1
|
Tong T, Zhai PS, Qin X, Zhang Z, Li CW, Guo HY, Ma HL. Nuclear TOP1MT Confers Cisplatin Resistance via Pseudogene in HNSCC. J Dent Res 2024; 103:1238-1248. [PMID: 39382100 DOI: 10.1177/00220345241272017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Cisplatin resistance is one of the major causes of treatment failure in head and neck squamous cell carcinoma (HNSCC). There is an urgent need to uncover the underlying mechanism for developing effective treatment strategies. A quantitative proteomics assay was used to identify differential proteins in cisplatin-resistant cells. Mitochondrial topoisomerase I (TOP1MT) localization was determined using laser confocal microscopy and nucleocytoplasmic separation assay. Chromatin immunoprecipitation sequencing, dual-luciferase reporter assay, and RNA immunoprecipitation were used to identify the interaction between pseudogenes, miRNAs, and real genes. In vivo experiments verified the interaction between TOP1MT and pseudogenes on cisplatin resistance. TOP1MT was identified as a driving factor of cisplatin resistance in vitro, in vivo, and in HNSCC patients. Moreover, TOP1MT exceptionally translocated to the nucleus in cisplatin-resistant HNSCC cells in a signal peptide-dependent manner. Nuclear TOP1MT (nTOP1MT) transcriptionally regulated the mitochondrial functional pseudogene MTATP6P1, which bound to miR-137 and miR-491-5p as a competing endogenous RNA (ceRNA) and promoted the expression of MTATP6. An increase in MTATP6 enhanced mitochondrial oxidative phosphorylation (OXPHOS), which conferred cisplatin resistance in HNSCC. Our findings revealed that nTOP1MT transcriptionally activated MTAPT6P1 and increased MTATP6 expression via ceRNA, which facilitated OXPHOS and cisplatin resistance. These results provide novel insight for overcoming cisplatin resistance in HNSCC.
Collapse
Affiliation(s)
- T Tong
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine, PR China
- College of Stomatology, Shanghai Jiao Tong University, PR China
- National Center for Stomatology, PR China
- National Clinical Research Center for Oral Diseases, PR China
- Shanghai Key Laboratory of Stomatology, Shanghai, PR China
- Department of Oral and Maxillofacial Surgery, Shanghai Stomatological Hospital, Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, PR China
| | - P S Zhai
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine, PR China
- College of Stomatology, Shanghai Jiao Tong University, PR China
- National Center for Stomatology, PR China
- National Clinical Research Center for Oral Diseases, PR China
- Shanghai Key Laboratory of Stomatology, Shanghai, PR China
| | - X Qin
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine, PR China
- College of Stomatology, Shanghai Jiao Tong University, PR China
- National Center for Stomatology, PR China
- National Clinical Research Center for Oral Diseases, PR China
- Shanghai Key Laboratory of Stomatology, Shanghai, PR China
| | - Z Zhang
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine, PR China
- College of Stomatology, Shanghai Jiao Tong University, PR China
- National Center for Stomatology, PR China
- National Clinical Research Center for Oral Diseases, PR China
- Shanghai Key Laboratory of Stomatology, Shanghai, PR China
| | - C W Li
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine, PR China
- College of Stomatology, Shanghai Jiao Tong University, PR China
- National Center for Stomatology, PR China
- National Clinical Research Center for Oral Diseases, PR China
- Shanghai Key Laboratory of Stomatology, Shanghai, PR China
| | - H Y Guo
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine
| | - H L Ma
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine, PR China
- College of Stomatology, Shanghai Jiao Tong University, PR China
- National Center for Stomatology, PR China
- National Clinical Research Center for Oral Diseases, PR China
- Shanghai Key Laboratory of Stomatology, Shanghai, PR China
| |
Collapse
|
2
|
Moreira F, Arenas M, Videira A, Pereira F. Evolution of TOP1 and TOP1MT Topoisomerases in Chordata. J Mol Evol 2023; 91:192-203. [PMID: 36651963 PMCID: PMC10081982 DOI: 10.1007/s00239-022-10091-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023]
Abstract
Type IB topoisomerases relax the torsional stress associated with DNA metabolism in the nucleus and mitochondria and constitute important molecular targets of anticancer drugs. Vertebrates stand out among eukaryotes by having two Type IB topoisomerases acting specifically in the nucleus (TOP1) and mitochondria (TOP1MT). Despite their major importance, the origin and evolution of these paralogues remain unknown. Here, we examine the molecular evolutionary processes acting on both TOP1 and TOP1MT in Chordata, taking advantage of the increasing number of available genome sequences. We found that both TOP1 and TOP1MT evolved under strong purifying selection, as expected considering their essential biological functions. Critical active sites, including those associated with resistance to anticancer agents, were found particularly conserved. However, TOP1MT presented a higher rate of molecular evolution than TOP1, possibly related with its specialized activity on the mitochondrial genome and a less critical role in cells. We could place the duplication event that originated the TOP1 and TOP1MT paralogues early in the radiation of vertebrates, most likely associated with the first round of vertebrate tetraploidization (1R). Moreover, our data suggest that cyclostomes present a specialized mitochondrial Type IB topoisomerase. Interestingly, we identified two missense mutations replacing amino acids in the Linker region of TOP1MT in Neanderthals, which appears as a rare event when comparing the genome of both species. In conclusion, TOP1 and TOP1MT differ in their rates of evolution, and their evolutionary histories allowed us to better understand the evolution of chordates.
Collapse
Affiliation(s)
- Filipa Moreira
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos S/N 4450-208, Matosinhos, Portugal
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Miguel Arenas
- Department of Biochemistry, Genetics and Immunology, University of Vigo, 36310, Vigo, Spain
- CINBIO, Universidade de Vigo, 36310, Vigo, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur), 36310, Vigo, Spain
| | - Arnaldo Videira
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Filipe Pereira
- IDENTIFICA Genetic Testing, Rua Simão Bolívar 259 3º Dir Tras, 4470-214, Maia, Portugal.
- Centre for Functional Ecology, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456, Coimbra, Portugal.
| |
Collapse
|
3
|
Dimethylmyricacene: An In Vitro and In Silico Study of a Semisynthetic Non-Camptothecin Derivative Compound, Targeting Human DNA Topoisomerase 1B. Cells 2022; 11:cells11213486. [DOI: 10.3390/cells11213486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Human topoisomerase 1B regulates the topological state of supercoiled DNA enabling all fundamental cell processes. This enzyme, which is the unique molecular target of the natural anticancer compound camptothecin, acts by nicking one DNA strand and forming a transient protein–DNA covalent complex. The interaction of human topoisomerase 1B and dimethylmyricacene, a compound prepared semisynthetically from myricanol extracted from Myrica cerifera root bark, was investigated using enzymatic activity assays and molecular docking procedures. Dimethylmyricacene was shown to inhibit both the cleavage and the religation steps of the enzymatic reaction, and cell viability of A-253, FaDu, MCF-7, HeLa and HCT-116 tumor cell lines.
