1
|
Flor AC, Wolfgeher DJ, Kron SJ. Noncanonical inhibition of topoisomerase II alpha by oxidative stress metabolites. Redox Biol 2025; 80:103504. [PMID: 39879737 PMCID: PMC11810846 DOI: 10.1016/j.redox.2025.103504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
During its catalytic cycle, the homodimeric ATPase topoisomerase II alpha (TOP2A) cleaves double stranded DNA and remains covalently bound to 5' ends via tyrosine phosphodiester bonds. After passing a second, intact duplex through, TOP2A rejoins the break and releases from the DNA. Thereby, TOP2A can relieve strain accumulated during transcription, replication and chromatin remodeling and disentangle sister chromatids for mitosis. Chemotherapy agents such as etoposide are poisons that trap TOP2A mid-cycle, covalently bound to cleaved DNA, leaving behind DNA double strand breaks and activating DNA damage response. While etoposide has been proposed to stabilize the TOP2A-DNA cleavage complex (TOP2Acc) via interfacial inhibition, we have elucidated a complementary mechanism mediated by the ability of etoposide and other TOP2A poisons to induce oxidative stress. Consequently, lipid peroxidation and accumulation of lipid-derived electrophiles such as 4-hydroxynonenal (HNE) results in covalent modification of TOP2A, both blocking ATPase activity and trapping TOP2Acc. HNE modifies multiple sites on human TOP2A in vitro, including alkylating Cys216 in the ATPase domain in a DNA-dependent fashion. Taken together, our data suggest an underappreciated role for TOP2A as a redox sensor in tumor cells, connecting oxidative stress to DNA damage signaling and thereby creating a target for redox-active drugs.
Collapse
Affiliation(s)
- Amy C Flor
- University of Chicago, Department of Molecular Genetics and Cell Biology, 929 E. 57th Street, Chicago, IL, 60637, USA
| | - Donald J Wolfgeher
- University of Chicago, Department of Molecular Genetics and Cell Biology, 929 E. 57th Street, Chicago, IL, 60637, USA
| | - Stephen J Kron
- University of Chicago, Department of Molecular Genetics and Cell Biology, 929 E. 57th Street, Chicago, IL, 60637, USA.
| |
Collapse
|
2
|
Dougherty A, Hawaz MG, Hoang KG, Trac J, Keck JM, Ayes C, Deweese JE. Exploration of the Role of the C-Terminal Domain of Human DNA Topoisomerase IIα in Catalytic Activity. ACS OMEGA 2021; 6:25892-25903. [PMID: 34660952 PMCID: PMC8515377 DOI: 10.1021/acsomega.1c02083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 09/15/2021] [Indexed: 06/13/2023]
Abstract
Human topoisomerase IIα (TOP2A) is a vital nuclear enzyme involved in resolving knots and tangles in DNA during replication and cell division. TOP2A is a homodimer with a symmetrical, multidomain structure. While the N-terminal and core regions of the protein are well-studied, the C-terminal domain is poorly understood but is involved in enzyme regulation and is predicted to be intrinsically disordered. In addition, it appears to be a major region of post-translational modification and includes several Ser and Thr residues, many of which have not been studied for biochemical effects. Therefore, we generated a series of human TOP2A mutants where we changed specific Ser and Thr residues in the C-terminal domain to Ala, Gly, or Ile residues. We designed, purified, and examined 11 mutant TOP2A enzymes. The amino acid changes were made between positions 1272 and 1525 with 1-7 residues changed per mutant. Several mutants displayed increased levels of DNA cleavage without displaying any change in plasmid DNA relaxation or DNA binding. For example, mutations in the regions 1272-1279, 1324-1343, 1351-1365, and 1374-1377 produced 2-3 times more DNA cleavage in the presence of etoposide than wild-type TOP2A. Further, several mutants displayed changes in relaxation and/or decatenation activity. Together, these results support previous findings that the C-terminal domain of TOP2A influences catalytic activity and interacts with the substrate DNA. Furthermore, we hypothesize that it may be possible to regulate the enzyme by targeting positions in the C-terminal domain. Because the C-terminal domain differs between the two human TOP2 isoforms, this strategy may provide a means for selectively targeting TOP2A for therapeutic inhibition. Additional studies are warranted to explore these results in more detail.
