1
|
El Abbouchi A, Mkhayar K, Elkhattabi S, El Brahmi N, Hiebel MA, Bignon J, Guillaumet G, Suzenet F, El Kazzouli S. Design, Synthesis, Computational Studies, and Anti-Proliferative Evaluation of Novel Ethacrynic Acid Derivatives Containing Nitrogen Heterocycle, Urea, and Thiourea Moieties as Anticancer Agents. Molecules 2024; 29:1437. [PMID: 38611717 PMCID: PMC11013014 DOI: 10.3390/molecules29071437] [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: 02/12/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
In the present work, the synthesis of new ethacrynic acid (EA) derivatives containing nitrogen heterocyclic, urea, or thiourea moieties via efficient and practical synthetic procedures was reported. The synthesised compounds were screened for their anti-proliferative activity against two different cancer cell lines, namely, HL60 (promyelocytic leukaemia) and HCT116 (human colon carcinoma). The results of the in vitro tests reveal that compounds 1-3, 10, 16(a-c), and 17 exhibit potent anti-proliferative activity against the HL60 cell line, with values of the percentage of cell viability ranging from 20 to 35% at 1 μM of the drug and IC50 values between 2.37 μM and 0.86 μM. Compounds 2 and 10 showed a very interesting anti-proliferative activity of 28 and 48% at 1 μM, respectively, against HCT116. Two PyTAP-based fluorescent EA analogues were also synthesised and tested, showing good anti-proliferative activity. A test on the drug-likeness properties in silico of all the synthetised compounds was performed in order to understand the mechanism of action of the most active compounds. A molecular docking study was conducted on two human proteins, namely, glutathione S-transferase P1-1 (pdb:2GSS) and caspase-3 (pdb:4AU8) as target enzymes. The docking results show that compounds 2 and 3 exhibit significant binding modes with these enzymes. This finding provides a potential strategy towards developing anticancer agents, and most of the synthesised and newly designed compounds show good drug-like properties.
Collapse
Affiliation(s)
- Abdelmoula El Abbouchi
- Euromed Research Center, Euromed Faculty of Pharmacy, School of Engineering in Biomedical and Biotechnology, Euromed University of Fes (UEMF), Meknes Road, Fez 30000, Morocco; (A.E.A.); (N.E.B.)
- Institut de Chimie Organique et Analytique, Université d’Orléans, UMR CNRS 7311, BP 6759, CEDEX 2, 45067 Orléans, France; (M.-A.H.); (F.S.)
| | - Khaoula Mkhayar
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, Fez 30040, Morocco; (K.M.); (S.E.)
| | - Souad Elkhattabi
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, Fez 30040, Morocco; (K.M.); (S.E.)
| | - Nabil El Brahmi
- Euromed Research Center, Euromed Faculty of Pharmacy, School of Engineering in Biomedical and Biotechnology, Euromed University of Fes (UEMF), Meknes Road, Fez 30000, Morocco; (A.E.A.); (N.E.B.)
| | - Marie-Aude Hiebel
- Institut de Chimie Organique et Analytique, Université d’Orléans, UMR CNRS 7311, BP 6759, CEDEX 2, 45067 Orléans, France; (M.-A.H.); (F.S.)
| | - Jérôme Bignon
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France;
| | - Gérald Guillaumet
- Euromed Research Center, Euromed Faculty of Pharmacy, School of Engineering in Biomedical and Biotechnology, Euromed University of Fes (UEMF), Meknes Road, Fez 30000, Morocco; (A.E.A.); (N.E.B.)
- Institut de Chimie Organique et Analytique, Université d’Orléans, UMR CNRS 7311, BP 6759, CEDEX 2, 45067 Orléans, France; (M.-A.H.); (F.S.)
| | - Franck Suzenet
- Institut de Chimie Organique et Analytique, Université d’Orléans, UMR CNRS 7311, BP 6759, CEDEX 2, 45067 Orléans, France; (M.-A.H.); (F.S.)
| | - Saïd El Kazzouli
- Euromed Research Center, Euromed Faculty of Pharmacy, School of Engineering in Biomedical and Biotechnology, Euromed University of Fes (UEMF), Meknes Road, Fez 30000, Morocco; (A.E.A.); (N.E.B.)
| |
Collapse
|
2
|
Zhang J, Ye ZW, Morgenstern R, Townsend DM, Tew KD. Microsomal glutathione transferase 1 in cancer and the regulation of ferroptosis. Adv Cancer Res 2023; 160:107-132. [PMID: 37704286 PMCID: PMC10586476 DOI: 10.1016/bs.acr.2023.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Microsomal glutathione transferase 1 (MGST1) is a member of the MAPEG family (membrane associated proteins in eicosanoid and glutathione metabolism), defined according to enzymatic activities, sequence motifs, and structural properties. MGST1 is a homotrimer which can bind three molecules of glutathione (GSH), with one modified to a thiolate anion displaying one-third-of-sites-reactivity. MGST1 has both glutathione transferase and peroxidase activities. Each is based on stabilizing the GSH thiolate in the same active site. MGST1 is abundant in the liver and displays a broad subcellular distribution with high levels in endoplasmic reticulum and mitochondrial membranes, consistent with a physiological role in protection from reactive electrophilic intermediates and oxidative stress. In this review paper, we particularly focus on recent advances made in understanding MGST1 activation, induction, broad subcellular distribution, and the role of MGST1 in apoptosis, ferroptosis, cancer progression, and therapeutic responses.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States.
| | - Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Ralf Morgenstern
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet, Stockholm, Sweden
| | - Danyelle M Townsend
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
3
|
Li Y, Xu X, Wang X, Zhang C, Hu A, Li Y. MGST1 Expression Is Associated with Poor Prognosis, Enhancing the Wnt/β-Catenin Pathway via Regulating AKT and Inhibiting Ferroptosis in Gastric Cancer. ACS OMEGA 2023; 8:23683-23694. [PMID: 37426275 PMCID: PMC10323946 DOI: 10.1021/acsomega.3c01782] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND The role of microsomal glutathione S-transferase 1 (MGST1) underlying gastric cancer (GC) is unclear. The purpose of this research was to study the expression level and biological functions of MGST1 in GC cells. METHODS Expression of MGST1 was detected by RT-qPCR, Western blot (WB), and immunohistochemical staining. MGST1 was knockdown and overexpression by short hairpin RNA lentivirus in GC cells. Cell proliferation was evaluated by the CCK-8 assay and EDU assay. The cell cycle was detected by flow cytometry. The TOP-Flash reporter assay was used to examine the activity of T-cell factor/lymphoid enhancer factor transcription based on β-catenin. WB was performed to assess the protein levels involved in the cell signaling pathway and ferroptosis. The MAD assay and C11 BODIPY 581/591 lipid peroxidation probe assay were performed to determine the reactive oxygen species lipid level in GC cells. RESULTS MGST1 expression was upregulated in GC and it was correlated with poor overall survival of GC patients. MGST1 knockdown significantly inhibited GC cell proliferation and cell cycle by regulating the AKT/GSK-3β/β-catenin axis. In addition, we found that MGST1 inhibits ferroptosis in GC cells. CONCLUSION These findings suggested that MGST1 played a confirmed role in promoting GC development and serving as a possible independent prognostic factor for GC.
Collapse
Affiliation(s)
- Yaxian Li
- General
Surgery Department, The First Affiliated
Hospital of Anhui Medical University, Hefei 230000, China
| | - Xin Xu
- General
Surgery Department, The First Affiliated
Hospital of Anhui Medical University, Hefei 230000, China
| | - Xiaodong Wang
- General
Surgery Department, The First Affiliated
Hospital of Anhui Medical University, Hefei 230000, China
- The
Robert Bosch Center for Tumor Diseases (RBCT), Stuttgart 70376, Germany
| | - Chaoyang Zhang
- General
Surgery Department, The First Affiliated
Hospital of Anhui Medical University, Hefei 230000, China
| | - Asheng Hu
- General
Surgery Department, The First Affiliated
Hospital of Anhui Medical University, Hefei 230000, China
| | - Yongxiang Li
- General
Surgery Department, The First Affiliated
Hospital of Anhui Medical University, Hefei 230000, China
| |
Collapse
|
4
|
Cebula M, Morgenstern R. Enzymology of reactive intermediate protection: kinetic analysis and temperature dependence of the mesophilic membrane protein catalyst MGST1. FEBS J 2023. [PMID: 36808476 DOI: 10.1111/febs.16754] [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: 11/14/2022] [Revised: 01/25/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023]
Abstract
Glutathione transferases (GSTs) are a class of phase II detoxifying enzymes catalysing the conjugation of glutathione (GSH) to endogenous and exogenous electrophilic molecules, with microsomal glutathione transferase 1 (MGST1) being one of its key members. MGST1 forms a homotrimer displaying third-of-the-sites-reactivity and up to 30-fold activation through modification of its Cys-49 residue. It has been shown that the steady-state behaviour of the enzyme at 5 °C can be accounted for by its pre-steady-state behaviour if the presence of a natively activated subpopulation (~ 10%) is assumed. Low temperature was used as the ligand-free enzyme is unstable at higher temperatures. Here, we overcame enzyme lability through stop-flow limited turnover analysis, whereby kinetic parameters at 30 °C were obtained. The acquired data are more physiologically relevant and enable confirmation of the previously established enzyme mechanism (at 5 °C), yielding parameters relevant for in vivo modelling. Interestingly, the kinetic parameter defining toxicant metabolism, kcat /KM , is strongly dependent on substrate reactivity (Hammett value 4.2), underscoring that glutathione transferases function as efficient and responsive interception catalysts. The temperature behaviour of the enzyme was also analysed. Both the KM and KD values decreased with increasing temperature, while the chemical step k3 displayed modest temperature dependence (Q10 : 1.1-1.2), mirrored in that of the nonenzymatic reaction (Q10 : 1.1-1.7). Unusually high Q10 values for GSH thiolate anion formation (k2 : 3.9), kcat (2.7-5.6) and kcat /KM (3.4-5.9) support that large structural transitions govern GSH binding and deprotonation, which limits steady-state catalysis.
