1
|
Ogbodo UC, Enejoh OA, Okonkwo CH, Gnanasekar P, Gachanja PW, Osata S, Atanda HC, Iwuchukwu EA, Achilonu I, Awe OI. Computational identification of potential inhibitors targeting cdk1 in colorectal cancer. Front Chem 2023; 11:1264808. [PMID: 38099190 PMCID: PMC10720044 DOI: 10.3389/fchem.2023.1264808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction: Despite improved treatment options, colorectal cancer (CRC) remains a huge public health concern with a significant impact on affected individuals. Cell cycle dysregulation and overexpression of certain regulators and checkpoint activators are important recurring events in the progression of cancer. Cyclin-dependent kinase 1 (CDK1), a key regulator of the cell cycle component central to the uncontrolled proliferation of malignant cells, has been reportedly implicated in CRC. This study aimed to identify CDK1 inhibitors with potential for clinical drug research in CRC. Methods: Ten thousand (10,000) naturally occurring compounds were evaluated for their inhibitory efficacies against CDK1 through molecular docking studies. The stability of the lead compounds in complex with CDK1 was evaluated using molecular dynamics simulation for one thousand (1,000) nanoseconds. The top-scoring candidates' ADME characteristics and drug-likeness were profiled using SwissADME. Results: Four hit compounds, namely, spiraeoside, robinetin, 6-hydroxyluteolin, and quercetagetin were identified from molecular docking analysis to possess the least binding scores. Molecular dynamics simulation revealed that robinetin and 6-hydroxyluteolin complexes were stable within the binding pocket of the CDK1 protein. Discussion: The findings from this study provide insight into novel candidates with specific inhibitory CDK1 activities that can be further investigated through animal testing, clinical trials, and drug development research for CRC treatment.
Collapse
Affiliation(s)
| | - Ojochenemi A. Enejoh
- Genomics and Bioinformatics Department, National Biotechnology Development Agency, Abuja, Nigeria
| | - Chinelo H. Okonkwo
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
| | | | - Pauline W. Gachanja
- Department of Biochemistry and Biotechnology, Pwani University, Kilifi, Kenya
| | - Shamim Osata
- Department of Biochemistry, University of Nairobi, Nairobi, Kenya
| | - Halimat C. Atanda
- Biotechnology Department, Federal University of Technology, Akure, Nigeria
| | - Emmanuel A. Iwuchukwu
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, Faculty of Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Ikechukwu Achilonu
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, Faculty of Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Olaitan I. Awe
- Department of Computer Science, University of Ibadan, Ibadan, Nigeria
- African Society for Bioinformatics and Computational Biology, Cape Town, South Africa
| |
Collapse
|
2
|
Bono A, La Monica G, Alamia F, Mingoia F, Gentile C, Peri D, Lauria A, Martorana A. In Silico Mixed Ligand/Structure-Based Design of New CDK-1/PARP-1 Dual Inhibitors as Anti-Breast Cancer Agents. Int J Mol Sci 2023; 24:13769. [PMID: 37762072 PMCID: PMC10531453 DOI: 10.3390/ijms241813769] [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: 08/12/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
CDK-1 and PARP-1 play crucial roles in breast cancer progression. Compounds acting as CDK-1 and/or PARP-1 inhibitors can induct cell death in breast cancer with a selective synthetic lethality mechanism. A mixed treatment by means of CDK-1 and PARP-1 inhibitors resulted in radical breast cancer cell growth reduction. Inhibitors with a dual target mechanism of action could arrest cancer progression by simultaneously blocking the DNA repair mechanism and cell cycle, resulting in advantageous monotherapy. To this aim, in the present work, we identified compound 645656 with a significant affinity for both CDK-1 and PARP-1 by a mixed ligand- and structure-based virtual screening protocol. The Biotarget Predictor Tool was used at first in a Multitarget mode to filter the large National Cancer Institute (NCI) database. Then, hierarchical docking studies were performed to further screen the compounds and evaluate the ligands binding mode, whose putative dual-target mechanism of action was investigated through the correlation between the antiproliferative activity data and the target proteins' (CDK-1 and PARP-1) expression pattern. Finally, a Molecular Dynamics Simulation confirmed the high stability of the most effective selected compound 645656 in complex with both PARP-1 and CDK-1.
Collapse
Affiliation(s)
- Alessia Bono
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche “STEBICEF”, University of Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy; (A.B.); (G.L.M.); (F.A.); (C.G.); (A.M.)
| | - Gabriele La Monica
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche “STEBICEF”, University of Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy; (A.B.); (G.L.M.); (F.A.); (C.G.); (A.M.)
| | - Federica Alamia
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche “STEBICEF”, University of Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy; (A.B.); (G.L.M.); (F.A.); (C.G.); (A.M.)
| | - Francesco Mingoia
- Istituto per lo Studio dei Materiali Nanostrutturati (ISMN), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy;
| | - Carla Gentile
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche “STEBICEF”, University of Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy; (A.B.); (G.L.M.); (F.A.); (C.G.); (A.M.)
| | - Daniele Peri
- Dipartimento di Ingegneria dell’Innovazione Industriale e Digitale, Università degli Studi di Palermo, Viale 10 delle Scienze Ed. 6, 90128 Palermo, Italy;
| | - Antonino Lauria
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche “STEBICEF”, University of Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy; (A.B.); (G.L.M.); (F.A.); (C.G.); (A.M.)
| | - Annamaria Martorana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche “STEBICEF”, University of Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy; (A.B.); (G.L.M.); (F.A.); (C.G.); (A.M.)
