1
|
Radwan IT, El-Sherbiny IM, Selim AM, Metwally NH. Design, synthesis of some novel coumarins and their nanoformulations into lipid-chitosan nanocapsule as unique antimicrobial agents. Sci Rep 2024; 14:30598. [PMID: 39715779 DOI: 10.1038/s41598-024-79861-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Developing and creating novel antibiotics is one of the most important targets in treating infectious diseases. Novel coumarins were synthesized and characterized using different spectroscopic techniques such as Fourier Transform Infrared (FTIR), Nuclear magnetic resonance1H and 13C and mass spectroscopy (MS). All of the synthesized compounds have been tested for activity and sensitivity against the microbial strains of B. subtilis, S. aureus, E. coli, P. aeruginosa, S. typhi, and C. albicans. All compounds showed substantial results against the tested microbes except S. typhi, which was not affected in any way by these coumarins. Exceptional results were shown by compounds 4, 6d, and 8b, which made them the best candidates for loading to the vicinity of nanostructure lipid carrier and coated by chitosan nanocapsule (NLC-Cs). Transmission electron microscope (TEM) confirmed spherical morphology with particles size less than 500 nm. Also, dynamic light scattering (DLS) were utilized to measure the average particle size (between 100 and 200 nm) and the stability assessed by zeta potential were found to be more positive confirming the chitosan encapsulation. Antimicrobial activity assessments were performed for both synthetic compounds and their NLCs analogues. The nanoformulation of 4-NLC-Cs, 6d-NLC-Cs, and 8b-NLC-Cs manifested unique biological results, especially 8b-NLC-Cs, which revealed powerful effects over all the tested organisms including S. typhi. The increasing biological effect of the drugs in their nanoscale form is reflected in the increasing value of inhibition zone diameter and suppressing the value of MIC to reach record levels like 8b-NLC-Cs disclosed MIC = 0.48 and 0.24 µg/ml against S. aureus and C. albicans, respectively, by the mean 8b-NLC-Cs nanoformulation suppressed the MIC by 65 folds of its initial value before nano. In continuation, it was proven that the compounds 4, 6d and 8b were found to make noticeable changes on the DNA-Gyrase levels with reduced IC50 values particularly 8b showed excellent inhibitory effect with IC50 = 4.56 µM. TEM was used to pursue the morphological changes that occur in bacterial cells of P. aeruginosa. The weakness of the cell wall in most bacterial cells treated with nanomaterials, 8b-NLC-Cs, has reached the point of the cell wall rupture and the cell components spilling out of the cells causing necrotic cell death.
Collapse
Affiliation(s)
- Ibrahim Taha Radwan
- Supplementary General Sciences Department, Faculty of Oral and Dental Medicine, Future University in Egypt, Cairo, 11835, Egypt
| | - Ibrahim M El-Sherbiny
- Center for Materials Science (CMS), Zewail City of Science and Technology, 6th October City, 12578, Giza, Egypt
| | - Abdelfattah M Selim
- Department of Animal Medicine (Infectious Diseases), College of Veterinary Medicine, Benha University, Toukh, 13736, Egypt
| | | |
Collapse
|
2
|
Natarajan R, Sivaperuman A, Samuel A, Patel DH, Jain N, Veerappan M, Kumar NK. 2D QSAR, Design, and in Silico Analysis of Thiophene-Tethered Lactam Derivatives as Antimicrobial Agents. Chem Biodivers 2023; 20:e202300331. [PMID: 37337355 DOI: 10.1002/cbdv.202300331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND A very high rate of resistance causes health-care-associated and community-acquired infections. E. coli is one of the nine pathogens of highest concern to most of the antibiotics and other class of antimicrobials. OBJECTIVE The objective of the present study is to develop novel thiophene derivatives using 2D QSAR and in silico approach for E. coli resistance. METHODS Substituted thiophene series reported by Nishu Singla et al., were taken for QSAR analysis. From the results, a set of 15 new compounds were designed. A complete in silico analysis has been done using PADEL, Autodock vina, Swiss ADME, Protox II software. RESULTS The designed compounds obey the Lipinski's rule of five and were known to have excellent inhibitory action (pIC50 values -0.87 to -1.46) which is similar to the most active compound of the data set (pIC50 -0.69) taken for the study. The bioavailability score (0.65) with no toxicity representing that the designed compounds are suitable for oral administration. CONCLUSION The designed compounds are inactive for mutagenicity and cytotoxicity and ADMET studies states that these molecules are likely to be orally bioavailable and could be easily transported, diffused, and absorbed. So, the designed compounds will definitely serve as a lead antibacterial agent for E. coli resistance.
Collapse
Affiliation(s)
- Ramalakshmi Natarajan
- Department of Pharmaceutical Chemistry, C.L.BaidMetha College of Pharmacy, Thorapakkam-600097, Chennai, India
| | - Amuthalakshmi Sivaperuman
- Department of Pharmaceutical Chemistry, C.L.BaidMetha College of Pharmacy, Thorapakkam-600097, Chennai, India
| | - Abiseik Samuel
- Department of Pharmaceutical Chemistry, C.L.BaidMetha College of Pharmacy, Thorapakkam-600097, Chennai, India
| | - Dinesh Hansaram Patel
- Department of Pharmaceutical Chemistry, C.L.BaidMetha College of Pharmacy, Thorapakkam-600097, Chennai, India
| | - Nikhil Jain
- Department of Pharmaceutical Chemistry, C.L.BaidMetha College of Pharmacy, Thorapakkam-600097, Chennai, India
| | - Manigandan Veerappan
- Department of Pharmaceutical Chemistry, C.L.BaidMetha College of Pharmacy, Thorapakkam-600097, Chennai, India
| | - Nikhil Kushal Kumar
- Department of Pharmaceutical Chemistry, C.L.BaidMetha College of Pharmacy, Thorapakkam-600097, Chennai, India
| |
Collapse
|
3
|
Jaswal S, Nehra B, Kumar S, Monga V. Recent advancements in the medicinal chemistry of bacterial type II topoisomerase inhibitors. Bioorg Chem 2020; 104:104266. [PMID: 33142421 DOI: 10.1016/j.bioorg.2020.104266] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/14/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
Abstract
Replication proteins are sought as a potential targets for antimicrobial agents. Despite their promising target characteristics, only topoisomerase II inhibitors targeting DNA gyrase and/or topoisomerase IV have reached clinical use. Topoisomerases are the enzymes that are essential for cellular functions and various biological activities. A wide range of natural and synthetic compounds have been identified as potential topoisomerase inhibitors but the resistance is most commonly found in these drugs. The emergence of FQ resistance has increased the need for the development of novel topoisomerase inhibitors with efficacy and high potency against FQ-resistant strains. Besides structural modifications of existing FQ scaffolds, novel non-quinolone topoisomerase II inhibitors, known as novel bacterial topoisomerase inhibitors, have been developed which showed remarkable inhibitory activity against DNA gyrase/topoisomerase IV or both with an improved spectrum of antibacterial potency including drug-resistant strains. This review aims to summarize various recent advancements in the medicinal chemistry of topoisomerase inhibitors with the following objectives: (1) To represent inclusive data on types of topoisomerases and various marketed topoisomerase inhibitors as drugs; (2) To discuss the recent advances in the medicinal chemistry of various topoisomerase inhibitors (DNA gyrase and topo IV) belonging to different structural classes as potential antibacterial agents; (3) To summarizes the structure activity relationship (SAR) including in silico and mechanistic studies to afford ideas and to provide focused direction for the development of new chemical entities which are effective against drug-resistant bacterial pathogens and biofilms.
