1
|
Rendic SP, Guengerich FP. Formation of potentially toxic metabolites of drugs in reactions catalyzed by human drug-metabolizing enzymes. Arch Toxicol 2024; 98:1581-1628. [PMID: 38520539 DOI: 10.1007/s00204-024-03710-9] [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: 12/01/2023] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
Data are presented on the formation of potentially toxic metabolites of drugs that are substrates of human drug metabolizing enzymes. The tabular data lists the formation of potentially toxic/reactive products. The data were obtained from in vitro experiments and showed that the oxidative reactions predominate (with 96% of the total potential toxication reactions). Reductive reactions (e.g., reduction of nitro to amino group and reductive dehalogenation) participate to the extent of 4%. Of the enzymes, cytochrome P450 (P450, CYP) enzymes catalyzed 72% of the reactions, myeloperoxidase (MPO) 7%, flavin-containing monooxygenase (FMO) 3%, aldehyde oxidase (AOX) 4%, sulfotransferase (SULT) 5%, and a group of minor participating enzymes to the extent of 9%. Within the P450 Superfamily, P450 Subfamily 3A (P450 3A4 and 3A5) participates to the extent of 27% and the Subfamily 2C (P450 2C9 and P450 2C19) to the extent of 16%, together catalyzing 43% of the reactions, followed by P450 Subfamily 1A (P450 1A1 and P450 1A2) with 15%. The P450 2D6 enzyme participated in an extent of 8%, P450 2E1 in 10%, and P450 2B6 in 6% of the reactions. All other enzymes participate to the extent of 14%. The data show that, of the human enzymes analyzed, P450 enzymes were dominant in catalyzing potential toxication reactions of drugs and their metabolites, with the major role assigned to the P450 Subfamily 3A and significant participation of the P450 Subfamilies 2C and 1A, plus the 2D6, 2E1 and 2B6 enzymes contributing. Selected examples of drugs that are activated or proposed to form toxic species are discussed.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, USA
| |
Collapse
|
2
|
Rehman ZU, Saini P, Kumar S. Synthesis and Biological Evaluation of Some New Chalcone Derivatives as Anti-inflammatory Agents. Curr Drug Discov Technol 2023; 20:42-66. [PMID: 35702807 DOI: 10.2174/1570163819666220613153225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/20/2022] [Accepted: 05/07/2022] [Indexed: 11/22/2022]
Abstract
AIM The present research work aims to prepare a series of 1-(4-(2-(1H-indol-1-yl)-2- oxoethoxy)phenyl)-3-phenylprop-2-en-1-one derivatives. METHODS The major compound was achieved by the reaction of indole with chloroacetylchloride in benzene afforded 2-chloro-1-(indoline-1-yl) ethanone which reacts o- hydroxy acetophenone in presence of acetonitrile to form 2-(4-acetylphenoxy)-1-(1H-indol-1-yl)ethan-1-one then goes through aldol condensation to give various final derivatives. RESULTS AND CONCLUSION After the synthesis of compounds, the synthesized compounds were characterized by checking their solubility, melting point, thin layer chromatography, IR, 1HNMR spectral data and elemental analysis. All of the prepared derivatives were evaluated for their anti-inflammatory activity on wistar albino rats by following the carrageenan-induced Rat Hind Paw Edema model.
Collapse
Affiliation(s)
- Zia Ur Rehman
- School of Pharmaceutical Sciences, IFTM University Moradabad (244001), U.P, India
| | - Pooja Saini
- School of Pharmaceutical Sciences, IFTM University Moradabad (244001), U.P, India
| | - Sushil Kumar
- School of Pharmaceutical Sciences, IFTM University Moradabad (244001), U.P, India
| |
Collapse
|
3
|
Sanders J, Castiglione M, Shun T, Vollmer LL, Schurdak ME, Vogt A, Schwacha A. Validation of a high throughput screening assay to identify small molecules that target the eukaryotic replicative helicase. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:229-241. [PMID: 35058181 PMCID: PMC9196137 DOI: 10.1016/j.slasd.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mcm2-7 is the catalytic core of the eukaryotic replicative helicase, which together with CDC45 and the GINS complex unwind parental DNA to generate templates for DNA polymerase. Being a highly regulated and complex enzyme that operates via an incompletely understood multi-step mechanism, molecular probes of Mcm2-7 that interrogate specific mechanistic steps would be useful tools for research and potential future chemotherapy. Based upon a synthetic lethal approach, we previously developed a budding yeast multivariate cell-based high throughput screening (HTS) assay to identify putative Mcm inhibitors by their ability to specifically cause a growth defect in an mcm mutant relative to a wild-type strain[1]. Here, as proof of concept, we used this assay to screen a 1280-member compound library (LOPAC) for potential Mcm2-7 inhibitors. Primary screening and dose-dependent retesting identified twelve compounds from this library that specifically inhibited the growth of the Mcm mutant relative to the corresponding wild-type strain (0.9 % hit rate). Secondary assays were employed to rule out non-specific DNA damaging agents, establish direct protein-ligand interaction via biophysical methods, and verify in vivo DNA replication inhibition via fluorescence activated cell sorter analysis (FACS). We identified one agent (β-carboline-3-carboxylic acid N-methylamide, CMA) that physically bound to the purified Mcm2-7 complex (Kdapp119 µM), and at slightly higher concentrations specifically blocked S-phase cell cycle progression of the wild-type strain. In total, identification of Mcm2-7 as a CMA target validates our synthetic lethal HTS assay paradigm as a tool to identify chemical probes for the Mcm2-7 replicative helicase.
Collapse
Affiliation(s)
- Jordan Sanders
- The Department of Biological Sciences, University of Pittsburgh, Pittsburgh PA 15260
| | - Michael Castiglione
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260
| | - Tongying Shun
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260
| | - Laura L Vollmer
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260
| | - Mark E Schurdak
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260; Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh PA 15260
| | - Andreas Vogt
- Drug Discovery Institute, University of Pittsburgh Medical School, Pittsburgh PA 15260; Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh PA 15260.
| | - Anthony Schwacha
- The Department of Biological Sciences, University of Pittsburgh, Pittsburgh PA 15260.
| |
Collapse
|
4
|
Su XX, Chen YR, Wu JQ, Wu XZ, Li KT, Wang XN, Sun JW, Wang H, Ou TM. Design, synthesis, and evaluation of 9-(pyrimidin-2-yl)-9H-carbazole derivatives disrupting mitochondrial homeostasis in human lung adenocarcinoma. Eur J Med Chem 2022; 232:114200. [DOI: 10.1016/j.ejmech.2022.114200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/06/2022] [Accepted: 02/11/2022] [Indexed: 12/12/2022]
|
5
|
Li X, Qi SM, Wang Y, Jiang HW, Li YH, Zhao BC, Zhang T, Sun Y, Gao XZ, Quan SX, Liu PF, Li WS, Wu JH, Bao TY, Jiang HL. Antidepressant effect of electroacupuncture on modulating the expression of c-Fos/AP-1 through the JNK signaling pathway. Anat Rec (Hoboken) 2021; 304:2480-2493. [PMID: 34431619 DOI: 10.1002/ar.24740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 12/28/2022]
Abstract
The effectiveness and safety of electroacupuncture (EA) for depression have been identified by abundant clinical trials and experimental findings. The c-Jun-NH(2)-terminal kinase (JNK) signaling pathway is considered to be involved in the antidepressant mechanism of EA. However, the antidepressant effect of EA via modulating the expression of c-Fos/activator protein-1 (AP-1) under the condition of JNK inhibition remains unexplored. In this study, we investigated the antidepressant effect and possible mechanism of EA in regulating the expression of c-Fos/AP-1 under the condition of JNK inhibition by SP600125 in rats exposed to chronic unpredictable mild stress (CUMS). The depression-like behaviors were evaluated by the body weight, sucrose preference test (SPT), and open field test (OFT). The expression levels of c-Jun in the hypothalamus, c-Fos in the pituitary gland, and c-Fos and AP-1 in the serum of CUMS induced rat model of depression were detected by ELISA. The results indicated that treatment with EA and fluoxetine can reverse the CUMS-induced depression-like behaviors in rats and can up-regulate the expression levels of c-Jun in the hypothalamus, c-Fos in the pituitary gland, and c-Fos and AP-1 in the serum. Of note, the data demonstrated that SP600125, the inhibitor of JNK signaling pathway, can exert synergistic effect with EA in regulating CUMS-induced abnormal activation of the JNK signaling pathway. The antidepressant effect of EA might be mediated by modulating the expression of c-Fos/AP-1.
Collapse
Affiliation(s)
- Xiang Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.,Research Center of Mental and Neurological Disorders, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Si-Min Qi
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui-Wu Jiang
- School of Health Management, Xi'an Medical University, Xi'an, China
| | - Ya-Huan Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.,Research Center of Mental and Neurological Disorders, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Bing-Cong Zhao
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| | - Tao Zhang
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| | - Yang Sun
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.,Research Center of Mental and Neurological Disorders, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xing-Zhou Gao
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.,Research Center of Mental and Neurological Disorders, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Song-Xiao Quan
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Peng-Fei Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Wen-Shan Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ji-Hong Wu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Tu-Ya Bao
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.,Research Center of Mental and Neurological Disorders, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Hui-Li Jiang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.,Research Center of Mental and Neurological Disorders, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Tylińska B, Dobosz A, Spychała J, Cwynar-Zając Ł, Czyżnikowska Ż, Kuźniarski A, Gębarowski T. Evaluation of Interactions of Selected Olivacine Derivatives with DNA and Topoisomerase II. Int J Mol Sci 2021; 22:ijms22168492. [PMID: 34445198 PMCID: PMC8395211 DOI: 10.3390/ijms22168492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 11/16/2022] Open
Abstract
Olivacine and ellipticine are model anticancer drugs acting as topoisomerase II inhibitors. Here, we present investigations performed on four olivacine derivatives in light of their antitumor activity. The aim of this study was to identify the best antitumor compound among the four tested olivacine derivatives. The study was performed using CCRF/CEM and MCF-7 cell lines. Comet assay, polarography, inhibition of topoisomerase II activity, histone acetylation, and molecular docking studies were performed. Each tested compound displayed interaction with DNA and topoisomerase II, but did not cause histone acetylation. Compound 2 (9-methoxy-5,6-dimethyl-1-({[1-hydroxy-2-(hydroxymethyl)butan-2-yl]amino}methyl)-6H-pyrido[4,3-b]carbazole) was found to be the best candidate as an anticancer drug because it had the highest affinity for topoisomerase II and caused the least genotoxic damage in cells.