| |
Collapse
|
3
|
Mandal D, Patel P, Verma SK, Sahu BR, Parija T. Proximal discrepancy in intrinsic atomic interaction arrests G2/M phase by inhibiting Cyclin B1/CDK1 to infer molecular and cellular biocompatibility of D-limonene. Sci Rep 2022; 12:18184. [PMID: 36307489 PMCID: PMC9616896 DOI: 10.1038/s41598-022-21364-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 09/26/2022] [Indexed: 12/31/2022] Open
Abstract
The quest for different natural compounds for different biomedical applications especially in the treatment of cancer is at a high pace with increasing incidence of severity. D-limonene has been portrayed as one of the effective potential candidate centered to the context of breast cancer. The anticipation of its count as an effective biomedical agent required a detailed understanding of their molecular mechanism of biocompatibility. This study elucidates the mechanistic action of D-limonene channelized by the induction of apoptosis for controlling proliferation in breast cancer cells. The possible mechanism was explored through an experimental and computational approach to estimate cell proliferation inhibition, cell cycle phase distribution, apoptosis analysis using a flow cytometry, western blotting and molecular docking. The results showed reduced dose and time-dependent viability of MCF7 cells. The study suggested the arrest of the cell cycle at G2/M phase leading to apoptosis and other discrepancies of molecular activity mediated via significant alteration in protein expression pattern of anti-apoptotic proteins like Cyclin B1 and CDK1. Computational analysis showed firm interaction of D-limonene with Cyclin B1 and CDK1 proteins influencing their structural and functional integrity indicating the mediation of mechanism. This study concluded that D-limonene suppresses the proliferation of breast cancer cells by inducing G2/M phase arrest via deregulation of Cyclin B1/CDK1.
Collapse
Affiliation(s)
- Deepa Mandal
- grid.412122.60000 0004 1808 2016School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | - Paritosh Patel
- grid.412122.60000 0004 1808 2016School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | - Suresh K. Verma
- grid.412122.60000 0004 1808 2016School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | - Bikash Ranjan Sahu
- grid.412122.60000 0004 1808 2016School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | - Tithi Parija
- grid.412122.60000 0004 1808 2016School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| |
Collapse
|
4
|
Yan Q, Zhang B, Ling X, Zhu B, Mei S, Yang H, Zhang D, Huo J, Zhao Z. CTLA-4 Facilitates DNA Damage–Induced Apoptosis by Interacting With PP2A. Front Cell Dev Biol 2022; 10:728771. [PMID: 35281086 PMCID: PMC8907142 DOI: 10.3389/fcell.2022.728771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
Cytotoxic T-lymphocyte–associated protein 4 (CTLA-4) plays a pivotal role in regulating immune responses. It accumulates in intracellular compartments, translocates to the cell surface, and is rapidly internalized. However, the cytoplasmic function of CTLA-4 remains largely unknown. Here, we describe the role of CTLA-4 as an immunomodulator in the DNA damage response to genotoxic stress. Using isogenic models of murine T cells with either sufficient or deficient CTLA-4 expression and performing a variety of assays, including cell apoptosis, cell cycle, comet, western blotting, co-immunoprecipitation, and immunofluorescence staining analyses, we show that CTLA-4 activates ataxia–telangiectasia mutated (ATM) by binding to the ATM inhibitor protein phosphatase 2A into the cytoplasm of T cells following transient treatment with zeocin, exacerbating the DNA damage response and inducing apoptosis. These findings provide new insights into how T cells maintain their immune function under high-stress conditions, which is clinically important for patients with tumors undergoing immunotherapy combined with chemoradiotherapy.
Collapse
Affiliation(s)
- Qiongyu Yan
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xi Ling
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shenghui Mei
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hua Yang
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Dongjie Zhang
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiping Huo
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhigang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Zhigang Zhao,
| |
Collapse
|
5
|
Hu M, Varier KM, Li Z, Qin X, Rao Q, Song J, Hu A, Hang Y, Yuan C, Gajendran B, Shu L, Wen M, Li Y, Liu H. A natural acylphloroglucinol triggered antiproliferative possessions in HEL cells by impeding STAT3 signaling and attenuating angiogenesis in transgenic zebrafish model. Biomed Pharmacother 2021; 141:111877. [PMID: 34323693 DOI: 10.1016/j.biopha.2021.111877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/07/2021] [Accepted: 06/28/2021] [Indexed: 01/20/2023] Open
Abstract
Leukemia is responsible for a reason of death, globally. Even though there are several treatment regimens available in the clinics against this disease, a perfect chemotherapeutic agent for the same is still under investigation. Natural plant-derived secondary metabolites are used in clinics to treat leukemia for better benefits with reduced side-effects. Likely, several bioactive compounds from Callistemon sp. were reported for their bioactive benefits. Furthermore, acylphloroglucinol derivatives from Callistemon salignus, showed both antimicrobial and cytotoxic activities in various adherent human cancer cell lines. Thus, in the present study, a natural acylphloroglucinol (2,6-dihydroxy-4-methoxyisobutyrophenone, L72) was tested for its antiproliferative efficacy in HEL cells. The MTT and the cell cycle analysis study revealed that L72 treatment can offer antiproliferative effects, both time and dose-dependent manner, causing G2/M cell cycle arrest. The western blot analysis revealed that L72 treatment triggered intrinsic apoptotic machinery and activated p21. Likewise, L72 could downregulate the gene expressions of XIAP, FLT3, IDH2, and SOD2, which was demonstrated by qPCR analysis, thus promoting its antiproliferative action. The L72 could impede STAT3 expression, which was evidenced by insilico autodock analysis and western blot analysis using STAT3 inhibitor, Pimozide. The treatment of transgenic (Flk-1+/egfr+) zebrafish embryos resulted in the STAT3 gene inhibition, proving its anti-angiogenic effect, as well. Thus, the study revealed that L72 could act as an antiproliferative agent, by triggering caspase-dependent intrinsic apoptosis, reducing cell proliferation by attenuating STAT3, and activating an anti-angiogenic pathway via Flk-1inhibition.