Collapse
Affiliation(s)
- Marcus Cebula
- Division of Biochemical Toxicology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ralf Morgenstern
- Division of Biochemical Toxicology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
5
|
Zhao J, Gao Y, He W, Wang W, Hu W, Sun Y. Synthesis, characterization and biological evaluation of two cyclometalated iridium(III) complexes containing a glutathione S-transferase inhibitor. J Inorg Biochem 2023; 238:112050. [PMID: 36332411 DOI: 10.1016/j.jinorgbio.2022.112050] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/12/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
The cyclometalated iridium(III) compounds have been intensively studied for health-related applications due to their outstanding luminescent properties and multiple anticancer modes of action. Herein, two iridium(III) compounds Ir-1 and Ir-3 containing glutathione S-transferase inhibitor (GSTi) were developed and studied together with two unfunctionalized compounds Ir-2 and Ir-4 as a comparison. Biological study indicated that GSTi-bearing complexes Ir-1 and Ir-3 exert a synergistic effect on the inhibition of cancer cells. The photophysical properties of Ir-1 ∼ Ir-4 were investigated by UV/vis absorption and fluorescence spectroscopy and rationalized with TD-DFT calculations. As expected, GSTi-bearing complexes Ir-1 and Ir-3 exhibited considerable cytotoxicity against both A549 and cisplatin-resistant A549/cis cancer cells, much higher than the unfunctionalized iridium compounds Ir-2 and Ir-4. Further study indicated that Ir-1 and Ir-3 mainly localize in the mitochondria of tumor cells, and exert their cytotoxicity via generating ROS and inhibiting GST activity. The flow cytometry investigations demonstrated that Ir-1 and Ir-3 can arrest the cell cycle in S phase and induce the cell death through apoptosis process. Overall, the complexation of GST inhibitors with cyclometalated iridium(III) agents provides an effective way for potentiating the cytotoxicity of iridium(III) anticancer agents and resensitizing the efficacy against cisplatin resistant cancer cells.
Collapse
Affiliation(s)
- Jian Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China; Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Ya Gao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Weiyu He
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Wei Wang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Weiwei Hu
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Yanyan Sun
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
6
|
Kasparkova J, Kostrhunova H, Novohradsky V, Ma L, Zhu G, Milaeva ER, Shtill AA, Vinck R, Gasser G, Brabec V, Nazarov AA. Is antitumor Pt(IV) complex containing two axial lonidamine ligands a true dual- or multi-action prodrug? METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6618656. [PMID: 35759404 DOI: 10.1093/mtomcs/mfac048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/09/2022] [Indexed: 11/14/2022]
Abstract
This work studied the mechanism of action of a Pt(IV) complex 2 bearing two axial lonidamine ligands, which are selective inhibitors of aerobic glycolysis. The presence of two lonidamine ligands in 2 compared to the parent Pt(II) complex increased its antiproliferative activity, cellular accumulation, and changed its cell cycle profile and mechanism of cell death. In 3D cell culture, 2 showed exceptional antiproliferative activity with IC50 values as low as 1.6 μM in MCF7 cells. The study on the influence of the lonidamine ligands in the Pt complex on glycolysis showed only low potency of ligands to affect metabolic processes in cancer cells, making the investigated complex, not a dual- or multi-action prodrug. However, the Pt(IV) prodrug effectively delivers the cytotoxic Pt(II) complex into cancer cells.
Collapse
Affiliation(s)
- Jana Kasparkova
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Hana Kostrhunova
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Vojtech Novohradsky
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Lili Ma
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P. R. China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P. R. China
| | - Elena R Milaeva
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russian Federation
| | - Alexender A Shtill
- Blokhin Cancer Center, Russian Academy of Medical Sciences, 115478 Moscow, Russian Federation
| | - Robin Vinck
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, 75005 Paris, France
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, 75005 Paris, France
| | - Viktor Brabec
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Alexey A Nazarov
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russian Federation
| |
Collapse
|
7
|
Zhao L, Qu Y, Zhang F, Ma D, Gao H, Gan L, Zhang H, Zhang S, Fang J. Baylis–Hillman Adducts as a Versatile Module for Constructing Fluorogenic Release System. J Med Chem 2022; 65:6056-6069. [DOI: 10.1021/acs.jmedchem.1c01940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lanning Zhao
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yuan Qu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Fang Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Di Ma
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Hao Gao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Lu Gan
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Hong Zhang
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Shengxiang Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing 210094, China
| |
Collapse
|
8
|
Gobbo A, Pereira SAP, Biancalana L, Zacchini S, Saraiva MLMFS, Dyson PJ, Marchetti F. Anticancer ruthenium( ii) tris(pyrazolyl)methane complexes with bioactive co-ligands. Dalton Trans 2022; 51:17050-17063. [DOI: 10.1039/d2dt03009h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
New anticancer RuII-tpm complexes are presented, including a synthetic strategy to tether bioactive molecules to the metallic scaffold.
Collapse
Affiliation(s)
- Alberto Gobbo
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Sarah A. P. Pereira
- LAQV, REQUIMTE, Laboratório de Química Aplicada, Faculdade de Farmácia da Universidade do Porto, Portugal
| | - Lorenzo Biancalana
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Stefano Zacchini
- University of Bologna, Department of Industrial Chemistry “Toso Montanari”, Viale Risorgimento 4, I-40136 Bologna, Italy
| | - M. Lúcia M. F. S. Saraiva
- LAQV, REQUIMTE, Laboratório de Química Aplicada, Faculdade de Farmácia da Universidade do Porto, Portugal
| | - Paul J. Dyson
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH 1015, Switzerland
| | - Fabio Marchetti
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124 Pisa, Italy
| |
Collapse
|
9
|
Zhu W, Yu H, Qian X, Lu K, Zhao C, Zhang Y, Wang HY, Liu Y. Near-infrared frequency upconversion probe for revealing the relationship between glutathione S-transferase and drug-resistance. Anal Chim Acta 2021; 1181:338920. [PMID: 34556207 DOI: 10.1016/j.aca.2021.338920] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022]
Abstract
Drug resistance poses an enormous challenge for successful chemotherapy. Glutathione S-transferase (GST) has been confirmed to be involved in the progression of drug resistance to some anticancer drugs, thus revealing that the role of GST in anticancer drug resistance is necessary. Herein, by taking advantage of frequency upconversion luminescence (FUCL) technology, we reported an FUCL probe (NRh-NDs) that can detect GST based on a rhodamine derivative structure decorated with a 2,4-dinitrobenzenesulfonyl group (NDs). The NRh-NDs showed excellent sensitivity and high selectivity for GST and released the emissive dye NRh-NH2, which showed emission and excitation wavelengths in vitro of 820 nm and 850 nm, respectively. The NRh-NDs probe successfully tested endogenic GST in U87, MCF-7 and A549 cells. The cell data showed that the increased levels of GST were positively related to cisplatin resistance but not to 5-fluorouracil resistance. These results suggested that the probe could be used as a visual tool to reveal the cause of drug resistance for cisplatin resistance in cancer treatment. Furthermore, it may serve as an effective tool to confirm the mechanism of antitumor drug resistance.
Collapse
Affiliation(s)
- Wenchao Zhu
- School of Mechanical Engineering, Southeast University, Nanjing, 211189, China; School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Hui Yu
- School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoli Qian
- School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Kai Lu
- Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Chao Zhao
- School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuanyuan Zhang
- School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Hai-Yan Wang
- School of Mechanical Engineering, Southeast University, Nanjing, 211189, China.
| | - Yi Liu
- School of Engineering, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
10
|
El Abbouchi A, El Brahmi N, Hiebel MA, Bignon J, Guillaumet G, Suzenet F, El Kazzouli S. Synthesis and evaluation of a novel class of ethacrynic acid derivatives containing triazoles as potent anticancer agents. Bioorg Chem 2021; 115:105293. [PMID: 34426162 DOI: 10.1016/j.bioorg.2021.105293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/03/2021] [Accepted: 08/17/2021] [Indexed: 12/30/2022]
Abstract
For unmet clinical needs, a novel class of ethacrynic acid (EA) derivatives containing triazole moieties (3a-i and 8) were designed, synthesized and evaluated as new anticancer agents. The in vitro anti-proliferative activities were assessed first on HL60 cell line and in a second stage, the two selected compounds 3a and 3c were tested on a panel of human cancer cell lines (A549, MCF7, PC3, U87-MG, SKOV3 and HCT116) and on a normal cell line (MCR5). Compound3c exhibited very good antitumor activities with IC50 values of 20.2, 56.5 and 76.8 nM against A549, PC3 and U87-MG cell lines respectively, which is 2.8- and 1.3-fold more active than doxorubicin on A549 and U87-MG cancer cells, respectively. In addition, compound 3c displays a very good safety index (SI) of 82 fold for A549. Compound 3a showed also good IC50 values of 50 nM on both A549 and PC3 cells and lower selectivity compared to 3c for A549 and PC3 vs. MCR5 with SI of 33 and 18 fold, respectively. The measurement of mitochondrial membrane potential on HCT116 cells after treatments by either 3a or 3c showed that both compounds induced mitochondrial dysfunctions causing thus caspase-induced apoptosis.
