1
|
Herlah B, Goričan T, Benedik NS, Grdadolnik SG, Sosič I, Perdih A. Simulation- and AI-directed optimization of 4,6-substituted 1,3,5-triazin-2(1 H)-ones as inhibitors of human DNA topoisomerase IIα. Comput Struct Biotechnol J 2024; 23:2995-3018. [PMID: 39135887 PMCID: PMC11318567 DOI: 10.1016/j.csbj.2024.06.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/29/2024] [Accepted: 06/30/2024] [Indexed: 08/15/2024] Open
Abstract
The 4,6-substituted-1,3,5-triazin-2(1H)-ones are promising inhibitors of human DNA topoisomerase IIα. To further develop this chemical class targeting the enzyme´s ATP binding site, the triazin-2(1H)-one substitution position 6 was optimized. Inspired by binding of preclinical substituted 9H-purine derivative, bicyclic substituents were incorporated at position 6 and the utility of this modification was validated by a combination of molecular simulations, dynamic pharmacophores, and free energy calculations. Considering also predictions of Deepfrag, a software developed for structure-based lead optimization based on deep learning, compounds with both bicyclic and monocyclic substitutions were synthesized and investigated for their inhibitory activity. The SAR data showed that the bicyclic substituted compounds exhibited good inhibition of topo IIα, comparable to their mono-substituted counterparts. Further evaluation on a panel of human protein kinases showed selectivity for the inhibition of topo IIα. Mechanistic studies indicated that the compounds acted predominantly as catalytic inhibitors, with some exhibiting topo IIα poison effects at higher concentrations. Integration of STD NMR experiments and molecular simulations, provided insights into the binding model and highlighted the importance of the Asn120 interaction and hydrophobic interactions with substituents at positions 4 and 6. In addition, NCI-60 screening demonstrated cytotoxicity of the compounds with bicyclic substituents and identified sensitive human cancer cell lines, underlining the translational relevance of our findings for further preclinical development of this class of compounds. The study highlights the synergy between simulation and AI-based approaches in efficiently guiding molecular design for drug optimization, which has implications for further preclinical development of this class of compounds.
Collapse
Affiliation(s)
- Barbara Herlah
- National Institute of Chemistry, Hajdrihova 19, SI 1000 Ljubljana, Slovenia
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia
| | - Tjaša Goričan
- National Institute of Chemistry, Hajdrihova 19, SI 1000 Ljubljana, Slovenia
| | - Nika Strašek Benedik
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia
| | | | - Izidor Sosič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia
| | - Andrej Perdih
- National Institute of Chemistry, Hajdrihova 19, SI 1000 Ljubljana, Slovenia
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia
| |
Collapse
|
2
|
Kim Y, Kim S, Heo K, Lee S. Single-molecule FRET-based approach for protein-targeted drug discovery. Mol Cells 2024; 47:100150. [PMID: 39549747 DOI: 10.1016/j.mocell.2024.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
Many therapeutic drugs target various proteins involved in diverse biological processes. Among these proteins, type II topoisomerases are critical targets for anticancer and antibacterial chemotherapies, yet the action mechanisms of many type II topoisomerase-targeting drugs have not been fully elucidated. In this regard, the development of rapid and accurate methods to identify the mode of action of potential drug candidates is crucial to improve the efficiency of drug screening and discovery. Here, using type II topoisomerase as a model system, we present a single-molecule fluorescence resonance energy transfer-based drug screening method capable of delineating when and how the drug candidates participate in the entire reaction steps of the target protein. This unique capability has been demonstrated to be applicable to the identification of representative types of widely prescribed drugs targeting type II topoisomerase: etoposide which stabilizes the enzyme-DNA cleavage complex, and bisdioxopiperazines (ICRF-I93) which lock the N-terminal gate of the enzyme into the closed state. Based on this demonstration experiment, we expect that our proposed method will be extended to broad applications in the screening of potent drugs targeting various proteins.
Collapse
Affiliation(s)
- Yuyoung Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Surim Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kang Heo
- Strategic Development Team, Vieworks Company, Anyang-si, Gyeonggi-do 14055, Republic of Korea
| | - Sanghwa Lee
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea.
| |
Collapse
|
3
|
Cuevas D, Amigo R, Agurto A, Heredia AA, Guzmán C, Recabal-Beyer A, González-Pecchi V, Caprile T, Haigh JJ, Farkas C. The Role of Epithelial-to-Mesenchymal Transition Transcription Factors (EMT-TFs) in Acute Myeloid Leukemia Progression. Biomedicines 2024; 12:1915. [PMID: 39200378 PMCID: PMC11351244 DOI: 10.3390/biomedicines12081915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Acute myeloid leukemia (AML) is a diverse malignancy originating from myeloid progenitor cells, with significant genetic and clinical variability. Modern classification systems like those from the World Health Organization (WHO) and European LeukemiaNet use immunophenotyping, molecular genetics, and clinical features to categorize AML subtypes. This classification highlights crucial genetic markers such as FLT3, NPM1 mutations, and MLL-AF9 fusion, which are essential for prognosis and directing targeted therapies. The MLL-AF9 fusion protein is often linked with therapy-resistant AML, highlighting the risk of relapse due to standard chemotherapeutic regimes. In this sense, factors like the ZEB, SNAI, and TWIST gene families, known for their roles in epithelial-mesenchymal transition (EMT) and cancer metastasis, also regulate hematopoiesis and may serve as effective therapeutic targets in AML. These genes contribute to cell proliferation, differentiation, and extramedullary hematopoiesis, suggesting new possibilities for treatment. Advancing our understanding of the molecular mechanisms that promote AML, especially how the bone marrow microenvironment affects invasion and drug resistance, is crucial. This comprehensive insight into the molecular and environmental interactions in AML emphasizes the need for ongoing research and more effective treatments.
Collapse
Affiliation(s)
- Diego Cuevas
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción 4030000, Chile; (D.C.); (A.A.); (A.A.H.); (C.G.); (V.G.-P.)
| | - Roberto Amigo
- Laboratorio de Regulación Transcripcional, Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile;
| | - Adolfo Agurto
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción 4030000, Chile; (D.C.); (A.A.); (A.A.H.); (C.G.); (V.G.-P.)
| | - Adan Andreu Heredia
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción 4030000, Chile; (D.C.); (A.A.); (A.A.H.); (C.G.); (V.G.-P.)
| | - Catherine Guzmán
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción 4030000, Chile; (D.C.); (A.A.); (A.A.H.); (C.G.); (V.G.-P.)
| | - Antonia Recabal-Beyer
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile;
| | - Valentina González-Pecchi
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción 4030000, Chile; (D.C.); (A.A.); (A.A.H.); (C.G.); (V.G.-P.)
| | - Teresa Caprile
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile;
| | - Jody J. Haigh
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Carlos Farkas
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción 4030000, Chile; (D.C.); (A.A.); (A.A.H.); (C.G.); (V.G.-P.)
| |
Collapse
|
4
|
Lv C, Yang J, Zhao L, Zou Z, Kang C, Zhang Q, Wu C, Yang L, Cheng C, Zhao Y, Liao Q, Hu X, Li C, Sun X, Jin M. Bacillus subtilis partially inhibits African swine fever virus infection in vivo and in vitro based on its metabolites arctiin and genistein interfering with the function of viral topoisomerase II. J Virol 2023; 97:e0071923. [PMID: 37929962 PMCID: PMC10688316 DOI: 10.1128/jvi.00719-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/21/2023] [Indexed: 11/07/2023] Open
Abstract
IMPORTANCE African swine fever virus (ASFV) is a highly fatal swine disease that severely affects the pig industry. Although ASFV has been prevalent for more than 100 years, effective vaccines or antiviral strategies are still lacking. In this study, we identified four Bacillus subtilis strains that inhibited ASFV proliferation in vitro. Pigs fed with liquid biologics or powders derived from four B. subtilis strains mixed with pellet feed showed reduced morbidity and mortality when challenged with ASFV. Further analysis showed that the antiviral activity of B. subtilis was based on its metabolites arctiin and genistein interfering with the function of viral topoisomerase II. Our findings offer a promising new strategy for the prevention and control of ASFV that may significantly alleviate the economic losses in the pig industry.
Collapse
Affiliation(s)
- Changjie Lv
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Jingyu Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, China
| | - Li Zhao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, China
| | - Zhong Zou
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Chao Kang
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Qiang Zhang
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Chao Wu
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Li Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Chuxing Cheng
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Ya Zhao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Qi Liao
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Xiaotong Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Chengfei Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Xiaomei Sun
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Meilin Jin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
5
|
Kim Y, Lee HM. CRISPR-Cas System Is an Effective Tool for Identifying Drug Combinations That Provide Synergistic Therapeutic Potential in Cancers. Cells 2023; 12:2593. [PMID: 37998328 PMCID: PMC10670858 DOI: 10.3390/cells12222593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
Despite numerous efforts, the therapeutic advancement for neuroblastoma and other cancer treatments is still ongoing due to multiple challenges, such as the increasing prevalence of cancers and therapy resistance development in tumors. To overcome such obstacles, drug combinations are one of the promising applications. However, identifying and implementing effective drug combinations are critical for achieving favorable treatment outcomes. Given the enormous possibilities of combinations, a rational approach is required to predict the impact of drug combinations. Thus, CRISPR-Cas-based and other approaches, such as high-throughput pharmacological and genetic screening approaches, have been used to identify possible drug combinations. In particular, the CRISPR-Cas system (Clustered Regularly Interspaced Short Palindromic Repeats) is a powerful tool that enables us to efficiently identify possible drug combinations that can improve treatment outcomes by reducing the total search space. In this review, we discuss the rational approaches to identifying, examining, and predicting drug combinations and their impact.