Collapse
Affiliation(s)
- Mi Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China; Key Laboratory of Regenerative Medicine of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Krishnapriya M Varier
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China; School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Zhicao Li
- Key Laboratory of Regenerative Medicine of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Xujie Qin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, PR China
| | - Qing Rao
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Jingrui Song
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Anling Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Yubing Hang
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Chunmao Yuan
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Babu Gajendran
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China; School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| | - Liping Shu
- Key Laboratory of Regenerative Medicine of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, PR China.
| | - Min Wen
- Key Laboratory of Regenerative Medicine of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, PR China.
| | - Yanmei Li
- State Key Laboratory for Functions and Applications of Medicinal Plants/Department of Immunology, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China.
| | - Haiyang Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, PR China.
| |
Collapse
|
6
|
Xun R, Lu H, Wang X. Identification of CDC25C as a Potential Biomarker in Hepatocellular Carcinoma Using Bioinformatics Analysis. Technol Cancer Res Treat 2020; 19:1533033820967474. [PMID: 33111630 PMCID: PMC7607810 DOI: 10.1177/1533033820967474] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most aggressive type of gastrointestinal tumor, with a high rate of mortality. However, identifying biomarkers for the treatment of HCC remains to be developed. We aimed to determine whether cell division cycle 25C (CDC25C) could be used as a novel diagnostic and therapeutic biomarker in HCC. Expression of CDC25C in HCC was analyzed by using GEPIA (Gene Expression Profiling Interactive Analysis) and UALCAN databases. GEPIA and CBioPortal databases were applied to analyze patients’survival and CDC25C mutations, respectively. PPI (Protein-Protein Interaction) network was further built by STRING (Search Tool for the Retrieval of Interacting Genes) and Metascape Web portals. To the best of our knowledge, the novel observations identified in the present study reveal that the expression of CDC25C in HCC was significantly enhanced when compare to that in normal liver tissues (P < 0.001). A higher CDC25C expression resulted in a remarkably shorter disease free survival as well as overall survival. Moreover, the expression of CDC25C in HCC was related to HCC patients’grade and race, but not gender. The expression levels of CDC25C elevated gradually from stage 1 to 3 but decreased in stage 4. The specific gene mutations V41A, L87 H, N222 K and X309-splice of CDC25C occurred in HCC samples and these unique mutations were not detected in any other tumor tissues. Finally, PPI networks and GO enrichment analysis suggested that CDC25C might be associated with cell cycle and p53 signaling pathway. Taken together, bioinformatics analysis revealed that CDC25C might be a potential diagnostic predictor for HCC.
Collapse
Affiliation(s)
- Ruifeng Xun
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China.,Department of Orthopedic, Peoples Hospital of Linquan County, Fuyang, China
| | - Hougen Lu
- Department of Orthopedic, The Second School of Clinical Medicine & Jingzhou Central Hospital, Yangtze University, Jingzhou, China
| | - Xianwang Wang
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China
| |
Collapse
|
7
|
Lazzara V, Arizza V, Luparello C, Mauro M, Vazzana M. Bright Spots in The Darkness of Cancer: A Review of Starfishes-Derived Compounds and Their Anti-Tumor Action. Mar Drugs 2019; 17:E617. [PMID: 31671922 PMCID: PMC6891385 DOI: 10.3390/md17110617] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
The fight against cancer represents a great challenge for researchers and, for this reason, the search for new promising drugs to improve cancer treatments has become inevitable. Oceans, due to their wide diversity of marine species and environmental conditions have proven to be precious sources of potential natural drugs with active properties. As an example, in this context several studies performed on sponges, tunicates, mollusks, and soft corals have brought evidence of the interesting biological activities of the molecules derived from these species. Also, echinoderms constitute an important phylum, whose members produce a huge number of compounds with diverse biological activities. In particular, this review is the first attempt to summarize the knowledge about starfishes and their secondary metabolites that exhibited a significant anticancer effect against different human tumor cell lines. For each species of starfish, the extracted molecules, their effects, and mechanisms of action are described.
Collapse
Affiliation(s)
- Valentina Lazzara
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy.
| | - Vincenzo Arizza
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy.
| | - Claudio Luparello
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy.
| | - Manuela Mauro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy.
| | - Mirella Vazzana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy.