Collapse
Affiliation(s)
- Beata Tylińska
- Department of Organic Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Agnieszka Dobosz
- Department of Basic Medical Sciences, Wroclaw Medical University, 50-556 Wroclaw, Poland; (J.S.); (Ł.C.-Z.); (T.G.)
- Correspondence: ; Tel.: +48-717-840-482
| | - Jan Spychała
- Department of Basic Medical Sciences, Wroclaw Medical University, 50-556 Wroclaw, Poland; (J.S.); (Ł.C.-Z.); (T.G.)
| | - Łucja Cwynar-Zając
- Department of Basic Medical Sciences, Wroclaw Medical University, 50-556 Wroclaw, Poland; (J.S.); (Ł.C.-Z.); (T.G.)
| | - Żaneta Czyżnikowska
- Department of Inorganic Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Amadeusz Kuźniarski
- Department of Prosthetic Dentistry, Wroclaw Medical University, 50-425 Wroclaw, Poland;
| | - Tomasz Gębarowski
- Department of Basic Medical Sciences, Wroclaw Medical University, 50-556 Wroclaw, Poland; (J.S.); (Ł.C.-Z.); (T.G.)
| |
Collapse
|
7
|
Xu Y, Kong J, Hu P. Computational Drug Repurposing for Alzheimer's Disease Using Risk Genes From GWAS and Single-Cell RNA Sequencing Studies. Front Pharmacol 2021; 12:617537. [PMID: 34276354 PMCID: PMC8277916 DOI: 10.3389/fphar.2021.617537] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 06/15/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Traditional therapeutics targeting Alzheimer's disease (AD)-related subpathologies have so far proved ineffective. Drug repurposing, a more effective strategy that aims to find new indications for existing drugs against other diseases, offers benefits in AD drug development. In this study, we aim to identify potential anti-AD agents through enrichment analysis of drug-induced transcriptional profiles of pathways based on AD-associated risk genes identified from genome-wide association analyses (GWAS) and single-cell transcriptomic studies. Methods: We systematically constructed four gene lists (972 risk genes) from GWAS and single-cell transcriptomic studies and performed functional and genes overlap analyses in Enrichr tool. We then used a comprehensive drug repurposing tool Gene2Drug by combining drug-induced transcriptional responses with the associated pathways to compute candidate drugs from each gene list. Prioritized potential candidates (eight drugs) were further assessed with literature review. Results: The genomic-based gene lists contain late-onset AD associated genes (BIN1, ABCA7, APOE, CLU, and PICALM) and clinical AD drug targets (TREM2, CD33, CHRNA2, PRSS8, ACE, TKT, APP, and GABRA1). Our analysis identified eight AD candidate drugs (ellipticine, alsterpaullone, tomelukast, ginkgolide A, chrysin, ouabain, sulindac sulfide and lorglumide), four of which (alsterpaullone, ginkgolide A, chrysin and ouabain) have shown repurposing potential for AD validated by their preclinical evidence and moderate toxicity profiles from literature. These support the value of pathway-based prioritization based on the disease risk genes from GWAS and scRNA-seq data analysis. Conclusion: Our analysis strategy identified some potential drug candidates for AD. Although the drugs still need further experimental validation, the approach may be applied to repurpose drugs for other neurological disorders using their genomic information identified from large-scale genomic studies.
Collapse
Affiliation(s)
- Yun Xu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
8
|
Tylińska B, Wiatrak B. Bioactive Olivacine Derivatives-Potential Application in Cancer Therapy. BIOLOGY 2021; 10:564. [PMID: 34205757 PMCID: PMC8235335 DOI: 10.3390/biology10060564] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/15/2021] [Indexed: 11/18/2022]
Abstract
Olivacine and its derivatives are characterized by multidirectional biological activity. Noteworthy is their antiproliferative effect related to various mechanisms, such as inhibition of growth factors, enzymes, kinases and others. The activity of these compounds was tested on cell lines of various tumors. In most publications, the most active olivacine derivatives exceeded the effects of doxorubicin (a commonly used anticancer drug), so in the future, they may become the main new anticancer drugs. In this publication, we present the groups of the most active olivacine derivatives obtained. In this work, the in vitro and in vivo activity of olivacine and its most active derivatives are presented. We describe olivacine derivatives that have been in clinical trials. We conducted a structure-activity relationship (SAR) analysis that may be used to obtain new olivacine derivatives with better properties than the available anticancer drugs.
Collapse
Affiliation(s)
- Beata Tylińska
- Department of Organic Chemistry, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, 50-345 Wroclaw, Poland;
| |
Collapse
|
9
|
Tian LX, Li XY, Tang X, Zhou XY, Luo L, Ma XY, Tang WQ, Yu J, Ma W, Yang X, Yan J, Xu X, Liang HP. Ellipticine Conveys Protective Effects to Lipopolysaccharide-Activated Macrophages by Targeting the JNK/AP-1 Signaling Pathway. Inflammation 2019; 43:231-240. [DOI: 10.1007/s10753-019-01112-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
10
|
Dan VM, Varghese TS, Viswanathan G, Baby S. Ellipticine, its Derivatives: Re-evaluation of Clinical Suitability with the Aid of Drug Delivery Systems. Curr Cancer Drug Targets 2019; 20:33-46. [PMID: 31560288 DOI: 10.2174/1568009619666190927150131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/20/2019] [Accepted: 07/12/2019] [Indexed: 11/22/2022]
Abstract
Targeted drug delivery systems gave newer dimensions for safer and more effective use of therapeutic drugs, thus helping in circumventing the issues of toxicity and unintended drug accumulation. These ongoing developments in delivery systems can, in turn, bring back drugs that suffered various limitations, Ellipticine (EPT) being a candidate. EPT derivatives witnessed entry into clinical settings but failed to survive in clinics citing various toxic side effects. A large body of preclinical data deliberates the potency of drug delivery systems in increasing the efficiency of EPT/derivatives while decreasing their toxic side effects. Recent developments in drug delivery systems provide a platform to explore EPT and its derivatives as good clinical candidates in treating tumors. The present review deals with delivery mechanisms of EPT/EPT derivatives as antitumor drugs, in vitro and in vivo, and evaluates the suitability of EPT-carriers in clinical settings.
Collapse
Affiliation(s)
- Vipin Mohan Dan
- Microbiology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Pacha-Palode 695562, Thiruvananthapuram, Kerala, India
| | - Thania Sara Varghese
- Garden Management Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Pacha-Palode 695562, Thiruvananthapuram, Kerala, India
| | - Gayathri Viswanathan
- Phytochemistry and Phytopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Pacha-Palode 695562, Thiruvananthapuram, Kerala, India
| | - Sabulal Baby
- Phytochemistry and Phytopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Pacha-Palode 695562, Thiruvananthapuram, Kerala, India
| |
Collapse
|
11
|
Ellipticine-loaded apoferritin nanocarrier retains DNA adduct-based cytochrome P450-facilitated toxicity in neuroblastoma cells. Toxicology 2019; 419:40-54. [DOI: 10.1016/j.tox.2019.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/17/2022]
|
12
|
Sumalatha S, Namrata V, Lakshmi M, Sridhar G. Synthesis and Anticancer Activity of Oxadiazole Incorporated Ellipticine Derivatives. RUSS J GEN CHEM+ 2019. [DOI: 10.1134/s107036321903023x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
13
|
Willis AJ, Indra R, Wohak LE, Sozeri O, Feser K, Mrizova I, Phillips DH, Stiborova M, Arlt VM. The impact of chemotherapeutic drugs on the CYP1A1-catalysed metabolism of the environmental carcinogen benzo[a]pyrene: Effects in human colorectal HCT116 TP53(+/+), TP53(+/-) and TP53(-/-) cells. Toxicology 2018; 398-399:1-12. [PMID: 29471073 PMCID: PMC6593262 DOI: 10.1016/j.tox.2018.02.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/29/2018] [Accepted: 02/16/2018] [Indexed: 02/02/2023]
Abstract
Polycyclic aromatic hydrocarbons such as benzo[a]pyrene (BaP) can induce cytochrome P450 1A1 (CYP1A1) via a p53-dependent mechanism. The effect of different p53-activating chemotherapeutic drugs on CYP1A1 expression, and the resultant effect on BaP metabolism, was investigated in a panel of isogenic human colorectal HCT116 cells with differing TP53 status. Cells that were TP53(+/+), TP53(+/-) or TP53(-/-) were treated for up to 48 h with 60 μM cisplatin, 50 μM etoposide or 5 μM ellipticine, each of which caused high p53 induction at moderate cytotoxicity (60-80% cell viability). We found that etoposide and ellipticine induced CYP1A1 in TP53(+/+) cells but not in TP53(-/-) cells, demonstrating that the mechanism of CYP1A1 induction is p53-dependent; cisplatin had no such effect. Co-incubation experiments with the drugs and 2.5 μM BaP showed that: (i) etoposide increased CYP1A1 expression in TP53(+/+) cells, and to a lesser extent in TP53(-/-) cells, compared to cells treated with BaP alone; (ii) ellipticine decreased CYP1A1 expression in TP53(+/+) cells in BaP co-incubations; and (iii) cisplatin did not affect BaP-mediated CYP1A1 expression. Further, whereas cisplatin and etoposide had virtually no influence on CYP1A1-catalysed BaP metabolism, ellipticine treatment strongly inhibited BaP bioactivation. Our results indicate that the underlying mechanisms whereby etoposide and ellipticine regulate CYP1A1 expression must be different and may not be linked to p53 activation alone. These results could be relevant for smokers, who are exposed to increased levels of BaP, when prescribing chemotherapeutic drugs. Beside gene-environment interactions, more considerations should be given to potential drug-environment interactions during chemotherapy.