Collapse
Affiliation(s)
| | - Hyeong-Min Lee
- Department of Computational Biology, St. Jude Research Hospital, Memphis, TN 38105, USA;
| |
Collapse
|
6
|
Wang T, Shi S, Shi Y, Jiang P, Hu G, Ye Q, Shi Z, Yu K, Wang C, Fan G, Zhao S, Ma H, Chang ACY, Li Z, Bian Q, Lin CP. Chemical-induced phase transition and global conformational reorganization of chromatin. Nat Commun 2023; 14:5556. [PMID: 37689690 PMCID: PMC10492836 DOI: 10.1038/s41467-023-41340-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 08/29/2023] [Indexed: 09/11/2023] Open
Abstract
Chemicals or drugs can accumulate within biomolecular condensates formed through phase separation in cells. Here, we use super-resolution imaging to search for chemicals that induce phase transition within chromatin at the microscale. This microscopic screening approach reveals that adriamycin (doxorubicin) - a widely used anticancer drug that is known to interact with chromatin - specifically induces visible local condensation and global conformational change of chromatin in cancer and primary cells. Hi-C and ATAC-seq experiments systematically and quantitatively demonstrate that adriamycin-induced chromatin condensation is accompanied by weakened chromatin interaction within topologically associated domains, compartment A/B switching, lower chromatin accessibility, and corresponding transcriptomic changes. Mechanistically, adriamycin complexes with histone H1 and induces phase transition of H1, forming fibrous aggregates in vitro. These results reveal a phase separation-driven mechanism for a chemotherapeutic drug.
Collapse
Affiliation(s)
- Tengfei Wang
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Shuxiang Shi
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- Lingang Laboratory, 200031, Shanghai, China
| | - Yuanyuan Shi
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Peipei Jiang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Ganlu Hu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Qinying Ye
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Zhan Shi
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Kexin Yu
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- iHuman Institute, ShanghaiTech University, 201010, Shanghai, China
| | - Chenguang Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoping Fan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Suwen Zhao
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- iHuman Institute, ShanghaiTech University, 201010, Shanghai, China
| | - Hanhui Ma
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Alex C Y Chang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Li
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Qian Bian
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China.
| |
Collapse
|
7
|
Olatunde OZ, Yong J, Lu C, Ming Y. A Review on Shikonin and Its Derivatives as Potent Anticancer Agents Targeted against Topoisomerases. Curr Med Chem 2023; 31:CMC-EPUB-129356. [PMID: 36752292 DOI: 10.2174/0929867330666230208094828] [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: 09/14/2022] [Revised: 11/12/2022] [Accepted: 11/30/2022] [Indexed: 02/09/2023]
Abstract
The topoisomerases (TOPO) play indispensable roles in DNA metabolism, by regulating the topological state of DNA. Topoisomerase I and II are the well-established drug-targets for the development of anticancer agents and antibiotics. These drugs-targeting enzymes have been used to establish the relationship between drug-stimulated DNA cleavable complex formation and cytotoxicity. Some anticancer drugs (such as camptothecin, anthracyclines, mitoxantrone) are also widely used as Topo I and Topo II inhibitors, but the poor water solubility, myeloma suppression, dose-dependent cardiotoxicity, and multidrug resistance (MDR) limited their prolong use as therapeutics. Also, most of these agents displayed selective inhibition only against Topo I or II. In recent years, researchers focus on the design and synthesis of the dual Topo I and II inhibitors, or the discovery of the dual Topo I and II inhibitors from natural products. Shikonin (a natural compound with anthraquinone skeleton, isolated from the roots of Lithospermum erythrorhizon) has drawn much attention due to its wide spectrum of anticancer activities, especially due to its dual Topo inhibitive performance, and without the adverse side effects, and different kinds of shikonin derivatives have been synthesized as TOPO inhibitors for the development of anticancer agents. In this review, the progress of the shikonin and its derivatives together with their anticancer activities, anticancer mechanism, and their structure-activity relationship (SAR) was comprehensively summarized by searching the CNKI, PubMed, Web of Science, Scopus, and Google Scholar databases.
Collapse
Affiliation(s)
- Olagoke Zacchaeus Olatunde
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian,350002, China
| | - Jianping Yong
- Xiamen Institute of Rare-earth Materials, Chinese Academy of Sciences, Xiamen, Fujian, 361021, China
| | - Canzhong Lu
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian,350002, China
- Xiamen Institute of Rare-earth Materials, Chinese Academy of Sciences, Xiamen, Fujian, 361021, China
| | - Yanlin Ming
- Fujian Institute of Subtropical Botany, Xiamen, Fujian, 361006, China
| |
Collapse
|
8
|
Koirala N, Butnariu M, Panthi M, Gurung R, Adhikari S, Subba RK, Acharya Z, Popović-Djordjević J. Antibiotics in the management of tuberculosis and cancer. ANTIBIOTICS - THERAPEUTIC SPECTRUM AND LIMITATIONS 2023:251-294. [DOI: 10.1016/b978-0-323-95388-7.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Xu G, Li Z, Ding Y, Shen Y. Discovery of 1,2-diphenylethene derivatives as human DNA topoisomerase II catalytic inhibitors and antitumor agents. Eur J Med Chem 2022; 243:114706. [DOI: 10.1016/j.ejmech.2022.114706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/09/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022]
|
10
|
Ravikumar C, Selvan ST, Saminathan M, Safin DA. Crystal structure, quantum computational, molecular docking and in vitro anti-proliferative investigations of 1H‐imidazole‐2‐thione analogues derivative. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
11
|
Dougherty A, Hawaz MG, Hoang KG, Trac J, Keck JM, Ayes C, Deweese JE. Exploration of the Role of the C-Terminal Domain of Human DNA Topoisomerase IIα in Catalytic Activity. ACS OMEGA 2021; 6:25892-25903. [PMID: 34660952 PMCID: PMC8515377 DOI: 10.1021/acsomega.1c02083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 09/15/2021] [Indexed: 06/13/2023]
Abstract
Human topoisomerase IIα (TOP2A) is a vital nuclear enzyme involved in resolving knots and tangles in DNA during replication and cell division. TOP2A is a homodimer with a symmetrical, multidomain structure. While the N-terminal and core regions of the protein are well-studied, the C-terminal domain is poorly understood but is involved in enzyme regulation and is predicted to be intrinsically disordered. In addition, it appears to be a major region of post-translational modification and includes several Ser and Thr residues, many of which have not been studied for biochemical effects. Therefore, we generated a series of human TOP2A mutants where we changed specific Ser and Thr residues in the C-terminal domain to Ala, Gly, or Ile residues. We designed, purified, and examined 11 mutant TOP2A enzymes. The amino acid changes were made between positions 1272 and 1525 with 1-7 residues changed per mutant. Several mutants displayed increased levels of DNA cleavage without displaying any change in plasmid DNA relaxation or DNA binding. For example, mutations in the regions 1272-1279, 1324-1343, 1351-1365, and 1374-1377 produced 2-3 times more DNA cleavage in the presence of etoposide than wild-type TOP2A. Further, several mutants displayed changes in relaxation and/or decatenation activity. Together, these results support previous findings that the C-terminal domain of TOP2A influences catalytic activity and interacts with the substrate DNA. Furthermore, we hypothesize that it may be possible to regulate the enzyme by targeting positions in the C-terminal domain. Because the C-terminal domain differs between the two human TOP2 isoforms, this strategy may provide a means for selectively targeting TOP2A for therapeutic inhibition. Additional studies are warranted to explore these results in more detail.
Collapse
Affiliation(s)
- Ashley
C. Dougherty
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Mariam G. Hawaz
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Kristine G. Hoang
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Judy Trac
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Jacob M. Keck
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Carmen Ayes
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Joseph E. Deweese
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland
Avenue, Nashville, Tennessee 37232-0146, United States
| |
Collapse
|
12
|
Kozurkova M. Acridine derivatives as inhibitors/poisons of topoisomerase II. J Appl Toxicol 2021; 42:544-552. [PMID: 34514603 DOI: 10.1002/jat.4238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 12/18/2022]
Abstract
The potential of acridines (amsacrine) as a topoisomerase II inhibitor or poison was first discovered in 1984, and since then, a considerable number of acridine derivatives have been tested as topoisomerase inhibitors/poisons, containing different substituents on the acridine chromophore. This review will discuss a series of studies published over the course of the last decade, which have investigated various novel acridine derivatives against topoisomerase II activity.
Collapse
Affiliation(s)
- Maria Kozurkova
- Department of Biochemistry, Institute of Chemistry, Faculty of Science, P. J. Šafárik University, Kosice, Slovak Republic.,Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
13
|
Trapped topoisomerase-DNA covalent complexes in the mitochondria and their role in human diseases. Mitochondrion 2021; 60:234-244. [PMID: 34500116 DOI: 10.1016/j.mito.2021.08.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 11/22/2022]
Abstract
Topoisomerases regulate DNA topology, organization of the intracellular DNA, the transmission of genetic materials, and gene expressions. Other than the nuclear genome, mitochondria also harbor the small, circular DNA (mtDNA) that encodes a critical subset of proteins for the production of cellular ATP; however, mitochondria are solely dependent on the nucleus for all the mitochondrial proteins necessary for mtDNA replication, repair, and maintenance. Mitochondrial genome compiles topological stress from bidirectional transcription and replication, therefore imports four nuclear encoded topoisomerases (Top1mt, Top2α, Top2β, and Top3α) in the mitochondria to relax mtDNA supercoiling generated during these processes. Trapping of topoisomerase on DNA results in the formation of protein-linked DNA adducts (PDAs), which are widely exploited by topoisomerase-targeting anticancer drugs. Intriguingly mtDNA is potentially exposed to DNA damage that has been attributed to a variety of human diseases, including neurodegeneration, cancer, and premature aging. In this review, we focus on the role of different topoisomerases in the mitochondria and our current understanding of the mitochondrial DNA damage through trapped protein-DNA complexes, and the progress in the molecular mechanisms of the repair for trapped topoisomerase covalent complexes (Topcc). Finally, we have discussed how the pathological DNA lesions that cause mtDNA damage,trigger mitochondrial fission and mitophagy, which serve as quality control events for clearing damaged mtDNA.
