51
|
Solovyev AI, Mikheylis AV, Plyusnin VF, Shubin AA, Grivin VP, Larionov SV, Tkachenko NV, Lemmetyinen H. Photochemistry of dithiophosphinate Ni(S2P(i-Bu)2)2 complex in CCl4. Transient species and TD-DFT calculations. J Photochem Photobiol A Chem 2019. [DOI: 10.1016/j.jphotochem.2019.111857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
52
|
Wu W, Yu L, Jiang Q, Huo M, Lin H, Wang L, Chen Y, Shi J. Enhanced Tumor-Specific Disulfiram Chemotherapy by In Situ Cu2+ Chelation-Initiated Nontoxicity-to-Toxicity Transition. J Am Chem Soc 2019; 141:11531-11539. [DOI: 10.1021/jacs.9b03503] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Wencheng Wu
- The State Key
Laboratory of High Performance Ceramics and Superfine Microstructures,
Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People’s Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People’s Republic of China
| | - Luodan Yu
- The State Key
Laboratory of High Performance Ceramics and Superfine Microstructures,
Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People’s Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People’s Republic of China
| | - Quzi Jiang
- The State Key
Laboratory of High Performance Ceramics and Superfine Microstructures,
Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People’s Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People’s Republic of China
| | - Minfeng Huo
- The State Key
Laboratory of High Performance Ceramics and Superfine Microstructures,
Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People’s Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People’s Republic of China
| | - Han Lin
- The State Key
Laboratory of High Performance Ceramics and Superfine Microstructures,
Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People’s Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People’s Republic of China
| | - Liying Wang
- The State Key
Laboratory of High Performance Ceramics and Superfine Microstructures,
Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People’s Republic of China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, People’s Republic of China
| | - Yu Chen
- The State Key
Laboratory of High Performance Ceramics and Superfine Microstructures,
Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People’s Republic of China
| | - Jianlin Shi
- The State Key
Laboratory of High Performance Ceramics and Superfine Microstructures,
Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People’s Republic of China
| |
Collapse
|
53
|
Askari B, Rudbari HA, Micale N, Schirmeister T, Giannetto A, Lanza S, Bruno G, Mirkhani V. Synthesis, solution behaviour and potential anticancer activity of new trinuclear organometallic palladium(II) complex of {S}-1-phenylethyl dithiooxamide: Comparison with the trinuclear heterobimetallic platinum(II) analogue. Polyhedron 2019. [DOI: 10.1016/j.poly.2019.02.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
54
|
New macrocyclic Cu(II) complex with bridge terephthalate: Synthesis, spectral properties, in vitro cytotoxic and antimicrobial activity. Comparison with related complexes. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2018.10.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
55
|
Yang YY, Yang JH, Wang D. Three New Metal-Organic Coordination Complexes: Crystal Structures and Anticancer Activity in Multiple Myeloma. J STRUCT CHEM+ 2019. [DOI: 10.1134/s0022476619050184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
56
|
Spillier Q, Vertommen D, Ravez S, Marteau R, Thémans Q, Corbet C, Feron O, Wouters J, Frédérick R. Anti-alcohol abuse drug disulfiram inhibits human PHGDH via disruption of its active tetrameric form through a specific cysteine oxidation. Sci Rep 2019; 9:4737. [PMID: 30894617 PMCID: PMC6426982 DOI: 10.1038/s41598-019-41187-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 12/27/2018] [Indexed: 12/15/2022] Open
Abstract
Due to rising costs and the difficulty to identify new targets, drug repurposing appears as a viable strategy for the development of new anti-cancer treatments. Although the interest of disulfiram (DSF), an anti-alcohol drug, to treat cancer was reported for many years, it is only very recently that one anticancer mechanism-of-action was highlighted. This would involve the inhibition of the p97 segregase adaptor NPL4, which is essential for the turnover of proteins involved in multiple regulatory and stress-response intracellular pathways. However, recently DSF was also reported as one of the first phosphoglycerate dehydrogenase (PHGDH) inhibitors, a tetrameric enzyme catalyzing the initial step of the serine synthetic pathway that is highly expressed in numerous cancer types. Here, we investigated the structure-activity relationships (SAR) of PHGDH inhibition by disulfiram analogues as well as the mechanism of action of DSF on PHGDH via enzymatic and cell-based evaluation, mass spectrometric and mutagenesis experiments.
Collapse
Affiliation(s)
- Quentin Spillier
- Medicinal Chemistry Research Group (CMFA), Louvain Drug Research Institute (LDRI), Université Catholique de Louvain, B-1200, Brussels, Belgium
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, B-1200, Brussels, Belgium
| | - Didier Vertommen
- de Duve Institute, Université catholique de Louvain, B-1200, Brussels, Belgium
| | - Séverine Ravez
- UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Université de Lille, Inserm, CHU Lille, F-59000, Lille, France
| | - Romain Marteau
- Medicinal Chemistry Research Group (CMFA), Louvain Drug Research Institute (LDRI), Université Catholique de Louvain, B-1200, Brussels, Belgium
| | - Quentin Thémans
- Department of Chemistry, NAmur MEdicine & Drug Innovation Center (NAMEDIC-NARILIS), Université de Namur, 61 rue de Bruxelles, B-5000, Namur, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, B-1200, Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, B-1200, Brussels, Belgium
| | - Johan Wouters
- Department of Chemistry, NAmur MEdicine & Drug Innovation Center (NAMEDIC-NARILIS), Université de Namur, 61 rue de Bruxelles, B-5000, Namur, Belgium
| | - Raphaël Frédérick
- Medicinal Chemistry Research Group (CMFA), Louvain Drug Research Institute (LDRI), Université Catholique de Louvain, B-1200, Brussels, Belgium.
| |
Collapse
|
57
|
Cui JL, Guo ET. Two novel metal coordination polymers: anticancer activity against human osteogenic sarcoma cells. INORG NANO-MET CHEM 2019. [DOI: 10.1080/24701556.2019.1569055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Ji-Li Cui
- Department of Pathology, The First Affiliated Hospital of Henan University, Kaifeng, Henan, China
| | - Er-Tao Guo
- Department of Gastroenterology, The First Affiliated Hospital of Henan University, Kaifeng, Henan, China
| |
Collapse
|
58
|
Li X, Huang Q, Long H, Zhang P, Su H, Liu J. A new gold(I) complex-Au(PPh 3)PT is a deubiquitinase inhibitor and inhibits tumor growth. EBioMedicine 2018; 39:159-172. [PMID: 30527624 PMCID: PMC6354570 DOI: 10.1016/j.ebiom.2018.11.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 11/16/2022] Open
Abstract
Background Ubiquitin-proteasome system (UPS) is integral to cell survival by maintaining protein homeostasis, and its dysfunction has been linked to cancer and several other human diseases. Through counteracting ubiquitination, deubiquitinases (DUBs) can either positively or negatively regulate UPS function, thereby representing attractive targets of cancer therapies. Previous studies have shown that metal complexes can inhibit tumor growth through targeting the UPS; however, novel metal complexes with higher specificity for cancer therapy are still lacking. Methods We synthesized a new gold(I) complex, Au(PPh3)PT. The inhibitory activity of Au(PPh3)PT on the UPS and the growth of multiple cancer cell types were tested in vitro, ex vivo, and in vivo. Furthermore, we compared the efficacy of Au(PPh3)PT with other metal compounds in inhibition of UPS function and tumor growth. Findings Here we report that (i) a new gold(I) complex-pyrithione, i.e., Au(PPh3)PT, induced apoptosis in two lung cancer cell lines A549 and NCI-H1299; (ii) Au(PPh3)PT severely impaired UPS proteolytic function; (iii) Au(PPh3)PT selectively inhibited 19S proteasome-associated DUBs (UCHL5 and USP14) and other non-proteasomal DUBs with minimal effects on the function of 20S proteasome; (iv) Au(PPh3)PT induced apoptosis in cancer cells from acute myeloid leukemia patients; (v) Au(PPh3)PT effectively suppressed the growth of lung adenocarcinoma xenografts in nude mice; and (vi) Au (PPh3)PT elicited less cytotoxicity in normal cells than several other metal compounds. Interpretation Together, this study discovers a new gold(I) complex to be an effective inhibitor of the DUBs and a potential anti-cancer drug. Fund The National High Technology Research and Development Program of China, the project of Guangdong Province Natural Science Foundation, the projects from Foundation for Higher Education of Guangdong, the project from Guangzhou Medical University for Doctor Scientists, the Medical Scientific Research Foundation of Guangdong Province, and the Guangzhou Key Medical Discipline Construction Project Fund.
Collapse
Affiliation(s)
- Xiaofen Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, China; Protein Modification and Degradation Lab, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangdong, China
| | - Qingtian Huang
- Protein Modification and Degradation Lab, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangdong, China
| | - Huidan Long
- Protein Modification and Degradation Lab, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangdong, China
| | - Peiquan Zhang
- Protein Modification and Degradation Lab, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangdong, China
| | - Huabo Su
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, China; Protein Modification and Degradation Lab, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangdong, China; Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jinbao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, China; Protein Modification and Degradation Lab, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangdong, China.
| |
Collapse
|
59
|
Na CG, Alexanian EJ. A General Approach to Site-Specific, Intramolecular C-H Functionalization Using Dithiocarbamates. Angew Chem Int Ed Engl 2018; 57:13106-13109. [PMID: 30085389 PMCID: PMC6249686 DOI: 10.1002/anie.201806963] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/17/2018] [Indexed: 01/11/2023]
Abstract
Intramolecular hydrogen atom transfer is an established approach for the site-specific functionalization of unactivated, aliphatic C-H bonds. Transformations using this strategy typically require unstable intermediates formed using strong oxidants and have mainly targeted C-H halogenations or intramolecular aminations. Herein, we report a site-specific C-H functionalization that significantly increases the synthetic scope and convergency of reactions proceeding via intramolecular hydrogen atom transfer. Stable, isolable N-dithiocarbamates are used as precursors to amidyl radicals formed via either light or radical initiation to efficiently deliver highly versatile alkyl dithiocarbamates across a wide range of complex structures.