Collapse
Affiliation(s)
- Alexandra J Willis
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Radek Indra
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Laura E Wohak
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Osman Sozeri
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Kerstin Feser
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Iveta Mrizova
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic
| | - David H Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom; NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England, London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom; NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England, London, 150 Stamford Street, London SE1 9NH, United Kingdom.
| |
Collapse
|
14
|
Stiborova M. Formation of Covalent DNA Adducts by Enzymatically Activated Carcinogens and Drugs In Vitro and Their Determination by 32P-postlabeling. J Vis Exp 2018. [PMID: 29630053 DOI: 10.3791/57177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Covalent DNA adducts formed by chemicals or drugs with carcinogenic potency are judged as one of the most important factors in the initiation phase of carcinogenic processes. This covalent binding, which is considered the cause of tumorigenesis, is now evaluated as a central dogma of chemical carcinogenesis. Here, methods are described employing the reactions catalyzed by cytochrome P450 and additional biotransformation enzymes to investigate the potency of chemicals or drugs for their activation to metabolites forming these DNA adducts. Procedures are presented describing the isolation of cellular fractions possessing biotransformation enzymes (microsomal and cytosolic samples with cytochromes P450 or other biotransformation enzymes, i.e., peroxidases, NADPH:cytochrome P450 oxidoreductase, NAD(P)H:quinone oxidoreductase, or xanthine oxidase). Furthermore, methods are described that can be used for the metabolic activation of analyzed chemicals by these enzymes as well as those for isolation of DNA. Further, the appropriate methods capable of detecting and quantifying chemical/drug-derived DNA adducts, i.e., different modifications of the 32P-postlabeling technique and employment of radioactive-labeled analyzed chemicals, are shown in detail.
Collapse
|
15
|
Iqbal S, Khan MA, Javaid K, Sadiq R, Fazal-Ur-Rehman S, Choudhary MI, Basha FZ. New carbazole linked 1,2,3-triazoles as highly potent non-sugar α-glucosidase inhibitors. Bioorg Chem 2017; 74:72-81. [PMID: 28756277 DOI: 10.1016/j.bioorg.2017.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/18/2017] [Accepted: 07/14/2017] [Indexed: 10/19/2022]
Abstract
In the present study, a series of new carbazole linked 1H-1,2,3-triazoles (2-27) were synthesized via click reaction of N-propargyl-9H-carbazole (1) and azides of appropriate acetophenones and heterocycles. Synthesized carbazole triazoles including 7, 9, 10, 19, 20, and 23-26 (IC50=0.8±0.01-100.8±3.6μM), exhibited several folds more potent α-glucosidase inhibitory in vitro activity as compared to standard drug, acarbose. Compounds 2-5, 7-13, and 17-27 did not show any cytotoxicity against 3T3 cell lines, except triazoles 6, and 14-16. Among the series, carbazole triazoles 23 (IC50=1.0±0.057μM) and 25 (IC50=0.8±0.01μM) were found to be most active, and could serve as an attractive building block in the search of new non-sugar derivatives as anti-diabetic agents.
Collapse
Affiliation(s)
- Shazia Iqbal
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Maria Aqeel Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Kulsoom Javaid
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Rabia Sadiq
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | | | - M Iqbal Choudhary
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21412, Saudi Arabia
| | - Fatima Z Basha
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
16
|
|
17
|
Synthesis and biological evaluation of oxadiazole incorporated ellipticine derivatives as anticancer agents. MONATSHEFTE FUR CHEMIE 2016. [DOI: 10.1007/s00706-016-1790-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
18
|
RohitKumar HG, Asha KR, KiranKumar HN, Inamdar LS, Rao GMA. Cell Cycle Arrest and Induction of Apoptosis in Colon Adenocarcinoma Cells by a DNA Intercalative Quinoline Derivative, 4-Morpholinopyrimido [4',5':4,5] Selenolo (2,3-b) Quinoline. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2016; 34:525-43. [PMID: 26167663 DOI: 10.1080/15257770.2015.1030503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Circular dichroism, topological studies, molecular docking, absorbance, and fluorescence spectral titrations were employed to study the interaction of 4-morpholinopyrimido [4',5':4,5] selenolo (2,3-b) quinoline (MPSQ) with DNA. The association constants of MPSQ-DNA interactions were of the order of 10(4) M(-1). Melting temperature, topological, and docking studies confirmed that the mode of interaction was by intercalation with preference to d(GpC)-d(CpG) site of DNA. Cytotoxicity studies showed the MPSQ-induced dose-dependent inhibitory effect on the proliferation of different cancer cells. Colon adenocarcinoma (COLO 205) cells are more sensitive among the cell lines tested, with an IC50 value of 15 μM. Flow cytometry revealed that MPSQ affects the cell cycle progression by arresting at G2M phase. Further, Annexin V staining, mitochondrial membrane potential assay, and caspase-3 activity assay confirmed that MPSQ leads to mitochondria-mediated apoptotic cell death in COLO 205 cells.
Collapse
Affiliation(s)
- Heggodu G RohitKumar
- a Department of Biochemistry, Davangere University , Davangere , Karnataka , India
| | | | | | | | | |
Collapse
|
19
|
Rizza P, Pellegrino M, Caruso A, Iacopetta D, Sinicropi MS, Rault S, Lancelot JC, El-Kashef H, Lesnard A, Rochais C, Dallemagne P, Saturnino C, Giordano F, Catalano S, Andò S. 3-(Dipropylamino)-5-hydroxybenzofuro[2,3-f]quinazolin-1(2H)-one (DPA-HBFQ-1) plays an inhibitory role on breast cancer cell growth and progression. Eur J Med Chem 2015; 107:275-87. [PMID: 26599533 DOI: 10.1016/j.ejmech.2015.11.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 10/05/2015] [Accepted: 11/03/2015] [Indexed: 01/01/2023]
Abstract
A series of unknown 3-(alkyl(dialkyl)amino)benzofuro[2,3-f]quinazolin-1(2H)-ones 4-17 has been synthesized as new ellipticine analogs, in which the carbazole moiety and the pyridine ring were replaced by a dibenzofuran residue and a pyrimidine ring, respectively. The synthesis of these benzofuroquinazolinones 4-17 was performed in a simple one-pot reaction using 3-aminodibenzofuran or its 2-methoxy derivative, as starting materials. From 3-(dipropylamino)-5-methoxybenzofuro[2,3-f] quinazolin-1(2H)-one (13), we prepared 3-(dipropylamino)-5-hydroxybenzofuro[2,3-f]quinazolin-1(2H)-one (18), referred to as DPA-HBFQ-1. The cytotoxic activities of all the synthesized compounds, tested in different human breast cancer cell lines, revealed that DPA-HBFQ-1 was the most active compound. In particular, the latter was able to inhibit anchorage-dependent and -independent cell growth and to induce apoptosis in estrogen receptor alpha (ERα)-positive and -negative breast cancer cells. It did not affect proliferation and apoptotic responses in MCF-10A normal breast epithelial cells. The observed effects have been ascribed to an enhanced p21(Cip1/WAF1) expression in a p53-dependent manner of tumor suppressor and to a selective inhibition of human topoisomerase II. In addition, DPA-HBFQ-1 exerted growth inhibitory effects also in other cancer cell lines, even though with a lower cytotoxic activity. Our results indicate DPA-HBFQ-1 as a good candidate to be useful as cancer therapeutic agent, particularly for breast cancer.
Collapse
Affiliation(s)
- Pietro Rizza
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Michele Pellegrino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Anna Caruso
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy.
| | - Sylvain Rault
- Université de Caen Basse-Normandie, Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258, FR CNRS 3038 INC3M, Bd Becquerel, 14032 Caen Cedex, France.
| | - Jean Charles Lancelot
- Université de Caen Basse-Normandie, Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258, FR CNRS 3038 INC3M, Bd Becquerel, 14032 Caen Cedex, France
| | - Hussein El-Kashef
- Department of Chemistry, Faculty of Science, Assiut University, 71516 Assiut, Egypt
| | - Aurelien Lesnard
- Université de Caen Basse-Normandie, Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258, FR CNRS 3038 INC3M, Bd Becquerel, 14032 Caen Cedex, France
| | - Christophe Rochais
- Université de Caen Basse-Normandie, Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258, FR CNRS 3038 INC3M, Bd Becquerel, 14032 Caen Cedex, France
| | - Patrick Dallemagne
- Université de Caen Basse-Normandie, Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258, FR CNRS 3038 INC3M, Bd Becquerel, 14032 Caen Cedex, France
| | - Carmela Saturnino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy.