Collapse
|
14
|
Radaeva M, Ton AT, Hsing M, Ban F, Cherkasov A. Drugging the 'undruggable'. Therapeutic targeting of protein-DNA interactions with the use of computer-aided drug discovery methods. Drug Discov Today 2021; 26:2660-2679. [PMID: 34332092 DOI: 10.1016/j.drudis.2021.07.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/22/2021] [Accepted: 07/17/2021] [Indexed: 02/09/2023]
Abstract
Transcription factors (TFs) act as major oncodrivers in many cancers and are frequently regarded as high-value therapeutic targets. The functionality of TFs relies on direct protein-DNA interactions, which are notoriously difficult to target with small molecules. However, this prior view of the 'undruggability' of protein-DNA interfaces has shifted substantially in recent years, in part because of significant advances in computer-aided drug discovery (CADD). In this review, we highlight recent examples of successful CADD campaigns resulting in drug candidates that directly interfere with protein-DNA interactions of several key cancer TFs, including androgen receptor (AR), ETS-related gene (ERG), MYC, thymocyte selection-associated high mobility group box protein (TOX), topoisomerase II (TOP2), and signal transducer and activator of transcription 3 (STAT3). Importantly, these findings open novel and compelling avenues for therapeutic targeting of over 1600 human TFs implicated in many conditions including and beyond cancer.
Collapse
Affiliation(s)
- Mariia Radaeva
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Anh-Tien Ton
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Michael Hsing
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Fuqiang Ban
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| |
Collapse
|
15
|
Liu H, Lu C, Han L, Zhang X, Song G. Optical – Magnetic probe for evaluating cancer therapy. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
16
|
Vann KR, Oviatt AA, Osheroff N. Topoisomerase II Poisons: Converting Essential Enzymes into Molecular Scissors. Biochemistry 2021; 60:1630-1641. [PMID: 34008964 PMCID: PMC8209676 DOI: 10.1021/acs.biochem.1c00240] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The extensive length, compaction, and interwound nature of DNA, together with its controlled and restricted movement in eukaryotic cells, create a number of topological issues that profoundly affect all of the functions of the genetic material. Topoisomerases are essential enzymes that modulate the topological structure of the double helix, including the regulation of DNA under- and overwinding and the removal of tangles and knots from the genome. Type II topoisomerases alter DNA topology by generating a transient double-stranded break in one DNA segment and allowing another segment to pass through the DNA gate. These enzymes are involved in a number of critical nuclear processes in eukaryotic cells, such as DNA replication, transcription, and recombination, and are required for proper chromosome structure and segregation. However, because type II topoisomerases generate double-stranded breaks in the genetic material, they also are intrinsically dangerous enzymes that have the capacity to fragment the genome. As a result of this dualistic nature, type II topoisomerases are the targets for a number of widely prescribed anticancer drugs. This article will describe the structure and catalytic mechanism of eukaryotic type II topoisomerases and will go on to discuss the actions of topoisomerase II poisons, which are compounds that stabilize DNA breaks generated by the type II enzyme and convert these essential enzymes into "molecular scissors." Topoisomerase II poisons represent a broad range of structural classes and include anticancer drugs, dietary components, and environmental chemicals.
Collapse
Affiliation(s)
- Kendra R Vann
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Alexandria A Oviatt
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Neil Osheroff
- Departments of Biochemistry and Medicine (Hematology/Oncology), Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- VA Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United States
| |
Collapse
|
17
|
Mir RH, Mohi-ud-din R, Wani TU, Dar MO, Shah AJ, Lone B, Pooja C, Masoodi MH. Indole: A Privileged Heterocyclic Moiety in the Management of Cancer. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825666210208142108] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heterocyclic are a class of compounds that are intricately entwined into life processes.
Almost more than 90% of marketed drugs carry heterocycles. Synthetic chemistry, in
turn, allocates a cornucopia of heterocycles. Among the heterocycles, indole, a bicyclic structure
consisting of a six-membered benzene ring fused to a five-membered pyrrole ring with
numerous pharmacophores that generate a library of various lead molecules. Due to its profound
pharmacological profile, indole got wider attention around the globe to explore it fully
in the interest of mankind. The current review covers recent advancements on indole in the
design of various anti-cancer agents acting by targeting various enzymes or receptors, including
(HDACs), sirtuins, PIM kinases, DNA topoisomerases, and σ receptors.
Collapse
Affiliation(s)
- Reyaz Hassan Mir
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Roohi Mohi-ud-din
- Pharmacognosy Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, Kashmir, India
| | - Taha Umair Wani
- Pharmaceutics Lab, Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Mohammad Ovais Dar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Abdul Jaleel Shah
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Bashir Lone
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India
| | - Chawla Pooja
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, Moga-142001, India
| | - Mubashir Hussain Masoodi
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| |
Collapse
|
18
|
Lee KC, Yen CK, Chen CN, Chang SF, Lu YC, Huang WS. Drug Resistance of CPT-11 in Human DLD-1 Colorectal Cancer Cells through MutS Homolog 2 Upregulation. Int J Med Sci 2021; 18:1269-1276. [PMID: 33526988 PMCID: PMC7847627 DOI: 10.7150/ijms.52620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/18/2020] [Indexed: 11/20/2022] Open
Abstract
Colorectal cancers (CRCs) is the most commonly diagnosed and deadly cancer types in the world. Despite advances in chemotherapy for CRCs, drug resistance remains a major challenge to high incurable and eventually deadly rates for patients. CPT-11 is one of the current chemotherapy agents for CRC patients and the CPT-11 resistance development of CRCs is also inevitable. Recently, accumulating data has suggested that DNA repair system might be an inducer of chemotherapy resistance in cancer cells. Thus, this study was aimed to examine whether MutS homolog (MSH) 2, one member of DNA repair system, plays a role to affect the cytotoxicity of CPT-11 to CRCs. Human DLD-1 CRC cells were used in this study. It was shown that MSH2 gene and protein expression could be upregulated in DLD-1 cells under CPT-11 treatment and this upregulation subsequently attenuates the sensitivity of DLD-1 cells to CPT-11. Moreover, ERK1/2 and Akt signaling and AP-1 transcription factor have been found to modulate these effects. These results elucidate the drug resistance role of MSH2 upregulation in the CPT-11-treated DLD-1 CRC cells. Our findings may provide a useful thought for new adjuvant drug development by controlling the DNA repair system.
Collapse
Affiliation(s)
- Ko-Chao Lee
- Department of Colorectal Surgery, Department of Surgery, Chang Gung Memorial Hospital; Kaohsiung Medical Center, Kaohsiung 833, Taiwan
| | - Chia-Kung Yen
- Department of Food Science, National Chiayi University, Chiayi 600, Taiwan
| | - Cheng-Nan Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600, Taiwan
| | - Shun-Fu Chang
- Department of Medical Research and Development, Chiayi Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| | - Ying-Chen Lu
- Department of Food Science, National Chiayi University, Chiayi 600, Taiwan
| | - Wen-Shih Huang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan.,Division of Colon and Rectal Surgery, Department of Surgery, Chiayi Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| |
Collapse
|
19
|
Sagredou S, Dalezis P, Nikoleousakos N, Nikolaou M, Voura M, Almpanakis K, Panayiotidis MI, Sarli V, Trafalis DT. 3,6-Disubstituted 1,2,4-Triazolo[3,4- b]Thiadiazoles with Anticancer Activity Targeting Topoisomerase II Alpha. Onco Targets Ther 2020; 13:7369-7386. [PMID: 32801761 PMCID: PMC7395825 DOI: 10.2147/ott.s254856] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/30/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Topoisomerase IIα (topIIα) maintains the topology of DNA in order to ensure the proper functioning of numerous DNA processes. Inhibition of topIIα leads to the killing of cancer cells thus constituting such inhibitors as useful tools in cancer therapeutics. Triazolo[3,4-b]thiadiazole derivatives are known for their wide range of pharmacological activities while previous studies have documented their in vitro anticancer activity. The purpose of the current study was to investigate if these chemical compounds can act as topIIα inhibitors in cell-free and cell-based systems. MATERIALS AND METHODS The MTT assay was performed in DLD-1, HT-29, and LoVo cancer cells so as to evaluate the antiproliferative activity of KA25, KA26, and KA39 triazolo[3,4-b]thiadiazole derivatives. The KA39 compound was tested as a potential topIIα inhibitor using the plasmid-based topoisomerase II drug screening kit. The inhibitory effect of the three derivatives on topIIα phosphorylation was studied in HT-29 and LoVo cancer cells according to Human Phospho-TOP2A/Topoisomerase II Alpha Cell-Based Phosphorylation ELISA Kit. Moreover, flow cytometry was utilized in order to explore apoptotic induction and cell cycle growth arrest, upon treatment with KA39, in DLD-1 and HT-29 cells, respectively. In silico studies were also carried out for further investigation. RESULTS All three triazolo[3,4-b]thiadiazole derivatives showed an in vitro antiproliferative effect with the KA39 compound being the most potent one. Our results indicated that KA39 induced both early and late apoptosis as well as cell cycle growth arrest in S phase. In addition, the compound blocked the relaxation of supercoiled DNA while it also inhibited topIIα phosphorylation (upon treatment; P<0.001). CONCLUSION Among the three triazolo[3,4-b]thiadiazole derivatives, KA39 was shown to be the most potent anticancer agent and catalytic inhibitor of topIIα phosphorylation as well.