Collapse
Affiliation(s)
- Ashley
C. Dougherty
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Mariam G. Hawaz
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Kristine G. Hoang
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Judy Trac
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Jacob M. Keck
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Carmen Ayes
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Joseph E. Deweese
- Department
of Pharmaceutical Sciences, Lipscomb University
College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland
Avenue, Nashville, Tennessee 37232-0146, United States
| |
Collapse
|
3
|
Zhang W, Gou P, Dupret JM, Chomienne C, Rodrigues-Lima F. Etoposide, an anticancer drug involved in therapy-related secondary leukemia: Enzymes at play. Transl Oncol 2021; 14:101169. [PMID: 34243013 PMCID: PMC8273223 DOI: 10.1016/j.tranon.2021.101169] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 01/13/2023] Open
Abstract
Etoposide is a semi-synthetic glycoside derivative of podophyllotoxin, also known as VP-16. It is a widely used anticancer medicine in clinics. Unfortunately, high doses or long-term etoposide treatment can induce therapy-related leukemia. The mechanism by which etoposide induces secondary hematopoietic malignancies is still unclear. In this article, we review the potential mechanisms of etoposide induced therapy-related leukemia. Etoposide related leukemogenesis is known to depend on reactive oxidative metabolites of etoposide, notably etoposide quinone, which interacts with cellular proteins such as topoisomerases II (TOP2), CREB-binding protein (CREBBP), and T-Cell Protein Tyrosine Phosphatase (TCPTP). CYP3A4 and CYP3A5 metabolize etoposide to etoposide catechol, which readily oxidizes to etoposide quinone. As a poison of TOP2 enzymes, etoposide and its metabolites induce DNA double-stranded breaks (DSB), and the accumulation of DSB triggers cell apoptosis. If the cell survives, the DSB gives rise to the likelihood of faulty DNA repair events. The gene translocation could occur in mixed-lineage leukemia (MLL) gene, which is well-known in leukemogenesis. Recently, studies have revealed that etoposide metabolites, especially etoposide quinone, can covalently bind to cysteines residues of CREBBP and TCPTP enzymes, . This leads to enzyme inhibition and further affects histone acetylation and phosphorylation of the JAK-STAT pathway, thus putatively altering the proliferation and differentiation of hematopoietic stem cells (HSC). In brief, current studies suggest that etoposide and its metabolites contribute to etoposide therapy-related leukemia through TOP2 mediated DSB and impairs specific enzyme activity, such as CREBBP and TCPTP.
Collapse
Affiliation(s)
- Wenchao Zhang
- Université de Paris, BFA, UMR 8251, CNRS, Paris F-75013, France.
| | - Panhong Gou
- Inserm UMR-S1131, Université de Paris, IRSL, Hôpital Saint-Louis, Paris, France
| | | | - Christine Chomienne
- Inserm UMR-S1131, Université de Paris, IRSL, Hôpital Saint-Louis, Paris, France; Service de Biologie Cellulaire, Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Saint Louis, Paris, France
| | | |
Collapse
|
4
|
Vann KR, Oviatt AA, Osheroff N. Topoisomerase II Poisons: Converting Essential Enzymes into Molecular Scissors. Biochemistry 2021; 60:1630-1641. [PMID: 34008964 PMCID: PMC8209676 DOI: 10.1021/acs.biochem.1c00240] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The extensive length, compaction, and interwound nature of DNA, together with its controlled and restricted movement in eukaryotic cells, create a number of topological issues that profoundly affect all of the functions of the genetic material. Topoisomerases are essential enzymes that modulate the topological structure of the double helix, including the regulation of DNA under- and overwinding and the removal of tangles and knots from the genome. Type II topoisomerases alter DNA topology by generating a transient double-stranded break in one DNA segment and allowing another segment to pass through the DNA gate. These enzymes are involved in a number of critical nuclear processes in eukaryotic cells, such as DNA replication, transcription, and recombination, and are required for proper chromosome structure and segregation. However, because type II topoisomerases generate double-stranded breaks in the genetic material, they also are intrinsically dangerous enzymes that have the capacity to fragment the genome. As a result of this dualistic nature, type II topoisomerases are the targets for a number of widely prescribed anticancer drugs. This article will describe the structure and catalytic mechanism of eukaryotic type II topoisomerases and will go on to discuss the actions of topoisomerase II poisons, which are compounds that stabilize DNA breaks generated by the type II enzyme and convert these essential enzymes into "molecular scissors." Topoisomerase II poisons represent a broad range of structural classes and include anticancer drugs, dietary components, and environmental chemicals.