Collapse
|
4
|
Singh J, Suryan A, Kumar S, Sharma S. Phthalazinone Scaffold: Emerging Tool in the Development of Target Based Novel Anticancer Agents. Anticancer Agents Med Chem 2020; 20:2228-2245. [PMID: 32767957 DOI: 10.2174/1871520620666200807220146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 11/22/2022]
Abstract
Phthalazinones are important nitrogen-rich heterocyclic compounds which have been a topic of considerable medicinal interest because of their diversified pharmacological activities. This versatile scaffold forms a common structural feature for many bioactive compounds, which leads to the design and development of novel anticancer drugs with fruitful results. The current review article discusses the progressive development of novel phthalazinone analogues that are targets for various receptors such as PARP, EGFR, VEGFR-2, Aurora kinase, Proteasome, Hedgehog pathway, DNA topoisomerase and P-glycoprotein. It describes mechanistic insights into the anticancer properties of phthalazinone derivatives and also highlights various simple and economical techniques for the synthesis of phthalazinones.
Collapse
Affiliation(s)
- Jyoti Singh
- Chandigarh College of Pharmacy, Landran, Punjab, India
| | - Amruta Suryan
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | | | - Shweta Sharma
- Chandigarh College of Pharmacy, Landran, Punjab, India
| |
Collapse
|
5
|
Soren BC, Dasari JB, Ottaviani A, Iacovelli F, Fiorani P. Topoisomerase IB: a relaxing enzyme for stressed DNA. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:18-25. [PMID: 35582040 PMCID: PMC9094055 DOI: 10.20517/cdr.2019.106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/10/2019] [Accepted: 12/18/2019] [Indexed: 11/12/2022]
Abstract
DNA topoisomerase I enzymes relieve the torsional strain in DNA; they are essential for fundamental molecular processes such as DNA replication, transcription, recombination, and chromosome condensation; and act by cleaving and then religating DNA strands. Over the past few decades, scientists have focused on the DNA topoisomerases biological functions and established a unique role of Type I DNA topoisomerases in regulating gene expression and DNA chromosome condensation. Moreover, the human enzyme is being investigated as a target for cancer chemotherapy. The active site tyrosine is responsible for initiating two transesterification reactions to cleave and then religate the DNA backbone, allowing the release of superhelical tension. The different steps of the catalytic mechanism are affected by various inhibitors; some of them prevent the interaction between the enzyme and the DNA while others act as poisons, leading to TopI-DNA lesions, breakage of DNA, and eventually cellular death. In this review, our goal is to provide an overview of mechanism of human topoisomerase IB action together with the different types of inhibitors and their effect on the enzyme functionality.
Collapse
Affiliation(s)
- Bini Chhetri Soren
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Jagadish Babu Dasari
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy.,Present address: Department of Research and Application Development, Biogenex Life Sciences, Telangana 501510, India
| | - Alessio Ottaviani
- Institute of Translational Pharmacology, National Research Council, Rome 00133, Italy
| | - Federico Iacovelli
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Paola Fiorani
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy.,Institute of Translational Pharmacology, National Research Council, Rome 00133, Italy
| |
Collapse
|
6
|
Varghese SS, Ghosh SK. Stress-responsive Entamoeba topoisomerase II: a potential antiamoebic target. FEBS Lett 2019; 594:1005-1020. [PMID: 31724164 DOI: 10.1002/1873-3468.13677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/05/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022]
Abstract
Topoisomerases, the ubiquitous enzymes involved in all DNA processes across the biological world, are targets for various anticancer and antimicrobial agents. In Entamoeba histolytica, the causative agent of amebiasis, we found one of seven unexplored putative topoisomerases to be highly upregulated during heat shock and oxidative stress, and also during the late hours of encystation. Further analysis revealed the upregulated enzyme to be a eukaryotic type IIA topoisomerase (TopoII) with demonstrable activity in vitro. This enzyme is localized to newly forming nuclei during encystation. Gene silencing of the TopoII reduces viability and encystation efficiency. Notable susceptibility of Entamoeba TopoII to prokaryotic topoisomerase inhibitors opens up the possibility for exploring this enzyme as a new antiamoebic target.
Collapse
Affiliation(s)
- Sneha Susan Varghese
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Sudip Kumar Ghosh
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
7
|
Machine Learning Models Combined with Virtual Screening and Molecular Docking to Predict Human Topoisomerase I Inhibitors. Molecules 2019; 24:molecules24112107. [PMID: 31167344 PMCID: PMC6601036 DOI: 10.3390/molecules24112107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/20/2019] [Accepted: 05/28/2019] [Indexed: 12/15/2022] Open
Abstract
In this work, random forest (RF), support vector machine, k-nearest neighbor and C4.5 decision tree, were used to establish classification models for predicting whether an unknown molecule is an inhibitor of human topoisomerase I (Top1) protein. All these models have achieved satisfactory results, with total prediction accuracies from 89.70% to 97.12%. Through comparative analysis, it can be found that the RF model has the best forecasting effect. The parameters were further optimized to generate the best-performing RF model. At the same time, features selection was implemented to choose properties most relevant to the inhibition of Top1 from 189 molecular descriptors through a special RF procedure. Subsequently, a ligand-based virtual screening was performed from the Maybridge database by the optimal RF model and 596 hits were picked out. Then, 67 molecules with relative probability scores over 0.7 were selected based on the screening results. Next, the 67 molecules above were docked to Top1 using AutoDock Vina. Finally, six top-ranked molecules with binding energies less than −10.0 kcal/mol were screened out and a common backbone, which is entirely different from that of existing Top1 inhibitors reported in the literature, was found.
Collapse
|
8
|
Bansal S, Sur S, Tandon V. Benzimidazoles: Selective Inhibitors of Topoisomerase I with Differential Modes of Action. Biochemistry 2018; 58:809-817. [DOI: 10.1021/acs.biochem.8b01102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Sandhya Bansal
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Souvik Sur
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Vibha Tandon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
9
|
Sinha P, Kumari N, Singh K, Singh K, Mishra L. Homoleptic bisterpyridyl complexes: Synthesis, characterization, DNA binding, DNA cleavage and topoisomerase II inhibition activity. Inorganica Chim Acta 2015. [DOI: 10.1016/j.ica.2015.03.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
10
|
|
11
|
He X, Zeng L, Yang G, Xie L, Sun X, Tan L. DNA binding, photocleavage and topoisomerase inhibitory activity of polypyridyl ruthenium(II) complexes containing the same ancillary ligand and different main ligands. Inorganica Chim Acta 2013. [DOI: 10.1016/j.ica.2013.08.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
12
|
Lee CC, Chang DM, Huang KF, Chen CL, Chen TC, Lo Y, Guh JH, Huang HS. Design, synthesis and antiproliferative evaluation of fluorenone analogs with DNA topoisomerase I inhibitory properties. Bioorg Med Chem 2013; 21:7125-33. [PMID: 24094433 DOI: 10.1016/j.bmc.2013.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/03/2013] [Accepted: 09/03/2013] [Indexed: 11/29/2022]
Abstract
A series of 2,7-diamidofluorenones were designed, synthesized, and screened by SRB assay. Some synthesized compounds exhibited antitumor activities in submicromolar range. Ten compounds (3a, 3b, 3c, 3g, 3j, 3l, 4a, 4h, 4i, and 4j) were also selected by NCI screening system and 3c (GI50=1.66 μM) appeared to be the most active agent of this series. Furthermore, 3c attenuated topoisomerase I-mediated DNA relaxation at low micromolar concentrations. These results indicated that fluorenones have potential to be further developed into anticancer drugs.