Collapse
Affiliation(s)
- Sofia Sagredou
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens11527, Greece
| | - Panagiotis Dalezis
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens11527, Greece
| | - Nikolaos Nikoleousakos
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens11527, Greece
| | - Michail Nikolaou
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens11527, Greece
| | - Maria Voura
- Department of Chemistry, Aristotle University of Thessaloniki , Thessaloniki, 54124, Greece
| | | | - Mihalis I Panayiotidis
- Department of Electron Microscopy & Molecular Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia2371, Cyprus
- The Cyprus School of Molecular Medicine, Nicosia1683, Cyprus
| | - Vasiliki Sarli
- Department of Chemistry, Aristotle University of Thessaloniki , Thessaloniki, 54124, Greece
| | - Dimitrios T Trafalis
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens11527, Greece
| |
Collapse
|
20
|
Radaeva M, Dong X, Cherkasov A. The Use of Methods of Computer-Aided Drug Discovery in the Development of Topoisomerase II Inhibitors: Applications and Future Directions. J Chem Inf Model 2020; 60:3703-3721. [DOI: 10.1021/acs.jcim.0c00325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mariia Radaeva
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Xuesen Dong
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| |
Collapse
|
21
|
Zhang M, Xiao R, Liu G, Huang Y. Genotoxins exaggerate the stressed state of aneuploid embryonic stem cells via activation of autophagy. SCIENCE CHINA. LIFE SCIENCES 2020; 63:1026-1036. [PMID: 31872377 DOI: 10.1007/s11427-019-9666-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/01/2019] [Indexed: 01/19/2023]
Abstract
The cellular consequences of aneuploidy are largely dependent on the cell types examined. Aneuploid yeasts and mouse embryonic fibroblasts exhibit cell proliferation defects and can be selectively inhibited by compounds that cause proteotoxic or energy stress. By contrast, most aneuploid pluripotent stem cells proliferate rapidly and reach higher saturation densities. The responses of aneuploid pluripotent stem cells to the stress-inducing compounds remain uncharacterized. Here, we tested the response of aneuploid embryonic stem cells to several compounds that caused proteotoxic, energy and genotoxic stress using previously established mouse embryonic stem cell lines trisomic for chromosome 6, 8, 11, or 15. Not all trisomic embryonic stem cells were selectively inhibited by compounds that cause proteotoxic or energy stress. However, most of these cells exhibited increased sensitivity to genotoxins. They displayed elevated DNA damage response as characterized by increased γH2A.X foci under genotoxic stress. Further investigations indicated that elevated autophagy levels might contribute to the increased cytotoxic effects of genotoxins on trisomic embryonic stem cells. Our study laid the foundation for eliminating aneuploidy that might be an effective approach for controlling cancer progression.
Collapse
Affiliation(s)
- Meili Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China. .,Department of Medical Genetics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| | - Rong Xiao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.,Department of Medical Genetics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Guang Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.,Department of Medical Genetics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Yue Huang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China. .,Department of Medical Genetics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
22
|
Topoisomerase IIα is essential for maintenance of mitotic chromosome structure. Proc Natl Acad Sci U S A 2020; 117:12131-12142. [PMID: 32414923 DOI: 10.1073/pnas.2001760117] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Topoisomerase IIα (TOP2A) is a core component of mitotic chromosomes and important for establishing mitotic chromosome condensation. The primary roles of TOP2A in mitosis have been difficult to decipher due to its multiple functions across the cell cycle. To more precisely understand the role of TOP2A in mitosis, we used the auxin-inducible degron (AID) system to rapidly degrade the protein at different stages of the human cell cycle. Removal of TOP2A prior to mitosis does not affect prophase timing or the initiation of chromosome condensation. Instead, it prevents chromatin condensation in prometaphase, extends the length of prometaphase, and ultimately causes cells to exit mitosis without chromosome segregation occurring. Surprisingly, we find that removal of TOP2A from cells arrested in prometaphase or metaphase cause dramatic loss of compacted mitotic chromosome structure and conclude that TOP2A is crucial for maintenance of mitotic chromosomes. Treatments with drugs used to poison/inhibit TOP2A function, such as etoposide and ICRF-193, do not phenocopy the effects on chromosome structure of TOP2A degradation by AID. Our data point to a role for TOP2A as a structural chromosome maintenance enzyme locking in condensation states once sufficient compaction is achieved.
Collapse
|
23
|
Exploring the binding mode of triflamide derivatives at the active site of Topo I and Topo II enzymes: In silico analysis and precise molecular docking. J CHEM SCI 2020. [DOI: 10.1007/s12039-020-1750-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
24
|
Lima MA, Costa VA, Franco MA, de Oliveira GP, Deflon VM, Rocha FV. Palladium(II) complexes bearing thiosemicarbazone and phosphines as inhibitors of DNA-Topoisomerase II enzyme: Synthesis, characterizations and biological studies. INORG CHEM COMMUN 2020. [DOI: 10.1016/j.inoche.2019.107708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
25
|
Ting CP, Tschanen E, Jang E, Maimone TJ. Total synthesis of podophyllotoxin and select analog designs via C–H activation. Tetrahedron 2019. [DOI: 10.1016/j.tet.2019.04.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
26
|
Tarasov VV, Chubarev VN, Ashraf GM, Dostdar SA, Sokolov AV, Melnikova TI, Sologova SS, Grigorevskich EM, Makhmutovа A, Kinzirsky AS, Klochkov SG, Aliev G. How Cancer Cells Resist Chemotherapy: Design and Development of Drugs Targeting Protein-Protein Interactions. Curr Top Med Chem 2019; 19:394-412. [DOI: 10.2174/1568026619666190305130141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 09/20/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023]
Abstract
Background:Resistance toward chemotherapeutics is one of the main obstacles on the way to effective cancer treatment. Personalization of chemotherapy could improve clinical outcome. However, despite preclinical significance, most of the potential markers have failed to reach clinical practice partially due to the inability of numerous studies to estimate the marker’s impact on resistance properly.Objective:The analysis of drug resistance mechanisms to chemotherapy in cancer cells, and the proposal of study design to identify bona fide markers.Methods:A review of relevant papers in the field. A PubMed search with relevant keywords was used to gather the data. An example of a search request: drug resistance AND cancer AND paclitaxel.Results:We have described a number of drug resistance mechanisms to various chemotherapeutics, as well as markers to underlie the phenomenon. We also proposed a model of a rational-designed study, which could be useful in determining the most promising potential biomarkers.Conclusion:Taking into account the most reasonable biomarkers should dramatically improve clinical outcome by choosing the suitable treatment regimens. However, determining the leading biomarkers, as well as validating of the model, is a work for further investigations.
Collapse
Affiliation(s)
- Vadim V. Tarasov
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Vladimir N. Chubarev
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samira A. Dostdar
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Alexander V. Sokolov
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Tatiana I. Melnikova
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Susanna S. Sologova
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Ekaterina M. Grigorevskich
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Alfiya Makhmutovа
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Alexander S. Kinzirsky
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Sergey G. Klochkov
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Gjumrakch Aliev
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| |
Collapse
|
27
|
Murugavel S, Ravikumar C, Jaabil G, Alagusundaram P. Synthesis, computational quantum chemical study, in silico ADMET and molecular docking analysis, in vitro biological evaluation of a novel sulfur heterocyclic thiophene derivative containing 1,2,3-triazole and pyridine moieties as a potential human topoisomerase IIα inhibiting anticancer agent. Comput Biol Chem 2019; 79:73-82. [PMID: 30731361 DOI: 10.1016/j.compbiolchem.2019.01.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 01/17/2019] [Accepted: 01/25/2019] [Indexed: 02/07/2023]
Abstract
Computational quantum chemical study and biological evaluation of a synthesized novel sulfur heterocyclic thiophene derivative containing 1,2,3-triazole and pyridine moieties namely BTPT [2-(1-benzyl-5-methyl-1H-1,2,3-triazol-4-yl)-6-methoxy-4-(thiophen-2-yl) pyridine] was presented in this study. The crystal structure was determined by SCXRD method. For the title compound BTPT, spectroscopic characterization like 1H NMR, 13C NMR, FTIR, UV-vis were carried out theoretically by computational DFT method and compared with experimental data. Druglikeness parameters of BTPT were found through in silico pharmacological ADMET properties estimation. The molecular docking investigation was performed with human topoisomerase IIα (PDB ID:1ZXM) targeting ATP binding site. In vitro cytotoxicity activity of BTPT/doxorubicin were examined by MTT assay procedure against three human cancer cell lines A549, PC-3, MDAMB-231 with IC50 values of 0.68/0.70, 1.03/0.77 and 0.88/0.98 μM, respectively. Our title compound BTPT reveals notable cytotoxicity against breast cancer cell (MDAMB-231), moderate activity with human lung cancer cell (A-549) and less inhibition with human prostate cancer cell (PC-3) compared to familiar cancer medicine doxorubicin. From the results, BTPT could be observed as a potential candidate for novel anticancer drug development process.
Collapse
Affiliation(s)
- S Murugavel
- Department of Physics, Thanthai Periyar Government Institute of Technology, Vellore, 632002, Tamil Nadu, India.
| | - C Ravikumar
- Department of Physics, Thanthai Periyar EVR Government Polytechnic College, Vellore, 632002, Tamil Nadu, India
| | - G Jaabil
- Department of Organic Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, 625021, Tamil Nadu, India
| | - Ponnuswamy Alagusundaram
- Department of Organic Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, 625021, Tamil Nadu, India
| |
Collapse
|
28
|
Morris WH, Ngo L, Wilson JT, Medawala W, Brown AR, Conner JD, Fabunmi F, Cashman DJ, Lisic EC, Yu T, Deweese JE, Jiang X. Structural and Metal Ion Effects on Human Topoisomerase IIα Inhibition by α-(N)-Heterocyclic Thiosemicarbazones. Chem Res Toxicol 2018; 32:90-99. [DOI: 10.1021/acs.chemrestox.8b00204] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- William H. Morris
- Department of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Lana Ngo
- Department of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - James T. Wilson
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204-3951, United States
| | - Wathsala Medawala
- Department of Chemistry, Georgia College, Milledgeville, Georgia 31061, United States
| | - Anthony R. Brown
- Department of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Jennifer D. Conner
- Department of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Florence Fabunmi
- Department of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Derek J. Cashman
- Department of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Edward C. Lisic
- Department of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Tao Yu
- Department of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Joseph E. Deweese
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204-3951, United States
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Xiaohua Jiang
- Department of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| |
Collapse
|
29
|
Ribeiro CJA, Kankanala J, Xie J, Williams J, Aihara H, Wang Z. Triazolopyrimidine and triazolopyridine scaffolds as TDP2 inhibitors. Bioorg Med Chem Lett 2018; 29:257-261. [PMID: 30522956 DOI: 10.1016/j.bmcl.2018.11.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/12/2018] [Accepted: 11/21/2018] [Indexed: 01/09/2023]
Abstract
Tyrosyl-DNA phosphodiesterase 2 (TDP2) repairs topoisomerase II (TOP2) mediated DNA damages and causes cellular resistance to clinically used TOP2 poisons. Inhibiting TDP2 can potentially sensitize cancer cells toward TOP2 poisons. Commercial compound P10A10, to which the structure was assigned as 7-phenyl triazolopyrimidine analogue 6a, was previously identified as a TDP2 inhibitor hit in our virtual and fluorescence-based biochemical screening campaign. We report herein that the hit validation through resynthesis and structure elucidation revealed the correct structure of P10A10 (Chembridge ID 7236827) to be the 5-phenyl triazolopyrimidine regioisomer 7a. Subsequent structure-activity relationship (SAR) via the synthesis of a total of 47 analogues of both the 5-phenyl triazolopyrimidine scaffold (7) and its bioisosteric triazolopyridine scaffold (17) identified four derivatives (7a, 17a, 17e, and 17z) with significant TDP2 inhibition (IC50 < 50 µM), with 17z showing excellent cell permeability and no cytotoxicity.