Collapse
Affiliation(s)
- Kendra R Vann
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Alexandria A Oviatt
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Neil Osheroff
- Departments of Biochemistry and Medicine (Hematology/Oncology), Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- VA Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United States
| |
Collapse
|
5
|
Tremblay T, St-Gelais J, Houde M, Giguère D. Polyfluoroglycoside Synthesis via Simple Alkylation of an Anomeric Hydroxyl Group: Access to Fluoroetoposide Analogues. J Org Chem 2021; 86:4812-4824. [DOI: 10.1021/acs.joc.0c02841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Thomas Tremblay
- Département de Chimie, Université Laval, PROTEO, 1045 av. De la Médecine, Québec City, QC, Canada G1 V 0A6
| | - Jacob St-Gelais
- Département de Chimie, Université Laval, PROTEO, 1045 av. De la Médecine, Québec City, QC, Canada G1 V 0A6
| | - Maxime Houde
- Département de Chimie, Université Laval, PROTEO, 1045 av. De la Médecine, Québec City, QC, Canada G1 V 0A6
| | - Denis Giguère
- Département de Chimie, Université Laval, PROTEO, 1045 av. De la Médecine, Québec City, QC, Canada G1 V 0A6
| |
Collapse
|
6
|
Murphy MB, Kumar P, Bradley AM, Barton CE, Deweese JE, Mercer SL. Synthesis and evaluation of etoposide and podophyllotoxin analogs against topoisomerase IIα and HCT-116 cells. Bioorg Med Chem 2020; 28:115773. [PMID: 33035756 DOI: 10.1016/j.bmc.2020.115773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/18/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022]
Abstract
Etoposide is a widely-used anticancer agent that targets human type II topoisomerases. Evidence suggests that metabolism of etoposide in myeloid progenitor cells is associated with translocations involved in leukemia development. Previous studies suggest halogenation at the C-2' position of etoposide reduces metabolism. Halogens were introduced into the C-2' position by electrophilic aromatic halogenation onto etoposide (ETOP, 1), podophyllotoxin (PPT, 2), and 4-dimethylepipodophyllotoxin (DMEP, 3), and to bridge the gap of knowledge regarding the activity of these metabolically stable analogs. Five halogenated analogs (6-10) were synthesized. Analogs 8-10 displayed variable ability to inhibit DNA relaxation. Analog 9 was the only analog to show concentration-dependent enhancement of Top2-mediated DNA cleavage. Dose response assay results indicated that 8 and 10 were most effective at decreasing the viability of HCT-116 and A549 cancer cell lines in culture. Flow cytometry with 8 and 10 in HCT-116 cells provide evidence of sub-G1 cell populations indicative of apoptosis. Taken together, these results indicate C-2' halogenation of etoposide and its precursors, although metabolically stable, decreases overall activity relative to etoposide.
Collapse
Affiliation(s)
- Matthew B Murphy
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, One University, Park Drive, Nashville, TN 37204, USA
| | - Priyanka Kumar
- Department of Biology, Belmont University, 1900 Belmont Boulevard, Nashville, TN 37212, USA
| | - Amber M Bradley
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, One University, Park Drive, Nashville, TN 37204, USA
| | - Christopher E Barton
- Department of Biology, Belmont University, 1900 Belmont Boulevard, Nashville, TN 37212, USA
| | - Joseph E Deweese
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, One University, Park Drive, Nashville, TN 37204, USA; Departments of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37235, USA
| | - Susan L Mercer
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, One University, Park Drive, Nashville, TN 37204, USA; Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37235, USA.