Collapse
Affiliation(s)
- Chia-Chung Lee
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
13
|
A kinetic clutch governs religation by type IB topoisomerases and determines camptothecin sensitivity. Proc Natl Acad Sci U S A 2012; 109:16125-30. [PMID: 22991469 DOI: 10.1073/pnas.1206480109] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Type IB topoisomerases (Top1Bs) relax excessive DNA supercoiling associated with replication and transcription by catalyzing a transient nick in one strand to permit controlled rotation of the DNA about the intact strand. The natural compound camptothecin (CPT) and the cancer chemotherapeutics derived from it, irinotecan and topotecan, are highly specific inhibitors of human nuclear Top1B (nTop1). Previous work on vaccinia Top1B led to an elegant model that describes a straightforward dependence of rotation and religation on the torque caused by supercoiling. Here, we used a single-molecule DNA supercoil relaxation assay to measure the torque dependence of nTop1 and its inhibition by CPT. For comparison, we also examined mitochondrial Top1B and an N-terminal deletion mutant of nTop1. Despite substantial sequence homology in their core domains, nTop1 and mitochondrial Top1B exhibit dramatic differences in sensitivity to torque and CPT, with the N-terminal deletion mutant of nTop1 showing intermediate characteristics. In particular, nTop1 displays nearly torque-independent religation probability, distinguishing it from other Top1B enzymes studied to date. Kinetic modeling reveals a hitherto unobserved torque-independent transition linking the DNA rotation and religation phases of the enzymatic cycle. The parameters of this transition determine the torque sensitivity of religation and the efficiency of CPT binding. This "kinetic clutch" mechanism explains the molecular basis of CPT sensitivity and more generally provides a framework with which to interpret Top1B activity and inhibition.
Collapse
|
14
|
Synthesis, DNA-binding and topoisomerase inhibitory activity of ruthenium(II) polypyridyl complexes. Eur J Med Chem 2011; 46:1056-65. [DOI: 10.1016/j.ejmech.2011.01.019] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 01/02/2011] [Accepted: 01/12/2011] [Indexed: 11/15/2022]
|
15
|
Wang P, Leung CH, Ma DL, Lu W, Che CM. Organoplatinum(II) complexes with nucleobase motifs as inhibitors of human topoisomerase II catalytic activity. Chem Asian J 2011; 5:2271-80. [PMID: 20730852 DOI: 10.1002/asia.201000451] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Platinum(II) complexes bearing acetylide ligands containing nucleobase motifs are prepared and their impact on human topoisomerase II (TopoII) is evaluated. Both platinum(II) complexes [Pt(II)(C^N^N)(C≡CCH₂R)] (1a-c) and [Pt(II)(tBu₃terpy)(C≡CCH₂R)](+) (2a-c) (C^N^N=6-phenyl-2,2'-bipyridyl, tBu₃terpy=4,4',4''-tri-tert-butyl-2,2':6',2''-terpyridyl, and R=(a) adenine, (b) thymine, and (c) 2-amino-6-chloropurine) are stable in aqueous solutions for 48 hours at room temperature. The binding constants (K) for the platinum(II) complexes towards calf thymus DNA are in the order of 10⁵ dm³ mol⁻¹ as estimated by using UV/Vis absorption spectroscopy. Of the complexes examined, only complexes 1a-c are found to behave as intercalators. Both complexes 1a-c and 2a-c inhibit TopoII-induced relaxation of supercoiled DNA, while 2c is the most potent TopoII inhibitors among the tested compounds. Inhibition of DNA relaxation is detected at nanomolar concentrations of 2c. All of the platinum(II) complexes are cytotoxic to human cancer cells with IC₅₀ values of 0.5-13.7 μM, while they are less toxic against normal cells CCD-19 Lu.
Collapse
Affiliation(s)
- Ping Wang
- Department of Chemistry and Open Laboratory of Chemical Biology of the Institute of Molecular Technology for Drug Discovery and Synthesis, The University of Hong Kong, Pokfulam Road, Hong Kong, P.R. China
| | | | | | | | | |
Collapse
|
16
|
García-Estrada C, Prada CF, Fernández-Rubio C, Rojo-Vázquez F, Balaña-Fouce R. DNA topoisomerases in apicomplexan parasites: promising targets for drug discovery. Proc Biol Sci 2010; 277:1777-87. [PMID: 20200034 DOI: 10.1098/rspb.2009.2176] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The phylum Apicomplexa includes a large group of protozoan parasites responsible for a wide range of animal and human diseases. Destructive pathogens, such as Plasmodium falciparum and Plasmodium vivax, causative agents of human malaria, Cryptosporidium parvum, responsible of childhood diarrhoea, and Toxoplasma gondii, responsible for miscarriages and abortions in humans, are frequently associated with HIV immunosuppression in AIDS patients. The lack of effective vaccines, along with years of increasing pressure to eradicate outbreaks with the use of drugs, has favoured the formation of multi-drug resistant strains in endemic areas. Almost all apicomplexan of medical interest contain two endosymbiotic organelles that contain their own mitochondrial and apicoplast DNA. Apicoplast is an attractive target for drug testing because in addition to harbouring singular metabolic pathways absent in the host, it also has its own transcription and translation machinery of bacterial origin. Accordingly, apicomplexan protozoa contain an interesting mixture of enzymes to unwind DNA from eukaryotic and prokaryotic origins. On the one hand, the main mechanism of DNA unwinding includes the scission of one-type I-or both DNA strands-type II eukaryotic topoisomerases, establishing transient covalent bonds with the scissile end. These enzymes are targeted by camptothecin and etoposide, respectively, two natural drugs whose semisynthetic derivatives are currently used in cancer chemotherapy. On the other hand, DNA gyrase is a bacterial-borne type II DNA topoisomerase that operates within the apicoplast and is effectively targeted by bacterial antibiotics like fluoroquinolones and aminocoumarins. The present review is an update on the new findings concerning topoisomerases in apicomplexan parasites and the role of these enzymes as targets for therapeutic agents.