| |
Collapse
|
20
|
Żabka A, Winnicki K, Polit JT, Maszewski J. The effects of anti-DNA topoisomerase II drugs, etoposide and ellipticine, are modified in root meristem cells of Allium cepa by MG132, an inhibitor of 26S proteasomes. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2015; 96:72-82. [PMID: 26233708 DOI: 10.1016/j.plaphy.2015.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/17/2015] [Accepted: 07/17/2015] [Indexed: 06/04/2023]
Abstract
DNA topoisomerase II (Topo II), a highly specialized nuclear enzyme, resolves various entanglement problems concerning DNA that arise during chromatin remodeling, transcription, S-phase replication, meiotic recombination, chromosome condensation and segregation during mitosis. The genotoxic effects of two Topo II inhibitors known as potent anti-cancer drugs, etoposide (ETO) and ellipticine (EPC), were assayed in root apical meristem cells of Allium cepa. Despite various types of molecular interactions between these drugs and DNA-Topo II complexes at the chromatin level, which have a profound negative impact on the genome integrity (production of double-strand breaks, chromosomal bridges and constrictions, lagging fragments of chromosomes and their uneven segregation to daughter cell nuclei), most of the elicited changes were apparently similar, regarding both their intensity and time characteristics. No essential changes between ETO- and EPC-treated onion roots were noticed in the frequency of G1-, S-, G2-and M-phase cells, nuclear morphology, chromosome structures, tubulin-microtubule systems, extended distribution of mitosis-specific phosphorylation sites of histone H3, and the induction of apoptosis-like programmed cell death (AL-PCD). However, the important difference between the effects induced by the ETO and EPC concerns their catalytic activities in the presence of MG132 (proteasome inhibitor engaged in Topo II-mediated formation of cleavage complexes) and relates to the time-variable changes in chromosomal aberrations and AL-PCD rates. This result implies that proteasome-dependent mechanisms may contribute to the course of physiological effects generated by DNA lesions under conditions that affect the ability of plant cells to resolve topological problems that associated with the nuclear metabolic activities.
Collapse
Affiliation(s)
- Aneta Żabka
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland.
| | - Konrad Winnicki
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland.
| | - Justyna Teresa Polit
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland.
| | - Janusz Maszewski
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland.
| |
Collapse
|
21
|
HRABETA JAN, GROH TOMAS, KHALIL MOHAMEDASHRAF, POLJAKOVA JITKA, ADAM VOJTECH, KIZEK RENE, UHLIK JIRI, DOKTOROVA HELENA, CERNA TEREZA, FREI EVA, STIBOROVA MARIE, ECKSCHLAGER TOMAS. Vacuolar-ATPase-mediated intracellular sequestration of ellipticine contributes to drug resistance in neuroblastoma cells. Int J Oncol 2015; 47:971-80. [DOI: 10.3892/ijo.2015.3066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 06/08/2015] [Indexed: 11/06/2022] Open
|
22
|
Stiborová M, Černá V, Moserová M, Mrízová I, Arlt VM, Frei E. The anticancer drug ellipticine activated with cytochrome P450 mediates DNA damage determining its pharmacological efficiencies: studies with rats, Hepatic Cytochrome P450 Reductase Null (HRN™) mice and pure enzymes. Int J Mol Sci 2014; 16:284-306. [PMID: 25547492 PMCID: PMC4307247 DOI: 10.3390/ijms16010284] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 12/17/2014] [Indexed: 12/30/2022] Open
Abstract
Ellipticine is a DNA-damaging agent acting as a prodrug whose pharmacological efficiencies and genotoxic side effects are dictated by activation with cytochrome P450 (CYP). Over the last decade we have gained extensive experience in using pure enzymes and various animal models that helped to identify CYPs metabolizing ellipticine. In this review we focus on comparison between the in vitro and in vivo studies and show a necessity of both approaches to obtain valid information on CYP enzymes contributing to ellipticine metabolism. Discrepancies were found between the CYP enzymes activating ellipticine to 13-hydroxy- and 12-hydroxyellipticine generating covalent DNA adducts and those detoxifying this drug to 9-hydroxy- and 7-hydroellipticine in vitro and in vivo. In vivo, formation of ellipticine-DNA adducts is dependent not only on expression levels of CYP3A, catalyzing ellipticine activation in vitro, but also on those of CYP1A that oxidize ellipticine in vitro mainly to the detoxification products. The finding showing that cytochrome b5 alters the ratio of ellipticine metabolites generated by CYP1A1/2 and 3A4 explained this paradox. Whereas the detoxification of ellipticine by CYP1A and 3A is either decreased or not changed by cytochrome b5, activation leading to ellipticine-DNA adducts increased considerably. We show that (I) the pharmacological effects of ellipticine mediated by covalent ellipticine-derived DNA adducts are dictated by expression levels of CYP1A, 3A and cytochrome b5, and its own potency to induce these enzymes in tumor tissues, (II) animal models, where levels of CYPs are either knocked out or induced are appropriate to identify CYPs metabolizing ellipticine in vivo, and (III) extrapolation from in vitro data to the situation in vivo is not always possible, confirming the need for these animal models.
Collapse
Affiliation(s)
- Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, CZ-12843 Prague 2, Czech Republic.
| | - Věra Černá
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, CZ-12843 Prague 2, Czech Republic.
| | - Michaela Moserová
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, CZ-12843 Prague 2, Czech Republic.
| | - Iveta Mrízová
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, CZ-12843 Prague 2, Czech Republic.
| | - Volker M Arlt
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environmental & Health, King's College London, 150 Stamford Street, London SE1 9NH, UK.
| | - Eva Frei
- Division of Preventive Oncology, National Center for Tumor Diseases, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
23
|
Calabrò S, Alzoubi K, Bissinger R, Faggio C, Lang F. Stimulation of Suicidal Erythrocyte Death by Ellipticine. Basic Clin Pharmacol Toxicol 2014; 116:485-92. [DOI: 10.1111/bcpt.12350] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/07/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Salvatrice Calabrò
- Department of Physiology; University of Tübingen; Tübingen Germany
- Department of Biological and Environmental Sciences; University of Messina; S. Agata-Messina Italy
| | - Kousi Alzoubi
- Department of Physiology; University of Tübingen; Tübingen Germany
| | - Rosi Bissinger
- Department of Physiology; University of Tübingen; Tübingen Germany
| | - Caterina Faggio
- Department of Biological and Environmental Sciences; University of Messina; S. Agata-Messina Italy
| | - Florian Lang
- Department of Physiology; University of Tübingen; Tübingen Germany
| |
Collapse
|
24
|
Formation of DNA adducts by ellipticine and its micellar form in rats - a comparative study. SENSORS 2014; 14:22982-97. [PMID: 25479328 PMCID: PMC4299049 DOI: 10.3390/s141222982] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 11/27/2014] [Accepted: 11/28/2014] [Indexed: 01/11/2023]
Abstract
The requirements for early diagnostics as well as effective treatment of cancer diseases have increased the pressure on development of efficient methods for targeted drug delivery as well as imaging of the treatment success. One of the most recent approaches covering the drug delivery aspects is benefitting from the unique properties of nanomaterials. Ellipticine and its derivatives are efficient anticancer compounds that function through multiple mechanisms. Formation of covalent DNA adducts after ellipticine enzymatic activation is one of the most important mechanisms of its pharmacological action. In this study, we investigated whether ellipticine might be released from its micellar (encapsulated) form to generate covalent adducts analogous to those formed by free ellipticine. The 32P-postlabeling technique was used as a useful imaging method to detect and quantify covalent ellipticine-derived DNA adducts. We compared the efficiencies of free ellipticine and its micellar form (the poly(ethylene oxide)-block-poly(allyl glycidyl ether) (PAGE-PEO) block copolymer, P 119 nanoparticles) to form ellipticine-DNA adducts in rats in vivo. Here, we demonstrate for the first time that treatment of rats with ellipticine in micelles resulted in formation of ellipticine-derived DNA adducts in vivo and suggest that a gradual release of ellipticine from its micellar form might produce the enhanced permeation and retention effect of this ellipticine-micellar delivery system.
Collapse
|
25
|
Panno A, Sinicropi MS, Caruso A, El-Kashef H, Lancelot JC, Aubert G, Lesnard A, Cresteil T, Rault S. New Trimethoxybenzamides and Trimethoxyphenylureas Derived from Dimethylcarbazole as Cytotoxic Agents. Part I. J Heterocycl Chem 2014. [DOI: 10.1002/jhet.1951] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Antonella Panno
- Université de Caen Basse-Normandie; Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M, Bd Becquerel; 14032 Caen Cedex France
- Dipartimento di Scienze Farmaceutiche; Università della Calabria; 87036 Arcavacata di Rende Cosenza Italy
| | - Maria Stefania Sinicropi
- Dipartimento di Scienze Farmaceutiche; Università della Calabria; 87036 Arcavacata di Rende Cosenza Italy
| | - Anna Caruso
- Université de Caen Basse-Normandie; Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M, Bd Becquerel; 14032 Caen Cedex France
- Dipartimento di Scienze Farmaceutiche; Università della Calabria; 87036 Arcavacata di Rende Cosenza Italy
| | - Hussein El-Kashef
- Université de Caen Basse-Normandie; Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M, Bd Becquerel; 14032 Caen Cedex France
- Chemistry Department, Faculty of Science; Assiut University; 71516 Assiut Egypt
| | - Jean-Charles Lancelot
- Université de Caen Basse-Normandie; Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M, Bd Becquerel; 14032 Caen Cedex France
| | - Geneviève Aubert
- Institut de Chimie des Substances Naturelles, UPR 2301; CNRS; Avenue de la Terrasse 91198 Gif-sur-Yvette France
| | - Aurélien Lesnard
- Université de Caen Basse-Normandie; Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M, Bd Becquerel; 14032 Caen Cedex France
| | - Thierry Cresteil
- Institut de Chimie des Substances Naturelles, UPR 2301; CNRS; Avenue de la Terrasse 91198 Gif-sur-Yvette France
| | - Sylvain Rault
- Université de Caen Basse-Normandie; Centre d'Etudes et de Recherche sur le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M, Bd Becquerel; 14032 Caen Cedex France
| |
Collapse
|
26
|
Parisi OI, Morelli C, Puoci F, Saturnino C, Caruso A, Sisci D, Trombino GE, Picci N, Sinicropi MS. Magnetic molecularly imprinted polymers (MMIPs) for carbazole derivative release in targeted cancer therapy. J Mater Chem B 2014; 2:6619-6625. [DOI: 10.1039/c4tb00607k] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Magnetic Molecularly Imprinted Polymers (MMIPs) are synthesized with the aim to prepare novel devices for 9H-carbazole derivative sustained delivery in targeted cancer therapy.