Collapse
Affiliation(s)
- Shalini Jaswal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, Punjab, India
| | - Bhupender Nehra
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, Punjab, India
| | - Shubham Kumar
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, Punjab, India
| | - Vikramdeep Monga
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, Punjab, India.
| |
Collapse
|
4
|
Khan T, Sankhe K, Suvarna V, Sherje A, Patel K, Dravyakar B. DNA gyrase inhibitors: Progress and synthesis of potent compounds as antibacterial agents. Biomed Pharmacother 2018; 103:923-938. [DOI: 10.1016/j.biopha.2018.04.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/24/2018] [Accepted: 04/03/2018] [Indexed: 12/22/2022] Open
|
5
|
Sobek S, Boege F. DNA topoisomerases in mtDNA maintenance and ageing. Exp Gerontol 2014; 56:135-41. [PMID: 24440386 DOI: 10.1016/j.exger.2014.01.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/03/2014] [Accepted: 01/06/2014] [Indexed: 11/26/2022]
Abstract
DNA topoisomerases pass DNA strands through each other, a function essential for all DNA metabolic processes that create supercoils or entanglements of DNA. Topoisomerases play an ambivalent role in nuclear genome maintenance: Deficiency compromises gene transcription, replication and chromosome segregation, while the inherent DNA-cleavage activity of the enzymes endangers DNA integrity. Indeed, many DNA-damaging agents act through enhancing topoisomerase DNA cleavage. Mitochondrial DNA (mtDNA) clearly requires topoisomerase activity for transcription and replication, because it is a closed, double-stranded DNA molecule. Three topoisomerases have so far been found in mammalian mitochondria (I, IIβ, IIIα), but their precise role in mtDNA metabolism, mitochondrial maintenance and respiratory function remains mostly unclear. It is a reasonable surmise that these enzymes exhibit similar ambiguity with respect to genome maintenance and gene transcription as their nuclear counterparts. Here, we review what is known about the physiological roles of mitochondrial topoisomerases and draft three scenarios of how these enzymes possibly contribute to ageing-related mtDNA attrition and respiratory chain dysfunction. These scenarios are: mtDNA attrition by exogenously stimulated topoisomerase DNA cleavage, unbalancing of mitochondrial and nuclear transcription by direct effects on mitochondrial transcription, and contributions to enhanced mtDNA entanglement and recombination.
Collapse
Affiliation(s)
- Stefan Sobek
- Institute of Clinical Chemistry and Laboratory Diagnostics, Heinrich Heine University, Med. Faculty, Düsseldorf, Germany
| | - Fritz Boege
- Institute of Clinical Chemistry and Laboratory Diagnostics, Heinrich Heine University, Med. Faculty, Düsseldorf, Germany.
| |
Collapse
|
6
|
Kathiravan MK, Khilare MM, Chothe AS, Nagras MA. Design and development of topoisomerase inhibitors using molecular modelling studies. J Chem Biol 2012; 6:25-36. [PMID: 24078835 DOI: 10.1007/s12154-012-0079-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 07/24/2012] [Indexed: 11/24/2022] Open
Abstract
Topoisomerase inhibitors are used as anticancer and antibacterial agents. A series of novel 2,4,6-tri-substituted pyridine derivatives reported as topoisomerase inhibitors were used for quantitative structure-activity relationship (QSAR) study. In order to understand the structural requirement of these topoisomerase inhibitors, a ligand-based pharmacophore and atom-based 3D-QSAR model have been developed. A five-point pharmacophore with one hydrophobic group (H4), four aromatic rings (R5, R6, R7 and R8) was obtained. The pharmacophore hypothesis yielded a 3D-QSAR model with good partial least-square (PLS) statistic results. The training set correlation is characterized by PLS factors (r (2) = 0.7892, SD = 0.2948, F = 49.9, P = 1.379). The test set correlation is characterized by PLS factors (q (2) = 0.7776, root mean squared error = 0.2764, Pearson R = 0.8926). The docking study revealed the binding orientations of these inhibitors at active site amino acid residues of topoisomerases enzyme. The results of pharmacophore hypothesis and 3D-QSAR provided the detail structural insights as well as highlighted the important binding features of novel 2,4,6-tri-substituted pyridine derivatives and can be developed as potent topoisomerase inhibitors. FigureKey structural requirement for topoisomerase activity.
Collapse
Affiliation(s)
- Muthu K Kathiravan
- Department of Pharmaceutical Chemistry, P. G. Research Centre, Sinhgad College of Pharmacy, S.No.44/1, Vadgaon (Bk), Off Sinhgad Road, Pune, 411041 Maharashtra India
| | | | | | | |
Collapse
|
7
|
Kathiravan MK, Khilare MM, Nikoomanesh K, Chothe AS, Jain KS. Topoisomerase as target for antibacterial and anticancer drug discovery. J Enzyme Inhib Med Chem 2012; 28:419-35. [DOI: 10.3109/14756366.2012.658785] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Muthu K. Kathiravan
- Department of Pharmaceutical Chemistry, Sinhgad College of Pharmacy,
Maharashtra, India
| | - Madhavi M. Khilare
- Department of Pharmaceutical Chemistry, Sinhgad College of Pharmacy,
Maharashtra, India
| | - Kiana Nikoomanesh
- Department of Pharmaceutical Chemistry, Sinhgad College of Pharmacy,
Maharashtra, India
| | - Aparna S. Chothe
- Department of Pharmaceutical Chemistry, AISSMS College of Pharmacy,
Pune, Maharashtra, India
| | - Kishor S. Jain
- Department of Pharmaceutical Chemistry, Sinhgad College of Pharmacy,
Maharashtra, India
| |
Collapse
|
8
|
de Souza-Pinto NC, Mason PA, Hashiguchi K, Weissman L, Tian J, Guay D, Lebel M, Stevnsner TV, Rasmussen LJ, Bohr VA. Novel DNA mismatch-repair activity involving YB-1 in human mitochondria. DNA Repair (Amst) 2009; 8:704-19. [PMID: 19272840 DOI: 10.1016/j.dnarep.2009.01.021] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 01/24/2009] [Accepted: 01/31/2009] [Indexed: 01/03/2023]
Abstract
Maintenance of the mitochondrial genome (mtDNA) is essential for proper cellular function. The accumulation of damage and mutations in the mtDNA leads to diseases, cancer, and aging. Mammalian mitochondria have proficient base excision repair, but the existence of other DNA repair pathways is still unclear. Deficiencies in DNA mismatch repair (MMR), which corrects base mismatches and small loops, are associated with DNA microsatellite instability, accumulation of mutations, and cancer. MMR proteins have been identified in yeast and coral mitochondria; however, MMR proteins and function have not yet been detected in human mitochondria. Here we show that human mitochondria have a robust mismatch-repair activity, which is distinct from nuclear MMR. Key nuclear MMR factors were not detected in mitochondria, and similar mismatch-binding activity was observed in mitochondrial extracts from cells lacking MSH2, suggesting distinctive pathways for nuclear and mitochondrial MMR. We identified the repair factor YB-1 as a key candidate for a mitochondrial mismatch-binding protein. This protein localizes to mitochondria in human cells, and contributes significantly to the mismatch-binding and mismatch-repair activity detected in HeLa mitochondrial extracts, which are significantly decreased when the intracellular levels of YB-1 are diminished. Moreover, YB-1 depletion in cells increases mitochondrial DNA mutagenesis. Our results show that human mitochondria contain a functional MMR repair pathway in which YB-1 participates, likely in the mismatch-binding and recognition steps.