Collapse
Affiliation(s)
- Christina G. Na
- Department of Chemistry The University of North Carolina at Chapel Hill Chapel Hill, NC 27599 (USA)
| | - Erik J. Alexanian
- Department of Chemistry The University of North Carolina at Chapel Hill Chapel Hill, NC 27599 (USA)
| |
Collapse
|
60
|
Trace element concentrations in breast cancer patients. Breast 2018; 42:142-149. [PMID: 30296647 DOI: 10.1016/j.breast.2018.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 09/13/2018] [Accepted: 09/22/2018] [Indexed: 12/14/2022] Open
Abstract
Alterations in the circulating levels of trace elements have been observed in breast cancer (BC) patients. However, the relationships between these alterations and the metabolic and clinical consequences of BC are unknown. The treatment-of-choice of BC is surgery followed by radiation therapy (RT). The present study was aimed at investigating: 1) the concentrations of several trace elements in BC patients, and their relationships with the intrinsic molecular subtypes of tumors; 2) the toxicological effect of RT. We studied 49 women with BC who were scheduled to receive RT following excision of the tumor. Plasma samples were obtained before and after the irradiation procedure. The control group was composed of 49 healthy women. Patients had significantly lower pre-RT concentrations of B, Cu, and Zn, and significantly higher concentrations of Sr than the control group. Irradiation was associated with a striking increase in plasma B concentrations, while Cu, Fe, Sr and Zn concentrations were not significantly different from pre-RT levels, albeit Sr and Zn showed non-significant trends towards increases. The plasma concentrations of B, Cu, Fe, Sr, and Zn were associated with the tumor expression of hormone receptors, epidermal growth factor receptor 2, Ki67 antigen, as well as dermatitis and asthenia, all of which represent the main toxicological responses to RT.
Collapse
|
61
|
Na CG, Alexanian EJ. A General Approach to Site‐Specific, Intramolecular C−H Functionalization Using Dithiocarbamates. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201806963] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Christina G. Na
- Department of ChemistryThe University of North Carolina at Chapel Hill Chapel Hill NC 27599 USA
| | - Erik J. Alexanian
- Department of ChemistryThe University of North Carolina at Chapel Hill Chapel Hill NC 27599 USA
| |
Collapse
|
62
|
Patel K, Ahmed ZSO, Huang X, Yang Q, Ekinci E, Neslund-Dudas CM, Mitra B, Elnady FAEM, Ahn YH, Yang H, Liu J, Dou QP. Discovering proteasomal deubiquitinating enzyme inhibitors for cancer therapy: lessons from rational design, nature and old drug reposition. Future Med Chem 2018; 10:2087-2108. [PMID: 30066579 PMCID: PMC6123888 DOI: 10.4155/fmc-2018-0091] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/15/2018] [Indexed: 12/24/2022] Open
Abstract
The ubiquitin proteasome system has been validated as a target of cancer therapies evident by the US FDA approval of anticancer 20S proteasome inhibitors. Deubiquitinating enzymes (DUBs), an essential component of the ubiquitin proteasome system, regulate cellular processes through the removal of ubiquitin from ubiquitinated-tagged proteins. The deubiquitination process has been linked with cancer and other pathologies. As such, the study of proteasomal DUBs and their inhibitors has garnered interest as a novel strategy to improve current cancer therapies, especially for cancers resistant to 20S proteasome inhibitors. This article reviews proteasomal DUB inhibitors in the context of: discovery through rational design approach, discovery from searching natural products and discovery from repurposing old drugs, and offers a future perspective.
Collapse
Affiliation(s)
- Kush Patel
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Zainab SO Ahmed
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Giza 12613, Egypt
| | - Xuemei Huang
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- School of Life Science & Technology, Harbin Institute of Technology, Harbin 150001, PR China
| | - Qianqian Yang
- Protein Modification & Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou 510000, PR China
| | - Elmira Ekinci
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Christine M Neslund-Dudas
- Department of Public Health Sciences & Henry Ford Cancer Institute, Henry Ford Health System, One Ford Place, Suite 5C, Detroit, MI 48202, USA
| | - Bharati Mitra
- Department of Biochemistry, Microbiology & Immunology, Wayne State University School of Medicine, 540 E. Canfield Avenue, Detroit, MI 48201, USA
| | - Fawzy AEM Elnady
- Department of Anatomy & Embryology, Faculty of Veterinary Medicine, Cairo University, Giza, Giza 12613, Egypt
| | - Young-Hoon Ahn
- Department of Chemistry, Wayne State University, Detroit, MI 48202, USA
| | - Huanjie Yang
- School of Life Science & Technology, Harbin Institute of Technology, Harbin 150001, PR China
| | - Jinbao Liu
- Protein Modification & Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou 510000, PR China
| | - Qing Ping Dou
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Protein Modification & Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou 510000, PR China
| |
Collapse
|
63
|
Wehbe M, Leung AWY, Abrams MJ, Orvig C, Bally MB. A Perspective - can copper complexes be developed as a novel class of therapeutics? Dalton Trans 2018; 46:10758-10773. [PMID: 28702645 DOI: 10.1039/c7dt01955f] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Although copper-ligand complexes appear to be promising as a new class of therapeutics, other than the family of copper(ii) coordination compounds referred to as casiopeínas these compounds have yet to reach the clinic for human use. The pharmaceutical challenges associated with developing copper-based therapeutics will be presented in this article along with a discussion of the potential for high-throughput chemistry, computer-aided drug design, and nanotechnology to address the development of this important class of drug candidates.
Collapse
Affiliation(s)
- Mohamed Wehbe
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada.
| | | | | | | | | |
Collapse
|
64
|
Sathiyaraj E, Perumal MV, Nagarajan ER, Ramalingan C. Functionalized zinc(II) dithiocarbamate complexes: Synthesis, spectral and molecular structures of bis(N-cyclopropyl-N-4-methoxybenzyldithiocarbamato-S,S′)zinc(II) and (2,2′-bipyridine)bis(N-cyclopropyl-N-4-methoxybenzyldithiocarbamato-S,S′)zinc(II). JOURNAL OF SAUDI CHEMICAL SOCIETY 2018. [DOI: 10.1016/j.jscs.2017.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
65
|
Sonawane VK, Mahajan UB, Shinde SD, Chatterjee S, Chaudhari SS, Bhangale HA, Ojha S, Goyal SN, Kundu CN, Patil CR. A Chemosensitizer Drug: Disulfiram Prevents Doxorubicin-Induced Cardiac Dysfunction and Oxidative Stress in Rats. Cardiovasc Toxicol 2018; 18:459-470. [DOI: 10.1007/s12012-018-9458-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
66
|
Chen X, Yang Q, Xiao L, Tang D, Dou QP, Liu J. Metal-based proteasomal deubiquitinase inhibitors as potential anticancer agents. Cancer Metastasis Rev 2018; 36:655-668. [PMID: 29039082 PMCID: PMC5721122 DOI: 10.1007/s10555-017-9701-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Deubiquitinases (DUBs) play an important role in protein quality control in eukaryotic cells due to their ability to specifically remove ubiquitin from substrate proteins. Therefore, recent findings have focused on the relevance of DUBs to cancer development, and pharmacological intervention on these enzymes has become a promising strategy for cancer therapy. In particular, several DUBs are physically and/or functionally associated with the proteasome and are attractive targets for the development of novel anticancer drugs. The successful clinical application of cisplatin in cancer treatment has prompted researchers to develop various metal-based anticancer agents with new properties. Recently, we have reported that several metal-based drugs, such as the antirheumatic gold agent auranofin (AF), the antifouling paint biocides copper pyrithione (CuPT) and zinc pyrithione (ZnPT), and also our two synthesized complexes platinum pyrithione (PtPT) and nickel pyrithione (NiPT), can target the proteasomal DUBs UCHL5 and USP14. In this review, we summarize the recently reported small molecule inhibitors of proteasomal DUBs, with a focus on discussion of the unique nature of metal-based proteasomal DUB inhibitors and their anticancer activity.
Collapse
Affiliation(s)
- Xin Chen
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qianqian Yang
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lu Xiao
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Daolin Tang
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Q Ping Dou
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China.,The Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, USA.,Department of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, MI, 48201-2013, USA
| | - Jinbao Liu
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
67
|
Valle EMA, Maltarollo VG, Almeida MO, Honorio KM, dos Santos MC, Cerchiaro G. Time dependent-density functional theory (TD-DFT) and experimental studies of UV–Visible spectra and cyclic voltammetry for Cu(II) complex with Et2DTC. J Mol Struct 2018. [DOI: 10.1016/j.molstruc.2017.12.097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
68
|
Karlsson H, Fryknäs M, Strese S, Gullbo J, Westman G, Bremberg U, Sjöblom T, Pandzic T, Larsson R, Nygren P. Mechanistic characterization of a copper containing thiosemicarbazone with potent antitumor activity. Oncotarget 2018; 8:30217-30234. [PMID: 28415818 PMCID: PMC5444738 DOI: 10.18632/oncotarget.16324] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 03/08/2017] [Indexed: 01/05/2023] Open
Abstract
Background The thiosemicarbazone CD 02750 (VLX50) was recently reported as a hit compound in a phenotype-based drug screen in primary cultures of patient tumor cells. We synthesized a copper complex of VLX50, denoted VLX60, and characterized its antitumor and mechanistic properties. Materials and Methods The cytotoxic effects and mechanistic properties of VLX60 were investigated in monolayer cultures of multiple human cell lines, in tumor cells from patients, in a 3-D spheroid cell culture system and in vivo and were compared with those of VLX50. Results VLX60 showed ≥ 3-fold higher cytotoxic activity than VLX50 in 2-D cultures and, in contrast to VLX50, retained its activity in the presence of additional iron. VLX60 was effective against non-proliferative spheroids and against tumor xenografts in vivo in a murine model. In contrast to VLX50, gene expression analysis demonstrated that genes associated with oxidative stress were considerably enriched in cells exposed to VLX60 as was induction of reactive oxygen. VLX60 compromised the ubiquitin-proteasome system and was more active in BRAF mutated versus BRAF wild-type colon cancer cells. Conclusions The cytotoxic effects of the copper thiosemicarbazone VLX60 differ from those of VLX50 and shows interesting features as a potential antitumor drug, notably against BRAF mutated colorectal cancer.