| |
Collapse
|
8
|
Pan Z, Zhang X, Yu P, Chen X, Lu P, Li M, Liu X, Li Z, Wei F, Wang K, Zheng Q, Li D. Cinobufagin Induces Cell Cycle Arrest at the G2/M Phase and Promotes Apoptosis in Malignant Melanoma Cells. Front Oncol 2019; 9:853. [PMID: 31552178 PMCID: PMC6738445 DOI: 10.3389/fonc.2019.00853] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022] Open
Abstract
Emerging evidence has shown that cinobufagin, as an active ingredient of Venenum Bufonis, inhibits tumor development. The aim of this study was to investigate the inhibitory effects of cinobufagin on A375 human malignant melanoma cells. MTT and colony formation assays showed that cinobufagin significantly inhibited A375 cell proliferation and cell colony formation. Additional studies demonstrated that cinobufagin markedly increased the levels of ATM serine/threonine kinase (ATM) and checkpoint kinase 2 (Chk2) and decreased the levels of cell division cycle 25C (CDC25C), cyclin-dependent kinase 1 (CDK1), and cyclin B, subsequently inducing G2/M cell cycle arrest in A375 cells. Moreover, cinobufagin clearly inhibited the levels of phosphoinositide 3-kinase (PI3K), phosphorylated PI3K (p-PI3K), AKT, p-AKT, and B-cell lymphoma 2 (Bcl-2). By contrast, it increased the levels of Bcl-2-associated death promoter, Bcl-2-associated X, cytoplasmic cytochrome C, and apoptotic protease activating factor 1, leading to increased levels of cleaved caspase-9 and cleaved caspase-3, resulting in the apoptosis of A375 cells. Together, these results indicate that cinobufagin can induce cell cycle arrest at the G2/M phase and apoptosis, leading to inhibition of A375/B16 cell proliferation. Thus, cinobufagin may be useful for melanoma treatment.
Collapse
Affiliation(s)
- Zhaohai Pan
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Xin Zhang
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Pengfei Yu
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Xiaoyu Chen
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Peng Lu
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Minjing Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Xiaona Liu
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Zhipeng Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Fei Wei
- School of Public Health and Management, Binzhou Medical University, Yantai, China
| | - Kejun Wang
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Qiusheng Zheng
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China.,Key Laboratory of Xinjiang Endemic Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Defang Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| |
Collapse
|
9
|
Chiu HC, Huang WR, Liao TL, Chi PI, Nielsen BL, Liu JH, Liu HJ. Mechanistic insights into avian reovirus p17-modulated suppression of cell cycle CDK-cyclin complexes and enhancement of p53 and cyclin H interaction. J Biol Chem 2018; 293:12542-12562. [PMID: 29907572 DOI: 10.1074/jbc.ra118.002341] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/17/2018] [Indexed: 12/22/2022] Open
Abstract
The avian reovirus p17 protein is a nucleocytoplasmic shuttling protein. Although we have demonstrated that p17 causes cell growth retardation via activation of p53, the precise mechanisms remain unclear. This is the first report that avian reovirus p17 possesses broad inhibitory effects on cell cycle CDKs, cyclins, CDK-cyclin complexes, and CDK-activating kinase activity in various mammalian, avian, and cancer cell lines. Suppression of CDK activity by p17 occurs by direct binding to CDKs, cyclins, and CDK-cyclin complexes; transcriptional down-regulation of CDKs; cytoplasmic retention of CDKs and cyclins; and inhibition of CDK-activating kinase activity by promoting p53-cyclin H interaction. p17 binds to CDK-cyclin except for CDK1-cyclin B1 and CDK7-cyclin H complexes. We have determined that the negatively charged 151LAVXDVDA(E/D)DGADPN165 motif in cyclin B1 interacts with a positively charged region of CDK1. p17 mimics the cyclin B1 sequence to compete for CDK1 binding. The PSTAIRE motif is not required for interaction of CDK1-cyclin B1, but it is required for other CDK-cyclin complexes. p17 interacts with cyclins by its cyclin-binding motif, 125RXL127 Sequence and mutagenic analyses of p17 indicated that a 140WXFD143 motif and residues Asp-113 and Lys-122 in p17 are critical for CDK2 and CDK6 binding, leading to their sequestration in the cytoplasm. Exogenous expression of p17 significantly enhanced virus replication, whereas p17 mutants with low binding ability to cell cycle CDKs had no effect on virus yield, suggesting that p17 inhibits cell growth and the cell cycle, benefiting virus replication. An in vivo tumorigenesis assay also showed a significant reduction in tumor size.
Collapse
Affiliation(s)
- Hung-Chuan Chiu
- From the Institute of Molecular Biology.,iEGG and Animal Biotechnology Center
| | | | - Tsai-Ling Liao
- the Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan, and.,Rong Hsing Research Center for Translational Medicine
| | - Pei-I Chi
- From the Institute of Molecular Biology
| | - Brent L Nielsen
- the Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah 84602
| | | | - Hung-Jen Liu
- From the Institute of Molecular Biology, .,iEGG and Animal Biotechnology Center.,Rong Hsing Research Center for Translational Medicine.,the Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
10
|
Miladiyah I, Jumina J, Haryana SM, Mustofa M. Biological activity, quantitative structure-activity relationship analysis, and molecular docking of xanthone derivatives as anticancer drugs. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:149-158. [PMID: 29391779 PMCID: PMC5774476 DOI: 10.2147/dddt.s149973] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background Xanthone derivatives have a wide range of pharmacological activities, such as those involving antibacterial, antiviral, antimalarial, anthelmintic, anti-inflammatory, antiprotozoal, and anticancer properties. Among these, we investigated the anticancer properties of xanthone. This research aimed to analyze the biological activity of ten novel xanthone derivatives, to investigate the most contributing-descriptors for their cytotoxic activities, and to examine the possible mechanism of actions of xanthone compound through molecular docking. Materials and methods The cytotoxic tests were carried out on WiDR and Vero cell lines, by a 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay method. The structural features required for xanthone’s anticancer activity were conducted by using the semi-empirical Austin Model-1 method, and continued with quantitative structure-activity relationship (QSAR) analysis using BuildQSAR program. The study of the possible mechanism of actions of the selected xanthone compound was done through molecular docking with PLANTS. Results The three novel xanthone derivatives (compounds 5, 7, and 8) exhibited cytotoxic activity with compound 5 showed the highest degree of cytotoxicity at concentration 9.23 µg/mL (37.8 µM). The following best equation model was obtained from the BuildQSAR calculation: log 1/IC50 = −8.124 qC1 −35.088 qC2 −6.008 qC3 + 1.831 u + 0.540 logP −9.115 (n = 10, r = 0.976, s = 0.144, F = 15.920, Q2 = 0.651, SPRESS = 0.390). This equation model generated 15 proposed new xanthone compounds with better-predicted anticancer activities. A molecular docking study of compound 5 showed that xanthone formed binding interactions with some receptors involved in cancer pathology, including telomerase, tumor-promoting inflammation (COX-2), and cyclin-dependent kinase-2 (CDK2) inhibitor. Conclusion The results suggested that compound 5 showed the best cytotoxic activity among the xanthone derivatives tested. QSAR analysis showed that the descriptors contributed to xanthone’s cytotoxic activity were the net atomic charge at qC1, qC2, and qC3 positions, also dipole moment and logP. Compound 5 was suspected to be cytotoxic by its inhibition of telomerase, COX-2, and CDK2 receptors.