| |
Collapse
|
7
|
Machado NM, Ribeiro AB, Nicolella HD, Ozelin SD, Silva LHDD, Guissone APP, Rinaldi-Neto F, Lemos ILL, Furtado RA, Cunha WR, Rezende AAAD, Spanó MA, Tavares DC. Usnic acid attenuates genomic instability in Chinese hamster ovary (CHO) cells as well as chemical-induced preneoplastic lesions in rat colon. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2019; 82:401-410. [PMID: 31066341 DOI: 10.1080/15287394.2019.1613274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Usnic acid (UA) is one of the pharmacologically most important compounds produced by several lichen species. To better understand the mechanism of action (MOA) of this important substance, this study examined the genotoxicity attributed to UA and its influence on mutagens with varying MOA using the micronucleus (MN) test in Chinese hamster ovary cells (CHO). Additional experiments were conducted to investigate the effect of UA on colon carcinogenesis in Wistar rats employing the aberrant crypt focus (ACF) assay. In vitro studies showed a significant increase in the frequency of MN in cultures treated with the highest UA concentration tested (87.13 µM). In contrast, UA concentrations of 10.89, 21.78, or 43.56 µM produced an approximate 60% reduction in chromosomal damage induced by doxorubicin, hydrogen peroxide, and etoposide, indicating an antigenotoxic effect. In the ACF assay, male Wistar rats treated with different UA doses (3.125, 12.5, or 50 mg/kg b.w.) and with the carcinogen 1,2-dimethylhydrazine exhibited a significantly lower incidence of neoplastic lesions in the colon than animals treated only with the carcinogen. Data suggest that the MOA responsible for the chemopreventive effect of UA may be related to interaction with DNA topoisomerase II and/or the antioxidant potential of the compound.
Collapse
Affiliation(s)
- Nayane Moreira Machado
- a Institute of Biotechnology , Federal University of Uberlândia , Uberlândia , MG , Brazil
| | | | | | | | | | | | | | | | | | | | - Alexandre Azenha Alves De Rezende
- a Institute of Biotechnology , Federal University of Uberlândia , Uberlândia , MG , Brazil
- c Faculty of Integrated Sciences of Pontal , Federal University of Uberlândia , Ituiutaba , MG , Brazil
| | - Mário Antônio Spanó
- a Institute of Biotechnology , Federal University of Uberlândia , Uberlândia , MG , Brazil
| | | |
Collapse
|
8
|
Association of nephrotoxicity during platinum-etoposide doublet therapy with UGT1A1 polymorphisms in small cell lung cancer patients. Lung Cancer 2018; 126:156-161. [PMID: 30527181 DOI: 10.1016/j.lungcan.2018.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/30/2018] [Accepted: 11/04/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Etoposide is a key agent in the treatment of small cell lung cancer (SCLC). Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. We therefore examined the relation between UGT1A1 polymorphisms and toxicity profile during platinum-etoposide doublet therapy in SCLC patients. MATERIALS AND METHODS SCLC patients who underwent platinum-etoposide doublet therapy and molecular testing for UGT1A1 genotype were reviewed for the occurrence of adverse events during treatment. RESULTS A total of 41 SCLC patients received platinum-etoposide doublet therapy and were genotyped for UGT1A1*6 and UGT1A1*28 alleles. These alleles were detected in 15 (36.6%) patients, with the genotypes of *6/-, *6/*6, *28/-, *28/*28, or *6/*28 being observed in 9 (22.0%), 2 (4.9%), 2 (4.9%), 1 (2.4%), and 1 (2.4%) patients, respectively. The presence of these alleles was significantly associated with an increase in serum creatinine concentration of grade ≥2 (incidence of 66.7% for patients with the alleles versus 11.5% for those without, P < 0.001). Multivariate analysis also showed that these UGT1A1 alleles were significantly associated with therapy-induced nephrotoxicity (odds ratio of 19.30, 95% confidence interval of 2.50-149.00, P < 0.005). Although the differences did not achieve statistical significance, the incidence of other severe toxicities including febrile neutropenia was also slightly higher in patients with the UGT1A1*6 or UGT1A1*28 alleles than in those without them. CONCLUSION Our results reveal an association between UGT1A1 polymorphisms and toxicity of platinum-etoposide doublet therapy in SCLC patients, suggesting that close monitoring for toxicity, especially nephrotoxicity, is warranted for patients with such variant alleles receiving this treatment.