Collapse
Affiliation(s)
- Nadja C de Souza-Pinto
- Laboratory of Molecular Gerontology, National Institute on Aging/Intramural Research Program (NIA-IRP), National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Chua ST, Quek NM, Li M, Ng MTT, Yuan W, Chua ML, Guo J, Koh LE, Ye R, Li T. Nick-containing oligonucleotides as human topoisomerase I inhibitors. Bioorg Med Chem Lett 2008; 19:618-23. [PMID: 19131243 DOI: 10.1016/j.bmcl.2008.12.061] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 12/11/2008] [Accepted: 12/15/2008] [Indexed: 11/15/2022]
Abstract
A series of oligonucleotides with various lengths that contain nick and topoisomerase I-binding sites were designed. The interactions between these oligonucleotides and human topoisomerase I were investigated and the most efficient one among them has displayed IC(50) value of 6.3 nM. Our studies have also demonstrated that the position of the nick as well as the length of the oligonucleotides were crucial factors for the inhibition of this nuclear enzyme.
Collapse
Affiliation(s)
- Sock Teng Chua
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Walk, Blk 5 Level 3, Nanyang Technological University, Singapore 637616, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wang Y, Ng MTT, Zhou T, Li X, Tan CH, Li T. C3-Spacer-containing circular oligonucleotides as inhibitors of human topoisomerase I. Bioorg Med Chem Lett 2008; 18:3597-602. [PMID: 18490159 DOI: 10.1016/j.bmcl.2008.04.076] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2008] [Revised: 04/25/2008] [Accepted: 04/30/2008] [Indexed: 11/18/2022]
Abstract
Some dumbbell-shaped circular oligonucleotides containing internal C3-spacers and Topo I-binding sites were designed and synthesized which displayed high inhibitory efficiency on the activity of human Topo I as well as resisted the degradation by some DNA repair enzymes.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Chemistry, 3 Science Drive 3, National University of Singapore, Singapore 117543, Singapore
| | | | | | | | | | | |
Collapse
|
11
|
Abstract
The inhibition of DNA topoisomerase I (Top1) has proven to be a successful approach in the design of anticancer agents. However, despite the clinical successes of the camptothecin derivatives, a significant need for less toxic and more chemically stable Top1 inhibitors still persists. Here, we describe one of the most frequently used protocols to identify novel Top1 inhibitors. These methods use uniquely 3'-radiolabeled DNA substrates and denaturing polyacrylamide gel electrophoresis to provide evidence for the Top1-mediated DNA cleaving activity of potential Top1 inhibitors. These assays allow comparison of the effectiveness of different drugs in stabilizing the Top1-DNA intermediate or cleavage (cleavable) complex. A variation on these assays is also presented, which provides a suitable system for determining whether the inhibitor blocks the forward cleavage or religation reactions by measuring the reversibility of the drug-induced Top1-DNA cleavage complexes. This entire protocol can be completed in approximately 2 d.
Collapse
Affiliation(s)
- Thomas S Dexheimer
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
12
|
Mielke C, Kalfalah FM, Christensen MO, Boege F. Rapid and prolonged stalling of human DNA topoisomerase I in UVA-irradiated genomic areas. DNA Repair (Amst) 2007; 6:1757-63. [PMID: 17765665 DOI: 10.1016/j.dnarep.2007.06.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 06/22/2007] [Accepted: 06/25/2007] [Indexed: 12/31/2022]
Abstract
DNA topoisomerase I appears to be involved in DNA damage and repair in a complex manner. The enzyme is required for DNA maintenance and repair, but it may also damage DNA through its covalently DNA-bound, catalytic intermediate. The latter mechanism plays a role in tumor cell killing by camptothecins, but seems also involved in oxidative cell killing and certain stages of apoptosis. Stalling and/or suicidal DNA cleavage of topoisomerase I adjacent to nicks and modified DNA bases has been demonstrated in vitro. Here, we investigate the enzyme's interactions with UVA-induced DNA lesions inside living cells. We irradiated cells expressing GFP-tagged topoisomerase I with an UVA laser focused through a confocal microscope at confined areas of the nuclei. At irradiated sites, topoisomerase I accumulated within seconds, and accumulation lasted for more than 90 min. This effect was apparently due to reduced mobility, although the enzyme was not immobilized at the irradiated nuclear sites. Similar observations were made with mutant versions of topoisomerase I lacking the active site tyrosine or the N-terminal domain, but not with the N-terminal domain alone. Thus, accumulation of topoisomerase I at UVA-modified DNA sites is most likely due to non-covalent binding to damaged DNA, and not suicidal cleavage of such lesions. The rapid onset of accumulation suggests that topoisomerase I functions in this context as a component of DNA damage recognition and/or a cofactor of fast DNA-repair processes. However, the prolonged duration of accumulation suggests that it is also involved in more long-termed processes.
Collapse
Affiliation(s)
- Christian Mielke
- Institute of Clinical Chemistry and Laboratory Diagnostics, Heinrich-Heine-University, Medical School, Moorenstrasse 5, D-40225 Düsseldorf, Germany.
| | | | | | | |
Collapse
|
13
|
Li X, Tao Ng MT, Wang Y, Liu X, Li T. Dumbbell-shaped circular oligonucleotides as inhibitors of human topoisomerase I. Bioorg Med Chem Lett 2007; 17:4967-71. [PMID: 17591440 DOI: 10.1016/j.bmcl.2007.06.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 06/04/2007] [Accepted: 06/07/2007] [Indexed: 10/23/2022]
Abstract
A dumbbell-shaped circular oligonucleotide containing topoisomerase I-binding sites and two mismatched base pairs in its sequence has been designed and synthesized. Our further studies demonstrate that this particularly designed oligonucleotide displays an IC(50) value of 9 nM in its inhibition on the activity of human topoisomerase I, a magnitude smaller than that of camptothecin, an anticancer drug currently in clinical use.
Collapse
Affiliation(s)
- Xinming Li
- Department of Chemistry, 3 Science Drive 3, National University of Singapore, Singapore, Singapore
| | | | | | | | | |
Collapse
|
14
|
Yeung AT, Hattangadi D, Blakesley L, Nicolas E. Enzymatic mutation detection technologies. Biotechniques 2005; 38:749-58. [PMID: 15948293 DOI: 10.2144/05385rv01] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Mutation is as necessary for life as fidelity is in DNA replication. The study of mutations reveals the normal functions of genes, messages, proteins, the causes of many diseases, and the variability of responses among individuals. Indeed, recent mutations that have not yet become polymorphisms are often deleterious and pertinent to the disease history of afflicted individuals. This review discusses the principles behind a variety of methods for the detection of mutations and factors that should be considered in future methods design. One enzymatic approach in particular using orthologs of the CEL I nuclease that show high specificity for all mismatches, appears to be easy and robust. Further developments of this and other methods will allow mutation detection to become an integral component of individualized medicine.
Collapse
|
15
|
Thomas CJ, Rahier NJ, Hecht SM. Camptothecin: current perspectives. Bioorg Med Chem 2004; 12:1585-604. [PMID: 15028252 DOI: 10.1016/j.bmc.2003.11.036] [Citation(s) in RCA: 321] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2003] [Accepted: 11/28/2003] [Indexed: 01/20/2023]
Abstract
This review provides a detailed discussion of recent advances in the medicinal chemistry of camptothecin, a potent antitumor antibiotic. Two camptothecin analogues are presently approved for use in the clinic as antitumor agents and several others are in clinical trials. Camptothecin possesses a novel mechanism of action involving the inhibition of DNA relaxation by DNA topoisomerase I, and more specifically the stabilization of a covalent binary complex formed between topoisomerase I and DNA. This review summarizes the current status of studies of the mechanism of action of camptothecin, including topoisomerase I inhibition and additional cellular responses. Modern synthetic approaches to camptothecin and several of the semi-synthetic methods are also discussed. Finally, a systematic evaluation of novel and important analogues of camptothecin and their contribution to the current structure-activity profile are considered.