Collapse
Affiliation(s)
- Carlos J A Ribeiro
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Jayakanth Kankanala
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Jessica Williams
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States
| | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
30
|
Hu X, Wu X, Liu H, Cheng Z, Zhao Z, Xiang C, Feng X, Takeda S, Qing Y. Genistein-induced DNA damage is repaired by nonhomologous end joining and homologous recombination in TK6 cells. J Cell Physiol 2018; 234:2683-2692. [PMID: 30070703 DOI: 10.1002/jcp.27082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 06/28/2018] [Indexed: 02/05/2023]
Abstract
Genistein (GES), a phytoestrogen, has potential chemopreventive and chemotherapeutic effects on cancer. The anticancer mechanism of GES may be related with topoisomerase II associated DNA double-strand breaks (DSBs). However, the precise molecular mechanism remains elusive. Here, we performed genetic analyses using human lymphoblastoid TK6 cell lines to investigate whether non-homologous DNA end joining (NHEJ) and homologous recombination (HR), the two major repair pathways of DSBs, were involved in repairing GES-induced DNA damage. Our results showed that GES induced DSBs in TK6 cells. Cells lacking Ligase4, an NHEJ enzyme, are hypersensitive to GES. Furthermore, the sensitivity of Ligase4-/- cells was associated with enhanced DNA damage when comparing the accumulation of γ-H2AX foci and number of chromosomal aberrations (CAs) with WT cells. In addition, cells lacking Rad54, a HR enzyme, also presented hypersensitivity and increased DNA damages in response to GES. Meanwhile, Treatment of GES-lacking enhanced the accumulation of Rad51, an HR factor, in TK6 cells, especially in Ligase4-/- . These results provided direct evidence that GES induced DSBs in TK6 cells and clarified that both NHEJ and HR were involved in the repair of GES-induced DNA damage, suggesting that GES in combination with inhibition of NHEJ or HR would provide a potential anticancer strategy.
Collapse
Affiliation(s)
- Xiaoqing Hu
- State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Xiaohua Wu
- State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Hao Liu
- State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Ziyuan Cheng
- State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Zilu Zhao
- State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Cuifang Xiang
- State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyu Feng
- State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yong Qing
- State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
31
|
Indole in the target-based design of anticancer agents: A versatile scaffold with diverse mechanisms. Eur J Med Chem 2018; 150:9-29. [DOI: 10.1016/j.ejmech.2018.02.065] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/13/2018] [Accepted: 02/20/2018] [Indexed: 12/25/2022]
|
32
|
Ad’hiah AH, Al-Bederi ON, Al-Sammarrae KW. Cytotoxic effects of Agrimonia eupatoria L. against cancer cell lines in vitro. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.jaubas.2013.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Ali H. Ad’hiah
- Tropical-Biological Research Unit, College of Science, University of Baghdad, Baghdad, Iraq
| | | | | |
Collapse
|
33
|
New fluorescence-based high-throughput screening assay for small molecule inhibitors of tyrosyl-DNA phosphodiesterase 2 (TDP2). Eur J Pharm Sci 2018; 118:67-79. [PMID: 29574079 DOI: 10.1016/j.ejps.2018.03.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 01/03/2023]
Abstract
Tyrosyl-DNA phosphodiesterase 2 (TDP2) repairs topoisomerase II (TOP2) mediated DNA damages and causes resistance to TOP2-targeted cancer therapy. Inhibiting TDP2 could sensitize cancer cells toward TOP2 inhibitors. However, potent TDP2 inhibitors with favorable physicochemical properties are not yet reported. Therefore, there is a need to search for novel molecular scaffolds capable of inhibiting TDP2. We report herein a new simple, robust, homogenous mix-and-read fluorescence biochemical assay based using humanized zebrafish TDP2 (14M_zTDP2), which provides biochemical and molecular structure basis for TDP2 inhibitor discovery. The assay was validated by screening a preselected library of 1600 compounds (Z' ≥ 0.72) in a 384-well format, and by running in parallel gel-based assays with fluorescent DNA substrates. This library was curated via virtual high throughput screening (vHTS) of 460,000 compounds from Chembridge Library, using the crystal structure of the novel surrogate protein 14M_zTDP2. From this primary screening, we selected the best 32 compounds (2% of the library) to further assess their TDP2 inhibition potential, leading to the IC50 determination of 10 compounds. Based on the dose-response curve profile, pan-assay interference compounds (PAINS) structure identification, physicochemical properties and efficiency parameters, two hit compounds, 11a and 19a, were tested using a novel secondary fluorescence gel-based assay. Preliminary structure-activity relationship (SAR) studies identified guanidine derivative 12a as an improved hit with a 6.4-fold increase in potency over the original HTS hit 11a. This study highlights the importance of the development of combination approaches (biochemistry, crystallography and high throughput screening) for the discovery of TDP2 inhibitors.
Collapse
|
34
|
2-aryl benzimidazole conjugate induced apoptosis in human breast cancer MCF-7 cells through caspase independent pathway. Apoptosis 2018; 22:118-134. [PMID: 27770267 DOI: 10.1007/s10495-016-1290-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Apoptosis is a representative form of programmed cell death, which has been assumed to be critical for cancer prevention. Thus, any agent that can induce apoptosis may be useful for cancer treatment and apoptosis induction is arguably the most potent defense against cancer promotion. In our previous studies, 2-aryl benzimidazole conjugates were synthesized and evaluated for their antiproliferative activity and one of the new molecule (2f) was considered as a potential lead. This lead molecule showed significant antiproliferative activity against human breast cancer cell line, MCF-7. The results of the present study revealed that this compound arrested the cell cycle at G2/M phase. Topoisomerase II inhibition assay and Western blot analysis suggested that this compound effectively inhibits topoisomerase II activity which leads to apoptotic cell death. Apoptosis induction in MCF-7 cells was further confirmed by loss of mitochondrial membrane potential (∆Ψm), release of cytochrome c from mitochondria, an increase in the level of apoptosis inducing factor (AIF), generation of reactive oxygen species (ROS), up regulation of proapoptotic protein Bax and down regulation of anti apoptotic protein Bcl-2. Apoptosis assay using Annexin V-FITC assay also suggested that this compound induced cell death by apoptosis. However, compound 2f induced apoptosis could not be reversed by Z-VAD-FMK (a pan-caspase inhibitor) demonstrated that the 2f induced apoptosis was caspase independent. Further, 2f treatment did not activate caspase-7 and caspase-9 activity, suggesting that this compound induced apoptosis in breast cancer cells via a caspase independent pathway. Most importantly, this compound was less toxic towards non-tumorigenic breast epithelial cells, MCF-10A. Furthermore, docking studies also support the potentiality of this molecule to bind to the DNA topoisomerase II.
Collapse
|
35
|
Miyata S, Wang LY, Kitanaka S. 3EZ, 20Ac-ingenol induces cell-specific apoptosis in cyclin D1 over-expression through the activation of ATR and downregulation of p-Akt. Leuk Res 2017; 64:46-51. [PMID: 29179029 DOI: 10.1016/j.leukres.2017.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 10/19/2022]
Abstract
Acute lymphoblastic leukemia (ALL) samples exhibit an activated PI3K/Akt pathway, which suggests a general role of Akt in the development of leukemia. We have previously used western blot analysis to show that the catalytic topoisomerase (topo) inhibitor, 3EZ, 20Ac-ingenol, induced DNA damage response (DDR), which activated ATR, downregulated p-Akt through upregulation of PTEN level, and led to cell cycle arrest or apoptosis. In this study, we used ATR or PTEN siRNA and observed that the specific cell arrest and apoptosis of BALL-1 cells in DDR caused by 3EZ, 20Ac-ingenol was dependant on activation of ATR and downregulation of nuclear p-Akt through upregulation of PTEN. Moreover, some B cell lymphomas among ALLs overexpress cyclin D1. The DDR induced during the S-phase with 3EZ, 20Ac-ingenol treatment was increased by the intra S-phase checkpoint response that was triggered by the loss of nuclear cyclin D1 regulation in BALL-1 cells overexpressing cyclin D1. Although topo 1 catalytic inhibitors induce a decatenation checkpoint and subsequent G2/M phase arrest, the decatenation checkpoint caused by 3EZ, 20Ac-ingenol induced apoptosis only in the BALL-1 cells that accumulated cyclin D1.