Collapse
Affiliation(s)
- Ortensia Ilaria Parisi
- Department of Pharmacy
- Health and Nutritional Sciences
- University of Calabria
- 87036 Rende, Italy
- Department of Informatics
| | - Catia Morelli
- Department of Pharmacy
- Health and Nutritional Sciences
- University of Calabria
- 87036 Rende, Italy
| | - Francesco Puoci
- Department of Pharmacy
- Health and Nutritional Sciences
- University of Calabria
- 87036 Rende, Italy
| | - Carmela Saturnino
- Department of Pharmaceutical and Biomedical Sciences
- University of Salerno
- 84084 Fisciano, Italy
| | - Anna Caruso
- Department of Pharmacy
- Health and Nutritional Sciences
- University of Calabria
- 87036 Rende, Italy
- Department of Informatics
| | - Diego Sisci
- Department of Pharmacy
- Health and Nutritional Sciences
- University of Calabria
- 87036 Rende, Italy
| | - Giovanna Elvi Trombino
- Department of Pharmacy
- Health and Nutritional Sciences
- University of Calabria
- 87036 Rende, Italy
| | - Nevio Picci
- Department of Pharmacy
- Health and Nutritional Sciences
- University of Calabria
- 87036 Rende, Italy
| | | |
Collapse
|
27
|
Effects of ellipticine on ALDH1A1-expressing breast cancer stem cells--an in vitro and in silico study. Tumour Biol 2013; 35:723-37. [PMID: 23982874 DOI: 10.1007/s13277-013-1099-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 08/08/2013] [Indexed: 02/07/2023] Open
Abstract
Targeting breast cancer stem cells (BCSCs) offers a promising strategy for breast cancer treatment. We examined the plant alkaloid ellipticine for its efficacy to inhibit the expression of aldehyde dehydrogenase 1 class A1 (ALDH1A1)-positive BCSCs by in vitro and in silico methods. At 3 mM concentration, ellipticine decreased the expression of ALDH1A1-positive BCSCs by 62% (p = 0.073) in the MCF7 cell line and by 53% (p = 0.024) in the SUM159 cell line compared to vehicle-treated cultures. Ellipticine significantly reduced the formation of mammospheres, whereas paclitaxel enhanced mammosphere formation in both the treated cell lines. Interestingly, when treated with a combination of ellipticine and paclitaxel, the percentage of ALDH1A1-positive BCSCs dropped by several fold in vitro. A homology model of Homo sapiens ALDH1A1 was built using the crystal structure of NAD-bound sheep liver class I aldehyde dehydrogenase [PDB ID: 1BXS] as a template. Molecular simulation and docking studies revealed that the amino acids Asn-117 and Asn-121, Glu-249, Cys-302, and Gln-350, present in the active site of human ALDH1A1, played a vital role in interacting with the drug. The present study suggests that ellipticine reduces the proliferation and self-renewal ability of ALDH1A1-positive BCSCs and can be used in combination with a cytotoxic drug like paclitaxel for potential targeting of BCSCs.
Collapse
|
28
|
Sahu U, Sidhar H, Ghate PS, Advirao GM, Raghavan SC, Giri RK. A Novel Anticancer Agent, 8-Methoxypyrimido[4',5':4,5]thieno(2,3-b) Quinoline-4(3H)-One Induces Neuro 2a Neuroblastoma Cell Death through p53-Dependent, Caspase-Dependent and -Independent Apoptotic Pathways. PLoS One 2013; 8:e66430. [PMID: 23824039 PMCID: PMC3688904 DOI: 10.1371/journal.pone.0066430] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 05/05/2013] [Indexed: 01/04/2023] Open
Abstract
Neuroblastoma is the most common cancer in infants and fourth most common cancer in children. Despite recent advances in cancer treatments, the prognosis of stage-IV neuroblastoma patients continues to be dismal which warrant new pharmacotherapy. A novel tetracyclic condensed quinoline compound, 8-methoxypyrimido [4',5':4,5]thieno(2,3-b) quinoline-4(3H)-one (MPTQ) is a structural analogue of an anticancer drug ellipticine and has been reported to posses anticancer property. Study on MPTQ on neuroblastoma cells is very limited and mechanisms related to its cytotoxicity on neuroblastoma cells are completely unknown. Here, we evaluated the anticancer property of MPTQ on mouse neuro 2a and human SH-SY5Y neuroblastoma cells and investigated the mechanisms underlying MPTQ-mediated neuro 2a cell death. MPTQ-mediated neuro 2a and SH-SY5Y cell deaths were found to be dose and time dependent. Moreover, MPTQ induced cell death reached approximately 99.8% and 90% in neuro 2a and SH-SY5Y cells respectively. Nuclear oligonucleosomal DNA fragmentation and Terminal dUTP Nick End Labelling assays indicated MPTQ-mediated neuro 2a cell death involved apoptosis. MPTQ-mediated apoptosis is associated with increased phosphorylation of p53 at Ser15 and Ser20 which correlates with the hyperphosphorylation of Ataxia-Telangiectasia mutated protein (ATM). Immunocytochemical analysis demonstrated the increased level of Bax protein in MPTQ treated neuro 2a cells. MPTQ-mediated apoptosis is also associated with increased activation of caspase-9, -3 and -7 but not caspase-2 and -8. Furthermore, increased level of caspase-3 and cleaved Poly (ADP Ribose) polymerase were observed in the nucleus of MPTQ treated neuro 2a cells, suggesting the involvement of caspase-dependent intrinsic but not extrinsic apoptotic pathway. Increased nuclear translocation of apoptosis inducing factor suggests additional involvement of caspase-independent apoptosis pathway in MPTQ treated neuro 2a cells. Collectively, MPTQ-induced neuro 2a cell death is mediated by ATM and p53 activation, and Bax-mediated activation of caspase-dependent and caspase-independent mitochondrial apoptosis pathways.
Collapse
Affiliation(s)
- Upasana Sahu
- Division of Molecular and Cellular Neuroscience, National Brain Research Centre, Manesar, Haryana, India
| | - Himakshi Sidhar
- Division of Molecular and Cellular Neuroscience, National Brain Research Centre, Manesar, Haryana, India
| | - Pankaj S. Ghate
- Division of Molecular and Cellular Neuroscience, National Brain Research Centre, Manesar, Haryana, India
| | - Gopal M. Advirao
- Department of Biochemistry, Kuvempu University, Davanagere, Karnataka, India
| | - Sathees C. Raghavan
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Ranjit K. Giri
- Division of Molecular and Cellular Neuroscience, National Brain Research Centre, Manesar, Haryana, India
| |
Collapse
|
29
|
Vélez C, Cox O, Rosado-Berrios CA, Molina D, Arroyo L, Carro S, Filikov A, Kumar V, Malhotra SV, Cordero M, Zayas B. Novel Nitrobenzazolo[3,2- a]quinolinium Salts Induce Cell Death through a Mechanism Involving DNA Damage, Cell Cycle Changes, and Mitochondrial Permeabilization. OPEN JOURNAL OF APOPTOSIS 2013; 2:13-22. [PMID: 25243104 PMCID: PMC4169051 DOI: 10.4236/ojapo.2013.22002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This study reports the capacity of three nitro substituted benzazolo[3,2-a]quinolinium salts NBQs: NBQ 95 (NSC-763304), NBQ 38 (NSC 763305), and NBQ 97 (NSC-763306) as potential antitumor agents. NBQ's are unnatural alkaloids possessing a positive charge that could facilitate interaction with cell organelles. The anticancer activities of these compounds were evaluated through the National Cancer Institute (NCI) 60 cell line screening which represents diverse histologies. The screening was performed at 10 µM on all cell lines. Results from the NCI screening indicated cytotoxicity activity on six cell lines. In order to explore a possible mechanism of action, a detailed biological activity study of NBQ 95 and NBQ 38 was performed on A431 human epidermoid carcinoma cells to determine an apoptotic pathway involving, cell cycle changes, DNA fragmentation, mutations, mitochondrial membrane permeabilization and caspases activation. DNA fragmentation, cell cycle effects, mutagenesis, mitochondrial permeabilization and activation of caspases were determined by fluorimetry and differential imaging. Our data showed that A431 growth was inhibited with an average IC50 of 30 µM. In terms of the mechanism, these compounds interacted with DNA causing fragmentation and cell cycle arrest at sub G0/G1 stage. Mutagenesis was higher for NBQ 38 and moderate for NBQ 95 Mitochon-drial permeabilization was observed with NBQ 38 and slightly for NBQ 95. Both compounds caused activation of Caspases 3 and 7 suggesting an apoptotic cell death pathway through an intrinsic mechanism. This study reports evidence of the toxicity of these novel compounds with overlapping structural and mechanistic similarities to ellipticine, a known anti-tumor compound.