Collapse
Affiliation(s)
- Craig J Thomas
- Departments of Chemistry and Biology, University of Virginia, Charlottesville, VA 22901, USA
| | | | | |
Collapse
|
16
|
Yu D, Khan E, Khaleque MA, Lee J, Laco G, Kohlhagen G, Kharbanda S, Cheng YC, Pommier Y, Bharti A. Phosphorylation of DNA topoisomerase I by the c-Abl tyrosine kinase confers camptothecin sensitivity. J Biol Chem 2004; 279:51851-61. [PMID: 15448168 DOI: 10.1074/jbc.m404396200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
DNA topoisomerase I (topo I) is involved in the regulation of DNA supercoiling, gene transcription, recombination, and DNA repair. The anticancer agent camptothecin specifically targets topo I. The mechanisms responsible for the regulation of topo I in cells, however, are not known. This study demonstrates that c-Abl-dependent phosphorylation up-regulates topo I activity. The c-Abl SH3 domain bound directly to the N-terminal region of topo I. The results demonstrate that c-Abl phosphorylated topo I at Tyr268 in core subdomain II. c-Abl-mediated phosphorylation of topo I Tyr268 in vitro and in cells conferred activation of the topo I isomerase function. Moreover, activation of c-Abl by treatment of cells with ionizing radiation was associated with c-Abl-dependent phosphorylation of topo I and induction of topo I activity. The functional significance of the c-Abl/topo I interaction is supported by the findings that (i) mutant topo I(Y268F) exhibited loss of c-Abl-induced topo I activity, and (ii) c-Abl-/- cells were deficient in the accumulation of protein-linked DNA breaks. In addition, loss of topo I phosphorylation in c-Abl-deficient cells conferred resistance to camptothecin-induced apoptosis. These findings collectively support a model in which c-Abl-mediated phosphorylation of topo I is functionally important to topo I activity and sensitivity to topo I poisons.
Collapse
Affiliation(s)
- Donghui Yu
- Department of Adult Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Alessandri M, Beretta GL, Ferretti E, Mancia A, Khobta A, Capranico G. Enhanced CPT Sensitivity of Yeast Cells and Selective Relaxation of Gal4 Motif-containing DNA by Novel Gal4–Topoisomerase I Fusion Proteins. J Mol Biol 2004; 337:295-305. [PMID: 15003448 DOI: 10.1016/j.jmb.2004.01.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2003] [Revised: 01/14/2004] [Accepted: 01/20/2004] [Indexed: 10/26/2022]
Abstract
Human topoisomerase I-B (Top1) efficiently relaxes DNA supercoils during basic cellular processes, and can be transformed into a DNA-damaging agent by antitumour drugs, enzyme mutations and DNA lesions. Here, we describe Gal4-Top1 chimeric proteins (GalTop) with an N-terminal truncation of Top1, and mutations of the Gal4 Zn-cluster and/or Top1 domains that impair their respective DNA-binding activities. Expression levels of chimeras were similar in yeast cells, however, GalTop conferred an increased CPT sensitivity to RAD52- yeast cells as compared to a GalTop with mutations of the Gal4 domain, showing that a functional Gal4 domain can alter in vivo functions of Top1. In vitro enzyme activity was tested with a DNA relaxation assay using negatively supercoiled plasmids with 0 to 5 Gal4 consensus motifs. Only GalTop with a functional Gal4 domain could direct DNA relaxation activity of Top1 specifically to DNA molecules containing Gal4 motifs. By using a substrate competition assay, we could demonstrate that the Gal4-anchored Top1 remains functional and efficiently relax DNA substrates in cis. The enhanced CPT sensitivity of GalTop in yeast cells may then be due to alterations of the chromatin-binding activity of Top1. The GalTop chimeras may indeed mimic a normal mechanism by which Top1 is recruited to chromatin sites in living cells. Such hybrid Top1s may be helpful in further dissecting enzyme functions, and constitute a prototype of a site-specific DNA cutter endowed with high cell lethality.
Collapse
Affiliation(s)
- Marco Alessandri
- Alma Mater Studiorum-University of Bologna, G. Moruzzi Department of Biochemistry, via S. Giacomo 11, 40126 Bologna, Italy
| | | | | | | | | | | |
Collapse
|
18
|
Nitiss JL, Nitiss KC, Rose A, Waltman JL. Overexpression of Type I Topoisomerases Sensitizes Yeast Cells to DNA Damage. J Biol Chem 2001; 276:26708-14. [PMID: 11353773 DOI: 10.1074/jbc.m102674200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
DNA topoisomerases play essential roles in many DNA metabolic processes. It has been suggested that topoisomerases play an essential role in DNA repair. Topoisomerases can introduce DNA damage upon exposure to drugs that stabilize the covalent protein-DNA intermediate of the topoisomerase reaction. Lesions in DNA are also able to trap topoisomerase-DNA intermediates, suggesting that topoisomerases have the potential to either assist in DNA repair by locating sites of damage or exacerbating DNA damage by generation of additional damage at the site of a lesion. We have shown that overexpression of yeast topoisomerase I (TOP1) conferred hypersensitivity to methyl methanesulfonate and other DNA-damaging agents, whereas expression of a catalytically inactive enzyme did not. Overexpression of topoisomerase II did not change the sensitivity of cells to these DNA-damaging agents. Yeast cells lacking TOP1 were not more resistant to DNA damage than cells expressing wild type levels of the enzyme. Yeast topoisomerase I covalent complexes can be trapped efficiently on UV-damaged DNA. We suggest that TOP1 does not participate in the repair of DNA damage in yeast cells. However, the enzyme has the potential of exacerbating DNA damage by forming covalent DNA-protein complexes at sites of DNA damage.
Collapse
Affiliation(s)
- J L Nitiss
- Molecular Pharmacology Department, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA.
| | | | | | | |
Collapse
|
19
|
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
|
20
|
Strumberg D, Pilon AA, Smith M, Hickey R, Malkas L, Pommier Y. Conversion of topoisomerase I cleavage complexes on the leading strand of ribosomal DNA into 5'-phosphorylated DNA double-strand breaks by replication runoff. Mol Cell Biol 2000; 20:3977-87. [PMID: 10805740 PMCID: PMC85758 DOI: 10.1128/mcb.20.11.3977-3987.2000] [Citation(s) in RCA: 277] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Topoisomerase I cleavage complexes can be induced by a variety of DNA damages and by the anticancer drug camptothecin. We have developed a ligation-mediated PCR (LM-PCR) assay to analyze replication-mediated DNA double-strand breaks induced by topoisomerase I cleavage complexes in human colon carcinoma HT29 cells at the nucleotide level. We found that conversion of topoisomerase I cleavage complexes into replication-mediated DNA double-strand breaks was only detectable on the leading strand for DNA synthesis, which suggests an asymmetry in the way that topoisomerase I cleavage complexes are metabolized on the two arms of a replication fork. Extension by Taq DNA polymerase was not required for ligation to the LM-PCR primer, indicating that the 3' DNA ends are extended by DNA polymerase in vivo closely to the 5' ends of the topoisomerase I cleavage complexes. These findings suggest that the replication-mediated DNA double-strand breaks generated at topoisomerase I cleavage sites are produced by replication runoff. We also found that the 5' ends of these DNA double-strand breaks are phosphorylated in vivo, which suggests that a DNA 5' kinase activity acts on the double-strand ends generated by replication runoff. The replication-mediated DNA double-strand breaks were rapidly reversible after cessation of the topoisomerase I cleavage complexes, suggesting the existence of efficient repair pathways for removal of topoisomerase I-DNA covalent adducts in ribosomal DNA.