Collapse
Affiliation(s)
- Isnatin Miladiyah
- Pharmacology Department, Faculty of Medicine, Islamic University of Indonesia.,Doctorate Program of Medical Science and Health, Faculty of Medicine
| | - Jumina Jumina
- Chemistry Department, Faculty of Mathematics and Natural Sciences
| | | | - Mustofa Mustofa
- Pharmacology and Therapeutic Department, Faculty of Medicine, Gadjah Mada University, Yogyakarta, Indonesia
| |
Collapse
|
11
|
Hegde M, Vartak SV, Kavitha CV, Ananda H, Prasanna DS, Gopalakrishnan V, Choudhary B, Rangappa KS, Raghavan SC. A Benzothiazole Derivative (5g) Induces DNA Damage And Potent G2/M Arrest In Cancer Cells. Sci Rep 2017; 7:2533. [PMID: 28566733 PMCID: PMC5451441 DOI: 10.1038/s41598-017-02489-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 04/12/2017] [Indexed: 11/25/2022] Open
Abstract
Chemically synthesized small molecules play important role in anticancer therapy. Several chemical compounds have been reported to damage the DNA, either directly or indirectly slowing down the cancer cell progression by causing a cell cycle arrest. Direct or indirect reactive oxygen species formation causes DNA damage leading to cell cycle arrest and subsequent cell death. Therefore, identification of chemically synthesized compounds with anticancer potential is important. Here we investigate the effect of benzothiazole derivative (5g) for its ability to inhibit cell proliferation in different cancer models. Interestingly, 5g interfered with cell proliferation in both, cell lines and tumor cells leading to significant G2/M arrest. 5g treatment resulted in elevated levels of ROS and subsequently, DNA double-strand breaks (DSBs) explaining observed G2/M arrest. Consistently, we observed deregulation of many cell cycle associated proteins such as CDK1, BCL2 and their phosphorylated form, CyclinB1, CDC25c etc. Besides, 5g treatment led to decreased levels of mitochondrial membrane potential and activation of apoptosis. Interestingly, 5g administration inhibited tumor growth in mice without significant side effects. Thus, our study identifies 5g as a potent biochemical inhibitor to induce G2/M phase arrest of the cell cycle, and demonstrates its anticancer properties both ex vivo and in vivo.
Collapse
Affiliation(s)
- Mahesh Hegde
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India.,Department of Studies in Chemistry, University of Mysore, Mysuru, 570006, India
| | - Supriya V Vartak
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | | | - Hanumappa Ananda
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Doddakunche S Prasanna
- Department of Nanotechnology, Visvesvaraya Technological University, Center for Postgraduate Studies, Bengaluru Region, Muddenahalli, Chikkaballapur, Bangalore, 562101, India
| | - Vidya Gopalakrishnan
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India.,Institute of Bioinformatics and Applied Biotechnology, Electronics City, Bangalore, 560100, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Electronics City, Bangalore, 560100, India
| | | | - Sathees C Raghavan
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
12
|
|
13
|
5-Substituted 3-chlorokenpaullone derivatives are potent inhibitors of Trypanosoma brucei bloodstream forms. Bioorg Med Chem 2016; 24:3790-800. [DOI: 10.1016/j.bmc.2016.06.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/09/2016] [Accepted: 06/11/2016] [Indexed: 01/31/2023]
|
14
|
|
15
|
Miguel RB, Petersen PAD, Gonzales-Zubiate FA, Oliveira CC, Kumar N, do Nascimento RR, Petrilli HM, da Costa Ferreira AM. Inhibition of cyclin-dependent kinase CDK1 by oxindolimine ligands and corresponding copper and zinc complexes. J Biol Inorg Chem 2015; 20:1205-17. [PMID: 26411703 DOI: 10.1007/s00775-015-1300-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/21/2015] [Indexed: 12/11/2022]
Abstract
Oxindolimine-copper(II) and zinc(II) complexes that previously have shown to induce apoptosis, with DNA and mitochondria as main targets, exhibit here significant inhibition of kinase CDK1/cyclin B protein. Copper species are more active than the corresponding zinc, and the free ligand shows to be less active, indicating a major influence of coordination in the process, and a further modulation by the coordinated ligand. Molecular docking and classical molecular dynamics provide a better understanding of the effectiveness and kinase inhibition mechanism by these compounds, showing that the metal complex provides a stronger interaction than the free ligand with the ATP-binding site. The metal ion introduces charge in the oxindole species, giving it a more rigid conformation that then becomes more effective in its interactions with the protein active site. Analogous experiments resulted in no significant effect regarding phosphatase inhibition. These results can explain the cytotoxicity of these metal complexes towards different tumor cells, in addition to its capability of binding to DNA, and decreasing membrane potential of mitochondria.