Collapse
Affiliation(s)
| | - Mårten Fryknäs
- Department of Medical Sciences, Uppsala University, Sweden
| | - Sara Strese
- Department of Medical Sciences, Uppsala University, Sweden
| | - Joachim Gullbo
- Department of Medical Sciences, Uppsala University, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| | - Gunnar Westman
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Ulf Bremberg
- Department of Medicinal Chemistry, Uppsala University, Sweden
| | - Tobias Sjöblom
- Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| | - Tatjana Pandzic
- Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| | - Rolf Larsson
- Department of Medical Sciences, Uppsala University, Sweden
| | - Peter Nygren
- Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| |
Collapse
|
69
|
Viola-Rhenals M, Patel KR, Jaimes-Santamaria L, Wu G, Liu J, Dou QP. Recent Advances in Antabuse (Disulfiram): The Importance of its Metal-binding Ability to its Anticancer Activity. Curr Med Chem 2018; 25:506-524. [PMID: 29065820 PMCID: PMC6873226 DOI: 10.2174/0929867324666171023161121] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/05/2016] [Accepted: 12/13/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Considerable evidence demonstrates the importance of dithiocarbamates especially disulfiram as anticancer drugs. However there are no systematic reviews outlining how their metal-binding ability is related to their anticancer activity. This review aims to summarize chemical features and metal-binding activity of disulfiram and its metabolite DEDTC, and discuss different mechanisms of action of disulfiram and their contributions to the drug's anticancer activity. METHODS We undertook a disulfiram-related search on bibliographic databases of peerreviewed research literature, including many historic papers and in vitro, in vivo, preclinical and clinical studies. The selected papers were carefully reviewed and summarized. RESULTS More than five hundreds of papers were obtained in the initial search and one hundred eighteen (118) papers were included in the review, most of which deal with chemical and biological aspects of Disulfiram and the relationship of its chemical and biological properties. Eighty one (81) papers outline biological aspects of dithiocarbamates, and fifty seven (57) papers report biological activity of Disulfiram as an inhibitor of proteasomes or inhibitor of aldehyde dehydrogenase enzymes, interaction with other anticancer drugs, or mechanism of action related to reactive oxygen species. Other papers reviewed focus on chemical aspects of dithiocarbamates. CONCLUSION This review confirms the importance of chemical features of compounds such as Disulfiram to their biological activities, and supports repurposing DSF as a potential anticancer agent.
Collapse
Affiliation(s)
- Maricela Viola-Rhenals
- Biochemistry and Cell Biology of Cancer Group, Exacts and Natural Science Faculty, University of Cartagena, Cartagena, Colombia
| | - Kush R. Patel
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, United States
| | - Laura Jaimes-Santamaria
- Biochemistry and Cell Biology of Cancer Group, Exacts and Natural Science Faculty, University of Cartagena, Cartagena, Colombia
| | - Guojun Wu
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, United States
| | - Jinbao Liu
- Guangzhou Medical University, Protein Modification and Degradation Lab, Dongfeng Xi road 195#, Guangzhou, Guangdong 510182, China
| | - Q. Ping Dou
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, United States
- Guangzhou Medical University, Protein Modification and Degradation Lab, Dongfeng Xi road 195#, Guangzhou, Guangdong 510182, China
| |
Collapse
|
70
|
Skrott Z, Mistrik M, Andersen KK, Friis S, Majera D, Gursky J, Ozdian T, Bartkova J, Turi Z, Moudry P, Kraus M, Michalova M, Vaclavkova J, Dzubak P, Vrobel I, Pouckova P, Sedlacek J, Miklovicova A, Kutt A, Li J, Mattova J, Driessen C, Dou QP, Olsen J, Hajduch M, Cvek B, Deshaies RJ, Bartek J. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature 2017; 552:194-199. [PMID: 29211715 PMCID: PMC5730499 DOI: 10.1038/nature25016] [Citation(s) in RCA: 508] [Impact Index Per Article: 72.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/08/2017] [Indexed: 12/21/2022]
Abstract
Cancer incidence is rising and this global challenge is further exacerbated by tumour resistance to available medicines. A promising approach to such unmet need for improved cancer treatment is drug repurposing. Here we highlight the potential for repurposing disulfiram (Antabuse), an old alcohol-aversion drug effective against diverse cancer types in preclinical studies. Our nationwide epidemiological study reveals that patients who continuously used disulfiram have a lower risk of death from cancer compared to those who stopped using the drug at their diagnosis. Moreover, we identify ditiocarb-copper complex as the metabolite of disulfiram responsible for anticancer effects, and provide methods to detect its preferential accumulation in tumours and candidate biomarkers for impact in cells and tissues. Finally, our functional and biophysical analyses reveal the long-sought molecular target of disulfiram’s tumour suppressing effects as NPL4, an adapter of p97/VCP segregase essential for protein turnover involved in multiple regulatory and stress-response cellular pathways.
Collapse
Affiliation(s)
- Zdenek Skrott
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | | | - Søren Friis
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| | - Dusana Majera
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jan Gursky
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Tomas Ozdian
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jirina Bartkova
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark.,Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Zsofia Turi
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Pavel Moudry
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Marianne Kraus
- Kantonsspital St Gallen, Department Oncology/Hematology, St Gallen, Switzerland
| | - Martina Michalova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jana Vaclavkova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Petr Dzubak
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Ivo Vrobel
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Pavla Pouckova
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| | - Jindrich Sedlacek
- Department of Cell Biology & Genetics, Palacky University, Olomouc, Czech Republic
| | | | - Anne Kutt
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| | - Jing Li
- Division of Biology and Biological Engineering, Caltech, Pasadena, California 91125, USA
| | - Jana Mattova
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| | - Christoph Driessen
- Kantonsspital St Gallen, Department Oncology/Hematology, St Gallen, Switzerland
| | - Q Ping Dou
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan, USA.,School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Jørgen Olsen
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Boris Cvek
- Department of Cell Biology & Genetics, Palacky University, Olomouc, Czech Republic
| | - Raymond J Deshaies
- Division of Biology and Biological Engineering, Caltech, Pasadena, California 91125, USA.,Howard Hughes Medical Institute, Caltech, Pasadena, California 91125, USA
| | - Jiri Bartek
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark.,Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
71
|
Wehbe M, Lo C, Leung AWY, Dragowska WH, Ryan GM, Bally MB. Copper (II) complexes of bidentate ligands exhibit potent anti-cancer activity regardless of platinum sensitivity status. Invest New Drugs 2017; 35:682-690. [PMID: 28733701 PMCID: PMC5694505 DOI: 10.1007/s10637-017-0488-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/28/2017] [Indexed: 01/06/2023]
Abstract
Insensitivity to platinum, either through inherent or acquired resistance, is a major clinical problem in the treatment of many solid tumors. Here, we explored the therapeutic potential of diethyldithiocarbamate (DDC), pyrithione (Pyr), plumbagin (Plum), 8-hydroxyquinoline (8-HQ), clioquinol (CQ) copper complexes in a panel of cancer cell lines that differ in their sensitivity to platins (cisplatin/carboplatin) using a high-content imaging system. Our data suggest that the copper complexes were effective against both platinum sensitive (IC50 ~ 1 μM platinum) and insensitive (IC50 > 5 μM platinum) cell lines. Furthermore, copper complexes of DDC, Pyr and 8-HQ had greater therapeutic activity compared to the copper-free ligands in all cell lines; whereas the copper-dependent activities of Plum and CQ were cell-line specific. Four of the copper complexes (Cu(DDC)2, Cu(Pyr)2, Cu(Plum)2 and Cu(8-HQ)2) showed IC50 values less than that of cisplatin in all tested cell lines. The complex copper DDC (Cu(DDC)2) was selected for in vivo evaluation due to its low nano-molar range activity in vitro and the availability of an injectable liposomal formulation. Liposomal (Cu(DDC)2) was tested in a fast-growing platinum-resistant A2780-CP ovarian xenograft model and was found to achieve a statistically significant reduction (50%; p < 0.05) in tumour size. This work supports the potential use of copper-based therapeutics to treat cancers that are insensitive to platinum drugs.
Collapse
Affiliation(s)
- Mohamed Wehbe
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2146 East Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Cody Lo
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Ada W Y Leung
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Wieslawa H Dragowska
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Gemma M Ryan
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Marcel B Bally
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2146 East Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Center for Drug Research and Development, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
72
|
Soave CL, Guerin T, Liu J, Dou QP. Targeting the ubiquitin-proteasome system for cancer treatment: discovering novel inhibitors from nature and drug repurposing. Cancer Metastasis Rev 2017; 36:717-736. [PMID: 29047025 PMCID: PMC5722705 DOI: 10.1007/s10555-017-9705-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the past 15 years, the proteasome has been validated as an anti-cancer drug target and 20S proteasome inhibitors (such as bortezomib and carfilzomib) have been approved by the FDA for the treatment of multiple myeloma and some other liquid tumors. However, there are shortcomings of clinical proteasome inhibitors, including severe toxicity, drug resistance, and no effect in solid tumors. At the same time, extensive research has been conducted in the areas of natural compounds and old drug repositioning towards the goal of discovering effective, economical, low toxicity proteasome-inhibitory anti-cancer drugs. A variety of dietary polyphenols, medicinal molecules, metallic complexes, and metal-binding compounds have been found to be able to selectively inhibit tumor cellular proteasomes and induce apoptotic cell death in vitro and in vivo, supporting the clinical success of specific 20S proteasome inhibitors bortezomib and carfilzomib. Therefore, the discovery of natural proteasome inhibitors and researching old drugs with proteasome-inhibitory properties may provide an alternative strategy for improving the current status of cancer treatment and even prevention.