Collapse
Affiliation(s)
- Carlos García-Estrada
- Departamento de Ciencias Biomédicas (INTOXCAL), Universidad de León, , Campus de Vegazana s/n, León, Spain
| | | | | | | | | |
Collapse
|
17
|
Shen DQ, Wu N, Li YP, Wu ZP, Zhang HB, Huang ZS, Gu LQ, An LK. Design, Synthesis, and Cytotoxicity of Indolizinoquinoxaline-5,12-dione Derivatives, Novel DNA Topoisomerase IB Inhibitors. Aust J Chem 2010. [DOI: 10.1071/ch09580] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A series of new indolizinoquinoxaline-5,12-dione derivatives were designed and synthesized via a heterocyclization reaction of 6,7-dichloroquinoxaline-5,8-dione with active methylene reagents and pyridine derivatives. The synthesized compounds exhibited significant activity to inhibit the growth of four human tumour cell lines, including lung adenocarcinoma cell, large-cell lung carcinoma cell, breast carcinoma cell, and ardriamycin-resistant breast carcinoma cell at micromolar range. These compounds were also investigated for their inhibition to DNA topoisomerase IB activity. The results indicated that the indolizinoquinoxaline-5,12-dione structure might be a potential pharmacophore in anti-cancer drug design.
Collapse
|
18
|
Samor C, Guerrini A, Varchi G, Beretta GL, Fontana G, Bombardelli E, Carenini N, Zunino F, Bertucci C, Fiori J, Battaglia A. The Role of Polyamine Architecture on the Pharmacological Activity of Open Lactone Camptothecin−Polyamine Conjugates. Bioconjug Chem 2008; 19:2270-9. [DOI: 10.1021/bc800033r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Cristian Samor
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Andrea Guerrini
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Greta Varchi
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Giovanni Luca Beretta
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Gabriele Fontana
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Ezio Bombardelli
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Nives Carenini
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Franco Zunino
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Carlo Bertucci
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Jessica Fiori
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Arturo Battaglia
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
19
|
Das BB, Ganguly A, Majumder HK. DNA Topoisomerases of Leishmania: The Potential Targets for Anti-Leishmanial Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 625:103-15. [DOI: 10.1007/978-0-387-77570-8_9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
20
|
Role of a tryptophan anchor in human topoisomerase I structure, function and inhibition. Biochem J 2008; 411:523-30. [PMID: 18215123 DOI: 10.1042/bj20071436] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human Top1 (topoisomerase I) relaxes supercoiled DNA during cell division and transcription. Top1 is composed of 765 amino acids and contains an unstructured N-terminal domain of 200 amino acids, and a structured functional domain of 565 amino acids that binds and relaxes supercoiled DNA. In the present study we examined the region spanning the junction of the N-terminal domain and functional domain (junction region). Analysis of several published Top1 structures revealed that three tryptophan residues formed a network of aromatic stacking interactions and electrostatic interactions that anchored the N-terminus of the functional domain to sub-domains containing the nose cone and active site. Mutation of the three tryptophan residues (Trp(203)/Trp(205)/Trp(206)) to an alanine residue, either individually or together, in silico revealed that the individual tryptophan residue's contribution to the tryptophan 'anchor' was additive. When the three tryptophan residues were mutated to alanine in vitro, the resulting mutant Top1 differed from wild-type Top1 in that it lacked processivity, exhibited resistance to camptothecin and was inactivated by urea. The results indicated that the tryptophan anchor stabilized the N-terminus of the functional domain and prevented the loss of Top1 structure and function.
Collapse
|
21
|
Reguera RM, Redondo CM, Gutierrez de Prado R, Pérez-Pertejo Y, Balaña-Fouce R. DNA topoisomerase I from parasitic protozoa: A potential target for chemotherapy. ACTA ACUST UNITED AC 2006; 1759:117-31. [PMID: 16757380 DOI: 10.1016/j.bbaexp.2006.03.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2006] [Revised: 03/22/2006] [Accepted: 03/30/2006] [Indexed: 11/28/2022]
Abstract
The growing occurrence of drug resistant strains of unicellular prokaryotic parasites, along with insecticide-resistant vectors, are the factors contributing to the increased prevalence of tropical diseases in underdeveloped and developing countries, where they are endemic. Malaria, cryptosporidiosis, African and American trypanosomiasis and leishmaniasis threaten human beings, both for the high mortality rates involved and the economic loss resulting from morbidity. Due to the fact that effective immunoprophylaxis is not available at present; preventive sanitary measures and pharmacological approaches are the only sources to control the undesirable effects of such diseases. Current anti-parasitic chemotherapy is expensive, has undesirable side effects or, in many patients, is only marginally effective. Under this point of view molecular biology techniques and drug discovery must walk together in order to find new targets for chemotherapy intervention. The identification of DNA topoisomerases as a promising drug target is based on the clinical success of camptothecin derivatives as anticancer agents. The recent detection of substantial differences between trypanosome and leishmania DNA topoisomerase IB with respect to their homologues in mammals has provided a new lead in the study of the structural determinants that can be effectively targeted. The present report is an up to date review of the new findings on type IB DNA topoisomerase in unicellular parasites and the role of these enzymes as targets for therapeutic agents.
Collapse
Affiliation(s)
- R M Reguera
- Dpto. Farmacología y Toxicología (INTOXCAL), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | | | | | | | | |
Collapse
|
22
|
Ma J, Jones SH, Marshall R, Wu X, Hecht SM. DNA topoisomerase I inhibitors from Rinorea anguifera. Bioorg Med Chem Lett 2005; 15:813-6. [PMID: 15664863 DOI: 10.1016/j.bmcl.2004.10.095] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Accepted: 10/29/2004] [Indexed: 10/26/2022]
Abstract
An organic extract prepared from Rinorea anguifera was investigated in order to identify the natural principle(s) responsible for stabilization of a topoisomerase I-DNA covalent binary complex. Bioassay-guided fractionation resulted in the isolation of mauritianin and (+)-syringaresinol as new topoisomerase I inhibitors, and also of the known inhibitor camptothecin.
Collapse
Affiliation(s)
- Ji Ma
- Department of Chemistry and Department of Biology, University of Virginia, Charlottesville, VA 22901, USA
| | | | | | | | | |
Collapse
|
23
|
De P, Rodgers KK. Putting the pieces together: identification and characterization of structural domains in the V(D)J recombination protein RAG1. Immunol Rev 2005; 200:70-82. [PMID: 15242397 DOI: 10.1111/j.0105-2896.2004.00154.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
V(D)J recombination generates functional immunoglobulin and T-cell receptor genes in developing lymphocytes. The recombination-activating gene 1 (RAG1) and RAG2 proteins catalyze site-specific DNA cleavage in this recombination process. Biochemical studies have identified catalytically active regions of each protein, referred to as the core regions. Here, we review our progress in the identification and characterization, in biophysical and biochemical terms, of topologically independent domains within both the non-core and core regions of RAG1. Previous characterizations of a structural domain identified in the non-core region of RAG1 from residues 265-380, referred to as the zinc-binding dimerization domain, are discussed. This domain contains two zinc-binding motifs, a RING finger and a C2H2 zinc finger. Core RAG1 also consists of multiple domains, each of which functions individually in one or more of the essential macromolecular interactions formed by the intact core protein. Two structural domains referred to as the central and the C-terminal domains that include residues 528-760 and 761-979 of RAG1, respectively, have been identified. The interactions of the central and C-terminal domains in core RAG1 with the recombination signal sequence (RSS) have contributed additional insight to a developing model for the RAG1-RSS complex.