Collapse
Affiliation(s)
- D Strumberg
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4255, USA
| | | | | | | | | | | |
Collapse
|
21
|
Pourquier P, Takebayashi Y, Urasaki Y, Gioffre C, Kohlhagen G, Pommier Y. Induction of topoisomerase I cleavage complexes by 1-beta -D-arabinofuranosylcytosine (ara-C) in vitro and in ara-C-treated cells. Proc Natl Acad Sci U S A 2000; 97:1885-90. [PMID: 10677551 PMCID: PMC26531 DOI: 10.1073/pnas.97.4.1885] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/1999] [Indexed: 11/18/2022] Open
Abstract
1-beta-d-Arabinofuranosylcytosine (Ara-C) is a nucleoside analog commonly used in the treatment of leukemias. Ara-C inhibits DNA polymerases and can be incorporated into DNA. Its mechanism of cytotoxicity is not fully understood. Using oligonucleotides and purified human topoisomerase I (top1), we found a 4- to 6-fold enhancement of top1 cleavage complexes when ara-C was incorporated at the +1 position (immediately 3') relative to a unique top1 cleavage site. This enhancement was primarily due to a reversible inhibition of top1-mediated DNA religation. Because ara-C incorporation is known to alter base stacking and sugar puckering at the misincorporation site and at the neighboring base pairs, the observed inhibition of religation at the ara-C site suggests the importance of the alignment of the 5'-hydroxyl end for religation with the phosphate group of the top1 phosphotyrosine bond. This study also demonstrates that ara-C treatment and DNA incorporation trap top1 cleavage complexes in human leukemia cells. Finally, we report that camptothecin-resistant mouse P388/CPT45 cells with no detectable top1 are crossresistant to ara-C, which suggests that top1 poisoning is a potential mechanism for ara-C cytotoxicity.
Collapse
Affiliation(s)
- P Pourquier
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4255, USA
| | | | | | | | | | | |
Collapse
|
22
|
Christiansen K, Westergaard O. Mapping of eukaryotic DNA topoisomerase I catalyzed cleavage without concomitant religation in the vicinity of DNA structural anomalies. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1489:249-62. [PMID: 10673027 DOI: 10.1016/s0167-4781(99)00198-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Sensitive sites for covalent trapping of eukaryotic topoisomerase I at DNA structural anomalies were mapped by a new method using purified enzyme and defined DNA substrates. To insure that the obtained topoisomerase I trapping patterns were not influenced by DNA sequence variations, a single DNA imperfection was placed centrally within a homonucleotide track. Mapping of topoisomerase I-mediated irreversible cleavage sites on homopolymeric DNA substrates containing mismatches showed trapping of the enzyme in several positions in close vicinity of the DNA imperfection, with a strong preference for the 5' junction between the duplex DNA and the base-pairing anomaly. On homopolymeric DNA substrates containing a nick, sites of topoisomerase I-mediated cleavage on the intact strand were located just opposite to the nick and from one to ten nucleotides 5' to the nick. Sites of enzyme-mediated cleavage next to a nick and an immobile single-stranded branch were located 5' to the strand interruption in distances of two to six nucleotides and two to ten nucleotides, respectively. Taken together these findings suggest that covalent trapping of topoisomerase I proceeds at positions adjacent to mismatches, nicks and single-stranded branches, where the cleavage reaction is allowed and the ensuing ligation reaction prevented. In principle, the developed interference method might be of general utility to define topoisomerase-DNA interactions relative to different types of structural anomalies.
Collapse
Affiliation(s)
- K Christiansen
- Department of Molecular and Structural Biology, University of Aarhus, Aarhus C, Denmark
| | | |
Collapse
|
23
|
Gobert C, Skladanowski A, Larsen AK. The interaction between p53 and DNA topoisomerase I is regulated differently in cells with wild-type and mutant p53. Proc Natl Acad Sci U S A 1999; 96:10355-60. [PMID: 10468612 PMCID: PMC17892 DOI: 10.1073/pnas.96.18.10355] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
DNA topoisomerase I is a nuclear enzyme involved in transcription, recombination, and DNA damage recognition. Previous studies have shown that topoisomerase I interacts directly with the tumor-suppressor protein p53. p53 is a transcription factor that activates certain genes through binding to specific DNA sequences. We now report that topoisomerase I can be stimulated by both latent and activated wild-type p53 as well as by several mutant and truncated p53 proteins in vitro, indicating that sequence-specific DNA-binding and stimulation of topoisomerase I are distinct properties of p53. These assays also suggest that the binding site for topoisomerase I on p53 is between amino acids 302 and 321. In living cells, the interaction between p53 and topoisomerase I is strongly dependent on p53 status. In MCF-7 cells, which have wild-type p53, the association between the two proteins is tightly regulated in a spatial and temporal manner and takes place only during brief periods of genotoxic stress. In marked contrast, the two proteins are constitutively associated in HT-29 cells, which have mutant p53. These findings have important implications for both cellular stress response and genomic stability, given the ability of topoisomerase I to recognize DNA lesions as well as to cause illegitimate recombination.
Collapse
Affiliation(s)
- C Gobert
- Laboratory of Biology and Pharmacology of DNA Topoisomerases, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8532, Institut Gustave-Roussy, PR2, Villejuif 94805 Cedex, France
| | | | | |
Collapse
|
24
|
Kaur B, Fraser JL, Freyer GA, Davey S, Doetsch PW. A Uve1p-mediated mismatch repair pathway in Schizosaccharomyces pombe. Mol Cell Biol 1999; 19:4703-10. [PMID: 10373519 PMCID: PMC84268 DOI: 10.1128/mcb.19.7.4703] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UV damage endonuclease (Uve1p) from Schizosaccharomyces pombe was initially described as a DNA repair enzyme specific for the repair of UV light-induced photoproducts and proposed as the initial step in an alternative excision repair pathway. Here we present biochemical and genetic evidence demonstrating that Uve1p is also a mismatch repair endonuclease which recognizes and cleaves DNA 5' to the mispaired base in a strand-specific manner. The biochemical properties of the Uve1p-mediated mismatch endonuclease activity are similar to those of the Uve1p-mediated UV photoproduct endonuclease. Mutants lacking Uve1p display a spontaneous mutator phenotype, further confirming the notion that Uve1p plays a role in mismatch repair. These results suggest that Uve1p has a surprisingly broad substrate specificity and may function as a general type of DNA repair protein with the capacity to initiate mismatch repair in certain organisms.
Collapse
Affiliation(s)
- B Kaur
- Department of Biochemistry, Graduate Program in Biochemistry and Cell and Developmental Biology, Emory University, School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
25
|
Pourquier P, Ueng LM, Fertala J, Wang D, Park HJ, Essigmann JM, Bjornsti MA, Pommier Y. Induction of reversible complexes between eukaryotic DNA topoisomerase I and DNA-containing oxidative base damages. 7, 8-dihydro-8-oxoguanine and 5-hydroxycytosine. J Biol Chem 1999; 274:8516-23. [PMID: 10085084 DOI: 10.1074/jbc.274.13.8516] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We recently showed that abasic sites, uracil mismatches, nicks, and gaps can trap DNA topoisomerase I (top1) when these lesions are introduced in the vicinity of a top1 cleavage site (Pourquier, P., Ueng, L.-M., Kohlhagen, G., Mazumder, A., Gupta, M., Kohn, K. W., and Pommier, Y. (1997) J. Biol. Chem. 272, 7792-7796; Pourquier, P., Pilon, A. A., Kohlhagen, G., Mazumder, A., Sharma, A., and Pommier, Y. (1997) J. Biol. Chem. 26441-26447). In this study, we investigated the effects on top1 of an abundant base damage generated by various oxidative stresses: 7,8-dihydro-8-oxoguanine (8-oxoG). Using purified eukaryotic top1 and oligonucleotides containing the 8-oxoG modification, we found a 3-7-fold increase in top1-mediated DNA cleavage when 8-oxoG was present at the +1 or +2 position relative to the cleavage site. Another oxidative lesion, 5-hydroxycytosine, also enhanced top1 cleavage by 2-fold when incorporated at the +1 position of the scissile strand. 8-oxoG at the +1 position enhanced noncovalent top1 DNA binding and had no detectable effect on DNA religation or on the incision step. top1 trapping by 8-oxoG was markedly enhanced when asparagine adjacent to the catalytic tyrosine was mutated to histidine, suggesting a direct interaction between this residue and the DNA major groove immediately downstream from the top1 cleavage site. Altogether, these results demonstrate that oxidative base lesions can increase top1 binding to DNA and induce top1 cleavage complexes.