Collapse
Affiliation(s)
- Claire L Soave
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, 540.1 HWCRC, 4100 John R Road, Detroit, MI, 48201-2013, USA
| | - Tracey Guerin
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, 540.1 HWCRC, 4100 John R Road, Detroit, MI, 48201-2013, USA
| | - Jinbao Liu
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Q Ping Dou
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, 540.1 HWCRC, 4100 John R Road, Detroit, MI, 48201-2013, USA.
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| |
Collapse
|
73
|
Ly NH, Nguyen TD, Zoh KD, Joo SW. Interaction between Diethyldithiocarbamate and Cu(II) on Gold in Non-Cyanide Wastewater. SENSORS (BASEL, SWITZERLAND) 2017; 17:E2628. [PMID: 29140287 PMCID: PMC5713075 DOI: 10.3390/s17112628] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 01/12/2023]
Abstract
A surface-enhanced Raman scattering (SERS) detection method for environmental copper ions (Cu2+) was developed according to the vibrational spectral change of diethyldithiocarbamate (DDTC) on gold nanoparticles (AuNPs). The ultraviolet-visible (UV-Vis) absorption spectra indicated that DDTC formed a complex with Cu2+, showing a prominent peak at ~450 nm. We found Raman spectral changes in DDTC from ~1490 cm-1 to ~1504 cm-1 on AuNPs at a high concentration of Cu2+ above 1 μM. The other ions of Zn2+, Pb2+, Ni2+, NH₄⁺, Mn2+, Mg2+, K⁺, Hg2+, Fe2+, Fe3+, Cr3+, Co2+, Cd2+, and Ca2+ did not produce such spectral changes, even after they reacted with DDTC. The electroplating industrial wastewater samples were tested under the interference of highly concentrated ions of Fe3+, Ni2+, and Zn2+. The Raman spectroscopy-based quantification of Cu2+ ions was able to be achieved for the wastewater after treatment with alkaline chlorination, whereas the cyanide-containing water did not show any spectral changes, due to the complexation of the cyanide with the Cu2+ ions. A micromolar range detection limit of Cu2+ ions could be achieved by analyzing the Raman spectra of DDTC in the cyanide-removed water.
Collapse
Affiliation(s)
- Nguyễn Hoàng Ly
- Department of Chemistry, Soongsil University, Seoul 156-743, Korea.
| | - Thanh Danh Nguyen
- Department of Chemistry, Soongsil University, Seoul 156-743, Korea.
- Department of Information Communication, Materials, Chemistry Convergence Technology, Soongsil University, Seoul 156-743, Korea.
| | - Kyung-Duk Zoh
- Department of Environmental Health Sciences, School of Public Health, Seoul National University, Seoul 08826, Korea.
| | - Sang-Woo Joo
- Department of Chemistry, Soongsil University, Seoul 156-743, Korea.
- Department of Information Communication, Materials, Chemistry Convergence Technology, Soongsil University, Seoul 156-743, Korea.
| |
Collapse
|
74
|
Cadmium pyrithione suppresses tumor growth in vitro and in vivo through inhibition of proteasomal deubiquitinase. Biometals 2017; 31:29-43. [PMID: 29098502 DOI: 10.1007/s10534-017-0062-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 10/19/2017] [Indexed: 10/18/2022]
Abstract
The ubiquitin-proteasome system (UPS) is indispensable to the protein quality control in eukaryotic cells. Due to the remarkable clinical success of using proteasome inhibitors for clinical treatment of multiple myeloma, it is anticipated that targeting the UPS upstream of the proteasome step be an effective strategy for cancer therapy. Deubiquitinases (DUB) are proteases that remove ubiquitin from target proteins and therefore regulate multiple cellular processes including some signaling pathways altered in cancer cells. Thus, targeting DUB is a promising strategy for cancer drug discovery. Previously, we have reported that metal complexes, such as copper and gold complexes, can disrupt the UPS via suppressing the activity of 19S proteasome-associated DUBs and/or of the 20S proteasomes, thereby inducing cancer cell death. In this study, we found that cadmium pyrithione (CdPT) treatment led to remarkable accumulation of ubiquitinated proteins in cultured cancer cells and primary leukemia cells. CdPT potently inhibited the activity of proteasomal DUBs (USP14 and UCHL5), but slightly inhibited 20S proteasome activity. The anti-cancer activity of CdPT was associated with triggering apoptosis via caspase activation. Moreover, treatment with CdPT inhibited proteasome function and repressed tumor growth in animal xenograft models. Our results show that cadmium-containing complex CdPT may function as a novel proteasomal DUB inhibitor and suggest appealing prospects for cancer treatment.
Collapse
|
75
|
Wang T, Liu Y, Fu Y, Huang T, Yang Y, Li S, Li C. Antiproliferative activity of di-2-pyridylhydrazone dithiocarbamate acetate partly involved in p53 mediated apoptosis and autophagy. Int J Oncol 2017; 51:1909-1919. [PMID: 29039462 DOI: 10.3892/ijo.2017.4149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/04/2017] [Indexed: 11/05/2022] Open
Abstract
Cancer cells have higher demand of iron and copper ions for growth, disturbing the metal's homeostasis can inhibit proliferation of cancer cell. Dithiocarbamates possessing excellent metal chelating ability and antitumor activity are considered as candidates in chelation therapy, however, their antitumor molecular mechanisms remain to be elucidated. In the present study, a dithiocarbamate derivative, di-2-pyridylhydrazone dithiocarbamate s-acetic acid (DpdtaA) was prepared to address the issue whether the molecular mechanism behind biological behavior showed by dithiocarbamate was p53 mediated. The proliferation inhibition assay showed that DpdtaA exhibited excellent antiproliferative effect for hepatocellular carcinoma (IC50= 3.0±0.4 µM for HepG2, 6.1±0.6 µM for Bel-7402 cell). However, in the presence of copper ion, the antiproliferative activity of DpdtaA significantly attenuated (~3-fold for HepG2) due to formation of copper chelate. The ROS assay revealed that the antiproliferative activity of DpdtaA correlated with ROS generation. Western blotting demonstrated that DpdtaA could upregulate p53 via down-regulating the Mdm2, accordingly leading to changes of bcl family proteins, indicating that a p53-dependent intrinsic apoptosis was partly involved. Simulation from molecular docking hinted that DpdtaA could disrupt interaction between p53 and Mdm2, indicating the disruption might also contribute to the upregulation of p53. The alternations in lysosome membrane permeability and acidic vacuoles as well as LC3-II upregulation indicated that autophagy was involved. The copper addition led to significantly attenuate biological activity of DpdtaA, with few dithiocarbamates, but the mechanism in apoptosis induction was not altered except for weaker ability.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Youxun Liu
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Yun Fu
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Tengfei Huang
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Yun Yang
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Shaoshan Li
- Department of Surgery, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Changzheng Li
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
76
|
Ferreira GR, de Oliveira LFC. Synthesis, spectroscopic and structural studies of new azo dyes metal chelates derivated from 1-phenil-azo-2-naphthol. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.05.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
77
|
Yadav MK, Maurya AK, Rajput G, Manar KK, Vinayak M, Drew MGB, Singh N. Synthesis, characterization, DNA binding and cleavage activity of homoleptic zinc(II) β-oxodithioester chelate complexes. J COORD CHEM 2017. [DOI: 10.1080/00958972.2017.1377835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Manoj Kumar Yadav
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Akhilendra Kumar Maurya
- Biochemistry & Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Gunjan Rajput
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Krishna Kumar Manar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Manjula Vinayak
- Biochemistry & Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | | | - Nanhai Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
78
|
Zhao C, Chen X, Yang C, Zang D, Lan X, Liao S, Zhang P, Wu J, Li X, Liu N, Liao Y, Huang H, Shi X, Jiang L, Liu X, Dou QP, Wang X, Liu J. Repurposing an antidandruff agent to treating cancer: zinc pyrithione inhibits tumor growth via targeting proteasome-associated deubiquitinases. Oncotarget 2017; 8:13942-13956. [PMID: 28086217 PMCID: PMC5355152 DOI: 10.18632/oncotarget.14572] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 12/27/2016] [Indexed: 12/29/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) plays a central role in various cellular processes through selectively degrading proteins involved in critical cellular functions. Targeting UPS has been validated as a novel strategy for treating human cancer, as inhibitors of the 20S proteasome catalytic activity are currently in clinical use for treatment of multiple myeloma and other cancers, and the deubiquitinase activity associated with the proteasome is also a valid target for anticancer agents. Recent studies suggested that zinc pyrithione, an FDA-approved antidandruff agent, may have antitumor activity, but the detailed molecular mechanisms remain unclear. Here we report that zinc pyrithione (ZnPT) targets the proteasome-associated DUBs (USP14 and UCHL5) and inhibits their activities, resulting in a rapid accumulation of protein-ubiquitin conjugates, but without inhibiting the proteolytic activities of 20S proteasomes. Furthermore, ZnPT exhibits cytotoxic effects against various cancer cell lines in vitro, selectively kills bone marrow cells from leukemia patients ex vivo, and efficiently inhibits the growth of lung adenocarcinoma cancer cell xenografts in nude mice. This study has identified zinc pyrithione, an FDA-approved pharmacological agent with potential antitumor properties as a proteasomal DUB inhibitor.