Collapse
|
9
|
Bolton JL, Dunlap TL, Dietz BM. Formation and biological targets of botanical o-quinones. Food Chem Toxicol 2018; 120:700-707. [PMID: 30063944 PMCID: PMC6643002 DOI: 10.1016/j.fct.2018.07.050] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 01/12/2023]
Abstract
The formation of o-quinones from direct 2-electron oxidation of catechols and/or two successive one electron oxidations could explain the cytotoxic/genotoxic and/or chemopreventive effects of several phenolic botanical extracts. For example, poison ivy contains urushiol, an oily mixture, which is oxidized to various o-quinones likely resulting in skin toxicity through oxidative stress and alkylation mechanisms resulting in immune responses. Green tea contains catechins which are directly oxidized to o-quinones by various oxidative enzymes. Alternatively, phenolic botanicals could be o-hydroxylated by P450 to form catechols in vivo which are oxidized to o-quinones. Examples include, resveratrol which is oxidized to piceatannol and further oxidized to the o-quinone. Finally, botanical o-quinones can be formed by O-dealkylation of O-alkoxy groups or methylenedioxy rings resulting in catechols which are further oxidized to o-quinones. Examples include safrole, eugenol, podophyllotoxin and etoposide, as well as methysticin. Once formed these o-quinones have a variety of biological targets in vivo resulting in various biological effects ranging from chemoprevention - > no effect - > toxicity. This U-shaped biological effect curve has been described for a number of reactive intermediates including o-quinones. The current review summarizes the latest data on the formation and biological targets of botanical o-quinones.
Collapse
Affiliation(s)
- Judy L Bolton
- Department of Medicinal Chemistry and Pharmacognosy (M/C 781), College of Pharmacy, University of Illinois at Chicago, 833S. Wood Street, Chicago, IL, 60612-7231, United States.
| | - Tareisha L Dunlap
- Department of Medicinal Chemistry and Pharmacognosy (M/C 781), College of Pharmacy, University of Illinois at Chicago, 833S. Wood Street, Chicago, IL, 60612-7231, United States
| | - Birgit M Dietz
- Department of Medicinal Chemistry and Pharmacognosy (M/C 781), College of Pharmacy, University of Illinois at Chicago, 833S. Wood Street, Chicago, IL, 60612-7231, United States
| |
Collapse
|
10
|
Wilson JT, Fief CA, Jackson KD, Mercer SL, Deweese JE. HU-331 and Oxidized Cannabidiol Act as Inhibitors of Human Topoisomerase IIα and β. Chem Res Toxicol 2018; 31:137-144. [PMID: 29272108 DOI: 10.1021/acs.chemrestox.7b00302] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Topoisomerase II is a critical enzyme in replication, transcription, and the regulation of chromatin topology. Several anticancer agents target topoisomerases in order to disrupt cell growth. Cannabidiol is a major non-euphoriant, pharmacologically active component of cannabis. Previously, we examined the cannabidiol derivative HU-331 in order to characterize the mechanism of the compound against topoisomerase IIα. In this current work, we explore whether cannabidiol (CBD) impacts topoisomerase II activity, and we additionally examine the activity of these compounds against topoisomerase IIβ. CBD does not appear to strongly inhibit DNA relaxation and is not a poison of topoisomerase II DNA cleavage. However, oxidation of CBD allows this compound to inhibit DNA relaxation by topoisomerase IIα and β without poisoning DNA cleavage. Additionally, we found that oxidized CBD, similar to HU-331, inhibits ATP hydrolysis and can result in inactivation of topoisomerase IIα and β. We also determined that oxidized CBD and HU-331 are both able to stabilize the N-terminal clamp of topoisomerase II. Taken together, we conclude that while CBD does not have significant activity against topoisomerase II, both oxidized CBD and HU-331 are active against both isoforms of topoisomerase II. We hypothesize that oxidized CBD and HU-331 act against the enzyme through interaction with the N-terminal ATPase domain. According to the model we propose, topoisomerase II inactivation may result from a decrease in the ability of the enzyme to bind to DNA when the compound is bound to the N-terminus.