Collapse
Affiliation(s)
- P Pourquier
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, NCI, National Institutes of Health, Bethesda, Maryland 20892-4255, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Pourquier P, Bjornsti MA, Pommier Y. Induction of topoisomerase I cleavage complexes by the vinyl chloride adduct 1,N6-ethenoadenine. J Biol Chem 1998; 273:27245-9. [PMID: 9765247 DOI: 10.1074/jbc.273.42.27245] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We used purified mammalian topoisomerases I (top1) and oligonucleotides to study top1-mediated cleavage and religation in the presence of a potent carcinogenic adduct, 1,N6-ethenoadenosine (epsilonA) incorporated immediately downstream of a unique top1 cleavage site. We found tha epsilonA markedly enhanced top1 cleavage complexes when it was incorporated at the +1 position of the top1 cleavage. This enhancement was due to a reduction of the religation step of the top1 reaction. In addition, epsilonA reduced the top1-mediated cleavage and decreased binding of the enzyme to DNA. We also studied the effects of the epsilonA adduct on top1 trapping by camptothecin (CPT), a well known top1 inhibitor. CPT was inactive when epsilonA was present at the +1 position. Alkylation of the top1 cleavage complex by 7-chloromethyl-10,11-methylenedioxycamptothecin (7-ClMe-MDO-CPT) was also blocked by the epsilonA adduct. Altogether, these results demonstrate that the epsilonA carcinogenic adduct can efficiently trap human top1 and mimic CPT effects. Normal hydrogen bonding of the base pairs immediately downstream from the top1 cleavage site is probably essential for efficient DNA religation and binding of camptothecins in the top1 cleavage complex.
Collapse
Affiliation(s)
- P Pourquier
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, NCI, National Institutes of Health, Bethesda, Maryland 20892-4255, USA
| | | | | |
Collapse
|
27
|
Kingma PS, Osheroff N. The response of eukaryotic topoisomerases to DNA damage. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1400:223-32. [PMID: 9748592 DOI: 10.1016/s0167-4781(98)00138-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Beyond the known mutagenic properties of DNA lesions, recent evidence indicates that several forms of genomic damage dramatically influence the catalytic activities of DNA topoisomerases. Apurinic sites, apyrimidinic sites, base mismatches, and ultraviolet photoproducts all enhance topoisomerase I-mediated DNA cleavage when they are located in close proximity to the point of scission. Furthermore, when located between the points of scission of a topoisomerase II cleavage site, these same lesions (with the exception of ultraviolet photoproducts) greatly stimulate the cleavage activity of the type II enzyme. Thus, as found for anticancer drugs, lesions have the capacity to convert topoisomerases from essential cellular enzymes to potent DNA toxins. These findings raise exciting new questions regarding the mechanism of anticancer drugs, the physiological functions of topoisomerases, and the processing of DNA damage in the cell.
Collapse
Affiliation(s)
- P S Kingma
- Department of Biochemistry, 654 Medical Research Building I, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, USA
| | | |
Collapse
|
28
|
Pommier Y, Pourquier P, Fan Y, Strumberg D. Mechanism of action of eukaryotic DNA topoisomerase I and drugs targeted to the enzyme. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1400:83-105. [PMID: 9748515 DOI: 10.1016/s0167-4781(98)00129-8] [Citation(s) in RCA: 422] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
DNA topoisomerase I is essential for cellular metabolism and survival. It is also the target of a novel class of anticancer drugs active against previously refractory solid tumors, the camptothecins. The present review describes the topoisomerase I catalytic mechanisms with particular emphasis on the cleavage complex that represents the enzyme's catalytic intermediate and the site of action for camptothecins. Roles of topoisomerase I in DNA replication, transcription and recombination are also reviewed. Because of the importance of topoisomerase I as a chemotherapeutic target, we review the mechanisms of action of camptothecins and the other topoisomerase I inhibitors identified to date.
Collapse
Affiliation(s)
- Y Pommier
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, Bethesda, MD 20892-4255, USA.
| | | | | | | |
Collapse
|
29
|
Capranico G, Binaschi M. DNA sequence selectivity of topoisomerases and topoisomerase poisons. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1400:185-94. [PMID: 9748568 DOI: 10.1016/s0167-4781(98)00135-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chemical agents able to interfere with DNA topoisomerases are widespread in nature, and some of them have clinical efficacy as antitumor or antibacterial drugs. Drugs which have as a target DNA topoisomerases could be divided into two categories: poisons and catalytic inhibitors. Classical topoisomerase poisons stimulate cleavage in a sequence-selective manner, yielding drug-specific cleavage intensity pattern. The mechanisms of drug interaction with DNA topoisomerases, the DNA sequence selectivity of the action of topoisomerase II poisons and the identification of structural determinants of their activity have suggested that topoisomerase II poisons may fit into a specific pharmacophore, constituted by a planar ring system with DNA intercalation or intercalation-like properties, and protruding side chains interfering with the protein side of the covalent enzyme-DNA complex. The complete definition of the diverse pharmacophores of topoisomerase II poisons will certainly be of value for the design of new agents directed to specific genomic sites, and more effective in the treatment of human cancer.
Collapse
Affiliation(s)
- G Capranico
- Division of Experimental Oncology B, Istituto Nazionale per lo Studio e la Cura dei Tumori, via Venezian 1, 20133 Milan, Italy.
| | | |
Collapse
|
30
|
Bugreev DV, Vasyutina EL, Kolocheva TI, Buneva VN, Andoh T, Nevinsky GA. Interaction of human DNA topoisomerase I with specific sequence oligodeoxynucleotides. Biochimie 1998; 80:303-8. [PMID: 9672749 DOI: 10.1016/s0300-9084(98)80071-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The interaction of human DNA topoisomerase I (topo I) with specific sequence oligodeoxynucleotides (ODNs) of different length and structure has been investigated. All the ODNs used were shown to be effective enzyme inhibitors and to inhibit the topo I catalyzed relaxation of scDNA in a competitive manner. Among two DNA regions (A and B) required for topo I-mediated DNA cleavage, the former was found to display the higher affinity for the enzyme. The enzyme's affinity for ODNs corresponding to the scissile strand (five and nine nucleotide units in length) is about 2-4 orders of magnitude higher than that for non-specific ODNs of the same length. Topo I can efficiently recognize even extremely short specific ODNs containing only two or three bases (AGA and pAG, Ki = 15 and 60 microM, respectively): the sequence AAGA (Ki = 10 microM) is essential for tight DNA binding to topo I. The affinities of ODNs corresponding to the non-scissile strand are significantly lower. The ligand's affinity increases with its length. Additionally, about a ten-fold enhancement of specific sequence affinity occurs due to stable duplex formation during enzyme preincubation with ligands before addition of scDNA. We believe the possibility of using the short specific oligonucleotides and its derivatives as topoisomerase I-targeting drugs could not be excluded.