Collapse
Affiliation(s)
- Chong Zhao
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China.,Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China
| | - Xin Chen
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Changshan Yang
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Dan Zang
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Xiaoying Lan
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Siyan Liao
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Peiquan Zhang
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Jinjie Wu
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Xiaofen Li
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Ningning Liu
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China.,Guangzhou Research Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Yuning Liao
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Hongbiao Huang
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Xianping Shi
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Lili Jiang
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| | - Xiuhua Liu
- Institute of Environmental and Analytical Sciences, College of Chemistry and Chemical Engineering, Henan University, Kaifeng, Henan 475004, China
| | - Q Ping Dou
- The Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, Michigan 48201-2013, USA
| | - Xuejun Wang
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China.,Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota 57069, USA
| | - Jinbao Liu
- State Key Laboratory of Respiratory Disease, Protein Modification and Degradation Laboratory, Department of Pathophysiology, Guangzhou Medical University, Guangdong 510182, China
| |
Collapse
|
79
|
Hasinoff BB, Patel D. Disulfiram is a slow-binding partial noncompetitive inhibitor of 20S proteasome activity. Arch Biochem Biophys 2017; 633:23-28. [PMID: 28887129 DOI: 10.1016/j.abb.2017.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 08/31/2017] [Accepted: 09/02/2017] [Indexed: 01/10/2023]
Abstract
The alcohol abuse drug disulfiram has also been shown to exhibit potent cell growth inhibitory and anticancer activity. While a number of cellular and animal studies have suggested that disulfiram exhibits its anticancer activity through interaction with the proteasome, direct evidence for inhibition of proteasome activity is lacking. In this study we show that disulfiram potently inhibits the chymotrypsin-like activity of purified human 20S proteasome at low micromolar pharmacological concentrations. The enzyme progress curves displayed characteristics of a slow-binding reaction, similar to that observed for the FDA-approved proteasomal-targeted anticancer drugs bortezomib and carfilzomib. The apparent second order rate constant for reaction with 20s proteasome that was derived from an analysis of the progress curves was about 250-fold smaller than for bortezomib and carfilzomib. The concentration dependence of the enzyme kinetics was consistent with partial noncompetitive inhibition, whereby the putative disulfiram-proteasome adduct retains, partial but decreased enzyme activity. Disulfiram, which is known to have a high affinity for protein thiols, likely reacted with a non-critical cysteine residue, and not at the proteasome substrate binding site.
Collapse
Affiliation(s)
- Brian B Hasinoff
- College of Pharmacy, Apotex Centre, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada.
| | - Daywin Patel
- College of Pharmacy, Apotex Centre, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| |
Collapse
|
80
|
Poplaukhin P, Tiekink ERT. Crystal structure of bis-[ N-(2-hy-droxy-eth-yl)- N-methyl-dithio-carbamato-κ 2S, S'](pyridine)-zinc(II) pyridine monosolvate and its N-ethyl analogue. Acta Crystallogr E Crystallogr Commun 2017; 73:1246-1251. [PMID: 28932446 PMCID: PMC5598858 DOI: 10.1107/s2056989017010568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 12/03/2022]
Abstract
The common structural feature of the title compounds, [Zn(C4H8NOS2)2(C5H5N)]·C5H5N (I) and [Zn(C5H10NOS2)2(C5H5N)]·C5H5N (II), which differ by having di-thio-carbamate N-bound methyl (I) and ethyl (II) groups, is the coordination of each ZnII atom by two non-symmetrically chelating di-thio-carbamate ligands and by a pyridine ligand; in each case, the non-coordinating pyridine mol-ecule is connected to the Zn-containing mol-ecule via a (hy-droxy)O-H⋯N(pyridine) hydrogen bond. The resulting NS4 coordination geometry is closer to a square-pyramid than a trigonal bipyramid in the case of (I), but almost inter-mediate between the two extremes in (II). The mol-ecular packing features (hy-droxy)O-H⋯O(hy-droxy) hydrogen bonds, leading to supra-molecular chains with a zigzag arrangement along [10-1] (I) or a helical arrangement along [010] (II). In (I), π-π [inter-centroid distances = 3.4738 (10) and 3.4848 (10) Å] between coordinating and non-coordinating pyridine mol-ecules lead to stacks comprising alternating rings along the a axis. In (II), weaker π-π contacts occur between centrosymmetrically related pairs of coordinating pyridine mol-ecules [inter-centroid separation = 3.9815 (14) Å]. Further inter-actions, including C-H⋯π(chelate) inter-actions in (I), lead to a three-dimensional architecture in each case.
Collapse
Affiliation(s)
- Pavel Poplaukhin
- Chemical Abstracts Service, 2540 Olentangy River Rd, Columbus, Ohio 43202, USA
| | - Edward R. T. Tiekink
- Centre for Crystalline Materials, Faculty of Science and Technology, Sunway University, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
81
|
Wehbe M, Anantha M, Shi M, Leung AWY, Dragowska WH, Sanche L, Bally MB. Development and optimization of an injectable formulation of copper diethyldithiocarbamate, an active anticancer agent. Int J Nanomedicine 2017; 12:4129-4146. [PMID: 28615941 PMCID: PMC5459956 DOI: 10.2147/ijn.s137347] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Copper diethyldithiocarbamate (Cu(DDC)2) is the active anticancer agent generated when disulfiram (DSF) is provided in the presence of copper. To date, research directed toward repurposing DSF as an anticancer drug has focused on administration of DSF and copper in combination, efforts that have proven unsuccessful in clinical trials. This is likely due to the inability to form Cu(DDC)2 at relevant concentrations in regions of tumor growth. Little effort has been directed toward the development of Cu(DDC)2 because of the inherent aqueous insolubility of the complex. Here, we describe an injectable Cu(DDC)2 formulation prepared through a method that involves synthesis of Cu(DDC)2 inside the aqueous core of liposomes. Convection-enhanced delivery of a Cu(DDC)2 formulation prepared using 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC)/cholesterol liposomes into a rat model of F98 glioma engendered a 25% increase in median survival time relative to vehicle-treated animals. In a murine subcutaneous MV-4–11 model, treatment resulted in a 45% reduction in tumor burden when compared to controls. Pharmacokinetic studies indicated that the Cu(DDC)2 was rapidly eliminated after intravenous administration while the liposomes remained in circulation. To test whether liposomal lipid composition could increase Cu(DDC)2 circulation lifetime, a number of different formulations were evaluated. Studies demonstrated that liposomes composed of DSPC and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-n-(carboxy[polyethylene glycol]-2000) (95:5) enhanced Cu(DDC)2 concentrations in the circulation as reflected by a 4.2-fold increase in plasma AUC(0−∞) relative to the DSPC/cholesterol formulation. The anticancer activity of this Cu(DDC)2 formulation was subsequently evaluated in the MV-4–11 model. At its maximum tolerated dose, this formulation exhibited comparable activity to the DSPC/cholesterol formulation. This is the first report demonstrating the therapeutic effects of an injectable Cu(DDC)2 formulation in vivo.
Collapse
Affiliation(s)
- Mohamed Wehbe
- Experimental Therapeutics, British Columbia Cancer Agency.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC
| | | | - Minghan Shi
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Science.,Faculté de médecine et des sciences de la santé, Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec, QC
| | | | | | - Léon Sanche
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Science.,Faculté de médecine et des sciences de la santé, Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec, QC
| | - Marcel B Bally
- Experimental Therapeutics, British Columbia Cancer Agency.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC.,Department of Pathology and Laboratory Medicine, University of British Columbia.,Faculty of Medicine, Center for Drug Research and Development, Vancouver, BC, Canada
| |
Collapse
|
82
|
Aspartate aminotransferase is potently inhibited by copper complexes: Exploring copper complex-binding proteome. J Inorg Biochem 2017; 170:46-54. [DOI: 10.1016/j.jinorgbio.2017.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/28/2016] [Accepted: 02/10/2017] [Indexed: 12/11/2022]
|
83
|
Beniwal S, Chhimpa S, Gaur D, John P, Singh Y, Sharma J. Syntheses, characterization, antibacterial activity and molecular modelling of phenylantimony(III) heteroleptic derivatives containing substituted oximes and piperidine dithiocarbamate. Appl Organomet Chem 2017. [DOI: 10.1002/aoc.3725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Savita Beniwal
- Department of Chemistry; University of Rajasthan; Jaipur 302004 India
| | - Sunil Chhimpa
- Department of Zoology, Centre for Advanced Studies; University of Rajasthan; Jaipur 302004 India
| | - Deepti Gaur
- Department of Computer Science; North Cap University; Gurgaon 122017 India
| | - P.J. John
- Department of Zoology, Centre for Advanced Studies; University of Rajasthan; Jaipur 302004 India
| | - Yashpal Singh
- Department of Chemistry; University of Rajasthan; Jaipur 302004 India
| | - Jyoti Sharma
- Department of Chemistry; University of Rajasthan; Jaipur 302004 India
| |
Collapse
|
84
|
Bista R, Lee DW, Pepper OB, Azorsa DO, Arceci RJ, Aleem E. Disulfiram overcomes bortezomib and cytarabine resistance in Down-syndrome-associated acute myeloid leukemia cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:22. [PMID: 28143565 PMCID: PMC5286849 DOI: 10.1186/s13046-017-0493-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 01/26/2017] [Indexed: 02/02/2023]
Abstract