Collapse
Affiliation(s)
- James T Wilson
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Lipscomb University , Nashville, Tennessee 37204-3951, United States
| | - Cole A Fief
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Lipscomb University , Nashville, Tennessee 37204-3951, United States
| | - Klarissa D Jackson
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Lipscomb University , Nashville, Tennessee 37204-3951, United States
| | - Susan L Mercer
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Lipscomb University , Nashville, Tennessee 37204-3951, United States
| | - Joseph E Deweese
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Lipscomb University , Nashville, Tennessee 37204-3951, United States
| |
Collapse
|
11
|
Combination of Near Infrared Light-Activated Photodynamic Therapy Mediated by Indocyanine Green with Etoposide to Treat Non-Small-Cell Lung Cancer. Cancers (Basel) 2017; 9:cancers9060063. [PMID: 28587258 PMCID: PMC5483882 DOI: 10.3390/cancers9060063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/01/2017] [Indexed: 11/17/2022] Open
Abstract
Indocyanine green (ICG) has been reported as a potential near-infrared (NIR) photosensitizer for photodynamic therapy (PDT) of cancer. However the application of ICG-mediated PDT is both intrinsically and physiologically limited. Here we report a combination of ICG-PDT with a chemotherapy drug etoposide (VP-16), aiming to enhance the anticancer efficacy, to circumvent limitations of PDT using ICG, and to reduce side effects of VP-16. We found in controlled in vitro cell-based assays that this combination is effective in killing non-small-cell lung cancer cells (NSCLC, A549 cell line). We also found that the combination of ICG-PDT and VP-16 exhibits strong synergy in killing non-small-cell lung cancer cells partially through inducing more DNA double-strand breaks (DSBs), while it has a much weaker synergy in killing human normal cells (GM05757). Furthermore, by studying the treatment sequence dependence and the cytotoxicity of laser-irradiated mixtures of ICG and VP-16, we found that the observed synergy involves direct/indirect reactions between ICG and VP-16. We further propose that there exists an electron transfer reaction between ICG and VP-16 under irradiation. This study therefore shows the anticancer efficacy of ICG-PDT combined with VP-16. These findings suggest that ICG-mediated PDT may be applied in combination with the chemotherapy drug VP-16 to treat some cancers, especially the non-small-cell lung cancer.
Collapse
|
12
|
Tsend-Ayush A, Zhu X, Ding Y, Yao J, Yin L, Zhou J, Yao J. Lactobionic acid-conjugated TPGS nanoparticles for enhancing therapeutic efficacy of etoposide against hepatocellular carcinoma. NANOTECHNOLOGY 2017; 28:195602. [PMID: 28291743 DOI: 10.1088/1361-6528/aa66ba] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Many effective anti-cancer drugs have limited use in hepatocellular carcinoma (HCC) therapy due to the drug resistance mechanisms in liver cells. In recent years, tumor-targeted drug delivery and the inhibition of drug-resistance-related mechanisms has become an integrated strategy for effectively combating chemo-resistant cancer. Herein, lactobionic acid-conjugated d-α-tocopheryl polyethylene glycol 1000 succinate (TPGS-LA conjugate) has been developed as a potential asialoglycoprotein receptor (ASGPR)-targeted nanocarrier and an efficient inhibitor of P-glycoprotein (P-gp) to enhance etoposide (ETO) efficacy against HCC. The main properties of ETO-loaded TPGS-LA nanoparticles (NPs) were tested through in vitro and in vivo studies after being prepared using the nanoprecipitation method and characterized by dynamic light scattering (DLS). According to the results, smaller (∼141.43 nm), positively charged ETO-loaded TPGS-LA NPs were more suitable for providing efficient delivery to hepatoma cells by avoiding the clearance mechanisms. It was found that ETO-loaded TPGS-LA NPs were noticeably able to enhance the cytotoxicity of ETO in HepG2 cells. Besides this, markedly higher internalization by the ASGPR-overexpressed HepG2 cells and efficient accumulation at the tumor site in vivo were revealed in the TPGS-LA NP group. More importantly, animal studies confirmed that ETO-loaded TPGS-LA NPs achieved the highest therapeutic efficacy against HCC. Interestingly, ETO-loaded TPGS-LA NPs also exhibited a great inhibitory effect on P-gp compared to the ETO-loaded TPGS NPs. These results suggest that TPGS-LA NPs could be used as a potential ETO delivery system against HCC.