Collapse
Affiliation(s)
- Rodrigo Bernardi Miguel
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Philippe Alexandre Divina Petersen
- Departamento de Física dos Materiais e Mecânica, Instituto de Física, Universidade de São Paulo, Rua do Matão, Travessa R 187, São Paulo, 05508-090, SP, Brazil
| | - Fernando A Gonzales-Zubiate
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, 05508-000, SP, Brazil
| | - Carla Columbano Oliveira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, 05508-000, SP, Brazil
| | - Naresh Kumar
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Rafael Rodrigues do Nascimento
- Departamento de Física dos Materiais e Mecânica, Instituto de Física, Universidade de São Paulo, Rua do Matão, Travessa R 187, São Paulo, 05508-090, SP, Brazil
| | - Helena Maria Petrilli
- Departamento de Física dos Materiais e Mecânica, Instituto de Física, Universidade de São Paulo, Rua do Matão, Travessa R 187, São Paulo, 05508-090, SP, Brazil.
| | - Ana Maria da Costa Ferreira
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
16
|
|
17
|
Selective inhibitors of Plasmodium falciparum glycogen synthase-3 (PfGSK-3): New antimalarial agents? BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1644-9. [PMID: 25861860 DOI: 10.1016/j.bbapap.2015.03.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 03/25/2015] [Indexed: 01/19/2023]
Abstract
Plasmodium falciparum glycogen synthase kinase-3 (PfGSK-3) is one of the eukaryotic protein kinases that were identified as essential for the parasite causing malaria tropica. Although the physiological functions of PfGSK-3 are still unknown, it had been suggested as a putative target for novel antimalarial drugs. The high structural similarity of PfGSK-3 and its human orthologue HsGSK-3 makes the development of selective PfGSK-3 inhibitors a challenging task. Actually, established GSK-3 inhibitors are either unselective or are more potent for inhibition of the mammalian GSK-3. A high throughput screening campaign identified thieno[2,3-b]pyridines as a new class of PfGSK-3 inhibitors. Systematic variation of the substitution pattern at the parent scaffold led to compounds which selectively inhibited the plasmodial enzyme. These compounds also exhibited activity against erythrocyte stages of the parasites. A hypothetical explanation for the selectivity of the new antimalarial compounds was enunciated based on the results of docking a selective inhibitor into a PfGSK-3 homology model and by comparison of the results with an X-ray structure of HsGSK-3 co-crystallized with a similar but unselective compound. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.
Collapse
|
18
|
Maiwald F, Benítez D, Charquero D, Dar MA, Erdmann H, Preu L, Koch O, Hölscher C, Loaëc N, Meijer L, Comini MA, Kunick C. 9- and 11-Substituted 4-azapaullones are potent and selective inhibitors of African trypanosoma. Eur J Med Chem 2014; 83:274-83. [PMID: 24973661 DOI: 10.1016/j.ejmech.2014.06.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/04/2014] [Accepted: 06/10/2014] [Indexed: 10/25/2022]
Abstract
Trypanosomes from the "brucei" complex are pathogenic parasites endemic in sub-Saharan Africa and causative agents of severe diseases in humans and livestock. In order to identify new antitrypanosomal chemotypes against African trypanosomes, 4-azapaullones carrying α,β-unsaturated carbonyl chains in 9- or 11-position were synthesized employing a procedure with a Heck reaction as key step. Among the so prepared compounds, 5a and 5e proved to be potent antiparasitic agents with antitrypanosomal activity in the submicromolar range.
Collapse
Affiliation(s)
- Franziska Maiwald
- Technische Universität Braunschweig, Institut für Medizinische und Pharmazeutische Chemie, Beethovenstraße 55, D-38106 Braunschweig, Germany
| | - Diego Benítez
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, CP 11400 Montevideo, Uruguay
| | - Diego Charquero
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, CP 11400 Montevideo, Uruguay
| | - Mahin Abad Dar
- Forschungszentrum Borstel, Forschungsgruppe Infektionsimmunologie, Parkallee 22, D-23845 Borstel, Germany
| | - Hanna Erdmann
- Forschungszentrum Borstel, Forschungsgruppe Infektionsimmunologie, Parkallee 22, D-23845 Borstel, Germany
| | - Lutz Preu
- Technische Universität Braunschweig, Institut für Medizinische und Pharmazeutische Chemie, Beethovenstraße 55, D-38106 Braunschweig, Germany
| | - Oliver Koch
- Technische Universität Dortmund, Department of Chemistry and Chemical Biology, Technische Universität Dortmund, Otto-Hahn-Straße 6, 44227 Dortmund, Germany
| | - Christoph Hölscher
- Forschungszentrum Borstel, Forschungsgruppe Infektionsimmunologie, Parkallee 22, D-23845 Borstel, Germany
| | - Nadège Loaëc
- ManRos Therapeutics, Perharidy Research Center, 29680 Roscoff, France
| | - Laurent Meijer
- ManRos Therapeutics, Perharidy Research Center, 29680 Roscoff, France
| | - Marcelo A Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, CP 11400 Montevideo, Uruguay.