Collapse
Affiliation(s)
- Pallabi De
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | |
Collapse
|
24
|
Laco GS, Du W, Kohlhagen G, Sayer JM, Jerina DM, Burke TG, Curran DP, Pommier Y. Analysis of human topoisomerase I inhibition and interaction with the cleavage site +1 deoxyguanosine, via in vitro experiments and molecular modeling studies. Bioorg Med Chem 2004; 12:5225-35. [PMID: 15351405 DOI: 10.1016/j.bmc.2004.06.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2004] [Accepted: 06/09/2004] [Indexed: 10/26/2022]
Abstract
Human topoisomerase I (Top1) plays a pivotal role in cell replication and transcription, and therefore is an important anti-cancer target. Homocamptothecin is a lead compound for inhibiting Top1, and is composed of five conjugated planar rings (A-E). The homocamptothecin E-ring beta-hydroxylactone opens slowly to a carboxylate at pH>7.0. We analyzed, which form of homocamptothecin was biochemically relevant in the following ways: (1) the homocamptothecin carboxylate was tested for activity in vitro and found to be inactive; (2) homocamptothecin was incubated with Top1 and dsDNA, and we found that the homocamptothecin beta-hydroxylactone form was stabilized; (3) the homocamptothecin E-ring beta-hydroxylactone was modified to prevent opening, and the derivatives were either inactive or had low activity. These results indicated that the homocamptothecin beta-hydroxylactone was the active form, and that an E-ring carbonyl oxygen and adjacent unsubstituted/unprotonated ring atom were required for full activity. Homocamptothecin and derivatives were docked into a Top1/DNA active site model, in which the +1 deoxyguanosine was rotated out of the helix, in order to compare the interaction energies between the ligands and the Top1/DNA active site with the in vitro activities of the ligands. It was found that the ligand interaction energies and in vitro activities were correlated, while the orientations of the ligands in the Top1/DNA active site explained the importance of the E-ring beta-hydroxylactone independently of E-ring opening. An essential component of this Top1/DNA active site model is the rotated +1 deoxyguanosine, and in vitro experiments and molecular modeling studies supported rotation of the +1 deoxyguanosine out of the helix. These results allow for the rational design of more potent Top1 inhibitors through engineered interactions with as yet unutilized Top1 active-site residues including: Glu356, Asn430, and Lys751.
Collapse
Affiliation(s)
- Gary S Laco
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Andersen FF, Andersen KE, Kusk M, Frøhlich RF, Westergaard O, Andersen AH, Knudsen BR. Recombinogenic flap ligation mediated by human topoisomerase I. J Mol Biol 2003; 330:235-46. [PMID: 12823964 DOI: 10.1016/s0022-2836(03)00593-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Aberration of eukaryotic topoisomerase I catalysis leads to potentially recombinogenic pathways by allowing the joining of heterologous DNA strands. Recently, a new ligation pathway (flap ligation) was presented for vaccinia virus topoisomerase I, in which blunt end cleavage complexes ligate the recessed end of duplex acceptors having a single-stranded 3'-tail. This reaction was suggested to play an important role in the repair of topoisomerase I-induced DNA double-strand breaks. Here, we characterize flap ligation mediated by human topoisomerase I. We demonstrate that cleavage complexes containing the enzyme at a blunt end allow invasion of a 3'-acceptor tail matching the scissile strand of the donor, which facilitates ligation of the recessed 5'-hydroxyl end. However, the reaction was strictly dependent on the length of double-stranded DNA of the donor complexes, and longer stretches of base-pairing inhibited strand invasion. The stabilization of the DNA helix was most probably provided by the covalently bound enzyme itself, since deleting the N-terminal domain of human topoisomerase I stimulated flap ligation. We suggest that stabilization of the DNA duplex upon enzyme binding may play an important role during normal topoisomerase I catalysis by preventing undesired strand transfer reactions. For flap ligation to function in a repair pathway, factors other than topoisomerase I, such as helicases, would be necessary to unwind the DNA duplex and allow strand invasion.
Collapse
Affiliation(s)
- Félicie F Andersen
- Department of Molecular Biology, University of Aarhus, C.F. Møllers Allé, Building 130, DK-8000, C, Aarhus, Denmark
| | | | | | | | | | | | | |
Collapse
|
26
|
Buckner JC, Reid JM, Wright K, Kaufmann SH, Erlichman C, Ames M, Cha S, O'Fallon JR, Schaaf LJ, Miller LL. Irinotecan in the treatment of glioma patients: current and future studies of the North Central Cancer Treatment Group. Cancer 2003; 97:2352-8. [PMID: 12712456 DOI: 10.1002/cncr.11304] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Other than nitrosoureas (carmustine and lomustine) and temozolomide, no agents have consistently demonstrated clinically meaningful benefits for patients with gliomas. The active metabolite of irinotecan, 7-ethyl-10-hydroxy camptothecin (SN-38), exhibited promising antitumor effects in preclinical glioma models. Clinical trials using weekly or every 3 weeks dosing of irinotecan have been completed. Toxicity consisted primarily of mild to moderate neutropenia and diarrhea with both schedules, with occasional severe toxicity including one death from neutropenia and infection. Preliminary analyses have suggested imaging responses in 10-15% of patients. Preclinical models and our understanding of the mechanism of action suggest that irinotecan may sensitize glioma cells to the cytotoxic actions of radiation therapy and alkylating agents; clinical trials designed to assess the therapeutic benefit of combination therapy currently are in progress. There is substantial clinical evidence that the concurrent administration of irinotecan with certain anticonvulsants produces reduced exposure to SN-38. In the absence of anticonvulsants, there is also substantial interpatient variability in drug exposure, perhaps reflecting inherited differences in drug metabolism. Finally several mechanisms of tumor cell resistance to irinotecan have been hypothesized, but the clinical significance of these observations has not been confirmed. Correlative studies to address these pharmacokinetic, pharmacogenetic, and drug resistance questions are ongoing.