Collapse
Affiliation(s)
- Abdelmoula El Abbouchi
- Euromed Research Center, Engineering School of Biomedical and Biotechnology, Euromed University of Fes (UEMF)-Route de Meknès, 30000 Fez, Morocco; Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, BP 6759, Orléans cedex 2 54067, France
| | - Nabil El Brahmi
- Euromed Research Center, Engineering School of Biomedical and Biotechnology, Euromed University of Fes (UEMF)-Route de Meknès, 30000 Fez, Morocco
| | - Marie-Aude Hiebel
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, BP 6759, Orléans cedex 2 54067, France
| | - Jérôme Bignon
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Gérald Guillaumet
- Euromed Research Center, Engineering School of Biomedical and Biotechnology, Euromed University of Fes (UEMF)-Route de Meknès, 30000 Fez, Morocco; Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, BP 6759, Orléans cedex 2 54067, France.
| | - Franck Suzenet
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, BP 6759, Orléans cedex 2 54067, France.
| | - Saïd El Kazzouli
- Euromed Research Center, Engineering School of Biomedical and Biotechnology, Euromed University of Fes (UEMF)-Route de Meknès, 30000 Fez, Morocco.
| |
Collapse
|
11
|
Han T, Wu Y, Han W, Yan K, Zhao J, Sun Y. Antitumor Effect of Organometallic Half-Sandwich Ru(II)-Arene Complexes Bearing a Glutathione S-Transferase Inhibitor. Inorg Chem 2021; 60:13051-13061. [PMID: 34369147 DOI: 10.1021/acs.inorgchem.1c01482] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The facile modification of the ligands in organometallic Ru(II)-arene complexes offers more opportunities to optimize their pharmacological profiles. Herein, three Ru(II)-arene complexes containing a glutathione S-transferase (GST) inhibitor (NBDHEX) in chelate ligand have been designed and synthesized in this study. In vitro results indicated that the ligation with NBDHEX significantly increased the activities and selectivities of the organometallic Ru(II)-arene complexes against tumor cells, especially complex 3, which was the most active compound among the tested compounds. DFT calculations and hydrolysis results demonstrated that complex 3 with more alkyl groups in the arene ligand has increased electron density at the Ru(II) center as compared with complexes 1 and 2, thus resulting in the improved hydrolysis rate, which may be responsible for its higher anticancer activity. Further studies showed that complexes 1-3 can cause the loss of the mitochondrial membrane potential and upregulate the expression of Bcl-2 and Bax in A549 cells, suggesting that complexes 1-3-induced cell death may be mediated via the mitochondrial apoptotic pathway. Thus, these findings suggested that simultaneous modification of the chelate ligands and arene rings in the organometallic Ru(II)-arene complexes is an effective way to improve their pharmacological properties.
Collapse
Affiliation(s)
- Tianyu Han
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yuying Wu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Weinan Han
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Kaiwen Yan
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Jian Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yanyan Sun
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| |
Collapse
|
12
|
Steel TR, Walsh F, Wieczorek-Błauż A, Hanif M, Hartinger CG. Monodentately-coordinated bioactive moieties in multimodal half-sandwich organoruthenium anticancer agents. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213890] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Wang ZX, Chu HW, Yang KG, Zhao BF, Liang Z, Zhang LH, Zhang YK. Label-Free Quantitative Proteomics Analysis of the Sorafenib Resistance in HepG2 Cells. JOURNAL OF ANALYSIS AND TESTING 2021. [DOI: 10.1007/s41664-021-00176-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
14
|
Mutational Analysis of the Binding of Alternative Substrates and Inhibitors to the Active Site of Human Glutathione Transferase P1–1. Processes (Basel) 2020. [DOI: 10.3390/pr8101232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Glutathione transferases (GSTs) are enzymes that play a critical role in cellular detoxication by catalyzing the nucleophilic attack of glutathione on the electrophilic center of a number of xenobiotic compounds, including many therapeutic drugs. Mutations of amino acid residues in the glutathione-binding site of human glutathione transferase P1–1, namely W39C, K45A, Q52A, Q52K, and Q52E, have been engineered. The recombinant mutant proteins were expressed in Escherichia coli, but only mutants K45A, Q52A, and Q52K showed measurable activity. Steady-state kinetics comparing glutathione with the alternative thiol substrate γ-glutamylcysteine demonstrated the importance of the glycine residue in glutathione for high catalytic efficiency. Inhibition experiments with a set of glutathione analogs structurally related to the therapeutic drugs Telintra and Telcyta enabled determination of binding energies that were contributed by different substituents. The effects of substituting amino acid side chains in the glutathione-binding site of the enzyme on binding the glutathione derivatives and catalysis were evaluated.
Collapse
|
15
|
Nabiyeva T, Marschner C, Blom B. Synthesis, structure and anti-cancer activity of osmium complexes bearing π-bound arene substituents and phosphane Co-Ligands: A review. Eur J Med Chem 2020; 201:112483. [PMID: 32592914 DOI: 10.1016/j.ejmech.2020.112483] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/17/2020] [Accepted: 05/17/2020] [Indexed: 12/24/2022]
Abstract
While many examples of osmium complexes, as anti-cancer agents, have been reported and some reviews have been devoted to this topic, a particularly interesting and synthetically accessible sub-class of these compounds namely those bearing a π- bound arene and phosphane co-ligand have escaped review. These complexes have made a surprisingly late entry in the literature (2005) in terms of anti-cancer investigations. This is somewhat surprising considering the plethora of analogous complexes that have been reported for the lighter analogue, ruthenium. Herein we review all complexes, neutral and ionic, bearing the "(ƞ6-arene)Os(PR3)" moiety focusing on their synthesis, reactivity, structural features (by X-ray diffraction analysis) as well as anti-cancer biological activity. An attempt is made throughout the article to contrast these to each other and to analogous Ru systems, and a full summary of all existing in vitro biological data is presented.
Collapse
Affiliation(s)
- Tomiris Nabiyeva
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Kapoenstraat 2, PO Box 616, 6200, MD, Maastricht, the Netherlands
| | - Christoph Marschner
- Institut für Anorganische Chemie, Technische Universität Graz, Stremayrgasse 9, A-8010, Graz, Austria
| | - Burgert Blom
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Kapoenstraat 2, PO Box 616, 6200, MD, Maastricht, the Netherlands.
| |
Collapse
|
16
|
Chen H, Chen F, Wang X, Gou S. Multifunctional Pt(iv) complexes containing a glutathione S-transferase inhibitor lead to enhancing anticancer activity and preventing metastasis of osteosarcoma cells. Metallomics 2020; 11:317-326. [PMID: 30560252 DOI: 10.1039/c8mt00296g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cisplatin has been clinically applied in the treatment of osteosarcoma (OS), but its efficacy is severely limited due to drug resistance and metastasis. One of the chief culprits is the overexpression of glutathione S-transferases (GSTs) in cancer cells, which can accelerate the interaction of glutathione (GSH) with cisplatin, reducing its biological effects. In this study, three Pt(iv) complexes derived from cisplatin conjugated with a GST inhibitor (NBDHEX) were designed and synthesized. The stabilities and releasing capabilities of these complexes, as well as their abilities to inhibit GSTs, were investigated together with their in vitro anticancer activities toward osteosarcoma cells. Among them, complex 2, bearing one NBDHEX derivative and a hydroxyl group at the axial positions, could markedly kill human OS cells due to its suitable stability and prominent ability to inhibit GSTs. Meanwhile, it can prevent the metastasis of OS via down-regulating Akt. Thus, complex 2 has the potential for further research for the treatment of OS.
Collapse
Affiliation(s)
- Hong Chen
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, Southeast University, Nanjing 211189, China.
| | | | | | | |
Collapse
|
17
|
Narayanankutty A, Job JT, Narayanankutty V. Glutathione, an Antioxidant Tripeptide: Dual Roles in Carcinogenesis and Chemoprevention. Curr Protein Pept Sci 2020; 20:907-917. [PMID: 30727890 DOI: 10.2174/1389203720666190206130003] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/14/2019] [Accepted: 01/25/2019] [Indexed: 12/12/2022]
Abstract
Glutathione (GSH or reduced glutathione) is a tripeptide of gamma-Glutamyl-cysteinylglycine and the predominant intracellular antioxidant in many organisms including humans. GSH and associated enzymes are controlled by a transcription factor-nuclear factor-2 related erythroid factor-2 (Nrf2). In cellular milieu, GSH protects the cells essentially against a wide variety of free radicals including reactive oxygen species, lipid hydroperoxides, xenobiotic toxicants, and heavy metals. It has two forms, the reduced form or reduced glutathione (GSH) and oxidized form (GSSG), where two GSH moieties combine by sulfhydryl bonds. Glutathione peroxidase (GPx) and glutathione-s-transferase (GST) essentially perform the detoxification reactions using GSH, converting it into GSSG. Glutathione reductase (GR) operates the salvage pathway by converting GSSG to GSH with the expense of NADPH and restores the cellular GSH pool. Hence, GSH and GSH-dependent enzymes are necessary for maintaining the normal redox balance in the body and help in cell survival under stress conditions. In addition, GST removes various carcinogenic compounds offering a chemopreventive property, whereas the GSH system plays a significant role in regulating the cellular survival by offering redox stability in a variety of cancers including prostate, lung, breast, and colon cancer. Studies have also indicated that GSH inhibitors, such as buthionine sulfoximine, improve the chemo-sensitivity in cancer cells. In addition, GSH and dependent enzymes provide a survival advantage for cancer cells against chemotherapeutic drugs and radiotherapy.
Collapse
Affiliation(s)
- Arunaksharan Narayanankutty
- Postgraduate & Research Department of Zoology, St. Joseph's College (Autonomous), Devagiri (Affiliated to University of Calicut), Calicut- 673 019, Kerala, India
| | - Joice Tom Job
- Postgraduate & Research Department of Zoology, St. Joseph's College (Autonomous), Devagiri (Affiliated to University of Calicut), Calicut- 673 019, Kerala, India
| | | |
Collapse
|
18
|
Abstract
The mercapturic acid pathway is a major route for the biotransformation of xenobiotic and endobiotic electrophilic compounds and their metabolites. Mercapturic acids (N-acetyl-l-cysteine S-conjugates) are formed by the sequential action of the glutathione transferases, γ-glutamyltransferases, dipeptidases, and cysteine S-conjugate N-acetyltransferase to yield glutathione S-conjugates, l-cysteinylglycine S-conjugates, l-cysteine S-conjugates, and mercapturic acids; these metabolites constitute a "mercapturomic" profile. Aminoacylases catalyze the hydrolysis of mercapturic acids to form cysteine S-conjugates. Several renal transport systems facilitate the urinary elimination of mercapturic acids; urinary mercapturic acids may serve as biomarkers for exposure to chemicals. Although mercapturic acid formation and elimination is a detoxication reaction, l-cysteine S-conjugates may undergo bioactivation by cysteine S-conjugate β-lyase. Moreover, some l-cysteine S-conjugates, particularly l-cysteinyl-leukotrienes, exert significant pathophysiological effects. Finally, some enzymes of the mercapturic acid pathway are described as the so-called "moonlighting proteins," catalytic proteins that exert multiple biochemical or biophysical functions apart from catalysis.