| | - Conrad Kunick
- Technische Universität Braunschweig, Institut für Medizinische und Pharmazeutische Chemie, Beethovenstraße 55, D-38106 Braunschweig, Germany.
| |
Collapse
|
19
|
|
20
|
|
21
|
|
22
|
Campsteijn C, Ovrebo JI, Karlsen BO, Thompson EM. Expansion of Cyclin D and CDK1 Paralogs in Oikopleura dioica, a Chordate Employing Diverse Cell Cycle Variants. Mol Biol Evol 2011; 29:487-502. [DOI: 10.1093/molbev/msr136] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
23
|
Flohé L. The trypanothione system and the opportunities it offers to create drugs for the neglected kinetoplast diseases. Biotechnol Adv 2011; 30:294-301. [PMID: 21620942 DOI: 10.1016/j.biotechadv.2011.05.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 05/06/2011] [Accepted: 05/09/2011] [Indexed: 12/20/2022]
Abstract
Parasitic trypanosomatids (Kinetoplastida) are the causative agents of devastating and hard-to-treat diseases such as African sleeping sickness, Chagas disease and various forms of Leishmaniasis. Altogether they affect > 30 Million patients, account for half a million fatalities p.a. and cause substantial economical problems in the Third World due to human morbidity and life stock losses. The design of efficacious and safe drugs is expected from inhibition of metabolic pathways that are unique and essential to the parasite and absent in the host. In this respect, the trypanothione system first detected in the insect-pathogenic trypanosomatid Crithidia fasciculata qualified as an attractive drug target area. The existence of the system in pathogenic relatives was established by homology cloning and PCR. The vital importance of the system was verified in Trypanosoma brucei by dsRNA technology or knock-out in other trypanosomatids, respectively, and is explained by its pivotal role in the parasite's antioxidant defense and DNA synthesis. The key system component is the bis-glutathionyl derivative of spermidine, trypanothione. It is the proximal reductant of tryparedoxin which substitutes for thioredoxin-, glutaredoxin- and glutathione-dependent reactions. Heterologous expression, functional characterization and crystallization of recombinant system components finally enable structure-based rational inhibitor design.
Collapse
Affiliation(s)
- Leopold Flohé
- Institute of Chemistry, Otto-von-Guericke-Universität, Universitätsplatz 2, D-39106 Magdeburg, Germany.
| |
Collapse
|
24
|
Węsierska-Gądek J, Kryštof V. Selective Cyclin-Dependent Kinase Inhibitors Discriminating between Cell Cycle and Transcriptional Kinases. Ann N Y Acad Sci 2009; 1171:228-41. [DOI: 10.1111/j.1749-6632.2009.04726.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
25
|
Raffa D, Maggio B, Cascioferro S, Raimondi MV, Daidone G, Plescia S, Schillaci D, Cusimano MG, Titone L, Colomba C, Tolomeo M. N-(Indazolyl)benzamido Derivatives as CDK1 Inhibitors: Design, Synthesis, Biological Activity, and Molecular Docking Studies. Arch Pharm (Weinheim) 2009; 342:265-73. [DOI: 10.1002/ardp.200800159] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Lou H, Gao Y, Zhai M, Qi Y, Chen L, Lv H, Yu J, Li Y. A novel peptide from alpha5 helix of Asterina pectinifera cyclin B conjugated to HIV-Tat(49-57) with cytotoxic and apoptotic effects against human cancer cells. Bioorg Med Chem Lett 2008; 18:4633-7. [PMID: 18656352 DOI: 10.1016/j.bmcl.2008.07.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 06/30/2008] [Accepted: 07/07/2008] [Indexed: 11/30/2022]
Abstract
A series of novel peptides from various motifs of Asterina pectinifera cyclin B and their derivatives conjugated to HIV-Tat(49-57) were designed and synthesized. Their bioactivities on two human cancer cell lines were determined. Among them, Tat-a5 (KAQIRAMECNILGRKKRRQRRR) exhibited significant cytotoxic effects on cancer cell lines EC-9706 and HCT-116. Tat-a5 could arrest cancer cells at G(2)/M phase and make them apoptotic. Our results suggested that Tat-a5 could be a novel leading peptide with anticancer activity.
Collapse
Affiliation(s)
- Huiping Lou
- Department of Bioengineering, Zhengzhou University, Science Road 100, Zhengzhou 450001, China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Chen S, Chen L, Le NT, Zhao C, Sidduri A, Lou JP, Michoud C, Portland L, Jackson N, Liu JJ, Konzelmann F, Chi F, Tovar C, Xiang Q, Chen Y, Wen Y, Vassilev LT. Synthesis and activity of quinolinyl-methylene-thiazolinones as potent and selective cyclin-dependent kinase 1 inhibitors. Bioorg Med Chem Lett 2007; 17:2134-8. [PMID: 17303421 DOI: 10.1016/j.bmcl.2007.01.081] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Accepted: 01/29/2007] [Indexed: 12/13/2022]
Abstract
A novel series of quinolinyl-methylene-thiazolinones has been identified as potent and selective cyclin-dependent kinase 1 (CDK1) inhibitors. Their synthesis and structure activity relationships (SAR) are described. Representative compounds from this class reversibly inhibit CDK1 activity in vitro, and block cell cycle progression in human tumor cell lines, suggesting a potential use as antitumor agents.