Collapse
Affiliation(s)
- Jan C Buckner
- Division of Medical Oncology and Developmental Oncology Research, Cancer Center Statistics Unit, Mayo Clinic and Mayo Foundation, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yang Z, Champoux JJ. Reconstitution of enzymatic activity by the association of the cap and catalytic domains of human topoisomerase I. J Biol Chem 2002; 277:30815-23. [PMID: 12077150 DOI: 10.1074/jbc.m205302200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
When human topoisomerase I binds DNA, two opposing lobes in the enzyme, the cap region (amino acid, residues 175-433) and the catalytic domain (Deltacap, residues 433 to the COOH terminus) clamp tightly around the DNA helix to form the precleavage complex. Although Deltacap contains all of the residues known to be important for catalysis and binds DNA with an affinity similar to that of the intact enzyme, this fragment lacks catalytic activity. However, a mixture of Deltacap and topo31 (residues 175-433) reconstitutes enzymatic activity as measured by plasmid DNA relaxation and suicide cleavage assays. Although the formation of an active complex between topo31 and Deltacap is too unstable to be detected by pull-down experiments even in the presence of DNA, the association of topo31 with Deltacap persists and is detectable after the complex catalyzes the covalent attachment of the DNA to Deltacap by suicide cleavage. Removal of topo31 from Deltacap-DNA after suicide cleavage reveals that, unlike the cleavage reaction, religation does not require the cap region of the protein. These results suggest that activation of the catalytic domain of the enzyme for cleavage requires both DNA binding and the presence of the cap region of the protein.
Collapse
Affiliation(s)
- Zheng Yang
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA 98195-7242, USA
| | | |
Collapse
|
28
|
Affiliation(s)
- Jerrylaine V Walker
- Department of Molecular Pharmacology, St. Jude Children's Research Hospital, 332 N. Lauderdale Street, Memphis, TN 38105, USA
| | | |
Collapse
|
29
|
Abstract
DNA topoisomerases solve the topological problems associated with DNA replication, transcription, recombination, and chromatin remodeling by introducing temporary single- or double-strand breaks in the DNA. In addition, these enzymes fine-tune the steady-state level of DNA supercoiling both to facilitate protein interactions with the DNA and to prevent excessive supercoiling that is deleterious. In recent years, the crystal structures of a number of topoisomerase fragments, representing nearly all the known classes of enzymes, have been solved. These structures provide remarkable insights into the mechanisms of these enzymes and complement previous conclusions based on biochemical analyses. Surprisingly, despite little or no sequence homology, both type IA and type IIA topoisomerases from prokaryotes and the type IIA enzymes from eukaryotes share structural folds that appear to reflect functional motifs within critical regions of the enzymes. The type IB enzymes are structurally distinct from all other known topoisomerases but are similar to a class of enzymes referred to as tyrosine recombinases. The structural themes common to all topoisomerases include hinged clamps that open and close to bind DNA, the presence of DNA binding cavities for temporary storage of DNA segments, and the coupling of protein conformational changes to DNA rotation or DNA movement. For the type II topoisomerases, the binding and hydrolysis of ATP further modulate conformational changes in the enzymes to effect changes in DNA topology.
Collapse
Affiliation(s)
- J J Champoux
- Department of Microbiology, School of Medicine, University of Washington, Seattle, Washington 98195-7242, USA.
| |
Collapse
|
30
|
Affiliation(s)
- C Bailly
- INSERM U-524, and Laboratoire de Pharmacologie Antitumorale du Centre Oscar Lambret IRCL, 59045 Lille, France
| |
Collapse
|
31
|
Zhang H, Barceló JM, Lee B, Kohlhagen G, Zimonjic DB, Popescu NC, Pommier Y. Human mitochondrial topoisomerase I. Proc Natl Acad Sci U S A 2001; 98:10608-13. [PMID: 11526219 PMCID: PMC58513 DOI: 10.1073/pnas.191321998] [Citation(s) in RCA: 141] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2001] [Accepted: 07/16/2001] [Indexed: 11/18/2022] Open
Abstract
Tension generated in the circular mitochondrial genome during replication and transcription points to the need for mtDNA topoisomerase activity. Here we report a 601-aa polypeptide highly homologous to nuclear topoisomerase I. The N-terminal domain of this novel topoisomerase contains a mitochondrial localization sequence and lacks a nuclear localization signal. Therefore, we refer to this polypeptide as top1mt. The pattern of top1mt expression matches the requirement for high mitochondrial activity in specific tissues. top1mt is a type IB topoisomerase that requires divalent metal (Ca(2+) or Mg(2+)) and alkaline pH for optimum activity. The TOP1mt gene is highly homologous to the nuclear TOP1 gene and consists of 14 exons. It is localized on human chromosome 8q24.3.
Collapse
Affiliation(s)
- H Zhang
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4255, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Lin CF, Hsieh PC, Lu WD, Chiu HF, Wu MJ. A series of enediynes as novel inhibitors of topoisomerase I. Bioorg Med Chem 2001; 9:1707-11. [PMID: 11425571 DOI: 10.1016/s0968-0896(01)00081-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
A series of acyclic enediynes, 2-((6-substituted)-3-hexen-1,5-diynyl)benzonitriles (8--11), display potent inhibition against topoisomerase I without the formation of active biradical intermediates and show inhibitory activity against topoisomerase I at 10 microM, which is five times that of camptothecin from the results of agarose gel electrophoresis.
Collapse
Affiliation(s)
- C F Lin
- School of Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|
33
|
Abstract
Topoisomerase I is a ubiquitous and essential enzyme in multicellular organisms. It is involved in multiple DNA transactions including DNA replication, transcription, chromosome condensation and decondensation, and probably DNA recombination. Besides its activity of DNA relaxation necessary to eliminate torsional stresses associated with these processes, topoisomerase I may have other functions related to its interaction with other cellular proteins. Topoisomerase I is the target of the novel anticancer drugs, the camptothecins. Recently a broad range of physiological and environmentally-induced DNA modifications have also been shown to poison topoisomerases. This review summarizes the various factors that enhance or suppress top1 cleavage complexes and discusses the significance of such effects. We also review the different mechanisms that have been proposed for the repair of topoisomerase I-mediated DNA lesions.
Collapse
Affiliation(s)
- P Pourquier
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
34
|
Champoux JJ. Structure-based analysis of the effects of camptothecin on the activities of human topoisomerase I. Ann N Y Acad Sci 2001; 922:56-64. [PMID: 11193925 DOI: 10.1111/j.1749-6632.2000.tb07025.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The sole target for the anticancer drug camptothecin (CPT) is the type I topoisomerase. The drug poisons the topoisomerase by slowing the religation step of the reaction, thereby trapping the enzyme in a covalent complex on the DNA. In addition, CPT has been shown to inhibit plasmid DNA relaxation in vitro. The structural bases for these two activities of CPT are explored in relation to the recently published crystal structure of the enzyme with bound DNA.