Collapse
Affiliation(s)
| | - Osvaldo Cox
- Universidad Metropolitana, San Juan, Puerto Rico
| | | | | | - Luz Arroyo
- Universidad Metropolitana, San Juan, Puerto Rico
| | - Sujey Carro
- University of North Carolina, Chapel Hill, USA
| | - Anton Filikov
- Laboratory of Synthetic Chemistry-SAIC, Frederick National Laboratory for Cancer Research, Frederick, USA
| | - Vineet Kumar
- Laboratory of Synthetic Chemistry-SAIC, Frederick National Laboratory for Cancer Research, Frederick, USA
| | - Sanjay V. Malhotra
- Laboratory of Synthetic Chemistry-SAIC, Frederick National Laboratory for Cancer Research, Frederick, USA
| | | | | |
Collapse
|
30
|
Andrews WJ, Panova T, Normand C, Gadal O, Tikhonova IG, Panov KI. Old drug, new target: ellipticines selectively inhibit RNA polymerase I transcription. J Biol Chem 2013; 288:4567-82. [PMID: 23293027 DOI: 10.1074/jbc.m112.411611] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Transcription by RNA polymerase I (Pol-I) is the main driving force behind ribosome biogenesis, a fundamental cellular process that requires the coordinated transcription of all three nuclear polymerases. Increased Pol-I transcription and the concurrent increase in ribosome biogenesis has been linked to the high rates of proliferation in cancers. The ellipticine family contains a number of potent anticancer therapeutic agents, some having progressed to stage I and II clinical trials; however, the mechanism by which many of the compounds work remains unclear. It has long been thought that inhibition of Top2 is the main reason behind the drugs antiproliferative effects. Here we report that a number of the ellipticines, including 9-hydroxyellipticine, are potent and specific inhibitors of Pol-I transcription, with IC(50) in vitro and in cells in the nanomolar range. Essentially, the drugs did not affect Pol-II and Pol-III transcription, demonstrating a high selectivity. We have shown that Pol-I inhibition occurs by a p53-, ATM/ATR-, and Top2-independent mechanism. We discovered that the drug influences the assembly and stability of preinitiation complexes by targeting the interaction between promoter recognition factor SL1 and the rRNA promoter. Our findings will have an impact on the design and development of novel therapeutic agents specifically targeting ribosome biogenesis.
Collapse
Affiliation(s)
- William J Andrews
- School of Biological Sciences, The Queen’s University Belfast, Belfast BT9 7BL, United Kingdom
| | | | | | | | | | | |
Collapse
|
31
|
Stiborova M, Poljakova J, Eckschlager T, Kizek R, Frei E. Analysis of covalent ellipticine- and doxorubicin-derived adducts in DNA of neuroblastoma cells by the ³²P-postlabeling technique. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2012; 156:115-21. [PMID: 22837132 DOI: 10.5507/bp.2012.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Ellipticine and doxorubicin are antineoplastic agents, whose action is based mainly on DNA damage such as intercalation, inhibition of topoisomerase II and formation of covalent DNA adducts. The key target to resolve which of these mechanisms are responsible for ellipticine and doxorubicin anticancer effects is the development of suitable methods for identifying their individual DNA-damaging effects. Here, the (32)P-postlabeling method was tested to detect covalent DNA adducts formed by ellipticine and doxorubicin. METHODS The standard procedure of (32)P-postlabeling assay, this procedure under ATP-deficient conditions, the version using extraction of adducts with n-butanol and the nuclease P1 enrichment version were used to analyze ellipticineand/ or doxorubicin-derived DNA adducts. RESULTS Two covalent ellipticine-derived DNA adducts, which are associated with cytotoxicity of ellipticine to human UKF-NB-3 and UKF-NB-4 neuroblastoma cell lines, were detected by the (32)P-postlabeling method. These adducts are identical to those formed by the ellipticine metabolites, 13-hydroxy- and 12-hydroxyellipticine. In contrast, no covalent adducts formed by doxorubicin in DNA of these neuroblastoma cells and in DNA incubated with this drug and formaldehyde in vitro were detectable by the (32)P-postlabeling assay. CONCLUSIONS The results presented in this paper are the first to demonstrate that in contrast to covalent DNA adducts formed by ellipticine, the adducts generated by formaldehyde-mediated covalent binding of doxorubicin to DNA are not detectable by the (32)P-postlabeling assay. No DNA adducts were, detectable either in vitro, in incubations of DNA with doxorubicin or in DNA of neuroblastoma cells treated with this drug. The results also suggest that covalent binding of ellipticine to DNA of UKF-NB-3 and UKF-NB-4 neuroblastoma cell lines is the predominant mechanism responsible for the cytotoxicity of this drug. To understand the mechanisms of doxorubicin anticancer effects on neuroblastoma cells, development of novel methods for identifying covalent doxorubicin-derived DNA adducts is the major challenge for further research.
Collapse
Affiliation(s)
- Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic.
| | | | | | | | | |
Collapse
|
32
|
Ma W, Lu S, Pan P, Sadatmousavi P, Yuan Y, Chen P. Pharmacokinetics of peptide mediated delivery of anticancer drug ellipticine. PLoS One 2012; 7:e43684. [PMID: 22952737 PMCID: PMC3432029 DOI: 10.1371/journal.pone.0043684] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 07/26/2012] [Indexed: 11/19/2022] Open
Abstract
The amino acid pairing peptide EAK16-II (EAK) has shown the ability to stabilize the hydrophobic anticancer agent ellipticine (EPT) in aqueous solution. In this study, we investigate pharmacokinetics of the formulation of EAK-EPT complexes in vivo. The developed formulation can achieve a sufficiently high drug concentration required in vivo animal models. The nanostructure and surface properties of EAK-EPT complexes or nanoparticle were characterized by transmission electron microscopy (TEM) and zeta potential measurements, respectively. 12 healthy male SD rats were divided into EPT group and EAK-EPT group randomly. Rats in EPT group were tail intravenously injected with the EPT (20 mg/kg); rats in EAK-EPT group were injected with EAK-EPT complexes (EPT's concentration is 20 mg/kg). EPT was extracted from rat plasma with dexamethasone sodium phosphate as internal standards (IS). The pharmacokinetic parameters were obtained using high pressure liquid chromatography (HPLC). Significant differences in main pharmacokinetic parameters between EPT and EAK-EPT complexes were observed, demonstrating that the complexation with EAK prolongs the residence time of the drug and enlarges the area under the concentration-time curve (AUC). This means that EAK can serve as a suitable carrier to increase the bioavailability of EPT.
Collapse
Affiliation(s)
- Weina Ma
- Department of Pharmacy, No. 3 People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Sheng Lu
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Pei Pan
- Department of Pharmacy, No. 3 People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Parisa Sadatmousavi
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Yongfang Yuan
- Department of Pharmacy, No. 3 People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- * E-mail:
| | - P. Chen
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
33
|
Ellipticine oxidation and DNA adduct formation in human hepatocytes is catalyzed by human cytochromes P450 and enhanced by cytochrome b5. Toxicology 2012; 302:233-41. [PMID: 22917556 DOI: 10.1016/j.tox.2012.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 08/05/2012] [Accepted: 08/06/2012] [Indexed: 11/24/2022]
Abstract
Ellipticine is an antineoplastic agent considered a pro-drug, the pharmacological and genotoxic effects of which are dependent on cytochrome P450 (CYP)- and/or peroxidase-mediated activation to covalent DNA adducts. We investigated whether ellipticine-DNA adducts are formed in human hepatic microsomes and human hepatocytes. We then identified which human CYPs oxidize ellipticine to metabolites forming DNA adducts and the effect of cytochrome b(5) on this oxidation. 13-Hydroxyellipticine, the metabolite forming the major ellipticine-DNA adduct, was generated mainly by CYP3A4 and 1A1, followed by CYP2D6>2C19>1B1>1A2>2E1 and >2C9. Cytochrome b(5) increased formation of this metabolite by human CYPs, predominantly by CYP1A1, 3A4, 1A2 and 2C19. Formation of 12-hydroxyellipticine is generated mainly by CYP2C19, followed by CYP2C9>3A4>2D6>2E1 and >2A6. Other CYPs were less active (CYP2C8 and 2B6) or did not oxidize ellipticine to this metabolite (CYP1A1, 1A2 and 1B1). CYP2D6 was the most efficient enzyme generating ellipticine N(2)-oxide. CYP3A4 and 1A1 in the presence of cytochrome b(5) are mainly responsible for bioactivation of ellipticine to DNA adduct 1 (formed by ellipticine-13-ylium from 13-hydroxyellipticine), while 12-hydroxyellipticine generated during the CYP2C19-mediated ellipticine oxidation is the predominant metabolite forming ellipticine-12-ylium that generates ellipticine-DNA adduct 2. These ellipticine-DNA adducts were also generated by human hepatic microsomes and in primary human hepatocytes exposed to ellipticine. Ellipticine is toxic to these hepatocytes, decreasing their viability; the IC(50) value of ellipticine in these cells was 0.7 μM. In liver CYP3A4 is the predominant ellipticine activating CYP species, which is expected to result in efficient metabolism after oral ingestion of ellipticine in humans.
Collapse
|
34
|
Abstract
Considerable support exists for the roles of metabolism in modulating the carcinogenic properties of chemicals. In particular, many of these compounds are pro-carcinogens that require activation to electrophilic forms to exert genotoxic effects. We systematically analyzed the existing literature on the metabolism of carcinogens by human enzymes, which has been developed largely in the past 25 years. The metabolism and especially bioactivation of carcinogens are dominated by cytochrome P450 enzymes (66% of bioactivations). Within this group, six P450s--1A1, 1A2, 1B1, 2A6, 2E1, and 3A4--accounted for 77% of the P450 activation reactions. The roles of these P450s can be compared with those estimated for drug metabolism and should be considered in issues involving enzyme induction, chemoprevention, molecular epidemiology, interindividual variations, and risk assessment.