Collapse
Affiliation(s)
- D V Bugreev
- Novosibirsk Institute of Bioorganic Chemistry, Siberian Division of the Russian Academy of Sciences, Russia
| | | | | | | | | | | |
Collapse
|
31
|
Topoisomerase I-targeting drugs. ACTA ACUST UNITED AC 1998. [DOI: 10.1016/s1067-568x(98)80005-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
32
|
Boothman DA, Burrows HL, Yang CR, Davis TW, Wuerzberger SM, Planchon SM, Odegaard E, Lewis JE, Pink J, Meyers M, Patten CW, Sharda N, Kinsella TJ. Damage-sensing mechanisms in human cells after ionizing radiation. Stem Cells 1997; 15 Suppl 2:27-42. [PMID: 9368284 DOI: 10.1002/stem.5530150707] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Human cells have evolved several mechanisms for responding to damage created by ionizing radiation. Some of these responses involve the activation or suppression of the transcriptional machinery. Other responses involve the downregulation of enzymes, such as topoisomerase I, which appear to be necessary for DNA repair or apoptosis. Over the past five years, many studies have established links between DNA damage, activation of transcription factors that are coupled to DNA repair mechanisms, increased gene transcription and altered cell cycle regulation to allow for repair or cell death via apoptosis or necrosis. Together these factors determine whether a cell will survive with or without carcinogenic consequences. The immediate responses of human cells to ionizing radiation, in terms of sensing and responding to damage, are therefore, critical determinants of cell survival and carcinogenesis.
Collapse
Affiliation(s)
- D A Boothman
- Department of Human Oncology, University of Wisconsin-Madison
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Pourquier P, Pilon AA, Kohlhagen G, Mazumder A, Sharma A, Pommier Y. Trapping of mammalian topoisomerase I and recombinations induced by damaged DNA containing nicks or gaps. Importance of DNA end phosphorylation and camptothecin effects. J Biol Chem 1997; 272:26441-7. [PMID: 9334220 DOI: 10.1074/jbc.272.42.26441] [Citation(s) in RCA: 138] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We used purified mammalian topoisomerases I (top1) and oligonucleotides containing a unique top1 cleavage site to study top1-mediated cleavage and recombination in the presence of nicks and short gaps mimicking DNA damage. In general, top1 cleavage was not induced opposite to the nicks, and nicks upstream from the top1 cleavage site suppressed top1 activity. Irreversible top1 cleavage complexes ("suicide products" or "aborted complexes") were produced in DNA containing nicks or short gaps just opposite to the normal top1 cleavage site. Camptothecin enhanced the formation of the aborted top1 complexes only for nicks downstream from the cleavage site. These aborted (suicide) complexes can mediate DNA recombination and promote illegitimate recombination by catalyzing the ligation of nonhomologous DNA fragments (acceptors). We report for the first time that top1-mediated recombination is greatly enhanced by the presence of a phosphate at the 5' terminus of the top1 aborted complex (donor DNA). By contrast, phosphorylation of the 3' terminus of the gap did not affect recombination. At concentrations that strongly enhanced inhibition of intramolecular religation, resulting in an increase of top1 cleavable complexes, camptothecin did not reduce recombination (intermolecular religation). Nicks or gaps with 5'-phosphate termini would be expected to be produced directly by ionizing radiations or by processing of abasic sites and DNA lesions induced by carcinogens or drugs used in cancer chemotherapy. Thus, these results further demonstrate that DNA damage can efficiently trap top1-cleavable complexes and enhance top1-mediated DNA recombination.
Collapse
Affiliation(s)
- P Pourquier
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, NCI, National Institutes of Health, Bethesda, Maryland 20892-4255, USA
| | | | | | | | | | | |
Collapse
|
34
|
Parsels LA, Zellars RC, Loney TL, Parsels JD, Clarke MF, Merchant AK, Lawrence TS, Maybaum J. Prevention of fluorodeoxyuridine-induced cytotoxicity and DNA damage in HT29 colon carcinoma cells by conditional expression of wild-type p53 phenotype. Mol Pharmacol 1997; 52:600-5. [PMID: 9380022 DOI: 10.1124/mol.52.4.600] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We have examined the effects of conditionally expressing wild-type p53 activity in HT29 cells on DNA damage and cytotoxicity caused by exposure to fluorodeoxyuridine (FdUrd). Expression of wild-type p53 phenotype for 24 hr before FdUrd treatment provided HT29 cells with virtually complete protection from cytotoxicity caused by this drug. In addition, wild-type p53 expression also prevented FdUrd-induced DNA double-strand breaks and, unexpectedly, single-strand breaks in parental (mature) DNA. Temporary expression of wild-type p53 activity in the absence of drug treatment caused some loss of clonogenicity, although the magnitude of this cytotoxic effect was small compared with the level of cell kill obtained by treatment with cytotoxic drugs for similar periods of time, indicating that HT29 cells are not highly sensitive to induction of programmed cell death by wild-type p53. Because these observations conflict with previously suggested models for FdUrd-induced damage to parental DNA, we propose an alternative model to explain how incorporation of uracil into nascent DNA might result in single-strand breaks in the opposite (parental) strand and how these breaks might be converted to the double-strand breaks that produce cell death.
Collapse
Affiliation(s)
- L A Parsels
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0504, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Stewart L, Ireton GC, Champoux JJ. Reconstitution of human topoisomerase I by fragment complementation. J Mol Biol 1997; 269:355-72. [PMID: 9199405 DOI: 10.1006/jmbi.1997.1056] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Human topoisomerase I (topo I, 91 kDa) is composed of four major domains; the unconserved and highly charged "N-terminal" domain (24 kDa), the conserved "core" domain (54 kDa), a poorly conserved and positively charged "linker" region (5 kDa), and the highly conserved "C-terminal" domain (8 kDa) which contains the active site tyrosine at position 723. Here we demonstrate that human topo I activity can be reconstituted by mixing a 58 kDa recombinant core domain (residues Lys175 to Ala659) with any one of a series of recombinant C-terminal fragments that range in size from 12 kDa (linker and C-terminal domains, residues Leu658 to Phe765) to 6.3 kDa (C-terminal domain residues Gln713 to Phe765). The C-terminal fragments bind tightly to the core domain, forming a 1:1 complex that is stable irrespective of ionic strength (0.01 to 1 M). The reconstituted enzymes are active only over a relatively narrow range of salt concentrations (25 to 200 mM KCl) as compared to the intact topo70 enzyme (missing the N-terminal domain). Under physiological conditions (150 mM KCl and 10 mM Mg2+) they are much more distributive in their mode of action than topo70. The reconstituted enzyme binds DNA with an affinity that is approximately 20-fold lower than that of the intact topo70. In addition, the cleavage/religation equilibrium of the reconstituted enzyme appears to be biased towards religation relative to that of the intact enzyme. Despite differences in the cleavage/religation equilibrium and affinity for DNA, the reconstituted and intact enzymes have identical sequence specificities for the cleavage of duplex DNA or suicide cleavage of oligonucleotide substrates.
Collapse
Affiliation(s)
- L Stewart
- Department of Microbiology, School of Medicine, University of Washington, Seattle 98195-7242, USA
| | | | | |
Collapse
|
36
|
Pourquier P, Ueng LM, Kohlhagen G, Mazumder A, Gupta M, Kohn KW, Pommier Y. Effects of uracil incorporation, DNA mismatches, and abasic sites on cleavage and religation activities of mammalian topoisomerase I. J Biol Chem 1997; 272:7792-6. [PMID: 9065442 DOI: 10.1074/jbc.272.12.7792] [Citation(s) in RCA: 152] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Abasic sites and deamination of cytosine to uracil are probably the most common types of endogenous DNA damage. The effects of such lesions on DNA topoisomerase I (top1) activity were examined in oligonucleotides containing a unique top1 cleavage site. The presence of uracils and abasic sites within the first 4 bases immediately 5' to the cleavage site suppressed normal top1 cleavage and induced new top1 cleavage sites. Uracils immediately 3' to the cleavage site increased cleavage and produced a camptothecin mimicking effect. A mismatch with a bulge or abasic sites immediately 3' to the top1 cleavage site irreversibly trapped top1 cleavable complexes in the absence of camptothecin and produced a suicide cleavage complex. These results demonstrate that top1 activity is sensitive to physiological, environmental, and pharmacological DNA modifications and that top1 can act as a specific mismatch- and abasic site-nicking enzyme.