Collapse
Affiliation(s)
- J J Champoux
- Department of Microbiology, Box 357242, University of Washington, Seattle, Washington 98195-7242, USA.
| |
Collapse
|
35
|
Abstract
Based on the crystal structure of human topoisomerase I, we hypothesized that hydrogen bonding between the side chain of the highly conserved His(632) and one of the nonbridging oxygens of the scissile phosphate contributes to catalysis by stabilizing the transition state. This hypothesis has been tested by examining the effects of changing His(632) to glutamine, asparagine, alanine, and tryptophan. The change to glutamine reduced both the relaxation activity and single-turnover cleavage activity by approximately 100-fold, whereas the same change at three other conserved histidines (positions 222, 367, and 406) had no significant effect on the relaxation activity. The properties of the mutant protein containing asparagine instead of histidine at position 632 were similar to those of the glutamine mutant, whereas mutations to alanine or tryptophan reduced the activity by approximately 4 orders of magnitude. The reduction in activity for the mutants was not due to alterations in substrate binding affinities or changes in the cleavage specificities of the proteins. The above results for the glutamine mutation in conjunction with the similar effects of pH on the wild type and the H632Q mutant enzyme rule out the possibility that His(632) acts as a general acid to protonate the leaving 5'-oxygen during the cleavage reaction. Taken together, these data strongly support the hypothesis that the only role for His(632) is to stabilize the pentavalent transition state through hydrogen bonding to one of the nonbridging oxygens.
Collapse
Affiliation(s)
- Z Yang
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington 98195-7242, USA
| | | |
Collapse
|
36
|
Abstract
Protozoan parasites are responsible for a wide range of debilitating and fatal diseases that are proving notoriously difficult to treat. Many of the standard chemotherapies in use today are expensive, have toxic side effects and, in some cases have marginal efficacy because of the emergence of drug-resistant parasites. In the search for more effective treatments, protozoan topoisomerases are now being considered as potential drug targets, building on the clinical success of anticancer and antibacterial agents that target human and bacterial topoisomerases. In this review, Sandra Cheesman explores progress in this relatively new but potentially important field of research.
Collapse
Affiliation(s)
- S J Cheesman
- Institute of Cell and Molecular Biology, the University of Edinburgh, Darwin Building, King's Buildings, Mayfield Road, Edinburgh, UK EH9 3JR.
| |
Collapse
|
37
|
Fortune JM, Osheroff N. Topoisomerase II as a target for anticancer drugs: when enzymes stop being nice. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2000; 64:221-53. [PMID: 10697411 DOI: 10.1016/s0079-6603(00)64006-0] [Citation(s) in RCA: 259] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Topoisomerase II is an essential enzyme that plays a role in virtually every cellular DNA process. This enzyme interconverts different topological forms of DNA by passing one nucleic acid segment through a transient double-stranded break generated in a second segment. By virtue of its double-stranded DNA passage reaction, topoisomerase II is able to regulate DNA over- and underwinding, and can resolve knots and tangles in the genetic material. Beyond the critical physiological functions of the eukaryotic enzyme, topoisomerase II is the target for some of the most successful anticancer drugs used to treat human malignancies. These agents are referred to as topoisomerase II poisons, because they transform the enzyme into a potent cellular toxin. Topoisomerase II poisons act by increasing the concentration of covalent enzyme-cleaved DNA complexes that normally are fleeting intermediates in the catalytic cycle of topoisomerase II. As a result of their action, these drugs generate high levels of enzyme-mediated breaks in the genetic material of treated cells and ultimately trigger cell death pathways. Topoisomerase II is also the target for a second category of drugs referred to as catalytic inhibitors. Compounds in this category prevent topoisomerase II from carrying out its required physiological functions. Drugs from both categories vary widely in their mechanisms of actions. This review focuses on topoisomerase II function and how drugs alter the catalytic cycle of this important enzyme.
Collapse
Affiliation(s)
- J M Fortune
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
38
|
Lavrukhin OV, Fortune JM, Wood TG, Burbank DE, Van Etten JL, Osheroff N, Lloyd RS. Topoisomerase II from Chlorella virus PBCV-1. Characterization of the smallest known type II topoisomerase. J Biol Chem 2000; 275:6915-21. [PMID: 10702252 DOI: 10.1074/jbc.275.10.6915] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Type II topoisomerases, a family of enzymes that govern topological DNA interconversions, are essential to many cellular processes in eukaryotic organisms. Because no data are available about the functions of these enzymes in the replication of viruses that infect eukaryotic hosts, this led us to express and characterize the first topoisomerase II encoded by one of such viruses. Paramecium bursaria chlorella virus 1 (PBCV-1) infects certain chlorella-like green algae and encodes a 120-kDa protein with a similarity to type II topoisomerases. This protein was expressed in Saccharomyces cerevisiae and was highly active in relaxation of both negatively and positively supercoiled plasmid DNA, catenation of plasmid DNA, and decatenation of kinetoplast DNA networks. Its optimal activity was determined, and the omission of Mg(2+) or its replacement with other divalent cations abolished DNA relaxation. All activities of the recombinant enzyme were ATP dependent. Increasing salt concentrations shifted DNA relaxation from a normally processive mechanism to a distributive mode. Thus, even though the PBCV-1 enzyme is considerably smaller than other eukaryotic topoisomerase II enzymes (whose molecular masses are typically 160-180 kDa), it displays all the catalytic properties expected for a type II topoisomerase.
Collapse
Affiliation(s)
- O V Lavrukhin
- Department of Human Biological Chemistry and Genetics and Sealy Center for Molecular Science, University of Texas Medical Branch, Galveston, Texas 77555-1071, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Stewart L, Ireton GC, Champoux JJ. A functional linker in human topoisomerase I is required for maximum sensitivity to camptothecin in a DNA relaxation assay. J Biol Chem 1999; 274:32950-60. [PMID: 10551862 DOI: 10.1074/jbc.274.46.32950] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human topoisomerase I is composed of four major domains: the highly charged NH(2)-terminal region, the conserved core domain, the positively charged linker domain, and the highly conserved COOH-terminal domain. Near complete enzyme activity can be reconstituted by combining recombinant polypeptides that approximate the core and COOH-terminal domains, although DNA binding is reduced somewhat for the reconstituted enzyme (Stewart, L., Ireton, G. C., and Champoux, J. J. (1997) J. Mol. Biol. 269, 355-372). A reconstituted enzyme comprising the core domain plus a COOH-terminal fragment containing the complete linker region exhibits the same biochemical properties as a reconstituted enzyme lacking the linker altogether, and thus detachment of the linker from the core domain renders the linker non-functional. The rate of religation by the reconstituted enzyme is increased relative to the forms of the enzyme containing the linker indicating that in the intact enzyme the linker slows religation. Relaxation of plasmid DNA by full-length human topoisomerase I or a 70-kDa form of the enzyme that is missing only the non-essential NH(2)-terminal domain (topo70) is inhibited approximately 16-fold by the anticancer compound, camptothecin, whereas the reconstituted enzyme is nearly resistant to the inhibitory effects of the drug despite similar affinities for the drug by the two forms of the enzyme. Based on these results and in light of the crystal structure of human topoisomerase I, we propose that the linker plays a role in hindering supercoil relaxation during the normal relaxation reaction and that camptothecin inhibition of DNA relaxation depends on a direct effect of the drug on DNA rotation that is also dependent on the linker.