Collapse
|
35
|
Ryvolova M, Adam V, Eckschlager T, Stiborova M, Kizek R. Study of DNA-ellipticine interaction by capillary electrophoresis with laser-induced fluorescence detection. Electrophoresis 2012; 33:1545-9. [DOI: 10.1002/elps.201200056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
| | | | - Tomas Eckschlager
- Department of Paediatric Haematology and Oncology; 2; nd; Faculty of Medicine, Charles University and University Hospital Motol; Prague; Czech Republic
| | - Marie Stiborova
- Department of Biochemistry; Faculty of Science; Charles University; Prague; Czech Republic
| | | |
Collapse
|
36
|
Stiborová M, Indra R, Moserová M, Černá V, Rupertová M, Martínek V, Eckschlager T, Kizek R, Frei E. Cytochrome b5 Increases Cytochrome P450 3A4-Mediated Activation of Anticancer Drug Ellipticine to 13-Hydroxyellipticine Whose Covalent Binding to DNA Is Elevated by Sulfotransferases and N,O-Acetyltransferases. Chem Res Toxicol 2012; 25:1075-85. [DOI: 10.1021/tx3000335] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Marie Stiborová
- Department of Biochemistry,
Faculty of Science, Charles University,
Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Radek Indra
- Department of Biochemistry,
Faculty of Science, Charles University,
Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Michaela Moserová
- Department of Biochemistry,
Faculty of Science, Charles University,
Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Věra Černá
- Department of Biochemistry,
Faculty of Science, Charles University,
Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Martina Rupertová
- Department of Biochemistry,
Faculty of Science, Charles University,
Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Václav Martínek
- Department of Biochemistry,
Faculty of Science, Charles University,
Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Tomáš Eckschlager
- Department of Pediatric Hematology
and Oncology, Charles University and University Hospital Motol, Prague, Czech Republic
| | - René Kizek
- Department of Chemistry and Biochemistry,
Faculty of Agronomy, Mendel University,
Brno, Czech Republic
| | - Eva Frei
- Division
of Preventive Oncology,
National Center for Tumor Diseases, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
37
|
Miller CM, O'Sullivan EC, Devine KJ, McCarthy FO. Synthesis and biological evaluation of novel isoellipticine derivatives and salts. Org Biomol Chem 2012; 10:7912-21. [DOI: 10.1039/c2ob26181b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
38
|
Kizek R, Adam V, Hrabeta J, Eckschlager T, Smutny S, Burda JV, Frei E, Stiborova M. Anthracyclines and ellipticines as DNA-damaging anticancer drugs: Recent advances. Pharmacol Ther 2012; 133:26-39. [DOI: 10.1016/j.pharmthera.2011.07.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 07/22/2011] [Indexed: 12/21/2022]
|
39
|
Caruso A, Chimento A, El-Kashef H, Lancelot JC, Panno A, Pezzi V, Saturnino C, Sinicropi MS, Sirianni R, Rault S. Antiproliferative activity of some 1,4-dimethylcarbazoles on cells that express estrogen receptors: part I. J Enzyme Inhib Med Chem 2011; 27:609-13. [DOI: 10.3109/14756366.2011.603132] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Anna Caruso
- Department of Pharmaceutical Sciences, University of Calabria,
Arcavacata di Rende (CS), Italy
| | - Adele Chimento
- Department of Pharmaco-Biology, University of Calabria,
Arcavacata di Rende (CS), Italy
| | - Hussein El-Kashef
- Chemistry Department, Faculty of Science, Assiut University,
Assiut, Egypt
| | - Jean-Charles Lancelot
- Université de Caen Basse-Normandie, U.F.R. des Sciences Pharmaceutiques, Centre d’Etudes et de Recherche sur le Médicament de Normandie, Boulevard Becquerel,
Caen Cedex, France
| | - Antonella Panno
- Department of Pharmaceutical Sciences, University of Calabria,
Arcavacata di Rende (CS), Italy
| | - Vincenzo Pezzi
- Department of Pharmaco-Biology, University of Calabria,
Arcavacata di Rende (CS), Italy
| | - Carmela Saturnino
- Department of Pharmaceutical and Biomedical Sciences, University of Salerno,
Fisciano (SA), Italy
| | | | - Rosa Sirianni
- Department of Pharmaco-Biology, University of Calabria,
Arcavacata di Rende (CS), Italy
| | - Sylvain Rault
- Université de Caen Basse-Normandie, U.F.R. des Sciences Pharmaceutiques, Centre d’Etudes et de Recherche sur le Médicament de Normandie, Boulevard Becquerel,
Caen Cedex, France
| |
Collapse
|
40
|
Cytochrome b5 shifts oxidation of the anticancer drug ellipticine by cytochromes P450 1A1 and 1A2 from its detoxication to activation, thereby modulating its pharmacological efficacy. Biochem Pharmacol 2011; 82:669-80. [DOI: 10.1016/j.bcp.2011.06.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 06/03/2011] [Accepted: 06/03/2011] [Indexed: 01/19/2023]
|
41
|
Ellipticine cytotoxicity to cancer cell lines - a comparative study. Interdiscip Toxicol 2011; 4:98-105. [PMID: 21753906 PMCID: PMC3131681 DOI: 10.2478/v10102-011-0017-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Revised: 04/20/2011] [Accepted: 04/23/2011] [Indexed: 11/20/2022] Open
Abstract
Ellipticine is a potent antineoplastic agent exhibiting multiple mechanisms of action. This anticancer agent should be considered a pro-drug, whose pharmacological efficiency and/or genotoxic side effects are dependent on its cytochrome P450 (CYP)- and/or peroxidase-mediated activation to species forming covalent DNA adducts. Ellipticine can also act as an inhibitor or inducer of biotransformation enzymes, thereby modulating its own metabolism leading to its genotoxic and pharmacological effects. Here, a comparison of the toxicity of ellipticine to human breast adenocarcinoma MCF-7 cells, leukemia HL-60 and CCRF-CEM cells, neuroblastoma IMR-32, UKF-NB-3 and UKF-NB-4 cells and U87MG glioblastoma cells and mechanisms of its action to these cells were evaluated. Treatment of all cells tested with ellipticine resulted in inhibition of cell growth and proliferation. This effect was associated with formation of two covalent ellipticine-derived DNA adducts, identical to those formed by 13-hydroxy- and 12-hydroxyellipticine, the ellipticine metabolites generated by CYP and peroxidase enzymes, in MCF-7, HL-60, CCRF-CEM, UKF-NB-3, UKF-NB-4 and U87MG cells, but not in neuroblastoma UKF-NB-3 cells. Therefore, DNA adduct formation in most cancer cell lines tested in this comparative study might be the predominant cause of their sensitivity to ellipticine treatment, whereas other mechanisms of ellipticine action also contribute to its cytotoxicity to neuroblastoma UKF-NB-3 cells.
Collapse
|
42
|
DNA and histone deacetylases as targets for neuroblastoma treatment. Interdiscip Toxicol 2011; 3:47-52. [PMID: 21217872 PMCID: PMC2984128 DOI: 10.2478/v10102-010-0010-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 06/14/2010] [Accepted: 06/15/2010] [Indexed: 11/25/2022] Open
Abstract
Neuroblastoma, a tumor of the peripheral sympathetic nervous system, is the most frequent solid extra cranial tumor in children and is a major cause of death from neoplasia in infancy. Still little improvement in therapeutic options has been made, requiring a need for the development of new therapies. In our laboratory, we address still unsettled questions, which of mechanisms of action of DNA-damaging drugs both currently use for treatment of human neuroblastomas (doxorubicin, cis-platin, cyclophosphamide and etoposide) and another anticancer agent decreasing growth of neuroblastomas in vitro, ellipticine, are predominant mechanism(s) responsible for their antitumor action in neuroblastoma cell lines in vitro. Because hypoxia frequently occurs in tumors and strongly correlates with advanced disease and poor outcome caused by chemoresistance, the effects of hypoxia on efficiencies and mechanisms of actions of these drugs in neuroblastomas are also investigated. Since the epigenetic structure of DNA and its lesions play a role in the origin of human neuroblastomas, pharmaceutical manipulation of the epigenome may offer other treatment options also for neuroblastomas. Therefore, the effects of histone deacetylase inhibitors on growth of neuroblastoma and combination of these compounds with doxorubicin, cis-platin, etoposide and ellipticine as well as mechanisms of such effects in human neuroblastona cell lines in vitro are also investigated. Such a study will increase our knowledge to explain the proper function of these drugs on the molecular level, which should be utilized for the development of new therapies for neuroblastomas.
Collapse
|
43
|
Stiborová M, Rupertová M, Frei E. Cytochrome P450- and peroxidase-mediated oxidation of anticancer alkaloid ellipticine dictates its anti-tumor efficiency. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2011; 1814:175-85. [DOI: 10.1016/j.bbapap.2010.05.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 05/22/2010] [Accepted: 05/24/2010] [Indexed: 12/30/2022]
|
44
|
Natural products and their role in cancer therapy. Med Oncol 2010; 28:888-900. [PMID: 20414819 DOI: 10.1007/s12032-010-9528-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Accepted: 03/31/2010] [Indexed: 12/17/2022]
Abstract
This paper documented several natural products that have antineoplastic activities. Detailed descriptions of the mechanism of action of these products are given. Side effects of these natural products are also described. Based on these side effect profiles and their benefits, these natural products could be used as adjuvant cancer therapies. Traditional anticancer agents usually have debilitating side effects. These natural products would be of great benefit due to their anticancer activities. Additionally, some of these natural products can work proactively in preventing cancer formation with their chemopreventive properties. Incorporation into diet can be recommended after discussion with physician and other health care professionals.