Collapse
Affiliation(s)
- Altansukh Tsend-Ayush
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
13
|
|
14
|
Gibson EG, King MM, Mercer SL, Deweese JE. Two-Mechanism Model for the Interaction of Etoposide Quinone with Topoisomerase IIα. Chem Res Toxicol 2016; 29:1541-8. [DOI: 10.1021/acs.chemrestox.6b00209] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Elizabeth G. Gibson
- Department
of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - McKenzie M. King
- Department
of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Susan L. Mercer
- Department
of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| | - Joseph E. Deweese
- Department
of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, One University Park Drive, Nashville, Tennessee 37204-3951, United States
| |
Collapse
|
15
|
Wilson JT, Jiang X, McGill BC, Lisic EC, Deweese JE. Examination of the Impact of Copper(II) α-(N)-Heterocyclic Thiosemicarbazone Complexes on DNA Topoisomerase IIα. Chem Res Toxicol 2016; 29:649-58. [DOI: 10.1021/acs.chemrestox.5b00471] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- James T. Wilson
- Department
of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204-3951, United States
| | - Xiaohua Jiang
- Department
of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Bradley C. McGill
- Department
of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Edward C. Lisic
- Department
of Chemistry, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| | - Joseph E. Deweese
- Department
of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204-3951, United States
- Department
of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| |
Collapse
|
16
|
Regal KM, Mercer SL, Deweese JE. HU-331 is a catalytic inhibitor of topoisomerase IIα. Chem Res Toxicol 2014; 27:2044-51. [PMID: 25409338 DOI: 10.1021/tx500245m] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Topoisomerases are essential enzymes that are involved in DNA metabolism. Topoisomerase II generates transient DNA strand breaks that are stabilized by anticancer drugs, such as doxorubicin, causing an accumulation of DNA damage. However, doxorubicin causes cardiac toxicity and, like etoposide and other topoisomerase II-targeted agents, can induce DNA damage, resulting in secondary cancers. The cannabinoid quinone HU-331 has been identified as a potential anticancer drug that demonstrates more potency in cancer cells with less off-target toxicity than that of doxorubicin. Reports indicate that HU-331 does not promote cell death via apoptosis, cell cycle arrest, caspase activation, or DNA strand breaks. However, the precise mechanism of action is poorly understood. We employed biochemical assays to study the mechanism of action of HU-331 against purified topoisomerase IIα. These assays examined DNA binding, cleavage, ligation, relaxation, and ATPase activities of topoisomerase IIα. Our results demonstrate that HU-331 inhibits topoisomerase IIα-mediated DNA relaxation at micromolar levels. We find that HU-331 does not induce DNA strand breaks in vitro. When added prior to the DNA substrate, HU-331 blocks DNA cleavage and relaxation activities of topoisomerase IIα in a redox-sensitive manner. The action of HU-331 can be blocked, but not reversed, by the presence of dithiothreitol. Our results also show that HU-331 inhibits the ATPase activity of topoisomerase IIα using a noncompetitive mechanism. Preliminary binding studies also indicate that HU-331 decreases the ability of topoisomerase IIα to bind DNA. In summary, HU-331 inhibits relaxation activity without poisoning DNA cleavage. This action is sensitive to reducing agents and appears to involve noncompetitive inhibition of the ATPase activity and possibly inhibition of DNA binding. These studies provide a promising foundation for the exploration of HU-331 as a catalytic inhibitor of topoisomerase IIα.
Collapse
Affiliation(s)
- Kellie M Regal
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences , Nashville, Tennessee 37204-3951, United States
| | | | | |
Collapse
|
17
|
Lindsey RH, Pendleton M, Ashley RE, Mercer SL, Deweese JE, Osheroff N. Catalytic core of human topoisomerase IIα: insights into enzyme-DNA interactions and drug mechanism. Biochemistry 2014; 53:6595-602. [PMID: 25280269 PMCID: PMC4204876 DOI: 10.1021/bi5010816] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Coordination between the N-terminal gate and the catalytic core of topoisomerase II allows the proper capture, cleavage, and transport of DNA during the catalytic cycle. Because the activities of these domains are tightly linked, it has been difficult to discern their individual contributions to enzyme-DNA interactions and drug mechanism. To further address the roles of these domains, we analyzed the activity of the catalytic core of human topoisomerase IIα. The catalytic core and the wild-type enzyme both maintained higher levels of cleavage with negatively (as compared to positively) supercoiled plasmid, indicating that the ability to distinguish supercoil handedness is embedded within the catalytic core. However, the catalytic core alone displayed little ability to cleave DNA substrates that did not intrinsically provide the enzyme with a transport segment (i.e., substrates that did not contain crossovers). Finally, in contrast to interfacial topoisomerase II poisons, covalent poisons did not enhance DNA cleavage mediated by the catalytic core. This distinction allowed us to further characterize the mechanism of etoposide quinone, a drug metabolite that functions primarily as a covalent poison. Etoposide quinone retained some ability to enhance DNA cleavage mediated by the catalytic core, indicating that it still can function as an interfacial poison. These results further define the distinct contributions of the N-terminal gate and the catalytic core to topoisomerase II function. The catalytic core senses the handedness of DNA supercoils during cleavage, while the N-terminal gate is critical for capturing the transport segment and for the activity of covalent poisons.