Background Children with Down syndrome (DS) have increased risk for developing AML (DS-AMKL), and they usually experience severe therapy-related toxicities compared to non DS-AMKL. Refractory/relapsed disease has very poor outcome, and patients would benefit from novel, less toxic, therapeutic strategies that overcome resistance. Relapse/resistance are linked to cancer stem cells with high aldehyde dehydrogenase (ALDH) activity. The purpose of the present work was to study less toxic alternative therapeutic agents for relapsed/refractory DS-AMKL. Methods Fourteen AML cell lines including the DS-AMKL CMY and CMK from relapsed/refractory AML were used. Cytarabine (Ara-C), bortezomib (BTZ), disulfiram/copper (DSF/Cu2+) were evaluated for cytotoxicity, depletion of ALDH-positive cells, and resistance. BTZ-resistant CMY and CMK variants were generated by continuous BTZ treatment. Cell viability was assessed using CellTiter-Glo®, ALDH activity by ALDELUORTM, and proteasome inhibition by western blot of ubiquitinated proteins and the Proteasome-Glo™ Chymotrypsin-Like (CT-like) assay, apoptosis by Annexin V Fluos/Propidium iodide staining, and mutations were detected using PCR, cloning and sequencing. Results Ara-C-resistant AML cell lines were sensitive to BTZ and DSF/Cu2+. The Ara-C-resistant DS-AMKL CMY cells had a high percentage of ALDHbright “stem-like” populations that may underlie Ara-C resistance. One percent of these cells were still resistant to BTZ but sensitive to DSF/Cu2+. To understand the mechanism of BTZ resistance, BTZ resistant (CMY-BR) and (CMK-BR) were generated. A novel mutation PSMB5 Q62P underlied BTZ resistance, and was associated with an overexpression of the β5 proteasome subunit. BTZ-resistance conferred increased resistance to Ara-C due to G1 arrest in the CMY-BR cells, which protected the cells from S-phase damage by Ara-C. CMY-BR and CMK-BR cells were cross-resistant to CFZ and MG-132 but sensitive to DSF/Cu2+. In this setting, DSF/Cu2+ induced apoptosis and proteasome inhibition independent of CT-like activity inhibition. Conclusions We provide evidence that DSF/Cu2+ overcomes Ara-C and BTZ resistance in cell lines from DS-AMKL patients. A novel mutation underlying BTZ resistance was detected that may identify BTZ-resistant patients, who may not benefit from treatment with CFZ or Ara-C, but may be responsive to DSF/Cu2+. Our findings support the clinical development of DSF/Cu2+ as a less toxic efficacious treatment approach in patients with relapsed/refractory DS-AMKL. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0493-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ranjan Bista
- Institute of Molecular Medicine at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - David W Lee
- Institute of Molecular Medicine at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Biosciences Partnership Building (BSPB), 5th floor, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Oliver B Pepper
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Biosciences Partnership Building (BSPB), 5th floor, 475 N 5th Street, Phoenix, AZ, 85004, USA.,Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - David O Azorsa
- Institute of Molecular Medicine at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Biosciences Partnership Building (BSPB), 5th floor, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Robert J Arceci
- Institute of Molecular Medicine at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Biosciences Partnership Building (BSPB), 5th floor, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Eiman Aleem
- Institute of Molecular Medicine at Phoenix Children's Hospital, Phoenix, AZ, USA. .,Department of Child Health, University of Arizona College of Medicine-Phoenix, Biosciences Partnership Building (BSPB), 5th floor, 475 N 5th Street, Phoenix, AZ, 85004, USA. .,Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
85
|
Manar KK, Yadav CL, Tiwari N, Singh RK, Kumar A, Drew MGB, Singh N. Effect of functionalities on the crystal structures of new zinc(ii) dithiocarbamates: a combined anti-leishmanial and thermal decomposition study. CrystEngComm 2017. [DOI: 10.1039/c7ce00211d] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
86
|
Zhang DJ, Yang SY, Teo BK. One-Dimensional Helical Metal Coordination Polymer: Synthesis and Structure of Infinite Chain of [Ag7(S2CNEt2)6]
n
+
(as [SbF6]− salt) Composed of Ag6(S2CNEt2)6 Cluster Units Linked by Ag(I) Ions via Peculiar Chelating S–S Bites. J CLUST SCI 2016. [DOI: 10.1007/s10876-016-1112-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
87
|
Haleel A, Mahendiran D, Veena V, Sakthivel N, Rahiman AK. Antioxidant, DNA interaction, VEGFR2 kinase, topoisomerase I and in vitro cytotoxic activities of heteroleptic copper(II) complexes of tetrazolo[1,5-a]pyrimidines and diimines. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 68:366-382. [DOI: 10.1016/j.msec.2016.05.120] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/20/2016] [Accepted: 05/27/2016] [Indexed: 10/21/2022]
|
88
|
Sun RWY, Zhang M, Li D, Li M, Wong AST. Enhanced anti-cancer activities of a gold(III) pyrrolidinedithiocarbamato complex incorporated in a biodegradable metal-organic framework. J Inorg Biochem 2016; 163:1-7. [DOI: 10.1016/j.jinorgbio.2016.06.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/04/2016] [Accepted: 06/14/2016] [Indexed: 12/18/2022]
|
89
|
Wang T, Fu Y, Huang T, Liu Y, Wu M, Yuan Y, Li S, Li C. Copper Ion Attenuated the Antiproliferative Activity of Di-2-pyridylhydrazone Dithiocarbamate Derivative; However, There Was a Lack of Correlation between ROS Generation and Antiproliferative Activity. Molecules 2016; 21:molecules21081088. [PMID: 27556432 PMCID: PMC6273760 DOI: 10.3390/molecules21081088] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/08/2016] [Accepted: 08/16/2016] [Indexed: 12/16/2022] Open
Abstract
The use of chelators for cancer treatment has been an alternative option. Dithiocarbamates have recently attracted considerable attention owning to their diverse biological activities; thus, the preparation of new dithiocarbamate derivatives with improved antitumor activity and selectivity as well as probing the underlying molecular mechanism are required. In this study, di-2-pyridylhydrazone dithiocarbamate S-propionic acid (DpdtpA) and its copper complex were prepared and characterized, and its antiproliferative activity was evaluated. The proliferation inhibition assay showed that DpdtpA exhibited excellent antiproliferative effect in hepatocellular carcinoma (IC50 = 1.3 ± 0.3 μM for HepG2, and 2.5 ± 0.6 μM for Bel-7402). However, in the presence of copper ion, the antiproliferative activity of DpdtpA was dramatically attenuated (20–30 fold) owing to the formation of copper chelate. A preliminarily mechanistic study revealed that reactive oxygen species (ROS) generation mediated the antiproliferative activity of DpdtpA, and accordingly induced apoptosis, DNA cleavage, and autophagy. Surprisingly, the cytotoxicity of DpdtpA copper complex (DpdtpA–Cu) was also involved in ROS generation; however, a paradoxical relation between cellular ROS level and cytotoxicity was observed. Further investigation indicated that DpdtpA could induce cell cycle arrest at the S phase; however, DpdtpA–Cu lacked this effect, which explained the difference in their antiproliferative activity.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
- Henan Collaborative Innovation Center of Molecular Diagnostics and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Yun Fu
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Tengfei Huang
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Youxun Liu
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Meihao Wu
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Yanbin Yuan
- Department of Surgery, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Shaoshan Li
- Department of Surgery, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Changzheng Li
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
- Henan Collaborative Innovation Center of Molecular Diagnostics and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| |
Collapse
|
90
|
Vrzal R, Dvorak Z. The comparative effects of diethyldithiocarbamate-copper complex with established proteasome inhibitors on expression levels of CYP1A2/3A4 and their master regulators, aryl hydrocarbon and pregnane X receptor in primary cultures of human hepatocytes. Fundam Clin Pharmacol 2016; 30:585-595. [PMID: 27414036 DOI: 10.1111/fcp.12221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 06/20/2016] [Accepted: 07/12/2016] [Indexed: 12/16/2022]
Abstract
In the recent years, a therapeutic potential of disulfiram (Antabuse) complex with copper, as an anticancer drug, was recognized towards several cancer cell lines. The proteasome was suggested as one of the cellular targets for this compound. As the therapeutic use of diethyldithiocarbamate-copper complex (CuET) is expected to increase, it is of great interest to know whether this compound may be the source of drug-drug interactions via the induction of biotransformation enzymes, especially cytochromes P450 (CYPs). To this purpose, we examined the effect of CuET and compared it with typical inducers (rifampicin and dioxin) of CYPs and with well-established proteasome inhibitors (MG132 and bortezomib). Diethyldithiocarbamate-copper complex revealed inconsistent and rather modulatory effect on the expression of CYP1A2 and CYP3A4 in several cultures of human hepatocytes. Moreover, it was able to cause neither ubiquitin accumulation nor significant and dose-dependent inhibition of proteasome activity. It had no effect on essential transcription factors involved in regulation of selected CYPs, aryl hydrocarbon (AhR) nor pregnane X receptor (PXR). However, the AhR protein was increased in majority of examined hepatocyte cultures. The main finding of this study is that: (i) disulfiram-copper complex is not the cause of drug-drug interactions via CYP1A2/3A4 induction; (ii) proteasome inhibitors may have different impact on studied parameters in given in vitro system.
Collapse
Affiliation(s)
- Radim Vrzal
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 27, Olomouc, 783 71, Czech Republic
| | - Zdenek Dvorak
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 27, Olomouc, 783 71, Czech Republic
| |
Collapse
|
91
|
Chetioui S, Rouag DA, Djukic JP, Bochet CG, Touzani R, Bailly C, Crochet A, Fromm KM. Crystal structures of a copper(II) and the isotypic nickel(II) and palladium(II) complexes of the ligand (E)-1-[(2,4,6-tri-bromo-phen-yl)diazen-yl]naphthalen-2-ol. Acta Crystallogr E Crystallogr Commun 2016; 72:1093-8. [PMID: 27536389 PMCID: PMC4971848 DOI: 10.1107/s205698901601080x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 07/04/2016] [Indexed: 11/17/2022]
Abstract
In the copper(II) complex, bis-{(E)-1-[(2,4,6-tri-bromo-phen-yl)diazen-yl]naph-thalen-2-olato}copper(II), [Cu(C16H8Br3N2O)2], (I), the metal cation is coord-inated by two N atoms and two O atoms from two bidentate (E)-1-[(2,4,6-tri-bromo-phen-yl)diazen-yl]naphthalen-2-olate ligands, forming a slightly distorted square-planar environment. In one of the ligands, the tri-bromo-benzene ring is inclined to the naphthalene ring system by 37.4 (5)°, creating a weak intra-molecular Cu⋯Br inter-action [3.134 (2) Å], while in the other ligand, the tri-bromo-benzene ring is inclined to the naphthalene ring system by 72.1 (6)°. In the isotypic nickel(II) and palladium(II) complexes, namely bis-{(E)-1-[(2,4,6-tri-bromo-phen-yl)diazen-yl]naphthalen-2-olato}nickel(II), [Ni(C16H8Br3N2O)2], (II), and bis-{(E)-1-[(2,4,6-tri-bromo-phen-yl)diazen-yl]naphthalen-2-olato}palladium(II), [Pd(C16H8Br3N2O)2], (III), respectively, the metal atoms are located on centres of inversion, hence the metal coordination spheres have perfect square-planar geometries. The tri-bromo-benzene rings are inclined to the naphthalene ring systems by 80.79 (18)° in (II) and by 80.8 (3)° in (III). In the crystal of (I), mol-ecules are linked by C-H⋯Br hydrogen bonds, forming chains along [010]. The chains are linked by C-H⋯π inter-actions, forming sheets parallel to (011). In the crystals of (II) and (III), mol-ecules are linked by C-H⋯π inter-actions, forming slabs parallel to (10-1). For the copper(II) complex (I), a region of disordered electron density was corrected for using the SQUEEZE routine in PLATON [Spek (2015 ▸). Acta Cryst. C71, 9-18]. The formula mass and unit-cell characteristics of the disordered solvent mol-ecules were not taken into account during refinement.