Collapse
Affiliation(s)
- Patrick E Hanna
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
19
|
Zeng B, Ge C, Li R, Zhang Z, Fu Q, Li Z, Lin Z, Liu L, Xue Y, Xu Y, He J, Guo H, Li C, Huang W, Song X, Huang Y. Knockdown of microsomal glutathione S-transferase 1 inhibits lung adenocarcinoma cell proliferation and induces apoptosis. Biomed Pharmacother 2020; 121:109562. [DOI: 10.1016/j.biopha.2019.109562] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/04/2019] [Accepted: 10/20/2019] [Indexed: 12/12/2022] Open
|
20
|
Glutathione Transferase P1-1 an Enzyme Useful in Biomedicine and as Biomarker in Clinical Practice and in Environmental Pollution. Nutrients 2019; 11:nu11081741. [PMID: 31357662 PMCID: PMC6723968 DOI: 10.3390/nu11081741] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 12/19/2022] Open
Abstract
Glutathione transferase P1-1 (GSTP1-1) is expressed in some human tissues and is abundant in mammalian erythrocytes (here termed e-GST). This enzyme is able to detoxify the cell from endogenous and exogenous toxic compounds by using glutathione (GSH) or by acting as a ligandin. This review collects studies that propose GSTP1-1 as a useful biomarker in different fields of application. The most relevant studies are focused on GSTP1-1 as a biosensor to detect blood toxicity in patients affected by kidney diseases. In fact, this detoxifying enzyme is over-expressed in erythrocytes when unusual amounts of toxins are present in the body. Here we review articles concerning the level of GST in chronic kidney disease patients, in maintenance hemodialysis patients and to assess dialysis adequacy. GST is also over-expressed in autoimmune disease like scleroderma, and in kidney transplant patients and it may be used to check the efficiency of transplanted kidneys. The involvement of GSTP in the oxidative stress and in other human pathologies like cancer, liver and neurodegenerative diseases, and psychiatric disorders is also reported. Promising applications of e-GST discussed in the present review are its use for monitoring human subjects living in polluted areas and mammals for veterinary purpose.
Collapse
|
21
|
Gibson D. Multi-action Pt(IV) anticancer agents; do we understand how they work? J Inorg Biochem 2018; 191:77-84. [PMID: 30471522 DOI: 10.1016/j.jinorgbio.2018.11.008] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022]
Abstract
Pt(IV) complexes act as prodrugs that are activated inside cancer cells releasing cytotoxic Pt(II) drugs such as cisplatin as well as two axial ligands. These ligands can be used to confer favorable pharmacological properties to the prodrug. They can be innocent spectators, targeting agents or bioactive moieties. When the ligands are bioactive moieties such as enzyme inhibitors or antiproliferative agents, the prodrug attacks several cellular targets at the same time acting as a multi-action prodrug. These compounds are very potent and often overcome resistance to cisplatin. Despite solid rationalization and careful design, often there is no correlation between the ability of the bioactive ligand to inhibit the target enzyme and the cytotoxicity. This might be because most bioactive ligands affect several cellular functions and not only the ones they were designed to inhibit. Thus, even "dual action" prodrugs might in reality be multi-action prodrugs. This class of multi-action Pt(IV) prodrugs seems to have great potential in the attempts to overcome resistance.
Collapse
Affiliation(s)
- Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University, Jerusalem, 91120, Israel.
| |
Collapse
|
22
|
Whang CH, Yoo E, Hur SK, Kim KS, Kim D, Jo S. A highly GSH-sensitive SN-38 prodrug with an "OFF-to-ON" fluorescence switch as a bifunctional anticancer agent. Chem Commun (Camb) 2018; 54:9031-9034. [PMID: 30047958 DOI: 10.1039/c8cc05010d] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
SN-38 (7-ethyl-10-hydroxy-camptothecin) is an active metabolite of irinotecan (CPT-11) and the most potent camptothecin analogue. In this study, 2,4-dinitrobenzene sulfonyl (DNS) was covalently conjugated as a GSH-sensitive trigger to 10'-OH of SN-38 to yield a GSH-sensitive prodrug, denoted as DNS-SN38, with virtually quenched fluorescence due to donor-excited photo-induced electron transfer (d-PeT). By investigating DNS-SN38's activation properties upon fluorescence restoration and cytotoxic potency against ovarian cancer cell lines (A2780 and m-Cherry + OCSC1-F2), its potential applicability as a useful chemotherapeutic agent was demonstrated.
Collapse
Affiliation(s)
- Chang-Hee Whang
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38655, USA.
| | - Eunsoo Yoo
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Seong Kwon Hur
- Department of Molecular & Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Kyeong Soo Kim
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38655, USA.
| | - Dongin Kim
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Seongbong Jo
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38655, USA.
| |
Collapse
|
23
|
Allocati N, Masulli M, Di Ilio C, Federici L. Glutathione transferases: substrates, inihibitors and pro-drugs in cancer and neurodegenerative diseases. Oncogenesis 2018; 7:8. [PMID: 29362397 PMCID: PMC5833873 DOI: 10.1038/s41389-017-0025-3] [Citation(s) in RCA: 349] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/12/2017] [Indexed: 12/12/2022] Open
Abstract
Glutathione transferase classical GSH conjugation activity plays a critical role in cellular detoxification against xenobiotics and noxious compounds as well as against oxidative stress. However, this feature is also exploited by cancer cells to acquire drug resistance and improve their survival. As a result, various members of the family were found overexpressed in a number of different cancers. Moreover several GST polymorphisms, ranging from null phenotypes to point mutations, were detected in members of the family and found to correlate with the onset of neuro-degenerative diseases. In the last decades, a great deal of research aimed at clarifying the role played by GSTs in drug resistance, at developing inhibitors to counteract this activity but also at exploiting GSTs for prodrugs specific activation in cancer cells. Here we summarize some of the most important achievements reached in this lively area of research.
Collapse
Affiliation(s)
- Nerino Allocati
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy.
| | - Michele Masulli
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Carmine Di Ilio
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Luca Federici
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy.,CESI-MET, University "G. d'Annunzio", Chieti, Italy
| |
Collapse
|
24
|
Abstract
In this manuscript we focus on Pt(iv) anticancer prodrugs. We explore the main working hypotheses for the design of effective Pt(iv) prodrugs and note the exceptions to the common assumptions that are prevalent in the field. Special attention was devoted to the emerging class of "dual action" Pt(iv) prodrugs, where bioactive ligands are conjugated to the axial positions of platinum in order to obtain orthogonal or complementary effects that will increase the efficacy of killing the cancer cells. We discuss the rationale behind the design of the "dual action" prodrugs and the results of the pharmacological studies obtained. Simultaneous release of two bioactive moieties inside the cancer cells often triggers several processes that together determine the fate of the cell. Pt(iv) complexes provide many opportunities for applying new concepts in targeting, synergistic cell killing and exploiting novel nanodelivery systems.
Collapse
Affiliation(s)
- Dan Gibson
- Institute of Drug Research, School of Pharmacy, The Hebrew University, Jerusalem, Israel.
| |
Collapse
|
25
|
Abstract
Probes that detect specific biological materials are indispensable tools for deepening our understanding of various cellular phenomena. In live cell imaging, the probe must emit fluorescence only when a specific substance is detected. In this paper, we introduce a new probe we developed for live cell imaging. Glutathione S-transferase (GST) activity is higher in tumor cells than in normal cells and is involved in the development of resistance to various anticancer drugs. We previously reported the development of a general strategy for the synthesis of probes for detection of GST enzymes, including fluorogenic, bioluminogenic, and 19F-NMR probes. Arylsulfonyl groups were used as caging groups during probe design. The fluorogenic probes were successfully used to quantitate very low levels of GST activity in cell extracts and were also successfully applied to the imaging of microsomal MGST1 activity in living cells. The bioluminogenic and 19F-NMR probes were able to detect GST activity in Escherichia coli cells. Oligonucleotide-templated reactions are powerful tools for nucleic acid sensing. This strategy exploits the target strand as a template for two functionalized probes and provides a simple molecular mechanism for multiple turnover reactions. We developed a nucleophilic aromatic substitution reaction-triggered fluorescent probe. The probe completed its reaction within 30 s of initiation and amplified the fluorescence signal from 0.5 pM target oligonucleotide by 1500 fold under isothermal conditions. Additionally, we applied the oligonucleotide-templated reaction for molecular releasing and peptide detection.
Collapse
|
26
|
Yang L, Du C, Wu L, Yu J, An X, Yu W, Cao S, Li H, Ren X. Cytokine-Induced Killer Cells Modulates Resistance to Cisplatin in the A549/DDP Cell Line. J Cancer 2017; 8:3287-3295. [PMID: 29158802 PMCID: PMC5665046 DOI: 10.7150/jca.19426] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 08/30/2017] [Indexed: 12/21/2022] Open
Abstract
Background Cytokine-induced killer (CIK) cells can potentially enhance the tumor-killing activity of chemotherapy. Objective This study aimed to evaluate the effects of CIK cells on cisplatin (DDP) resistance in the human lung adenocarcinoma cell line A549/DDP. Methods The detect resistance index, drug resistance related-genes and cytokine secretion of A549/DDP co-cultured with CIK cells were assayed in vitro. ResultsAfter A549/DDP co-culture with CIK cells, the DDP resistance of A549/DDP significantly decreased in a time-dependent manner. The DDP resistance of A549/DDP co-cultured with CIK cells for 20 h decreased 4.93-fold compared with that of A549/DDP cells cultured alone (P<0.05). The mRNA and protein expression levels of the glutathione-S-transferase (GST) -π gene in A549/DDP significantly decreased after co-culture with CIK cells (P<0.05). The secretion of interferon (IFN)- γ significantly increased along with the co-culture time of A549/DDP with CIK cells. The expression of GST-π was restored by adding the neutralizing IFN-γ. ConclusionCIK cells can reverse the drug resistance of A549/DDP in a time-dependent manner by reducing GST-π expression to increase the accumulation of DDP. The effect of CIK cells on re-sensitizing lung cancer cells to the chemotherapy drug was partially dependent on the secretion of IFN-γ.