Collapse
Affiliation(s)
- P Pourquier
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, NCI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- R Kolodner
- Charles A. Dana Division of Human Cancer Genetics, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| |
Collapse
|
38
|
Lanza A, Tornaletti S, Rodolfo C, Scanavini MC, Pedrini AM. Human DNA topoisomerase I-mediated cleavages stimulated by ultraviolet light-induced DNA damage. J Biol Chem 1996; 271:6978-86. [PMID: 8636127 DOI: 10.1074/jbc.271.12.6978] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
DNA topoisomerases have been proposed as the proteins involved in the formation of the DNA-protein cross-links detected after ultraviolet light (UV) irradiation of cellular DNA. This possibility has been investigated by studying the effects of UV-induced DNA damage on human DNA topoisomerase I action. UV lesions impaired the enzyme's ability to relax negatively supercoiled DNA. Decreased relaxation activity correlated with the stimulation of cleavable complexes. Accumulation of cleavable complexes resulted from blockage of the rejoining step of the cleavage-religation reaction. Mapping of cleavage sites on the pAT153 genome indicated UV-induced cleavage at discrete positions corresponding to sites stimulated also by the topoisomerase I inhibitor camptothecin, except for one. Subsequent analysis at nucleotide level within the sequence encompassing the UV-specific cleavage site revealed the precise positions of sites stimulated by camptothecin with respect to those specific for UV irradiation. Interestingly, one of the UV-stimulated cleavage sites was formed within a sequence that did not contain dimerized pyrimidines, suggesting transmission of the distortion, caused by photodamage to DNA, into the neighboring sequences. These results support the proposal that DNA structural alterations induced by UV lesions can be sufficient stimulus to induce cross-linking of topoisomerase I to cellular DNA.
Collapse
Affiliation(s)
- A Lanza
- Istituto di Genetica Biochimica ed Evoluzionistica del CNR, Via Abbiategrasso, 207-27100 Pavia, Italy
| | | | | | | | | |
Collapse
|
39
|
O'Regan NE, Branch P, Macpherson P, Karran P. hMSH2-independent DNA mismatch recognition by human proteins. J Biol Chem 1996; 271:1789-96. [PMID: 8576184 DOI: 10.1074/jbc.271.3.1789] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Two distinct mismatch binding activities are detected using bandshift assays with human cell extracts and DNA with mispairs at defined positions. One requires hMSH2 protein and is absent from extracts of LoVo cells, which contain a partial deletion of the hMSH2 gene. The second activity is independent of hMSH2 and is present at normal levels in LoVo and three other cell lines, which are defective in in vitro hMSH2-dependent binding. The two mismatch recognition activities are distinguished by their sensitivity to polycations and can be resolved by chromatography on MonoQ. hMSH2-independent activity has been purified extensively from wild-type cells and from a cell line deficient in hMSH2-dependent binding. The purified material preferentially recognizes A-C, some pyrimidine-pyrimidine mismatches, and certain slipped mispaired structures. Binding exhibits some sequence preferences. The similar properties of the two mismatch binding activities suggest that they both contribute to mismatch repair.
Collapse
Affiliation(s)
- N E O'Regan
- Imperial Cancer Research Fund, Clare Hall Laboratories, South Mimms, United Kingdom
| | | | | | | |
Collapse
|
40
|
Bigioni M, Zunino F, Tinelli S, Austin CA, Willmore E, Capranico G. Position-specific effects of base mismatch on mammalian topoisomerase II DNA cleaving activity. Biochemistry 1996; 35:153-9. [PMID: 8555169 DOI: 10.1021/bi951736p] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
To further define the nucleic acid determinants of DNA site recognition by mammalian topoisomerase II, base mismatch effects on the enzyme DNA cleavage activity were determined in a 36-bp synthetic oligonucleotide corresponding to SV40 DNA. DNA cleavage sites induced by topoisomerase II without or with the antitumor drugs teniposide, idarubicin, or amsacrine were mapped using sequencing gels. Selected mismatches were studied, and always one of the two strands had the wild-type sequence. The effects of base mismatches were independent from the studied drugs. Mismatches introduced at the -4, -3, -2, or -1 positions, relative to the enzyme cleavage site, often abolished, or much reduced, DNA cleavage, whereas those at +1 and +2 positions often increased DNA breakage or were without influence. Mismatches at more distant positions, e.g., -7, -8, etc., had no effect. Those at positions -5 and -6 sometimes increased cleavage levels. These effects were always observed at sites already cleaved in the wild-type oligomer; new sites of cleavage were not induced by the studied mismatches. These results were obtained both for the native murine topoisomerase II and for the two recombinant human isozymes. No difference between topoisomerases II alpha(p170) and beta(p180) was seen in their response to mismatches. The results demonstrate that topoisomerase II recognition of the DNA site of cleavage requires fully paired nucleotides at the 3' terminus. Nevertheless, similarly to other DNA strand transferase enzymes, both topoisomerase II isoforms may have a sequence-specific nicking activity at the 5' side of unpaired bases.
Collapse
Affiliation(s)
- M Bigioni
- Division of Experimental Oncology B, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | | | | | | | | | | |
Collapse
|
41
|
Review. Clin Chem Lab Med 1996. [DOI: 10.1515/cclm.1996.34.11.873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
42
|
Nevinsky GA, Bugreev DV, Buneva VN, Yasui Y, Nishizawa M, Andoh T. High affinity interaction of mammalian DNA topoisomerase I with short single- and double-stranded oligonucleotides. FEBS Lett 1995; 368:97-100. [PMID: 7615096 DOI: 10.1016/0014-5793(95)00609-d] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The interaction of DNA topoisomerase I (topo I) with a set of single- and double-stranded oligonucleotides containing 5-27 mononucleotides was investigated. All single- and double-stranded oligonucleotides were found to inhibit competitively the supercoiled DNA relaxation reaction catalyzed by topo I. The enzyme affinity for specific sequence pentanucleotides of the scissile (GACTT, Ki = 2 microM) and non-cleaved chain (AAGTC, Ki = 110 microM) is about 2-4 orders of magnitude higher than that for non-specific oligonucleotides. This specific sequence affinity increases in several cases; lengthening of single-stranded oligonucleotides, formation of stable duplexes between complementary oligonucleotides and preincubation of the enzyme with ligands before addition of supercoiled DNA. We assume that oligonucleotides having a high affinity to the enzyme can offer a unique opportunity for rational design of topoisomerase-targeting drugs.
Collapse
Affiliation(s)
- G A Nevinsky
- Novosibirsk Institute of Bioorganic Chemistry, Siberian Division of the Russian Academy of Sciences
| | | | | | | | | | | |
Collapse
|
43
|
Gupta M, Fujimori A, Pommier Y. Eukaryotic DNA topoisomerases I. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1262:1-14. [PMID: 7772596 DOI: 10.1016/0167-4781(95)00029-g] [Citation(s) in RCA: 181] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- M Gupta
- Laboratory of Molecular Pharmacology, National Cancer Institute, National Institute of Health, Bethesda, MD 20892-4255, USA
| | | | | |
Collapse
|
44
|
Yao M, Kow YW. Strand-specific cleavage of mismatch-containing DNA by deoxyinosine 3'-endonuclease from Escherichia coli. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(18)31706-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|