Collapse
Affiliation(s)
- L Stewart
- Department of Microbiology, School of Medicine, University of Washington, Seattle, Washington 98195-7242, USA
| | | | | |
Collapse
|
40
|
Kong JY, Rabkin SW. Thapsigargin enhances camptothecin-induced apoptosis in cardiomyocytes. PHARMACOLOGY & TOXICOLOGY 1999; 85:212-20. [PMID: 10608483 DOI: 10.1111/j.1600-0773.1999.tb02011.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Topoisomerase I inhibitors are promising new chemotherapeutic agents for the treatment of certain malignancies. The present study investigated the impact of the topoisomerase I inhibitor camptothecin on cell death in cardiomyocytes and sought to determine whether the sesquiterpene gamma-lactone--thapsigargin, that alter sarcoplasmic reticulum calcium flux, modulates the effect of camptothecin on the cardiomyocyte. Camptothecin-induced cell death was demonstrated in cardiomyocytes maintained in culture, from 7 day old embryonic chick hearts, by the trypan blue and the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide) assay, two independent indicators of the loss of cell viability. The type of cell death was attributed to apoptosis based on cell structure, DNA fragmentation and flow cytometry studies. Camptothecin-treated cardiomyocytes were shrunken with membrane blebs and nuclear fragmentation. Camptothecin produced a dose-dependent increase in DNA fragments of 180 base pairs, or multiples thereof, which are characteristic of apoptosis. A two-fold increase in this type of DNA fragmentation was produced by camptothecin (10 microM) compared to control (diluent-treated) cells. Flow cytometry analysis of populations of 10,000 cardiomyocytes stained with propidium iodide demonstrated a significant increase in the proportion of the population with alterations of DNA content consistent with apoptosis. Pretreatment of cells with thapsigargin, which selectively inhibits sarcoplasmic reticulum and endoplasmic reticulum Ca+2-dependent ATPase, significantly augmented camptothecin-induced apoptosis. Exploring further the role of calcium in camptothecin-induced cell death, we found that the Ca+2 chelator EGTA decreased camptothecin-induced DNA fragmentation. These data indicate the potential for cardiotoxicity from camptothecin through the process of apoptosis and suggest that agents which affect cellular calcium regulation enhance camptothecin-induced apoptosis.
Collapse
Affiliation(s)
- J Y Kong
- University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
41
|
Abstract
DNA topoisomerases catalyze changes in the topology of DNA. Recently, other functions have also been reported for these enzymes. For example, topoisomerase I participates in transcription by RNA polymerases I, II, and III, and also has a kinase activity. Topoisomerase I binds directly to at least two helicases, nucleolin and SV40 T antigen, and mechanistic studies show that T antigen alters the function of topoisomerase I. Additional protein and nucleotide interactions for both topoisomerases I and II suggest that each protein is multifunctional. It may be that the multifunctional nature of these enzymes is the basis for the antitumor activity seen with inhibitors of these enzymes. Clinical trials with combinations of CPT-11 and 5-fluorouracil for the treatment of colon cancer, and preclinical studies with CPT-11 and vincristine are particularly encouraging. Protracted schedules of administration of topoisomerase inhibitors will likely have greater antitumor effect than more concentrated, higher dose exposures, but a systematic determination of optimal schedules of administration is needed.
Collapse
Affiliation(s)
- S M Guichard
- Laboratoire de Pharmacologie, Institut Claudius Regaud, Toulouse, France
| | | |
Collapse
|
42
|
Pommier Y, Pourquier P, Urasaki Y, Wu J, Laco GS. Topoisomerase I inhibitors: selectivity and cellular resistance. Drug Resist Updat 1999; 2:307-318. [PMID: 11504505 DOI: 10.1054/drup.1999.0102] [Citation(s) in RCA: 141] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Topoisomerase I (top1) inhibitors (camptothecins and other structurally diverse compounds) are effective and promising anticancer agents. Determinants of selectivity toward cancer cells and resistance are multifactorial. These factors can be separated in three groups. The first is related to alterations in drug distribution and metabolism. The second group includes both quantitative and qualitative (mutations) differences in top I. The third group includes resistance and sensitivity factors downstream from the cleavage complexes. They include DNA repair, cell cycle checkpoints and apoptosis, and are probably key to the relative selectivity of camptothecins toward cancer cells and to clinical resistance. Copyright 1999 Harcourt Publishers Ltd.
Collapse
Affiliation(s)
- Yves Pommier
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
43
|
Broccoli S, Marquis JF, Papadopoulou B, Olivier M, Drolet M. Characterization of a Leishmania donovani gene encoding a protein that closely resembles a type IB topoisomerase. Nucleic Acids Res 1999; 27:2745-52. [PMID: 10373592 PMCID: PMC148484 DOI: 10.1093/nar/27.13.2745] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In order to clone the gene encoding a type I DNA topoisomerase from Leishmania donovani, a PCR-amplified DNA fragment obtained with degenerate oligodeoxyribonucleotides was used to screen a genomic library from this parasite. An open reading frame of 1905 bases encoding a putative protein of 635 amino acid residues was isolated. A substantial part of the protein shares a significant degree of homology with the sequence of other known members of the IB topoisomerase family, in a highly conserved region of these enzymes termed the core domain. However, homology is completely lost after this conserved central core. Moreover, no conventional active tyrosine site could be identified. In fact, the protein expressed in Escherichia coli did not show any relaxation activity in vitro and was unable to complement a mutant deficient in topoisomerase I activity. The results of Southern blot experiments strongly suggested that the cloned gene was not a pseudogene. Northern analysis revealed that the gene was transcribed in its full length and also excluded the possibility that some form of splicing is necessary to produce a mature messenger. Furthermore, our results indicate that the gene is preferentially expressed in actively growing L.donovani promastigotes and that it is also expressed in other kinetoplastid parasites.
Collapse
Affiliation(s)
- S Broccoli
- Département de Microbiologie et Immunologie, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, Québec H3C 3J7, Canada and
| | | | | | | | | |
Collapse
|
44
|
Abstract
Topoisomerases relax the DNA superhelical tension that arises in cells as a result of several nuclear processes, including transcription, replication and recombination. Recently determined crystal structures of human topoisomerase I in complex with DNA and of the 27 kDa catalytic domain of the vaccinia virus topoisomerase have advanced our understanding of the eukaryotic type IB topoisomerases. These recent structural results provide insights into functional aspects of these topoisomerases, including their DNA binding, strand cleavage and religation activities, as well as the mechanism that these enzymes use to relax DNA superhelical tension. In addition, two proposed models of the anticancer drug camptothecin bound to a covalent complex of human topoisomerase I and DNA suggest a structural basis for the mode of action of the drug.
Collapse
Affiliation(s)
- M R Redinbo
- Department of Biological Structure and Biomolecular Structure Center, Howard Hughes Medical Institute, School of Medicine, University of Washington, Seattle, WA 98195, USA.
| | | | | |
Collapse
|