Collapse
Affiliation(s)
- Lili Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chunjuan Du
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei Wu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jinpu Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiumei An
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wenwen Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Shui Cao
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hui Li
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
27
|
Păunescu E, Soudani M, Martin P, Scopelliti R, Lo Bello M, Dyson PJ. Organometallic Glutathione S-Transferase Inhibitors. Organometallics 2017. [DOI: 10.1021/acs.organomet.7b00468] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Emilia Păunescu
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Mylène Soudani
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Paloma Martin
- Department
of Biology, University of Rome “Tor Vergata”, Via
della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Rosario Scopelliti
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Mario Lo Bello
- Department
of Biology, University of Rome “Tor Vergata”, Via
della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Paul J. Dyson
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
28
|
Dead-end complex, lipid interactions and catalytic mechanism of microsomal glutathione transferase 1, an electron crystallography and mutagenesis investigation. Sci Rep 2017; 7:7897. [PMID: 28801553 PMCID: PMC5554250 DOI: 10.1038/s41598-017-07912-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/04/2017] [Indexed: 01/01/2023] Open
Abstract
Microsomal glutathione transferase 1 (MGST1) is a detoxification enzyme belonging to the Membrane Associated Proteins in Eicosanoid and Glutathione Metabolism (MAPEG) superfamily. Here we have used electron crystallography of two-dimensional crystals in order to determine an atomic model of rat MGST1 in a lipid environment. The model comprises 123 of the 155 amino acid residues, two structured phospholipid molecules, two aliphatic chains and one glutathione (GSH) molecule. The functional unit is a homotrimer centered on the crystallographic three-fold axes of the unit cell. The GSH substrate binds in an extended conformation at the interface between two subunits of the trimer supported by new in vitro mutagenesis data. Mutation of Arginine 130 to alanine resulted in complete loss of activity consistent with a role for Arginine 130 in stabilizing the strongly nucleophilic GSH thiolate required for catalysis. Based on the new model and an electron diffraction data set from crystals soaked with trinitrobenzene, that forms a dead-end Meisenheimer complex with GSH, a difference map was calculated. The map reveals side chain movements opening a cavity that defines the second substrate site.
Collapse
|
29
|
Biancalana L, Batchelor LK, De Palo A, Zacchini S, Pampaloni G, Dyson PJ, Marchetti F. A general strategy to add diversity to ruthenium arene complexes with bioactive organic compounds via a coordinated (4-hydroxyphenyl)diphenylphosphine ligand. Dalton Trans 2017. [PMID: 28627572 DOI: 10.1039/c7dt02062g] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Esterification of (4-hydroxyphenyl)diphenylphosphine, coordinated to the [Ru(η6-p-cymene)Cl2] fragment, allows a series of bioactive carboxylic acids to be introduced directly into the organometallic molecule. Evaluation of the compounds on human ovarian cancer cells reveals synergistic enhancements in their antiproliferative activity relative to their bioactive organic and organometallic precursors.
Collapse
Affiliation(s)
- Lorenzo Biancalana
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via Moruzzi 13, I-56124 Pisa, Italy.
| | - Lucinda K Batchelor
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Alice De Palo
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via Moruzzi 13, I-56124 Pisa, Italy.
| | - Stefano Zacchini
- Dipartimento di Chimica Industriale "Toso Montanari", Università di Bologna, Viale Risorgimento 4, I-40136 Bologna, Italy
| | - Guido Pampaloni
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via Moruzzi 13, I-56124 Pisa, Italy.
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Fabio Marchetti
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via Moruzzi 13, I-56124 Pisa, Italy.
| |
Collapse
|
30
|
Georgakis ND, Karagiannopoulos DA, Thireou TN, Eliopoulos EE, Labrou NE, Tsoungas PG, Koutsilieris MN, Clonis YD. Concluding the trilogy: The interaction of 2,2'-dihydroxy-benzophenones and their carbonyl N-analogues with human glutathione transferase M1-1 face to face with the P1-1 and A1-1 isoenzymes involved in MDR. Chem Biol Drug Des 2017; 90:900-908. [PMID: 28440951 DOI: 10.1111/cbdd.13011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/27/2017] [Accepted: 04/14/2017] [Indexed: 02/04/2023]
Abstract
A series of 2,2'-dihydroxybenzophenones and their carbonyl N-analogues were studied as potential inhibitors against human glutathione transferase M1-1 (hGSTM1-1) purified from recombinant E. coli. Their screening revealed an inhibition against hGSTM1-1 within a range of 0-42% (25 μM). The IC50 values for the two stronger ones, 16 and 13, were 53.5 ± 5.6 μΜ and 28.5 ± 2.5 μΜ, respectively. The results were compared with earlier ones for isoenzymes hGSTP1-1 and hGSTA1-1 involved in MDR. All but one bind more strongly to A1-1, than M1-1 and P1-1, the latter being a poor binder. An order of potency A1-1 > > M1-1 > P1-1 meritted 13, 14 and 16 as the most potent inhibitors with hGSTM1-1. Enzyme kinetics with hGSTM1-1 (Km(CDNB) 213 ± 10 μΜ and Km(GSH) 303 ± 11 μΜ) revealed a competitive modality for 16 (Ki(16) = 22.3 ± 1.1 μΜ) and a mixed one for 13 versus CDNB (Ki(13) = 33.3 ± 1.6 μM for the free enzyme and Ki(13) ' = 17.7 ± 1.7 μM for the enzyme-CDNB complex). 5- or 5'-Bromo- or phenyl-substituted (but not in combination) inhibitors, having a H-bonded oxime weakly acidic group of a small volume, are optimal candidates for binding hGSTM1-1. The outcome of the isoenzyme trilogy identified good binder leads for the investigated GSTs involved in MDR.
Collapse
Affiliation(s)
- Nikolaos D Georgakis
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | | | - Trias N Thireou
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Elias E Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Nikolaos E Labrou
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Petros G Tsoungas
- Laboratory of Biochemistry, Hellenic Pasteur Institute, Athens, Greece
| | - Michael N Koutsilieris
- Department of Physiology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Yannis D Clonis
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| |
Collapse
|
31
|
Zompra A, Georgakis N, Pappa E, Thireou T, Eliopoulos E, Labrou N, Cordopatis P, Clonis Y. Glutathione analogues as substrates or inhibitors that discriminate between allozymes of the MDR-involved human glutathione transferase P1-1. Biopolymers 2017; 106:330-44. [PMID: 27037874 DOI: 10.1002/bip.22844] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/16/2016] [Accepted: 03/28/2016] [Indexed: 02/02/2023]
Abstract
Glutathione (GSH) structure-guided tripeptide analogues were designed and synthesized by solid phase technology, purified (≥95%) by RP and/or GF column chromatography, to identify those that, compared with GSH, exhibited similar or higher binding and catalytic efficiency toward the MDR-involved human GSTP1-1 isoenzyme, and could discriminate between the allozymic expression products of the polymorphic human GSTP1 gene locus, designated as hGSTP1*A (Ile(104) /Ala(113) ), hGSTP1*B (Val(104) /Ala(113) ), and hGSTP1*C (Val(104) /Val(113) ). The analogues bear single amino acid alterations as well as alterations in more than one position. Some analogues showed remarkable allozyme selectivity, binding catalytically to A (I, II, IV, XII), to C (V and XVI), to A and C (III, VII, XIV) or to all three allozymes (XV). A heterocyclic substituent at positions 1 or 2 of GSH favors inhibition of A, whereas a small hydrophobic/hydrophilic amide substituent at position 2 (Cys) favors inhibition of B and C. Heterocyclic substituents at position 1, only, produce catalytic analogues for A, whereas less bulky and more flexible hydrophobic/hydrophilic substituents, at positions 1 or 3, lead to effective substrates with C. When such substituents were introduced simultaneously at positions 1 and 3, the analogues produced have no catalytic potential but showed appreciable inhibitory effects, instead, with all allozymes. It is anticipated that when GSH analogues with selective inhibitory or catalytic binding, were conjugated to allozyme-selective inhibitors of hGSTP1-1, the derived leads would be useful for the designing of novel chimeric inhibitors against the MDR-involved hGSTP1-1 allozymes. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 106: 330-344, 2016.
Collapse
Affiliation(s)
- Aikaterini Zompra
- Laboratory Οf Pharmacognosy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Rio, Greece
| | - Nikolaos Georgakis
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Eleni Pappa
- Laboratory Οf Pharmacognosy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Rio, Greece
| | - Trias Thireou
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Elias Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Nikolaos Labrou
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Paul Cordopatis
- Laboratory Οf Pharmacognosy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Rio, Greece
| | - Yannis Clonis
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| |
Collapse
|
32
|
Biancalana L, Pratesi A, Chiellini F, Zacchini S, Funaioli T, Gabbiani C, Marchetti F. Ruthenium arene complexes with triphenylphosphane ligands: cytotoxicity towards pancreatic cancer cells, interaction with model proteins, and effect of ethacrynic acid substitution. NEW J CHEM 2017. [DOI: 10.1039/c7nj02300f] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The anticancer behaviour of Ru arene complexes can be tuned by an appropriate choice of the site and linkage of the bioactive group to the phosphane ligand.
