellet
- Laboratory of Medicinal Chemistry, CHU de Québec - Research Center (CHUL, T4), 2705 Laurier Boulevard, Québec, QC, G1V 4G2, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, CHU de Québec - Research Center (CHUL, T4), 2705 Laurier Boulevard, Québec, QC, G1V 4G2, Canada
| | - Étienne Ouellet
- Laboratory of Medicinal Chemistry, CHU de Québec - Research Center (CHUL, T4), 2705 Laurier Boulevard, Québec, QC, G1V 4G2, Canada
| | - Xavier Barbeau
- Département de chimie, Institut de biologie intégrative et des systèmes (IBIS), Centre de recherche sur la fonction, la structure et l'ingénierie des protéines (PROTEO), Université Laval, Québec City, QC, Canada
| | - Patrick Lagüe
- Département de biochimie microbiologie et bio-informatique, Institut de biologie intégrative et des systèmes (IBIS), Centre de recherche sur la fonction, la structure et l'ingénierie des protéines (PROTEO), Université Laval, Québec City, QC, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, CHU de Québec - Research Center (CHUL, T4), 2705 Laurier Boulevard, Québec, QC, G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|