Collapse
|
45
|
Caruso A, Lancelot JC, El-Kashef H, Sinicropi MS, Legay R, Lesnard A, Rault S. A rapid and versatile synthesis of novel pyrimido[5,4-b]carbazoles. Tetrahedron 2009. [DOI: 10.1016/j.tet.2009.10.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Byler KG, Wang C, Setzer WN. Quinoline alkaloids as intercalative topoisomerase inhibitors. J Mol Model 2009; 15:1417-26. [PMID: 19424733 DOI: 10.1007/s00894-009-0501-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 03/06/2009] [Indexed: 11/30/2022]
Abstract
Quinoline alkaloids are abundant in the Rutaceae, and many have exhibited cytotoxic activity. Because structurally related antitumor alkaloids such as camptothecin and fagaronine are known to function as intercalative topoisomerase poisons, it is hypothesized that cytotoxic Stauranthus alkaloids may also serve as intercalative topoisomerase inhibitors. To test this hypothesis theoretically, ten Stauranthus quinoline alkaloids were examined for potential intercalation into DNA using a molecular docking approach. Four of the alkaloids (stauranthine, skimmianine, 3',6'-dihydroxy-3',6'-dihydrostauranthine, and trans-3',4'-dihydroxy-3',4'-dihydrostauranthine) were able to intercalatively dock consistently into DNA. In order to probe the intermolecular interactions that may be responsible for intercalation of these quinoline alkaloids, density functional calculations have been carried out using both the B3LYP and M06 functionals. M06 calculations indicated favorable pi-pi interactions between either skimmianine or stauranthine and the guanine-cytosine base pair. Furthermore, the lowest-energy face-to-face orientation of stauranthine with guanine is consistent with favorable dipole-dipole orientations, favorable electrostatic interactions, and favorable frontier molecular orbital interactions. Likewise, the lowest-energy face-to-face orientation of stauranthine with the guanine-cytosine base pair reveals favorable electrostatic interactions as well as frontier molecular orbital interactions. Thus, not only can quinoline alkaloids dock intercalatively into DNA, but the docked orientations are also electronically favorable.
Collapse
Affiliation(s)
- Kendall G Byler
- Department of Chemistry, University of Alabama in Huntsville, 35899, USA
| | | | | |
Collapse
|
47
|
The mechanism of cytotoxicity and DNA adduct formation by the anticancer drug ellipticine in human neuroblastoma cells. Biochem Pharmacol 2009; 77:1466-79. [PMID: 19426684 DOI: 10.1016/j.bcp.2009.01.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 01/26/2009] [Accepted: 01/26/2009] [Indexed: 11/20/2022]
Abstract
Ellipticine is an antineoplastic agent, whose mode of action is based mainly on DNA intercalation, inhibition of topoisomerase II and formation of covalent DNA adducts mediated by cytochromes P450 and peroxidases. Here, the molecular mechanism of DNA-mediated ellipticine action in human neuroblastoma IMR-32, UKF-NB-3 and UKF-NB-4 cancer cell lines was investigated. Treatment of neuroblastoma cells with ellipticine resulted in apoptosis induction, which was verified by the appearance of DNA fragmentation, and in inhibition of cell growth. These effects were associated with formation of two covalent ellipticine-derived DNA adducts, identical to those formed by the cytochrome P450- and peroxidase-mediated ellipticine metabolites, 13-hydroxy- and 12-hydroxyellipticine. The expression of these enzymes at mRNA and protein levels and their ability to generate ellipticine-DNA adducts in neuroblastoma cells were proven, using the real-time polymerase chain reaction, Western blotting analyses and by analyzing ellipticine-DNA adducts in incubations of this drug with neuroblastoma S9 fractions, enzyme cofactors and DNA. The levels of DNA adducts correlated with toxicity of ellipticine to IMR-32 and UKF-NB-4 cells, but not with that to UKF-NB-3 cells. In addition, hypoxic cell culture conditions resulted in a decrease in ellipticine toxicity to IMR-32 and UKF-NB-4 cells and this correlated with lower levels of DNA adducts. Both these cell lines accumulated in S phase, suggesting that ellipticine-DNA adducts interfere with DNA replication. The results demonstrate that among the multiple modes of ellipticine antitumor action, formation of covalent DNA adducts by ellipticine is the predominant mechanism of cytotoxicity to IMR-32 and UKF-NB-4 neuroblastoma cells.
Collapse
|
48
|
Aimová D, Poljaková J, Kotrbová V, Moserová M, Frei E, Arlt VM, Stiborová M. Ellipticine and benzo(a)pyrene increase their own metabolic activation via modulation of expression and enzymatic activity of cytochromes P450 1A1 and 1A2. Interdiscip Toxicol 2008; 1:160-8. [PMID: 21218107 PMCID: PMC2993483 DOI: 10.2478/v10102-010-0033-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Revised: 08/07/2008] [Accepted: 08/18/2008] [Indexed: 11/21/2022] Open
Abstract
Two compounds known to covalently bind to DNA after their activation with cytochromes P450 (CYPs), carcinogenic benzo(a)pyrene (BaP) and an antineoplastic agent ellipticine, were investigated for their potential to induce CYP and NADPH:CYP reductase (POR) enzymes in rodent livers, the main target organ for DNA adduct formation. Two animal models were used in the study: (i) rats as animals mimicking the fate of ellipticine in humans and (ii) mice, especially wild-type (WT) and hepatic POR null (HRN™) mouse lines. Ellipticine and BaP induce expression of CYP1A enzymes in livers of experimental models, which leads to increase in their enzymatic activity. In addition, both compounds are capable of generating DNA adducts, predominantly in livers of studied organisms. As determined by (32)P postlabelling analysis, levels of ellipticine-derived DNA adducts formed in vivo in the livers of HRN™ mice were reduced (by up to 65%) relative to levels in WT mice, indicating that POR mediated CYP enzyme activity is important for the activation of ellipticine. In contrast to these results, 6.4 fold higher DNA binding of BaP was observed in the livers of HRN™ mice than in WT mice. This finding suggests a detoxication role of CYP1A in BaP metabolism in vivo. In in vitro experiments, DNA adduct formation in calf thymus DNA was up to 25 fold higher in incubations of ellipticine or BaP with microsomes from pretreated animals than with controls. This stimulation effect was attributed to induction of CYP1A1/2 enzymes, which are responsible for oxidative activation of both compounds to the metabolites generating major DNA adducts in vitro. Taken together, these results demonstrate that by inducing CYP1A1/2, ellipticine and BaP modulate their own enzymatic metabolic activation and detoxication, thereby modulating their either pharmacological (ellipticine) and/or genotoxic potential (both compounds).
Collapse
Affiliation(s)
- Dagmar Aimová
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Albertov 2030, 128 40 Prague 2, CZECH REPUBLIC
| | - Jitka Poljaková
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Albertov 2030, 128 40 Prague 2, CZECH REPUBLIC
| | - Věra Kotrbová
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Albertov 2030, 128 40 Prague 2, CZECH REPUBLIC
| | - Michaela Moserová
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Albertov 2030, 128 40 Prague 2, CZECH REPUBLIC
| | - Eva Frei
- Division of Molecular Toxicology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, GERMANY
| | - Volker M. Arlt
- Section of Molecular Carcinogenesis, Institute of Cancer Research, Brookes Lawley Building, Sutton, Surrey SM2 5NG, UNITED KINGDOM
| | - Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Albertov 2030, 128 40 Prague 2, CZECH REPUBLIC
| |
Collapse
|
49
|
Holt PA, Chaires JB, Trent JO. Molecular docking of intercalators and groove-binders to nucleic acids using Autodock and Surflex. J Chem Inf Model 2008; 48:1602-15. [PMID: 18642866 DOI: 10.1021/ci800063v] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The molecular docking tools Autodock and Surflex accurately reproduce the crystallographic structures of a collection of small molecule ligands that have been shown to bind nucleic acids. Docking studies were performed with the intercalators daunorubicin and ellipticine and the minor groove binders distamycin and pentamidine. Autodock and Surflex dock daunorubicin and distamycin to their nucleic acid targets within a resolution of approximately 2 A, which is similar to the limit of the crystal structure resolution. However, for the top ranked poses, Autodock and Surflex both dock ellipticine into the correct site but in a different orientation compared to the crystal structure. This appears not only to be partly related to the symmetry of the target nucleic acid, as ellipticine is able to dock from either side of the intercalation site, but also due to the shape of the ligand and docking accuracy. Surflex docks pentamidine in a symmetrically equivalent orientation relative to the crystal structure, while Autodock was able to dock this molecule in the original orientation. In the case of the Surflex docking of pentamidine, the initial rmsd is misleading, given the symmetrical structure of pentamidine. Importantly, the ranking functions of both of these programs are able to return a top pose within approximately 2 A rmsd for daunorubicin, distamycin, and pentamidine and approximately 3 A rmsd for ellipticine compared to their respective crystal structures. Some docking challenges and potential pitfalls are explored, such as the importance of hydrogen treatment on ligands as well as the scoring functions of Autodock and Surflex. Overall for this set of complexes, Surflex is preferred over Autodock for virtual screening, as although the results are comparable, Surflex has significantly faster performance and ease of use under the optimal software conditions tested. These experiments show that molecular docking techniques can be successfully extended to include nucleic acid targets, a finding which has important implications for virtual screening applications and in the design of new small molecules to target therapeutically relevant morphologies of nucleic acids.
Collapse
Affiliation(s)
- Patrick A Holt
- James Graham Brown Cancer Center and the Department of Medicine, University of Louisville, 5239 S. Jackson Street, Louisville, KY 40202, USA
| | | | | |
Collapse
|
50
|
Purnapatre K, Khattar SK, Saini KS. Cytochrome P450s in the development of target-based anticancer drugs. Cancer Lett 2008; 259:1-15. [DOI: 10.1016/j.canlet.2007.10.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Revised: 10/16/2007] [Accepted: 10/17/2007] [Indexed: 11/16/2022]
|