Collapse
Affiliation(s)
- Souheyla Chetioui
- Unité de Recherche de Chimie de l’Environnement et Moléculaire Structurale (CHEMS), Faculté des Sciences Exactes, Département de Chimie, Université des Frères Mentouri Constantine, Constantine 25000, Algeria
| | - Djamil-Azzeddine Rouag
- Unité de Recherche de Chimie de l’Environnement et Moléculaire Structurale (CHEMS), Faculté des Sciences Exactes, Département de Chimie, Université des Frères Mentouri Constantine, Constantine 25000, Algeria
| | - Jean-Pierre Djukic
- Laboratoire de Chimie et Systémique Organométallique (LCSOM), Institut de Chimie, Université de Strasbourg, UMR 7177., 4 rue Blaise Pascal, F-67070 Strasbourg Cedex, France
| | - Christian G. Bochet
- Chemistry Department, University of Fribourg, Chemin du Musee 9, CH-1700 Fribourg, Switzerland
| | - Rachid Touzani
- Laboratoire de Chimie Appliquée et Environnement, LCAE-URAC18, COSTE, Faculté des Sciences, Université Mohamed Premier, BP524, 60000 Oujda, Morocco
- Faculté Pluridisciplinaire Nador BP 300, Selouane 62702, Nador, Morocco
| | - Corinne Bailly
- Service de Radiocristallographie, Institut de Chimie, Université de Strasbourg, UMR 7177, 67008 Strasbourg Cedex, France
| | - Aurélien Crochet
- Fribourg Center for Nanomaterials, FriMat, University of Fribourg, Chemin du Musee 9, CH-1700 Fribourg, Switzerland
| | - Katharina M. Fromm
- Chemistry Department, University of Fribourg, Chemin du Musee 9, CH-1700 Fribourg, Switzerland
| |
Collapse
|
92
|
Zhao C, Chen X, Zang D, Lan X, Liao S, Yang C, Zhang P, Wu J, Li X, Liu N, Liao Y, Huang H, Shi X, Jiang L, Liu X, He Z, Wang X, Liu J. Platinum-containing compound platinum pyrithione is stronger and safer than cisplatin in cancer therapy. Biochem Pharmacol 2016; 116:22-38. [PMID: 27381943 PMCID: PMC5287571 DOI: 10.1016/j.bcp.2016.06.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/30/2016] [Indexed: 01/09/2023]
Abstract
DNA is the well-known molecular target of current platinum-based anticancer drugs; consequently, their clinical use is severely restricted by their systemic toxicities and drug resistance originating from non-selective DNA damage. Various strategies have been developed to circumvent the shortcomings of platinum-based chemotherapy but the inherent problem remains unsolved. Here we report that platinum pyrithione (PtPT), a chemically well-characterized synthetic complex of platinum, inhibits proteasome function and thereby exhibits greater and more selective cytotoxicity to multiple cancer cells than cisplatin, without showing discernible DNA damage both in vitro and in vivo. Moreover, unlike the classical proteasome inhibitor bortezomib/Velcade which inhibits the proteasome via blocking the peptidase activity of 20S proteasomes, PtPT primarily deactivates 26S proteasome-associated deubiquitinases USP14 and UCHL5. Furthermore, PtPT can selectively induce cytotoxicity and proteasome inhibition in cancer cells from leukemia patients but not peripheral blood mononuclear cells from healthy humans. In nude mice, PtPT also remarkably inhibited tumor xenograft growth, without showing the adverse effects that were induced by cisplatin. Hence, we have discovered a new platinum-based anti-tumor agent PtPT which targets 26S proteasome-associated deubiquitinases rather than DNA in the cell and thereby exerts safer and more potent anti-tumor effects, identifying a highly translatable new platinum-based anti-cancer strategy.
Collapse
Affiliation(s)
- Chong Zhao
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Xin Chen
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Dan Zang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Xiaoying Lan
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Siyan Liao
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Changshan Yang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Peiquan Zhang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Jinjie Wu
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Xiaofen Li
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Ningning Liu
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China.,Guangzhou Research Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Yuning Liao
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Hongbiao Huang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Xianping Shi
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Lili Jiang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Xiuhua Liu
- Institute of Environmental and Analytical Sciences, College of Chemistry and Chemical Engineering, Henan University, Kaifeng, Henan 475004, China.,Key Lab of Natural Drug and Immune Engineering of Henan Province, Kaifeng, Henan 475004, China
| | - Zhimin He
- Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou, Guangdong 510095, China
| | - Xuejun Wang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China.,Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD 57069, USA
| | - Jinbao Liu
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| |
Collapse
|
93
|
Bahrami H, Farhadi S, Siadatnasab F. Non-bonding interactions and non-covalent delocalization effects play a critical role in the relative stability of group 12 complexes arising from interaction of diethanoldithiocarbamate with the cations of transition metals Zn(II), Cd(II), and Hg(II): a theoretical study. J Mol Model 2016; 22:155. [PMID: 27299888 DOI: 10.1007/s00894-016-3008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 05/18/2016] [Indexed: 11/26/2022]
Abstract
The chelating properties of diethanoldithiocarbamate (DEDC) and π-electron flow from the nitrogen atom to the sulfur atom via a plane-delocalized π-orbital system (quasi ring) was studied using a density functional theory method. The molecular structure of DEDC and its complexes with Zn(II), Cd(II), and Hg(II) were also considered. First, the geometries of this ligand and DEDC-Zn(II), DEDC-Cd(II), and DEDC-Hg(II) were optimized, and the formation energies of these complexes were then calculated based on the electronic energy, or sum of electronic energies, with the zero point energy of each species. Formation energies indicated the DEDC-Zn(II) complex as the most stable complex, and DEDC-Cd(II) as the least stable. Structural data showed that the N1-C2 π-bond was localized in the complexes rather than the ligand, and a delocalized π-bond over S7-C2-S8 was also present. The stability of DEDC-Zn(II), DEDC-Cd(II), and DEDC-Hg(II) complexes increased in the presence of the non-specific effects of the solvent (PCM model), and their relative stability did not change. There was π-electron flow or resonance along N1-C2-S7 and along S7-C2-S8 in the ligand. The π-electron flow or resonance along N1-C2-S7 was abolished when the metal interacted with sulfur atoms. Energy belonging to van der Waals interactions and non-covalent delocalization effects between the metal and sulfur atoms of the ligand was calculated for each complex. The results of nucleus-independent chemical shift (NICS) indicated a decreasing trend as Zn(II) < Cd(II) < Hg(II) for the aromaticity of the quasi-rings. Finally, by ignoring van der Waals interactions and non-covalent delocalization effects between the metal and sulfur atoms of the ligand, the relative stability of the complexes was changed as follows:[Formula: see text] Graphical Abstract Huge electronic cloud localized on Hg(II) in the Hg(II)-DEDC complex.
Collapse
Affiliation(s)
- Homayoon Bahrami
- Department of Chemistry, Lorestan University, Khoramabad, 68135-465, Iran.
| | - Saeed Farhadi
- Department of Chemistry, Lorestan University, Khoramabad, 68135-465, Iran.
| | | |
Collapse
|
94
|
Wehbe M, Chernov L, Chen K, Bally MB. PRCosomes: pretty reactive complexes formed in liposomes. J Drug Target 2016; 24:787-796. [DOI: 10.1080/1061186x.2016.1186169] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mohamed Wehbe
- Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lina Chernov
- Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kent Chen
- Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, Canada
- Department of Interdisciplinary Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Marcel B. Bally
- Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Interdisciplinary Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
- Center for Drug Research and Development, Vancouver, BC, Canada
| |
Collapse
|
95
|
A novel nickel complex works as a proteasomal deubiquitinase inhibitor for cancer therapy. Oncogene 2016; 35:5916-5927. [PMID: 27086925 DOI: 10.1038/onc.2016.114] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/09/2015] [Accepted: 01/19/2016] [Indexed: 12/12/2022]
Abstract
Based on the central role of the ubiquitin-proteasome system (UPS) in the degradation of cellular proteins, proteasome inhibition has been considered an attractive approach for anticancer therapy. Deubiquitinases (DUBs) remove ubiquitin conjugates from diverse substrates; therefore, they are essential regulators of the UPS. DUB inhibitors, especially the inhibitors of proteasomal DUBs are becoming a research hotspot in targeted cancer therapy. Previous studies have shown that metal complexes, such as copper and zinc complexes, can induce cancer cell apoptosis through inhibiting UPS function. Moreover, we have found that copper pyrithione inhibits both 19S proteasome-associated DUBs and 20S proteasome activity with a mechanism distinct from that of the classical 20S proteasome inhibitor bortezomib. In the present study, we reveal that (i) nickel pyrithione complex (NiPT) potently inhibits the UPS via targeting the 19S proteasome-associated DUBs (UCHL5 and USP14), without effecting on the 20S proteasome; (ii) NiPT selectively induces proteasome inhibition and apoptosis in cultured tumor cells and cancer cells from acute myeloid leukemia human patients; and (iii) NiPT inhibits proteasome function and tumor growth in nude mice. This study, for the first time, uncovers a nickel complex as an effective inhibitor of the 19S proteasomal DUBs and suggests a potentially new strategy for cancer treatment.