Collapse
Affiliation(s)
- Lorenzo Biancalana
- Dipartimento di Chimica e Chimica Industriale
- Università di Pisa
- I-56124 Pisa
- Italy
| | - Alessandro Pratesi
- MetMed
- Dipartimento di Chimica “Ugo Schiff”
- Università di Firenze
- I-50019 Sesto Fiorentino
- Italy
| | - Federica Chiellini
- Dipartimento di Chimica e Chimica Industriale
- Università di Pisa
- I-56124 Pisa
- Italy
| | - Stefano Zacchini
- Dipartimento di Chimica Industriale “Toso Montanari”
- Università di Bologna
- I-40136 Bologna
- Italy
| | - Tiziana Funaioli
- Dipartimento di Chimica e Chimica Industriale
- Università di Pisa
- I-56124 Pisa
- Italy
| | - Chiara Gabbiani
- Dipartimento di Chimica e Chimica Industriale
- Università di Pisa
- I-56124 Pisa
- Italy
| | - Fabio Marchetti
- Dipartimento di Chimica e Chimica Industriale
- Università di Pisa
- I-56124 Pisa
- Italy
| |
Collapse
|
33
|
Sun YG, Sun YN, You LX, Liu YN, Ding F, Ren BY, Xiong G, Dragutan V, Dragutan I. Novel mononuclear Pt2+ and Pd2+ complexes containing (2,3-f)pyrazino(1,10)phenanthroline-2,3-dicarboxylic acid as a multi-donor ligand. Synthesis, structure, interaction with DNA, in vitro cytotoxicity, and apoptosis. J Inorg Biochem 2016; 164:129-140. [DOI: 10.1016/j.jinorgbio.2016.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 09/02/2016] [Accepted: 09/13/2016] [Indexed: 12/13/2022]
|
34
|
Mignani S, El Brahmi N, El Kazzouli S, Eloy L, Courilleau D, Caron J, Bousmina MM, Caminade AM, Cresteil T, Majoral JP. A novel class of ethacrynic acid derivatives as promising drug-like potent generation of anticancer agents with established mechanism of action. Eur J Med Chem 2016; 122:656-673. [PMID: 27448922 DOI: 10.1016/j.ejmech.2016.05.063] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/09/2016] [Accepted: 05/29/2016] [Indexed: 10/21/2022]
Abstract
The well-known diuretic Ethacrynic acid (EA, Edecrin), showing low anti-proliferative activities, was chemically modified at different positions. The new EA derivatives have been tested in vitro in anti-proliferative assays on both tumor KB (epidermal carcinoma) and leukemia HL60 (promyelocytic) cells suitable targets for anticancer activity. Reduction of the α-β double bond of EA completely abolished anti-cancer activities, whereas introduction of either 2-(4-substituted phenyl)ethanamine (series A) or 4-(4-substituted phenyl)piperazine (series B) moieties generated compounds showing moderate to strong anti-proliferative activities against human cancer cell lines. Several substitutions on the phenyl of these two moieties are tolerated. The mechanism of action of the EA derivatives prepared in this study is more complex than the inhibition of glutathione S-transferase π ascribed as unique effect to EA and might help to overcome tumor resistances.
Collapse
Affiliation(s)
- Serge Mignani
- Laboratoire de Chimie et de Biochimie pharmacologiques et toxicologique, CNRS UMR 860, Université Paris Descartes, PRES Sorbonne Paris Cité, 45, rue des Saints Pères, 75006, Paris, France.
| | - Nabil El Brahmi
- Euromed Research Institute, Engineering Division, Euro-Mediterranean University of Fes, Fès-Shore, Route de Sidi Hrazem, 30070, Fès, Morocco; Laboratoire de Chimie de Coordination CNRS, 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France; Université de Toulouse, UPS, INPT, F-31077, Toulouse Cedex4, France
| | - Saïd El Kazzouli
- Euromed Research Institute, Engineering Division, Euro-Mediterranean University of Fes, Fès-Shore, Route de Sidi Hrazem, 30070, Fès, Morocco.
| | - Laure Eloy
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Avenue de la terrasse, 91190, Gif sur Yvette, France
| | - Delphine Courilleau
- IPSIT Faculté de Pharmacie, Université Paris Sud, 92290, Chatenay-Malabry, France
| | - Joachim Caron
- Euromed Research Institute, Engineering Division, Euro-Mediterranean University of Fes, Fès-Shore, Route de Sidi Hrazem, 30070, Fès, Morocco; Laboratoire de Chimie de Coordination CNRS, 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France; Université de Toulouse, UPS, INPT, F-31077, Toulouse Cedex4, France
| | - Mosto M Bousmina
- Euromed Research Institute, Engineering Division, Euro-Mediterranean University of Fes, Fès-Shore, Route de Sidi Hrazem, 30070, Fès, Morocco
| | - Anne-Marie Caminade
- Laboratoire de Chimie de Coordination CNRS, 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France; Université de Toulouse, UPS, INPT, F-31077, Toulouse Cedex4, France
| | - Thierry Cresteil
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Avenue de la terrasse, 91190, Gif sur Yvette, France; IPSIT Faculté de Pharmacie, Université Paris Sud, 92290, Chatenay-Malabry, France.
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination CNRS, 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France; Université de Toulouse, UPS, INPT, F-31077, Toulouse Cedex4, France.
| |
Collapse
|
35
|
van Gisbergen MW, Cebula M, Zhang J, Ottosson-Wadlund A, Dubois L, Lambin P, Tew KD, Townsend DM, Haenen GRMM, Drittij-Reijnders MJ, Saneyoshi H, Araki M, Shishido Y, Ito Y, Arnér ESJ, Abe H, Morgenstern R, Johansson K. Chemical Reactivity Window Determines Prodrug Efficiency toward Glutathione Transferase Overexpressing Cancer Cells. Mol Pharm 2016; 13:2010-25. [PMID: 27093577 DOI: 10.1021/acs.molpharmaceut.6b00140] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glutathione transferases (GSTs) are often overexpressed in tumors and frequently correlated to bad prognosis and resistance against a number of different anticancer drugs. To selectively target these cells and to overcome this resistance we previously have developed prodrugs that are derivatives of existing anticancer drugs (e.g., doxorubicin) incorporating a sulfonamide moiety. When cleaved by GSTs, the prodrug releases the cytostatic moiety predominantly in GST overexpressing cells, thus sparing normal cells with moderate enzyme levels. By modifying the sulfonamide it is possible to control the rate of drug release and specifically target different GSTs. Here we show that the newly synthesized compounds, 4-acetyl-2-nitro-benzenesulfonyl etoposide (ANS-etoposide) and 4-acetyl-2-nitro-benzenesulfonyl doxorubicin (ANS-DOX), function as prodrugs for GSTA1 and MGST1 overexpressing cell lines. ANS-DOX, in particular, showed a desirable cytotoxic profile by inducing toxicity and DNA damage in a GST-dependent manner compared to control cells. Its moderate conversion of 500 nmol/min/mg, as catalyzed by GSTA1, seems hereby essential since the more reactive 2,4-dinitrobenzenesulfonyl doxorubicin (DNS-DOX) (14000 nmol/min/mg) did not display a preference for GSTA1 overexpressing cells. DNS-DOX, however, effectively killed GSTP1 (20 nmol/min/mg) and MGST1 (450 nmol/min/mg) overexpressing cells as did the less reactive 4-mononitrobenzenesulfonyl doxorubicin (MNS-DOX) in a MGST1-dependent manner (1.5 nmol/min/mg) as shown previously. Furthermore, we show that the mechanism of these prodrugs involves a reduction in GSH levels as well as inhibition of the redox regulatory enzyme thioredoxin reductase 1 (TrxR1) by virtue of their electrophilic sulfonamide moiety. TrxR1 is upregulated in many tumors and associated with resistance to chemotherapy and poor patient prognosis. Additionally, the prodrugs potentially acted as a general shuttle system for DOX, by overcoming resistance mechanisms in cells. Here we propose that GST-dependent prodrugs require a conversion rate "window" in order to selectively target GST overexpressing cells, while limiting their effects on normal cells. Prodrugs are furthermore a suitable system to specifically target GSTs and to overcome various drug resistance mechanisms that apply to the parental drug.