Collapse
Affiliation(s)
- Shohei Miyata
- Department of Chemistry, College of Humanities and Sciences, Nihon University, Tokyo 156-8550, Japan.
| | - Li-Yan Wang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | | |
Collapse
|
36
|
Rjiba-Touati K, Amara I, Bousabbeh M, Salem IB, Azzebi A, Guedri Y, Achour A, Bacha H, Abid S. Recombinant human erythropoietin prevents etoposide- and methotrexate-induced toxicity in kidney and liver tissues via the regulation of oxidative damage and genotoxicity in Wistar rats. Hum Exp Toxicol 2017; 37:848-858. [PMID: 29069929 DOI: 10.1177/0960327117733553] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Etoposide (ETO) and methotrexate (MTX) are two effective chemotherapeutic drugs. However, the clinical use of these drugs is limited by its toxicity in normal tissues, especially in kidney and in liver tissues. Recombinant human erythropoietin (rhEPO), erythropoietin hormone, has also been shown to exert tissue protective effects. The purpose of this study was to explore the protective effect of rhEPO against oxidative stress and genotoxicity induced by ETO and MTX in vivo. Adult male Wistar rats were divided into 10 groups (6 animals each): control group, rhEPO alone group, ETO alone group, MTX alone group and rhEPO + ETO/MTX groups. In rhEPO + ETO/MTX groups, three doses of pretreatment with rhEPO were performed: 1000, 3000 and 6000 IU/kg. Our results showed that rhEPO pretreatment protects liver and kidney tissues against oxidative stress induced by the anticancer drugs. The glycoprotein decreased malondialdehyde (MDA) levels, reduced catalase activity and ameliorated glutathione depletion. Furthermore, we showed that rhEPO administration prevented drug-induced DNA damage accessed by comet test. Altogether, our results suggested a protective role of rhEPO, especially at 3000 IU/kg, against ETO- and MTX-induced oxidative stress and genotoxicity in vivo.
Collapse
Affiliation(s)
- K Rjiba-Touati
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - I Amara
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - M Bousabbeh
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - I Ben Salem
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - A Azzebi
- 2 Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - Y Guedri
- 2 Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - A Achour
- 2 Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - H Bacha
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - S Abid
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| |
Collapse
|
37
|
Arba M, Ihsan S, Tri Wahyudi S, Tjahjono DH. Molecular modeling of cationic porphyrin-anthraquinone hybrids as DNA topoisomerase IIβ inhibitors. Comput Biol Chem 2017; 71:129-135. [PMID: 29153891 DOI: 10.1016/j.compbiolchem.2017.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/14/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022]
Abstract
Human DNA Topoisomerase II has been regarded as a promising target in anticancer drug discovery. In the present study, we designed six porphyrin-anthraquinone hybrids bearing pyrazole or pyridine group as meso substituents and evaluated their potentials as DNA Topoisomerase IIβ inhibitor. First, we investigated the binding orientation of porphyrin hybrids into DNA topoisomerase IIβ employing AutoDock 4.2 and then performed 20-ns molecular dynamics simulations to see the dynamic stability of each porphyrin-Topo IIβ complex using Amber 14. We found that the binding of porphyrin hybrids occured through intercalation and groove binding mode in addition interaction with the amino acid residues constituting the active cavity of Topo IIβ. Each porphyrin-Topo IIβ complex was stabilized during 20-ns dynamics simulations. The MM-PBSA free energy calculation shows that the binding affinities of porphyrin hybrids were modified with the number of meso substituent. Interestingly, the affinity of all porphyrin hybrids to Topo IIβ was stronger than that of native ligand (EVP), indicating the potential of the designed porphyrin to be considered in experimental research.
Collapse
Affiliation(s)
- Muhammad Arba
- Faculty of Pharmacy, Halu Oleo University, Kendari, 93231, Indonesia.
| | - Sunandar Ihsan
- Faculty of Pharmacy, Halu Oleo University, Kendari, 93231, Indonesia
| | | | - Daryono H Tjahjono
- School of Pharmacy, Bandung Institute of Technology, Bandung, 40312, Indonesia
| |
Collapse
|
38
|
Novel xanthone-polyamine conjugates as catalytic inhibitors of human topoisomerase IIα. Bioorg Med Chem Lett 2017; 27:4687-4693. [PMID: 28919339 DOI: 10.1016/j.bmcl.2017.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 08/31/2017] [Accepted: 09/04/2017] [Indexed: 01/03/2023]
Abstract
It has been proposed that xanthone derivatives with anticancer potential act as topoisomerase II inhibitors because they interfere with the ability of the enzyme to bind its ATP cofactor. In order to further characterize xanthone mechanism and generate compounds with potential as anticancer drugs, we synthesized a series of derivatives in which position 3 was substituted with different polyamine chains. As determined by DNA relaxation and decatenation assays, the resulting compounds are potent topoisomerase IIα inhibitors. Although xanthone derivatives inhibit topoisomerase IIα-catalyzed ATP hydrolysis, mechanistic studies indicate that they do not act at the ATPase site. Rather, they appear to function by blocking the ability of DNA to stimulate ATP hydrolysis. On the basis of activity, competition, and modeling studies, we propose that xanthones interact with the DNA cleavage/ligation active site of topoisomerase IIα and inhibit the catalytic activity of the enzyme by interfering with the DNA strand passage step.
Collapse
|
39
|
Jamieson GC, Fox JA, Poi M, Strickland SA. Molecular and Pharmacologic Properties of the Anticancer Quinolone Derivative Vosaroxin: A New Therapeutic Agent for Acute Myeloid Leukemia. Drugs 2017; 76:1245-1255. [PMID: 27484675 PMCID: PMC4989016 DOI: 10.1007/s40265-016-0614-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Vosaroxin is a first-in-class anticancer quinolone derivative that targets topoisomerase II and induces site-selective double-strand breaks in DNA, leading to tumor cell apoptosis. Vosaroxin has chemical and pharmacologic characteristics distinct from other topoisomerase II inhibitors due to its quinolone scaffold. The efficacy and safety of vosaroxin in combination with cytarabine were evaluated in patients with relapsed/refractory acute myeloid leukemia (AML) in a phase III, randomized, multicenter, double-blind, placebo-controlled study (VALOR). In this study, the addition of vosaroxin produced a 1.4-month improvement in median overall survival (OS; 7.5 months with vosaroxin/cytarabine vs. 6.1 months with placebo/cytarabine; hazard ratio [HR] 0.87, 95 % confidence interval [CI] 0.73−1.02; unstratified log-rank p\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=$$\end{document}= 0.061; stratified log-rank p\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=$$\end{document}=0.024), with the greatest OS benefit observed in patients ≥60 years of age (7.1 vs. 5.0 months; HR 0.75, 95 % CI 0.62−0.92; p\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=$$\end{document}=0.003) and patients with early relapse (6.7 vs. 5.2 months; HR 0.77, 95 % CI 0.59−1.00; p\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=$$\end{document}= 0.039), two AML patient groups that typically have poor prognosis. Here we review the chemical and pharmacologic properties of vosaroxin, how these properties are distinct from those of currently available topoisomerase II inhibitors, how they may contribute to the efficacy and safety profile observed in the VALOR trial, and the status of clinical development of vosaroxin for treatment of AML.
Collapse
Affiliation(s)
| | - Judith A Fox
- Sunesis Pharmaceuticals, Inc., South San Francisco, CA, USA
| | - Ming Poi
- College of Pharmacy, Ohio State University, Columbus, OH, USA
| | - Stephen A Strickland
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN, 37232, USA.
| |
Collapse
|
40
|
Paubelle E, Zylbersztejn F, Thomas X. The preclinical discovery of vosaroxin for the treatment of acute myeloid leukemia. Expert Opin Drug Discov 2017; 12:747-753. [PMID: 28504025 DOI: 10.1080/17460441.2017.1331215] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) represents a disease with a very poor outcome and remains an area of significant unmet need necessitating novel therapeutic strategies. Among novel therapeutic agents, vosaroxin is a first-in-class anticancer quinolone derivative that targets topoisomerase II and induces site-selective double-strand breaks in DNA, leading to tumor cell apoptosis. Areas covered: Herein, the authors provide a comprehensive review of the preclinical development of vosaroxin. This includes coverage of vosaroxin's mechanism of action in addition to its pharmacology and of the main studies reported over the past few years with vosaroxin when used to treat adult AML. Expert opinion: Given that vosaroxin is associated with fewer potential side effects, it may be of benefit to elderly patients with relapsed/refractory AML and to those with additional comorbidities who have previously received an anthracycline and cytarabine combination. Furthermore, vosaroxin also was seen to be active in multidrug-resistant preclinical models. However, further studies have to be performed to better evaluate its place in the armamentarium against AML.
Collapse
Affiliation(s)
- Etienne Paubelle
- a Hospices Civils de Lyon, Hematology Department , Lyon-Sud Hospital , Pierre-Bénite , France
| | | | - Xavier Thomas
- a Hospices Civils de Lyon, Hematology Department , Lyon-Sud Hospital , Pierre-Bénite , France
| |
Collapse
|
41
|
Cuya SM, Bjornsti MA, van Waardenburg RCAM. DNA topoisomerase-targeting chemotherapeutics: what's new? Cancer Chemother Pharmacol 2017; 80:1-14. [PMID: 28528358 DOI: 10.1007/s00280-017-3334-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 05/03/2017] [Indexed: 02/05/2023]
Abstract
To resolve the topological problems that threaten the function and structural integrity of nuclear and mitochondrial genomes and RNA molecules, human cells encode six different DNA topoisomerases including type IB enzymes (TOP1 and TOP1mt), type IIA enzymes (TOP2α and TOP2β) and type IA enzymes (TOP3α and TOP3β). DNA entanglements and the supercoiling of DNA molecules are regulated by topoisomerases through the introduction of transient enzyme-linked DNA breaks. The covalent topoisomerase-DNA complexes are the cellular targets of a diverse group of cancer chemotherapeutics, which reversibly stabilize these reaction intermediates. Here we review the structure-function and catalytic mechanisms of each family of eukaryotic DNA topoisomerases and the topoisomerase-targeting agents currently approved for patient therapy or in clinical trials, and highlight novel developments and challenges in the clinical development of these agents.