Collapse
Affiliation(s)
- Shaoqing Chen
- Roche Research Center, Hoffmann-La Roche Inc., Nutley, NJ 07110, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Dabydeen DA, Burnett JC, Bai R, Verdier-Pinard P, Hickford SJH, Pettit GR, Blunt JW, Munro MHG, Gussio R, Hamel E. Comparison of the activities of the truncated halichondrin B analog NSC 707389 (E7389) with those of the parent compound and a proposed binding site on tubulin. Mol Pharmacol 2006; 70:1866-75. [PMID: 16940412 DOI: 10.1124/mol.106.026641] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The complex marine natural product halichondrin B was compared with NSC 707389 (E7389), a structurally simplified, synthetic macrocyclic ketone analog, which has been selected for clinical trials in human patients. NSC 707389 was invariably more potent than halichondrin B in its interactions with tubulin. Both compounds inhibited tubulin assembly, inhibited nucleotide exchange on beta-tubulin, and were noncompetitive inhibitors of the binding of radiolabeled vinblastine and dolastatin 10 to tubulin. Neither compound seemed to induce an aberrant tubulin assembly reaction, as occurs with vinblastine (tight spirals) or dolastatin 10 (aggregated rings and spirals). We modeled the two compounds into a shared binding site on tubulin consistent with their biochemical properties. Of the two tubulin structures available, we selected for modeling the complex of a stathmin fragment with two tubulin heterodimers with two bound colchicinoid molecules and a single bound vinblastine between the two heterodimers (Nature (Lond) 435:519-522, 2005). Halichondrin B and NSC 707389 fit snugly between the two heterodimers adjacent to the exchangeable site nucleotide. Fitting the compounds into this site, which was also close to the vinblastine site, resulted in enough movement of amino acid residues at the vinblastine site to cause the latter compound to bind less well to tubulin. The model suggests that halichondrin B and NSC 707389 most likely form highly unstable, small aberrant tubulin polymers rather than the massive stable structures observed with vinca alkaloids and antimitotic peptides.
Collapse
Affiliation(s)
- Donnette A Dabydeen
- Toxicology and Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute at Frederick, National Institutes of Health, Frederick Maryland, USA 21702
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sridhar J, Akula N, Pattabiraman N. Selectivity and potency of cyclin-dependent kinase inhibitors. AAPS JOURNAL 2006; 8:E204-21. [PMID: 16584130 PMCID: PMC2751441 DOI: 10.1208/aapsj080125] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Members of the cyclin-dependent kinase (CDK) family play key roles in various cellular processes. There are 11 members of the CDK family known till now. CDKs are activated by forming noncovalent complexes with cyclins such as A-, B-, C-, D- (D1, D2, and D3), and E-type cyclins. Each isozyme of this family is responsible for particular aspects (cell signaling, transcription, etc) of the cell cycle, and some of the CDK isozymes are specific to certain kinds of tissues. Aberrant expression and overexpression of these kinases are evidenced in many disease conditions. Inhibition of isozymes of CDKs specifically can yield beneficiary treatment modalities with minimum side effects. More than 80 3-dimensional structures of CDK2, CDK5, and CDK6 complexed with inhibitors have been published. This review provides an understanding of the structural aspects of CDK isozymes and binding modes of various known CDK inhibitors so that these kinases can be better targeted for drug discovery and design. The amino acid residues that constitute the cyclin binding region, the substrate binding region, and the area around the adenosine triphosphate (ATP) binding site have been compared for CDK isozymes. Those amino acids at the ATP binding site that could be used to improve the potency and subtype specificity have been described.
Collapse
Affiliation(s)
- Jayalakshmi Sridhar
- />Laboratory for In-silico Biology and Drug Discovery, Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Room W417, 3970 Reservoir Rd NW, 20005 Washington, DC
| | - Nagaraju Akula
- />Laboratory for In-silico Biology and Drug Discovery, Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Room W417, 3970 Reservoir Rd NW, 20005 Washington, DC
| | - Nagarajan Pattabiraman
- />Laboratory for In-silico Biology and Drug Discovery, Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Room W417, 3970 Reservoir Rd NW, 20005 Washington, DC
- />Department of Biochemistry & Molecular Biology, Georgetown University, Washington DC
| |
Collapse
|
30
|
Braña MF, Cacho M, García ML, Mayoral EP, López B, de Pascual-Teresa B, Ramos A, Acero N, Llinares F, Muñoz-Mingarro D, Lozach O, Meijer L. Pyrazolo[3,4-c]pyridazines as novel and selective inhibitors of cyclin-dependent kinases. J Med Chem 2005; 48:6843-54. [PMID: 16250643 DOI: 10.1021/jm058013g] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pyrazolopyridazine 1a was identified in a high-throughput screening carried out by BASF Bioresearch Corp. (Worcester, MA) as a potent inhibitor of CDK1/cyclin B and shown to have selectivity for the CDK family. Analogues of the lead compound have been synthesized and their antitumor activities have been tested. A molecular model of the complex between the lead compound and the CDK2 ATP binding site has been built using a combination of conformational search and automated docking techniques. The stability of the resulting complex has been assessed by molecular dynamics simulations and the experimental results obtained for the synthesized analogues have been rationalized on the basis of the proposed binding mode for compound 1a. As a result of the SAR study, monofuryl 1o has been synthesized and is one of the most active compounds against CDK1 of this series.
Collapse
Affiliation(s)
- Miguel F Braña
- Facultad de Farmacia, Universidad San Pablo CEU, Urbanización Montepríncipe, 28668-Boadilla del Monte, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|