Collapse
Affiliation(s)
- Marike W van Gisbergen
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden.,Department of Radiation Oncology (MaastRO Lab), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Marcus Cebula
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden
| | - Jie Zhang
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden.,Departments of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina 29425, United States
| | - Astrid Ottosson-Wadlund
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden
| | - Ludwig Dubois
- Department of Radiation Oncology (MaastRO Lab), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MaastRO Lab), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Kenneth D Tew
- Departments of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina 29425, United States
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina , Charleston, South Carolina 29425, United States
| | - Guido R M M Haenen
- Department of Toxicology, NUTRIM-School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Marie-José Drittij-Reijnders
- Department of Toxicology, NUTRIM-School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Hisao Saneyoshi
- Nano Medical Engineering Laboratory, Discovery Research Institute, RIKEN 2-1 , Hirosawa, Wako-Shi, Saitama 351-0198, Japan
| | - Mika Araki
- Nano Medical Engineering Laboratory, Discovery Research Institute, RIKEN 2-1 , Hirosawa, Wako-Shi, Saitama 351-0198, Japan
| | - Yuko Shishido
- Department of Chemistry, Graduate School of Science, Nagoya University , Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, Discovery Research Institute, RIKEN 2-1 , Hirosawa, Wako-Shi, Saitama 351-0198, Japan
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , SE-171 77 Stockholm, Sweden
| | - Hiroshi Abe
- Nano Medical Engineering Laboratory, Discovery Research Institute, RIKEN 2-1 , Hirosawa, Wako-Shi, Saitama 351-0198, Japan.,Department of Chemistry, Graduate School of Science, Nagoya University , Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Ralf Morgenstern
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden
| | - Katarina Johansson
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , SE-171 77 Stockholm, Sweden
| |
Collapse
|
36
|
Agonigi G, Riedel T, Gay MP, Biancalana L, Oñate E, Dyson PJ, Pampaloni G, Păunescu E, Esteruelas MA, Marchetti F. Arene Osmium Complexes with Ethacrynic Acid-Modified Ligands: Synthesis, Characterization, and Evaluation of Intracellular Glutathione S-Transferase Inhibition and Antiproliferative Activity. Organometallics 2016. [DOI: 10.1021/acs.organomet.6b00197] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Gabriele Agonigi
- Dipartimento
di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi
13, I-56124 Pisa, Italy
| | - Tina Riedel
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - M. Pilar Gay
- Departamento
de Química Inorgánica, Instituto de Síntesis
Química y Catálisis Homogénea (ISQCH), Centro
de Innovación en Química Avanzada (ORFEO−CINQA), Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| | - Lorenzo Biancalana
- Dipartimento
di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi
13, I-56124 Pisa, Italy
| | - Enrique Oñate
- Departamento
de Química Inorgánica, Instituto de Síntesis
Química y Catálisis Homogénea (ISQCH), Centro
de Innovación en Química Avanzada (ORFEO−CINQA), Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| | - Paul J. Dyson
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Guido Pampaloni
- Dipartimento
di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi
13, I-56124 Pisa, Italy
| | - Emilia Păunescu
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Miguel A. Esteruelas
- Departamento
de Química Inorgánica, Instituto de Síntesis
Química y Catálisis Homogénea (ISQCH), Centro
de Innovación en Química Avanzada (ORFEO−CINQA), Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| | - Fabio Marchetti
- Dipartimento
di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi
13, I-56124 Pisa, Italy
| |
Collapse
|
37
|
Agonigi G, Riedel T, Zacchini S, Păunescu E, Pampaloni G, Bartalucci N, Dyson PJ, Marchetti F. Synthesis and Antiproliferative Activity of New Ruthenium Complexes with Ethacrynic-Acid-Modified Pyridine and Triphenylphosphine Ligands. Inorg Chem 2015; 54:6504-12. [DOI: 10.1021/acs.inorgchem.5b00802] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Gabriele Agonigi
- Dipartimento
di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi
13, I-56124 Pisa, Italy
| | - Tina Riedel
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Stefano Zacchini
- Dipartimento
di Chimica Industriale “Toso Montanari”, Università di Bologna, Viale Risorgimento 4, I-40136 Bologna, Italy
| | - Emilia Păunescu
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Guido Pampaloni
- Dipartimento
di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi
13, I-56124 Pisa, Italy
| | - Niccolò Bartalucci
- Dipartimento
di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi
13, I-56124 Pisa, Italy
| | - Paul J. Dyson
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Fabio Marchetti
- Dipartimento
di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi
13, I-56124 Pisa, Italy
| |
Collapse
|
38
|
Thiazolides promote apoptosis in colorectal tumor cells via MAP kinase-induced Bim and Puma activation. Cell Death Dis 2015; 6:e1778. [PMID: 26043078 PMCID: PMC4669824 DOI: 10.1038/cddis.2015.137] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/30/2015] [Accepted: 04/01/2015] [Indexed: 12/21/2022]
Abstract
While many anticancer therapies aim to target the death of tumor cells, sophisticated resistance mechanisms in the tumor cells prevent cell death induction. In particular enzymes of the glutathion-S-transferase (GST) family represent a well-known detoxification mechanism, which limit the effect of chemotherapeutic drugs in tumor cells. Specifically, GST of the class P1 (GSTP1-1) is overexpressed in colorectal tumor cells and renders them resistant to various drugs. Thus, GSTP1-1 has become an important therapeutic target. We have recently shown that thiazolides, a novel class of anti-infectious drugs, induce apoptosis in colorectal tumor cells in a GSTP1-1-dependent manner, thereby bypassing this GSTP1-1-mediated drug resistance. In this study we investigated in detail the underlying mechanism of thiazolide-induced apoptosis induction in colorectal tumor cells. Thiazolides induce the activation of p38 and Jun kinase, which is required for thiazolide-induced cell death. Activation of these MAP kinases results in increased expression of the pro-apoptotic Bcl-2 homologs Bim and Puma, which inducibly bind and sequester Mcl-1 and Bcl-xL leading to the induction of the mitochondrial apoptosis pathway. Of interest, while an increase in intracellular glutathione levels resulted in increased resistance to cisplatin, it sensitized colorectal tumor cells to thiazolide-induced apoptosis by promoting increased Jun kinase activation and Bim induction. Thus, thiazolides may represent an interesting novel class of anti-tumor agents by specifically targeting tumor resistance mechanisms, such as GSTP1-1.
Collapse
|
39
|
Pouliou FM, Thireou TN, Eliopoulos EE, Tsoungas PG, Labrou NE, Clonis YD. Isoenzyme- and allozyme-specific inhibitors: 2,2'-dihydroxybenzophenones and their carbonyl N-analogues that discriminate between human glutathione transferase A1-1 and P1-1 allozymes. Chem Biol Drug Des 2015; 86:1055-63. [PMID: 25891019 DOI: 10.1111/cbdd.12574] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/01/2015] [Accepted: 04/03/2015] [Indexed: 11/28/2022]
Abstract
The selectivity of certain benzophenones and their carbonyl N-analogues was investigated towards the human GSTP1-1 allozymes A, B and C involved in MDR. The allozymes were purified from extracts derived from E. coli harbouring the plasmids pEXP5-CT/TOPO-TA-hGSTP1*A, pOXO4-hGSTP1*B or pOXO4-hGSTP1*C. Compound screening with each allozyme activity indicated three compounds with appreciable inhibitory potencies, 12 and 13 with P1-1A 62% and 67%, 11 and 12 with P1-1C 51% and 70%, whereas that of 15 fell behind with P1-1B (41%). These findings were confirmed by IC50 values (74-125 μm). Enzyme inhibition kinetics, aided by molecular modelling and docking, revealed that there is competition with the substrate CDNB for the same binding site on the allozyme (Ki(13/A) = 63.6 ± 3.0 μm, Ki(15/B) = 198.6 ± 14.3 μm, and Ki(11/C) = 16.5 ± 2.7 μm). These data were brought into context by an in silico structural comparative analysis of the targeted proteins. Although the screened compounds showed moderate inhibitory potency against hGSTP1-1, remarkably, some of them demonstrated absolute isoenzyme and/or allozyme selectivity.
Collapse
Affiliation(s)
- Foteini M Pouliou
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Trias N Thireou
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Elias E Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Petros G Tsoungas
- Laboratory of Biochemistry, Hellenic Pasteur Institute, Athens, Greece
| | - Nikolaos E Labrou
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Yannis D Clonis
- Laboratory of Enzyme Technology, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| |
Collapse
|
40
|
de Oliveira DM, de Farias MT, Teles ALB, Dos Santos Junior MC, de Cerqueira MD, Lima RMF, El-Bachá RS. 8-Methoxypsoralen is a competitive inhibitor of glutathione S-transferase P1-1. Front Cell Neurosci 2014; 8:308. [PMID: 25324722 PMCID: PMC4179718 DOI: 10.3389/fncel.2014.00308] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/12/2014] [Indexed: 11/23/2022] Open
Abstract
The blood-brain barrier (BBB) is known to protect healthy brain cells from potentially dangerous chemical agents, but there are many evidences supporting the idea that this protective action is extended to tumor cells. Since the process of angiogenesis in brain tumors leads to BBB breakdown, biochemical characteristics of the BBB seem to be more relevant than physical barriers to protect tumor cells from chemotherapy. In fact, a number of resistance related factors were already demonstrated to be component of both BBB and tumor cells. The enzyme glutathione S-transferases (GST) detoxify electrophilic xenobiotics and endogenous secondary metabolites formed during oxidative stress. A role has been attributed to GST in the resistance of cancer cells to chemotherapeutic agents. This study characterized 8-methoxypsoralen (8-MOP) as a human GST P1-1 (hGST P1-1) inhibitor. To identify and characterize the potential inhibitory activity of 8-MOP, we studied the enzyme kinetics of the conjugation of 1-chloro-2,4-dinitrobenzene (CDNB) with GSH catalyzed by hGST P1-1. We report here that 8-MOP competitively inhibited hGST P1-1 relative to CDNB, but there was an uncompetitive inhibition relative to GSH. Chromatographic analyses suggest that 8-MOP is not a substrate. Molecular docking simulations suggest that 8-MOP binds to the active site, but its position prevents the GSH conjugation. Thus, we conclude that 8-MOP is a promising prototype for new GST inhibitors pharmacologically useful in the treatment of neurodegenerative disorders and the resistance of cancer to chemotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Martins Dias de Cerqueira
- Laboratory of Natural Products, Institute of Chemistry, Federal University of Bahia Salvador, Brazil
| | - Rute Maria Ferreira Lima
- Laboratory of Neurochemistry and Cell Biology, Institute of Health Sciences, Federal University of Bahia Salvador, Brazil
| | - Ramon Santos El-Bachá
- Laboratory of Neurochemistry and Cell Biology, Institute of Health Sciences, Federal University of Bahia Salvador, Brazil
| |
Collapse
|
41
|
Singh S. Cytoprotective and regulatory functions of glutathione S-transferases in cancer cell proliferation and cell death. Cancer Chemother Pharmacol 2014; 75:1-15. [PMID: 25143300 DOI: 10.1007/s00280-014-2566-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/04/2014] [Indexed: 01/05/2023]
Abstract
PURPOSE Glutathione S-transferases (GSTs) family of enzymes is best known for their cytoprotective role and their involvement in the development of anticancer drug resistance. Recently, emergence of non-detoxifying properties of GSTs has provided them with significant biological importance. Addressing the complex interactions of GSTs with regulatory kinases will help in understanding its precise role in tumor pathophysiology and in designing GST-centered anticancer strategies. METHODS We reviewed all published literature addressing the detoxification and regulatory roles of GSTs in the altered biology of cancer and evaluating novel agents targeting GSTs for cancer therapy. RESULTS The role of GSTs, especially glutathione S-transferase P1 isoform in tumoral drug resistance, has been the cause of intense debate. GSTs have been demonstrated to interact with different protein partners and modulate signaling pathways that control cell proliferation, differentiation and apoptosis. These specific functions of GSTs could lead to the development of new therapeutic approaches and to the identification of some interesting candidates for preclinical and clinical development. This review focuses on the crucial role played by GSTs in the development of resistance to anticancer agents and the major findings regarding the different modes of action of GSTs to regulate cell signaling.