Collapse
|
96
|
Wehbe M, Anantha M, Backstrom I, Leung A, Chen K, Malhotra A, Edwards K, Bally MB. Nanoscale Reaction Vessels Designed for Synthesis of Copper-Drug Complexes Suitable for Preclinical Development. PLoS One 2016; 11:e0153416. [PMID: 27055237 PMCID: PMC4824478 DOI: 10.1371/journal.pone.0153416] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/29/2016] [Indexed: 11/18/2022] Open
Abstract
The development of copper-drug complexes (CDCs) is hindered due to their very poor aqueous solubility. Diethyldithiocarbamate (DDC) is the primary metabolite of disulfiram, an approved drug for alcoholism that is being repurposed for cancer. The anticancer activity of DDC is dependent on complexation with copper to form copper bis-diethyldithiocarbamate (Cu(DDC)2), a highly insoluble complex that has not been possible to develop for indications requiring parenteral administration. We have resolved this issue by synthesizing Cu(DDC)2 inside liposomes. DDC crosses the liposomal lipid bilayer, reacting with the entrapped copper; a reaction that can be observed through a colour change as the solution goes from a light blue to dark brown. This method is successfully applied to other CDCs including the anti-parasitic drug clioquinol, the natural product quercetin and the novel targeted agent CX-5461. Our method provides a simple, transformative solution enabling, for the first time, the development of CDCs as viable candidate anticancer drugs; drugs that would represent a brand new class of therapeutics for cancer patients.
Collapse
Affiliation(s)
- Mohamed Wehbe
- Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| | - Malathi Anantha
- Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Ian Backstrom
- Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Ada Leung
- Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kent Chen
- Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Armaan Malhotra
- Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Katarina Edwards
- Department of Chemistry, University of Uppsala, 3 Husargatan (B7), Uppsala, Sweden
| | - Marcel B. Bally
- Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Center for Drug Research and Development, Vancouver, British Columbia, Canada
| |
Collapse
|
97
|
Fujie T, Murakami M, Yoshida E, Tachinami T, Shinkai Y, Fujiwara Y, Yamamoto C, Kumagai Y, Naka H, Kaji T. Copper diethyldithiocarbamate as an activator of Nrf2 in cultured vascular endothelial cells. J Biol Inorg Chem 2016; 21:263-73. [PMID: 26825804 PMCID: PMC4801994 DOI: 10.1007/s00775-016-1337-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/08/2016] [Indexed: 12/21/2022]
Abstract
The interest in organic-inorganic hybrid molecules as molecular probes for biological systems has been growing rapidly. Such hybrid molecules exhibit unique biological activities. Herein, copper(II) bis(diethyldithiocarbamate) (Cu10) was found to activate the transcription factor NF-E2-related factor 2 (Nrf2), which is responsible for regulating antioxidant and phase II xenobiotic enzymes, in vascular endothelial cells. The copper complex rapidly accumulated within cells and induced nuclear translocation of Nrf2, leading to upregulation of the expression of downstream proteins without cytotoxic effects. However, while copper bis(2-hydroxyethyl)dithiocarbamate activated Nrf2, copper ion, diethyldithiocarbamate ligand with or without zinc or iron failed to exhibit this activity. Intracellular accumulation of Cu10 was higher than that of Cu(II) and Cu(I). While the accumulation of copper(II) bis(dimethyldithiocarbamate) was reduced by small interfering RNA (siRNA)-mediated knockdown of the copper transporter CTR1, the knockdown did not affect Cu10 accumulation, indicating that Cu10 rapidly enters vascular endothelial cells via CTR1-independent mechanisms. In addition, copper and iron complexes with other ligands tested could not activate Nrf2, suggesting that the intramolecular interaction between copper and dithiocarbamate ligand is important for the activation of the transcription factor. Cu10 induced the expression of heme oxygenase-1, NAD(P)H quinone oxidoreductase 1, and γ-glutamylcysteine synthetase, downstream proteins of Nrf2. It was suggested that Cu10-induced activation of Nrf2 was due to proteasome inhibition as well as binding to Kelch-like ECH-associated protein 1. Since the effects of Cu10 on vascular endothelial cells are unique and diverse, the copper complex may be a good molecular probe to analyze the functions of the cells.
Collapse
Affiliation(s)
- Tomoya Fujie
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, 278-8510, Japan
| | - Masaki Murakami
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, 278-8510, Japan
| | - Eiko Yoshida
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, 278-8510, Japan
| | - Tadashi Tachinami
- Graduate School of Science and Research Center for Materials Science, Nagoya University, Chikusa, Nagoya, 464-8602, Japan
| | - Yasuhiro Shinkai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Yasuyuki Fujiwara
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, 192-0392, Japan
| | - Chika Yamamoto
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama, Funabashi, 274-8510, Japan
| | - Yoshito Kumagai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Hiroshi Naka
- Graduate School of Science and Research Center for Materials Science, Nagoya University, Chikusa, Nagoya, 464-8602, Japan.
| | - Toshiyuki Kaji
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, 278-8510, Japan.
| |
Collapse
|
98
|
Lee YM, Duh Y, Wang ST, Lai MMC, Yuan HS, Lim C. Using an Old Drug to Target a New Drug Site: Application of Disulfiram to Target the Zn-Site in HCV NS5A Protein. J Am Chem Soc 2016; 138:3856-62. [PMID: 26928525 DOI: 10.1021/jacs.6b00299] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In viral proteins, labile Zn-sites, where Zn(2+) is crucial for maintaining the native protein structure but the Zn-bound cysteines are reactive, are promising drug targets. Here, we aim to (i) identify labile Zn-sites in viral proteins using guidelines established from our previous work and (ii) assess if clinically safe Zn-ejecting agents could eject Zn(2+) from the predicted target site and thus inhibit viral replication. As proof-of-concept, we identified a labile Zn-site in the hepatitis C virus (HCV) NS5A protein and showed that the antialcoholism drug, disulfiram, could inhibit HCV replication to a similar extent as the clinically used antiviral agent, ribavirin. The discovery of a novel viral target and a new role for disulfiram in inhibiting HCV replication will enhance the therapeutic armamentarium against HCV. The strategy presented can also be applied to identify labile sites in other bacterial or viral proteins that can be targeted by disulfiram or other clinically safe Zn-ejectors.
Collapse
Affiliation(s)
- Yu-Ming Lee
- Institute of Biomedical Sciences, Academia Sinica , Taipei 115, Taiwan
| | - Yulander Duh
- Institute of Molecular Biology, Academia Sinica , Taipei 115, Taiwan
| | - Shih-Ting Wang
- Institute of Molecular Biology, Academia Sinica , Taipei 115, Taiwan
| | - Michael M C Lai
- Institute of Molecular Biology, Academia Sinica , Taipei 115, Taiwan
| | - Hanna S Yuan
- Institute of Molecular Biology, Academia Sinica , Taipei 115, Taiwan
| | - Carmay Lim
- Institute of Biomedical Sciences, Academia Sinica , Taipei 115, Taiwan.,Department of Chemistry, National Tsing Hua University , Hsinchu 300, Taiwan
| |
Collapse
|
99
|
Huang J, Campian JL, Gujar AD, Tran DD, Lockhart AC, DeWees TA, Tsien CI, Kim AH. A phase I study to repurpose disulfiram in combination with temozolomide to treat newly diagnosed glioblastoma after chemoradiotherapy. J Neurooncol 2016; 128:259-66. [DOI: 10.1007/s11060-016-2104-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 03/05/2016] [Indexed: 12/31/2022]
|
100
|
Liao W, Lai T, Chen L, Fu J, Sreenivasan ST, Yu Z, Ren J. Synthesis and Characterization of a Walnut Peptides-Zinc Complex and Its Antiproliferative Activity against Human Breast Carcinoma Cells through the Induction of Apoptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:1509-1519. [PMID: 26878665 DOI: 10.1021/acs.jafc.5b04924] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The walnut peptides and zinc ions were combined to generate a walnut peptides-zinc complex (WP1-Zn) with enhanced antiproliferative ability as well as reduced toxicity. The result indicated that Zn ions were successfully combined with WP1 through Zn-N and Zn-O covalent bonds. WP1-Zn compounds exhibited strong antiproliferative ability against the selected human cell lines, especially MCF-7 cells, whose survival rate reduced to 20.02% after exposure to 300 μg/mL of WP1-Zn for 48 h. WP1-Zn inhibited MCF-7 cell proliferation through inducing cell apoptosis and cell cycle arrest. The results indicated that WP1-Zn induced MCF-7 cell apoptosis via the ROS triggered mitochondrial-mediated pathway and cell surface receptor-mediated pathway. Our work is the first attempt to elucidate the synergic effect of novel walnut peptides and Zn and with the hope of better understanding the antiproliferative action of bioactive peptides and a zinc complex and support the potential application of WP1-Zn as a functional food ingredient or complementary medicine.
Collapse
Affiliation(s)
- Wenzhen Liao
- Department of Food Science and Technology, South China University of Technology , Wushan Road 381, Guangzhou 510640, China
| | - Ting Lai
- Department of Food Science and Technology, South China University of Technology , Wushan Road 381, Guangzhou 510640, China
| | - Luying Chen
- Department of Food Science and Technology, South China University of Technology , Wushan Road 381, Guangzhou 510640, China
| | - Junning Fu
- Department of Food Science and Engineering, Jinan University , Guangzhou 510623, China
| | - Sreeprasad T Sreenivasan
- Department of Civil and Environmental Engineering, Rice University , 6100 Main Street, Houston, Texas 77005, United States
| | - Zhiqiang Yu
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University , Guangzhou, Guangdong 510515, China
| | - Jiaoyan Ren
- Department of Food Science and Technology, South China University of Technology , Wushan Road 381, Guangzhou 510640, China
| |
Collapse
|