Collapse
Affiliation(s)
- Selma M Cuya
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 155 Volker Hall, 1720 2nd Ave. S., Birmingham, AL, 35294-0019, USA
| | - Mary-Ann Bjornsti
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 155 Volker Hall, 1720 2nd Ave. S., Birmingham, AL, 35294-0019, USA
| | - Robert C A M van Waardenburg
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 155 Volker Hall, 1720 2nd Ave. S., Birmingham, AL, 35294-0019, USA.
| |
Collapse
|
42
|
Topoisomerase II Inhibitors Induce DNA Damage-Dependent Interferon Responses Circumventing Ebola Virus Immune Evasion. mBio 2017; 8:mBio.00368-17. [PMID: 28377530 PMCID: PMC5380843 DOI: 10.1128/mbio.00368-17] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ebola virus (EBOV) protein VP35 inhibits production of interferon alpha/beta (IFN) by blocking RIG-I-like receptor signaling pathways, thereby promoting virus replication and pathogenesis. A high-throughput screening assay, developed to identify compounds that either inhibit or bypass VP35 IFN-antagonist function, identified five DNA intercalators as reproducible hits from a library of bioactive compounds. Four, including doxorubicin and daunorubicin, are anthracycline antibiotics that inhibit topoisomerase II and are used clinically as chemotherapeutic drugs. These compounds were demonstrated to induce IFN responses in an ATM kinase-dependent manner and to also trigger the DNA-sensing cGAS-STING pathway of IFN induction. These compounds also suppress EBOV replication in vitro and induce IFN in the presence of IFN-antagonist proteins from multiple negative-sense RNA viruses. These findings provide new insights into signaling pathways activated by important chemotherapy drugs and identify a novel therapeutic approach for IFN induction that may be exploited to inhibit RNA virus replication. Ebola virus and other emerging RNA viruses are significant but unpredictable public health threats. Therapeutic approaches with broad-spectrum activity could provide an attractive response to such infections. We describe a novel assay that can identify small molecules that overcome Ebola virus-encoded innate immune evasion mechanisms. This assay identified as hits cancer chemotherapeutic drugs, including doxorubicin. Follow-up studies provide new insight into how doxorubicin induces interferon (IFN) responses, revealing activation of both the DNA damage response kinase ATM and the DNA sensor cGAS and its partner signaling protein STING. The studies further demonstrate that the ATM and cGAS-STING pathways of IFN induction are a point of vulnerability not only for Ebola virus but for other RNA viruses as well, because viral innate immune antagonists consistently fail to block these signals. These studies thereby define a novel avenue for therapeutic intervention against emerging RNA viruses.
Collapse
|
43
|
Meier C, Steinhauer TN, Koczian F, Plitzko B, Jarolim K, Girreser U, Braig S, Marko D, Vollmar AM, Clement B. A Dual Topoisomerase Inhibitor of Intense Pro-Apoptotic and Antileukemic Nature for Cancer Treatment. ChemMedChem 2017; 12:347-352. [DOI: 10.1002/cmdc.201700026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Christopher Meier
- Department of Pharmaceutical and Medicinal Chemistry; Pharmaceutical Institute of the Christian Albrechts University in Kiel; Gutenbergstraße 76 24118 Kiel Germany
| | - Tamara N. Steinhauer
- Department of Pharmaceutical and Medicinal Chemistry; Pharmaceutical Institute of the Christian Albrechts University in Kiel; Gutenbergstraße 76 24118 Kiel Germany
| | - Fabian Koczian
- Department of Pharmacy, Center for Drug Research; Pharmaceutical Biology; University of Munich; Butenandtstraße 5-13 81377 Munich Germany
| | - Birte Plitzko
- Department of Pharmaceutical and Medicinal Chemistry; Pharmaceutical Institute of the Christian Albrechts University in Kiel; Gutenbergstraße 76 24118 Kiel Germany
| | - Katharina Jarolim
- Department of Food Chemistry and Toxicology; University of Vienna; Währinger Straße 38 1090 Vienna Austria
| | - Ulrich Girreser
- Department of Pharmaceutical and Medicinal Chemistry; Pharmaceutical Institute of the Christian Albrechts University in Kiel; Gutenbergstraße 76 24118 Kiel Germany
| | - Simone Braig
- Department of Pharmacy, Center for Drug Research; Pharmaceutical Biology; University of Munich; Butenandtstraße 5-13 81377 Munich Germany
| | - Doris Marko
- Department of Food Chemistry and Toxicology; University of Vienna; Währinger Straße 38 1090 Vienna Austria
| | - Angelika M. Vollmar
- Department of Pharmacy, Center for Drug Research; Pharmaceutical Biology; University of Munich; Butenandtstraße 5-13 81377 Munich Germany
| | - Bernd Clement
- Department of Pharmaceutical and Medicinal Chemistry; Pharmaceutical Institute of the Christian Albrechts University in Kiel; Gutenbergstraße 76 24118 Kiel Germany
| |
Collapse
|
44
|
Statins in anthracycline-induced cardiotoxicity: Rac and Rho, and the heartbreakers. Cell Death Dis 2017; 8:e2564. [PMID: 28102848 PMCID: PMC5386353 DOI: 10.1038/cddis.2016.418] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/02/2016] [Indexed: 01/06/2023]
Abstract
Cancer patients receiving anthracycline-based chemotherapy are at risk to develop life-threatening chronic cardiotoxicity with the pathophysiological mechanism of action not fully understood. Besides the most common hypothesis that anthracycline-induced congestive heart failure (CHF) is mainly caused by generation of reactive oxygen species, recent data point to a critical role of topoisomerase II beta (TOP2B), which is a primary target of anthracycline poisoning, in the pathophysiology of CHF. As the use of the only clinically approved cardioprotectant dexrazoxane has been limited by the FDA in 2011, there is an urgent need for alternative cardioprotective measures. Statins are anti-inflammatory and anti-oxidative drugs that are clinically well established for the prevention of cardiovascular diseases. They exhibit pleiotropic beneficial properties beyond cholesterol-lowering effects that most likely rest on the indirect inhibition of small Ras homologous (Rho) GTPases. The Rho GTPase Rac1 has been shown to be a major factor in the regulation of the pro-oxidative NADPH oxidase as well as in the regulation of type II topoisomerase. Both are discussed to play an important role in the pathophysiology of anthracycline-induced CHF. Therefore, off-label use of statins or novel Rac1 inhibitors might represent a promising pharmacological approach to gain control over chronic cardiotoxicity by interfering with key mechanisms of anthracycline-induced cardiomyocyte cell death.
Collapse
|
45
|
Ranganathan P, Kashyap T, Yu X, Meng X, Lai TH, McNeil B, Bhatnagar B, Shacham S, Kauffman M, Dorrance AM, Blum W, Sampath D, Landesman Y, Garzon R. XPO1 Inhibition using Selinexor Synergizes with Chemotherapy in Acute Myeloid Leukemia by Targeting DNA Repair and Restoring Topoisomerase IIα to the Nucleus. Clin Cancer Res 2016; 22:6142-6152. [PMID: 27358488 PMCID: PMC5161584 DOI: 10.1158/1078-0432.ccr-15-2885] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 05/19/2016] [Accepted: 06/20/2016] [Indexed: 12/21/2022]
Abstract
PURPOSE Selinexor, a selective inhibitor of XPO1, is currently being tested as single agent in clinical trials in acute myeloid leukemia (AML). However, considering the molecular complexity of AML, it is unlikely that AML can be cured with monotherapy. Therefore, we asked whether adding already established effective drugs such as topoisomerase (Topo) II inhibitors to selinexor will enhance its anti-leukemic effects in AML. EXPERIMENTAL DESIGN The efficacy of combinatorial drug treatment using Topo II inhibitors (idarubicin, daunorubicin, mitoxantrone, etoposide) and selinexor was evaluated in established cellular and animal models of AML. RESULTS Concomitant treatment with selinexor and Topo II inhibitors resulted in therapeutic synergy in AML cell lines and patient samples. Using a xenograft MV4-11 AML mouse model, we show that treatment with selinexor and idarubicin significantly prolongs survival of leukemic mice compared with each single therapy. CONCLUSIONS Aberrant nuclear export and cytoplasmic localization of Topo IIα has been identified as one of the mechanisms leading to drug resistance in cancer. Here, we show that in a subset of patients with AML that express cytoplasmic Topo IIα, selinexor treatment results in nuclear retention of Topo IIα protein, resulting in increased sensitivity to idarubicin. Selinexor treatment of AML cells resulted in a c-MYC-dependent reduction of DNA damage repair genes (Rad51 and Chk1) mRNA and protein expression and subsequent inhibition of homologous recombination repair and increased sensitivity to Topo II inhibitors. The preclinical data reported here support further clinical studies using selinexor and Topo II inhibitors in combination to treat AML. Clin Cancer Res; 22(24); 6142-52. ©2016 AACR.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Animals
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Nucleus/drug effects
- DNA Damage/drug effects
- DNA Repair/drug effects
- DNA Topoisomerases, Type II/metabolism
- Drug Resistance, Neoplasm/drug effects
- Female
- Humans
- Hydrazines/pharmacology
- Karyopherins/antagonists & inhibitors
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Mice, SCID
- Proto-Oncogene Proteins c-myc/metabolism
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Topoisomerase II Inhibitors/pharmacology
- Triazoles/pharmacology
- Exportin 1 Protein
Collapse
Affiliation(s)
| | | | - Xueyan Yu
- The Ohio State University, Columbus, Ohio
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zhao W, Jiang G, Bi C, Li Y, Liu J, Ye C, He H, Li L, Song D, Shao R. The dual topoisomerase inhibitor A35 preferentially and specially targets topoisomerase 2α by enhancing pre-strand and post-strand cleavage and inhibiting DNA religation. Oncotarget 2016; 6:37871-94. [PMID: 26462155 PMCID: PMC4741971 DOI: 10.18632/oncotarget.5680] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 09/25/2015] [Indexed: 01/14/2023] Open
Abstract
DNA topoisomerases play a key role in tumor proliferation. Chemotherapeutics targeting topoisomerases have been widely used in clinical oncology, but resistance and side effects, particularly cardiotoxicity, usually limit their application. Clinical data show that a decrease in topoisomerase (top) levels is the primary factor responsible for resistance, but in cells there is compensatory effect between the levels of top1 and top2α. Here, we validated cyclizing-berberine A35, which is a dual top inhibitor and preferentially targets top2α. The impact on the top2α catalytic cycle indicated that A35 could intercalate into DNA but did not interfere with DNA-top binding and top2α ATPase activity. A35 could facilitate DNA-top2α cleavage complex formation by enhancing pre-strand and post-strand cleavage and inhibiting religation, suggesting this compound can be a topoisomerase poison and had a district mechanism from other topoisomerase inhibitors. TARDIS and comet assays showed that A35 could induce cell DNA breakage and DNA-top complexes but had no effect on the cardiac toxicity inducer top2β. Silencing top1 augmented DNA break and silencing top2α decreased DNA break. Further validation in H9c2 cardiac cells showed A35 did not disturb cell proliferation and mitochondrial membrane potency. Additionally, an assay with nude mice further demonstrated A35 did not damage the heart. Our work identifies A35 as a novel skeleton compound dually inhibits topoisomerases, and predominantly and specially targets top2α by interfering with all cleavage steps and its no cardiac toxicity was verified by cardiac cells and mice heart. A35 could be a novel and effective targeting topoisomerase agent.