Collapse
Affiliation(s)
- Simendra Singh
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Gautam Buddha Nagar, Greater Noida, UP, India,
| |
Collapse
|
42
|
2,2′-Dihydroxybenzophenones and their carbonyl N-analogues as inhibitor scaffolds for MDR-involved human glutathione transferase isoenzyme A1-1. Bioorg Med Chem 2014; 22:3957-70. [DOI: 10.1016/j.bmc.2014.06.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/20/2014] [Accepted: 06/04/2014] [Indexed: 12/21/2022]
|
43
|
Zhuo R, Kosak KM, Sankar S, Wiles ET, Sun Y, Zhang J, Ayello J, Prestwich GD, Shami PJ, Cairo MS, Lessnick SL, Luo W. Targeting Glutathione S-transferase M4 in Ewing sarcoma. Front Pediatr 2014; 2:83. [PMID: 25147782 PMCID: PMC4123608 DOI: 10.3389/fped.2014.00083] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/23/2014] [Indexed: 12/26/2022] Open
Abstract
Ewing sarcoma is a malignant pediatric bone and soft tissue tumor. Although the 5-year survival rate of localized disease approaches 75%, the prognosis of metastatic and/or therapy-resistant disease remains dismal despite the wide use of aggressive therapeutic strategies. We previously reported that high expression of glutathione S-transferase M4 (GSTM4) in primary tumors correlates with poor patient outcomes. GSTM4 is required for oncogenic transformation and mediates resistance to chemotherapeutic drugs in Ewing sarcoma cells. Here, we performed RNA-sequencing analyses of Ewing sarcoma cells and combined our results with publicly available datasets to demonstrate that GSTM4 is a major GST specifically expressed in Ewing sarcoma. Pharmacological inhibition of GSTM4 activity using a pan GST inhibitor, 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio) hexanol (NBDHEX), significantly limited cellular proliferation and oncogenic transformation of Ewing sarcoma cells. Moreover, combined use of NBDHEX and etoposide synergistically increased cytotoxicity, suggesting a role for GSTM4 as an inhibitor of apoptosis. Mechanistic studies revealed that GSTM4 limits apoptosis owing to its ability to interact with Apoptosis Signal-regulating Kinase 1 (ASK1) and inhibit signaling via the c-Jun N-terminal Kinase axis. To exploit our observation that GSTM4 expression is specifically up-regulated in Ewing sarcoma, we tested the effect of a GSTM4-activated anti-cancer agent, O(2)-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate or JS-K, on tumor growth and survival. We found that JS-K robustly decreased Ewing sarcoma cell viability and xenograft tumor growth and improved overall survival of xenograft mice. Our data suggest that GSTM4 is a novel therapeutic target for the treatment of high GSTM4-expressing Ewing sarcoma. Strategies that combine standard chemotherapy with agents that inhibit GSTM4, that are activated by GSTM4, or that block GSTM4/ASK1 interactions, can potentially be more specific and/or efficacious than standard therapeutic approaches.
Collapse
Affiliation(s)
- Rupeng Zhuo
- Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah , Salt Lake City, UT , USA
| | - Kenneth M Kosak
- Department of Internal Medicine, Division of Hematology and Hematologic Malignancies, University of Utah , Salt Lake City, UT , USA
| | - Savita Sankar
- Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah , Salt Lake City, UT , USA
| | - Elizabeth T Wiles
- Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah , Salt Lake City, UT , USA
| | - Ying Sun
- Microarray and Genomic Analysis Core Facility, Huntsman Cancer Institute, University of Utah , Salt Lake City, UT , USA
| | - Jianxing Zhang
- Department of Medicinal Chemistry, University of Utah , Salt Lake City, UT , USA
| | - Janet Ayello
- Department of Pediatrics, New York Medical College , Valhalla, NY , USA
| | - Glenn D Prestwich
- Department of Medicinal Chemistry, University of Utah , Salt Lake City, UT , USA
| | - Paul J Shami
- Department of Internal Medicine, Division of Hematology and Hematologic Malignancies, University of Utah , Salt Lake City, UT , USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College , Valhalla, NY , USA ; Department of Medicine, New York Medical College , Valhalla, NY , USA ; Department of Microbiology and Immunology, New York Medical College , Valhalla, NY , USA ; Department of Cell Biology and Anatomy, New York Medical College , Valhalla, NY , USA ; Department of Pathology, New York Medical College , Valhalla, NY , USA
| | - Stephen L Lessnick
- Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah , Salt Lake City, UT , USA ; Division of Pediatric Hematology/Oncology, School of Medicine, University of Utah , Salt Lake City, UT , USA
| | - Wen Luo
- Department of Pediatrics, New York Medical College , Valhalla, NY , USA ; Department of Pathology, New York Medical College , Valhalla, NY , USA
| |
Collapse
|
44
|
Djukic TI, Savic-Radojevic AR, Pekmezovic TD, Matic MG, Pljesa-Ercegovac MS, Coric VM, Radic TM, Suvakov SR, Krivic BN, Dragicevic DP, Simic TP. Glutathione S-transferase T1, O1 and O2 polymorphisms are associated with survival in muscle invasive bladder cancer patients. PLoS One 2013; 8:e74724. [PMID: 24040330 PMCID: PMC3770566 DOI: 10.1371/journal.pone.0074724] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/02/2013] [Indexed: 11/26/2022] Open
Abstract
Objective To examine the association of six glutathione transferase (GST) gene polymorphisms (GSTT1, GSTP1/rs1695, GSTO1/rs4925, GSTO2/rs156697, GSTM1, GSTA1/rs3957357) with the survival of patients with muscle invasive bladder cancer and the genotype modifying effect on chemotherapy. Patients and Methods A total of 105 patients with muscle invasive bladder cancer were included in the study. The follow-up lasted 5 years. The effect of GSTs polymorphisms on predicting mortality was analyzed by the Cox proportional hazard models, while Kaplan-Meier analysis was performed to assess differences in survival. Results GSTT1 active, GSTO1 Asp140Asp or GSTO2 Asp142Asp genotypes were independent predictors of a higher risk of death among bladder cancer patients (HR = 2.5, P = 0.028; HR = 2.9, P = 0.022; HR = 3.9, P = 0.001; respectively) and significantly influenced the overall survival. There was no association between GSTP1, GSTM1 and GSTA1 gene variants with overall mortality. Only GSTO2 polymorphism showed a significant effect on the survival in the subgroup of patients who received chemotherapy (P = 0.006). Conclusion GSTT1 active genotype and GSTO1 Asp140Asp and GSTO2 Asp142Asp genotypes may have a prognostic/pharmacogenomic role in patients with muscle invasive bladder cancer.
Collapse
Affiliation(s)
- Tatjana I. Djukic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ana R. Savic-Radojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tatjana D. Pekmezovic
- Institute of Epidemiology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marija G. Matic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marija S. Pljesa-Ercegovac
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vesna M. Coric
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tanja M. Radic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Sonja R. Suvakov
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Dejan P. Dragicevic
- Clinic of Urology, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tatjana P. Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- * E-mail:
| |
Collapse
|
45
|
Koutsoumpli GE, Dimaki VD, Thireou TN, Eliopoulos EE, Labrou NE, Varvounis GI, Clonis YD. Synthesis and study of 2-(pyrrolesulfonylmethyl)-N-arylimines: a new class of inhibitors for human glutathione transferase A1-1. J Med Chem 2012; 55:6802-13. [PMID: 22849615 DOI: 10.1021/jm300385f] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Overexpression of human GSTA1-1 in tumor cells is part of MDR mechanisms. We report on the synthesis of 11 pyrrole derivatives as hGSTA1-1 inhibitors starting from 1-methyl-2-[(2-nitrobenzylsulfanyl]-1H-pyrrole. Molecular modeling revealed two locations in the enzyme H binding site: the catalytic primary one accommodating shorter and longer derivatives and the secondary one, where shorter derivatives can occupy. Derivative 9, displaying the highest inhibition and bearing a p-nitroarylimino moiety, and derivative 4, lacking this moiety, were studied kinetically. Derivative 9 binds (K(i(9)) = 71 ± 4 μM) at the primary site competitively vs CDNB. Derivative 4 binds (K(i(4)) = 135 ± 27 μM) at the primary and secondary sites, allowing the binding of a second molecule (4 or CDNB) leading to formation of unreactive and reactive complexes, respectively. The arylmethylsulfonylpyrrole core structure is a new pharmacophore for hGSTA1-1, whereas its derivative 9 may serve as a lead structure.
Collapse
Affiliation(s)
- Georgia E Koutsoumpli
- Laboratory of Enzyme Technology, Department of Agricultural Biotechnology, Agricultural University of Athens , 75 Iera Odos Street, GR-118 55 Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
46
|
Nazarov AA, Risse J, Ang WH, Schmitt F, Zava O, Ruggi A, Groessl M, Scopelitti R, Juillerat-Jeanneret L, Hartinger CG, Dyson PJ. Anthracene-Tethered Ruthenium(II) Arene Complexes as Tools To Visualize the Cellular Localization of Putative Organometallic Anticancer Compounds. Inorg Chem 2012; 51:3633-9. [DOI: 10.1021/ic202530j] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Alexey A. Nazarov
- Institute
of Chemical Sciences
and Engineering, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Julie Risse
- Institute
of Chemical Sciences
and Engineering, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore, Singapore
117543, Singapore
| | - Frederic Schmitt
- University Institute of Pathology Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland
| | - Olivier Zava
- Institute
of Chemical Sciences
and Engineering, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Albert Ruggi
- Institute
of Chemical Sciences
and Engineering, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Michael Groessl
- Institute
of Chemical Sciences
and Engineering, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Rosario Scopelitti
- Institute
of Chemical Sciences
and Engineering, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | | | | | - Paul J. Dyson
- Institute
of Chemical Sciences
and Engineering, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|