Collapse
Affiliation(s)
- R Hunter Lindsey
- Department of Biochemistry, ‡Department of Pharmacology, and §Department of Medicine (Hematology/Oncology), Vanderbilt University School of Medicine , Nashville, Tennessee 37232-0146, United States
| | | | | | | | | | | |
Collapse
|
18
|
Kocsis L, Brummond KM. Intramolecular dehydro-Diels-Alder reaction affords selective entry to arylnaphthalene or aryldihydronaphthalene lignans. Org Lett 2014; 16:4158-61. [PMID: 25061845 PMCID: PMC4136723 DOI: 10.1021/ol501853y] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Indexed: 01/20/2023]
Abstract
Intramolecular dehydro-Diels-Alder (DDA) reactions are performed affording arylnaphthalene or aryldihydronaphthalene lactones selectively as determined by choice of reaction solvent. This constitutes the first report of an entirely selective formation of arylnaphthalene lactones utilizing DDA reactions of styrene-ynes. The synthetic utility of the DDA reaction is demonstrated by the synthesis of taiwanin C, retrohelioxanthin, justicidin B, isojusticidin B, and their dihydronaphthalene derivatives. Computational methods for chemical shift assignment are presented that allow for regioisomeric lignans to be distinguished.
Collapse
Affiliation(s)
- Laura
S. Kocsis
- Department
of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Kay M. Brummond
- Department
of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
19
|
Smith NA, Byl JAW, Mercer SL, Deweese JE, Osheroff N. Etoposide quinone is a covalent poison of human topoisomerase IIβ. Biochemistry 2014; 53:3229-36. [PMID: 24766193 PMCID: PMC4033626 DOI: 10.1021/bi500421q] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Etoposide is a topoisomerase II poison
that is utilized to treat
a broad spectrum of human cancers. Despite its wide clinical use,
2–3% of patients treated with etoposide eventually develop
treatment-related acute myeloid leukemias (t-AMLs) characterized by
rearrangements of the MLL gene. The molecular basis
underlying the development of these t-AMLs is not well understood;
however, previous studies have implicated etoposide metabolites (i.e.,
etoposide quinone) and topoisomerase IIβ in the leukemogenic
process. Although interactions between etoposide quinone and topoisomerase
IIα have been characterized, the effects of the drug metabolite
on the activity of human topoisomerase IIβ have not been reported.
Thus, we examined the ability of etoposide quinone to poison human
topoisomerase IIβ. The quinone induced ∼4 times more
enzyme-mediated DNA cleavage than did the parent drug. Furthermore,
the potency of etoposide quinone was ∼2 times greater against
topoisomerase IIβ than it was against topoisomerase IIα,
and the drug reacted ∼2–4 times faster with the β
isoform. Etoposide quinone induced a higher ratio of double- to single-stranded
breaks than etoposide, and its activity was less dependent on ATP.
Whereas etoposide acts as an interfacial topoisomerase II poison,
etoposide quinone displayed all of the hallmarks of a covalent poison:
the activity of the metabolite was abolished by reducing agents, and
the compound inactivated topoisomerase IIβ when it was incubated
with the enzyme prior to the addition of DNA. These results are consistent
with the hypothesis that etoposide quinone contributes to etoposide-related
leukemogenesis through an interaction with topoisomerase IIβ.
Collapse
Affiliation(s)
- Nicholas A Smith
- Departments of †Biochemistry, ‡Medicine (Hematology/Oncology), and §Pharmacology, Vanderbilt University School of Medicine , Nashville, Tennessee 37232-0146, United States
| | | | | | | | | |
Collapse
|