Collapse
Affiliation(s)
- Wuli Zhao
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Guohua Jiang
- Analysis and Testing Center, Beijing Normal University, Beijing, China
| | - Chongwen Bi
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yangbiao Li
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jingbo Liu
- China Meitan General Hospital, Beijing, China
| | - Cheng Ye
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hongwei He
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Liang Li
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Danqing Song
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Rongguang Shao
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
47
|
Stefansdottir A, Johnston ZC, Powles-Glover N, Anderson RA, Adams IR, Spears N. Etoposide damages female germ cells in the developing ovary. BMC Cancer 2016; 16:482. [PMID: 27510889 PMCID: PMC4980800 DOI: 10.1186/s12885-016-2505-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 07/05/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND As with many anti-cancer drugs, the topoisomerase II inhibitor etoposide is considered safe for administration to women in the second and third trimesters of pregnancy, but assessment of effects on the developing fetus have been limited. The purpose of this research was to examine the effect of etoposide on germ cells in the developing ovary. Mouse ovary tissue culture was used as the experimental model, thus allowing us to examine effects of etoposide on all stages of germ cell development in the same way, in vitro. RESULTS Fetal ovaries from embryonic day 13.5 CD1 mice or neonatal ovaries from postnatal day 0 CD1 mice were cultured with 50-150 ng ml(-1) or 50-200 ng ml(-1) etoposide respectively, concentrations that are low relative to that in patient serum. When fetal ovaries were treated prior to follicle formation, etoposide resulted in dose-dependent damage, with 150 ng ml(-1) inducing a near-complete absence of healthy follicles. In contrast, treatment of neonatal ovaries, after follicle formation, had no effect on follicle numbers and only a minor effect on follicle health, even at 200 ng ml(-1). The sensitivity of female germ cells to etoposide coincided with topoisomerase IIα expression: in the developing ovary of both mouse and human, topoisomerase IIα was expressed in germ cells only prior to follicle formation. CONCLUSIONS Exposure of pre-follicular ovaries, in which topoisomerase IIα expression was germ cell-specific, resulted in a near-complete elimination of germ cells prior to follicle formation, with the remaining germ cells going on to form unhealthy follicles by the end of culture. In contrast, exposure to follicle-enclosed oocytes, which no longer expressed topoisomerase IIα in the germ cells, had no effect on total follicle numbers or health, the only effect seen specific to transitional follicles. Results indicate the potential for adverse effects on fetal ovarian development if etoposide is administered to pregnant women when germ cells are not yet enclosed within ovarian follicles, a process that starts at approximately 17 weeks gestation and is only complete towards the end of pregnancy.
Collapse
Affiliation(s)
- Agnes Stefansdottir
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Zoe C. Johnston
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD UK
- Present Address: Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, G61 1QH UK
| | | | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ UK
| | - Ian R. Adams
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Norah Spears
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD UK
| |
Collapse
|
48
|
A Chromatin-Focused siRNA Screen for Regulators of p53-Dependent Transcription. G3-GENES GENOMES GENETICS 2016; 6:2671-8. [PMID: 27334938 PMCID: PMC4978920 DOI: 10.1534/g3.116.031534] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The protein product of the Homo sapiens TP53 gene is a transcription factor (p53) that regulates the expression of genes critical for the response to DNA damage and tumor suppression, including genes involved in cell cycle arrest, apoptosis, DNA repair, metabolism, and a number of other tumorigenesis-related pathways. Differential transcriptional regulation of these genes is believed to alter the balance between two p53-dependent cell fates: cell cycle arrest or apoptosis. A number of previously identified p53 cofactors covalently modify and alter the function of both the p53 protein and histone proteins. Both gain- and loss-of-function mutations in chromatin modifiers have been strongly implicated in cancer development; thus, we sought to identify novel chromatin regulatory proteins that affect p53-dependent transcription and the balance between the expression of pro-cell cycle arrest and proapoptotic genes. We utilized an siRNA library designed against predicted chromatin regulatory proteins, and identified known and novel chromatin-related factors that affect both global p53-dependent transcription and gene-specific regulators of p53 transcriptional activation. The results from this screen will serve as a comprehensive resource for those interested in further characterizing chromatin and epigenetic factors that regulate p53 transcription.
Collapse
|
49
|
Park S, Hong E, Kwak SY, Jun KY, Lee ES, Kwon Y, Na Y. Synthesis and biological evaluation of C1-O-substituted-3-(3-butylamino-2-hydroxy-propoxy)-xanthen-9-one as topoisomerase IIα catalytic inhibitors. Eur J Med Chem 2016; 123:211-225. [PMID: 27484510 DOI: 10.1016/j.ejmech.2016.07.046] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/06/2016] [Accepted: 07/20/2016] [Indexed: 11/30/2022]
Abstract
Topoisomerase II poison blocks the transitorily generated DNA double-strand breaks (DSBs) from religation, thereby causes severe DNA damage and gene toxicity. While topoisomerase II catalytic inhibitor does not form cleavable DNA-enzyme complex because its function attributes to inhibition of the catalytic steps of the enzyme such as before generating DNA DSBs or in the last step of the catalytic cycle after religation. It has been reported that the stabilizing effect of etoposide on transient cleavable DNA-topoisomerase IIβ complex attributes to its secondary malignancy. Therefore, topoisomerase IIα has been considered as more attractive target than topoisomerase IIβ for the development of chemotherapeutic agents. In the previous work, we reported compounds I and II as novel topoisomerase IIα catalytic inhibitors targeting for ATP binding site of human topoisomerase IIα ATP-binding domain. As a continuous work, we have designed and synthesized 43 compounds of C1-O-alkyl and arylalkyl substitiuted compounds with or without methoxy group on ring A. In the topoisomerase IIα inhibitory test, among the tested C1-O-4-chlorophenethyl substituted compounds 37 and 47 were more active than others, and compound 37 showed strongest topoisomerase IIα inhibitory activity with 94.4% and 23.0% inhibition, respectively, at 100 and 20 μM. Compounds 37 and 47 have also showed much enhanced cytotoxic activity against T47D cells; IC50 (μM): 0.63 ± 0.01 and 0.19 ± 0.02, respectively, which are stronger than reference drugs. Band depletion assay and cleavage complex assay results showed compounds 37 and 47 were potential topoisomerase IIα catalytic inhibitor with low DNA damage.
Collapse
Affiliation(s)
- Seojeong Park
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea
| | - Eunji Hong
- College of Pharmacy, CHA University, Pocheon, 487-010, South Korea
| | - Soo Yeon Kwak
- College of Pharmacy, CHA University, Pocheon, 487-010, South Korea
| | - Kyu-Yeon Jun
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea
| | - Eung-Seok Lee
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, South Korea
| | - Youngjoo Kwon
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea.
| | - Younghwa Na
- College of Pharmacy, CHA University, Pocheon, 487-010, South Korea.
| |
Collapse
|
50
|
Liao G, Chen X, Wu J, Qian C, Wang Y, Ji L, Chao H. Ruthenium(II) polypyridyl complexes as dual inhibitors of telomerase and topoisomerase. Dalton Trans 2016; 44:15145-56. [PMID: 25604798 DOI: 10.1039/c4dt03585b] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
One novel ruthenium polypyridyl complex, [Ru(bpy)2(icip)](2+) (1), and two previously reported ruthenium polypyridyl complexes, [Ru(bpy)2(pdppz)](2+) ()2 and [Ru(bpy)2(tactp)](2+) (3) (bpy = 2,2'-bipyridine, icip = 2-(indeno[2,1-b]chromen-6-yl)-1H-imidazo[4,5-f][1,10]phenanthroline, pdppz = phenanthro[4,5-abc]dipyrido[3,2-h:2',3'-j]phenazine, tactp = 4,5,9,18-tetraazachryseno[9,10-b]-triphenylene), have been synthesised. As expected, these complexes show inhibition towards telomerase by inducing and stabilising the G-quadruplex structure, and behave as topoisomerase I/II poisons at the same time. Additionally, the acute and chronic cytotoxicities of the complexes are considered. Furthermore, cell apoptosis experiments are used to briefly study the mechanism. Because studies involving multi-target inhibition towards topoisomerase and telomerase of Ru(II) complexes have not been reported previously, the present research may help to develop innovative chemical strategies and therapies.
Collapse
Affiliation(s)
- Guoliang Liao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275, P. R. China.
| | | | | | | | | | | | | |
Collapse
|