ABSTRACT
We have conducted an experimental and computational evaluation of new doxorubicin (4a-c) and ß-lapachone (5a-c) analogs. These novel anticancer analogs were previously synthesized, but had not been tested or characterized until now. We have evaluated their antiproliferative and DNA cleavage inhibition properties using breast (MCF-7 and MDA-MB-231) and prostate (PC3) cancer cell lines. Additionally, cell cycle analysis was performed using flow cytometry. Computational studies, including molecular docking, pharmacokinetic properties, and an analysis of DFT and QTAIM chemical descriptors, were performed to gain insights into the electronic structure and elucidate the molecular binding of the new ß-lapachone and doxorubicin analogs with a DNA sequence and Topoisomerase II (Topo II)α. Our results show that 4a analog displays the highest antiproliferative activity in cancer cell lines by inducing cell death. We observed that stacking interactions and hydrogen bonding are essential to stabilize the molecule-DNA-Topo IIα complex. Moreover, 4a and 5a analogs inhibited Topo's DNA cleavage activity. Pharmacodynamic results indicated that studied molecules have favorable adsorption and permeability properties. The calculated chemical descriptors indicate that electron accumulation in quinone rings is relevant to the reactivity and biological activity. Based on our results, 4a is a strong candidate for becoming an anticancer drug.
Subject(s)
Antineoplastic Agents , Cell Proliferation , DNA Topoisomerases, Type II , Doxorubicin , Molecular Docking Simulation , Naphthoquinones , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , DNA Topoisomerases, Type II/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , MCF-7 Cells , Drug Screening Assays, Antitumor , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/metabolism , DNA Cleavage/drug effectsABSTRACT
In this work we will discuss the antiproliferative evaluation and the possible mechanisms of action of indole-thiosemicarbazone compounds LTs with anti-inflammatory activity, previously described in the literature. In this perspective, some analyzes were carried out, such as the study of binding to human serum albumin (HSA) and to biological targets: DNA and human topoisomerase IIα (topo). Antiproliferative study was performed with DU-145, Jukart, MCF-7 and T-47D tumor lines and J774A.1, besides HepG2 macrophages and hemolytic activity. In the HSA interaction tests, the highest binding constant was 3.70 × 106 M-1, referring to LT89 and in the fluorescence, most compounds, except for LT76 and LT87, promoted fluorescent suppression with the largest Stern-Volmer constant for the LT88 3.55 × 104. In the antiproliferative assay with DU-145 and Jurkat strains, compounds LT76 (0.98 ± 0.10/1.23 ± 0.32 µM), LT77 (0.94 ± 0.05/1.18 ± 0.08 µM) and LT87 (0.94 ± 0.12/0.84 ± 0.09 µM) stood out, due to their IC50 values mentioned above. With the MCF-7 and T-47D cell lines, the lowest IC50 was presented by LT81 with values of 0.74 ± 0.12 µM and 0.68 ± 0.10 µM, respectively, followed by the compounds LT76 and LT87. As well as the positive control amsacrine, the compounds LT76, LT81 and LT87 were able to inhibit the enzymatic action of human Topoisomerase IIα.
Subject(s)
Antineoplastic Agents , Thiosemicarbazones , Humans , Molecular Docking Simulation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Structure-Activity Relationship , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Cell Line, Tumor , Topoisomerase II Inhibitors/pharmacology , DNA/pharmacology , DNA Topoisomerases, Type II/metabolism , Indoles/pharmacology , Indoles/chemistry , Cell ProliferationABSTRACT
Topoisomerase inhibitors are extensively used in cancer chemotherapy. In the process of identifying novel anticancer compounds, biological evaluations are crucial and include, among others, the use of in silico and in vitro approaches. This work aimed to present recent research involving the obtainment and in silico and in vitro evaluation of topoisomerase I, II, and double inhibitors, of synthetic and natural origin, as potential compounds against tumor cells, in addition to proposing the construction of a desirable enzyme catalytic site. Therefore, it was observed that most Topoisomerase I inhibitors presented medium to large structures, with a rigid portion and a flexible region. In contrast, Topoisomerase IIα inhibitors showed medium and large structural characteristics, in addition to the planarity of the aromatic rings, which are mitigated due to flexible rings but may also present elements that restrict conformation. Most compounds that exhibit dual inhibitory activity had relatively long chains, in addition to a flat and rigid portion suggestive of affinity for Topo I and a flexible region characteristic of selective drugs for Topo II. Besides, it is noticed that most compounds that exhibit dual inhibitory showed similarities in the types of interactions and amino acids when compared to the selective compounds of Topo I and II. For instance, selective Topoisomerase I inhibitors interact with Arginine364 residues, and selective Topoisomerase II inhibitors interact with Arginine487 residues, as both residues are targets for dual compounds.
Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Antineoplastic Agents/chemistry , Cell Proliferation , DNA Topoisomerases, Type II/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Neoplasms/drug therapy , Structure-Activity Relationship , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/therapeutic use , Topoisomerase I Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/metabolismABSTRACT
In this study, eight natural isocoumarins (1-8) were isolated from a marine-derived Exserohilum sp. fungus. To explore their structure-activity relationship and discover potent antimalarial leads, a small library of 22 new derivatives (1a-1n, 2a, 3a-3c, 4a-4c, and 7a) were semisynthesized by varying the substituents of the aromatic ring and the aliphatic side chains. The natural compound (1) and three semisynthetic derivatives (1d, 1n, and 2a), possessing an all-cis stereochemistry, exhibited strong antiplasmodial activity with IC50 values of 1.1, 0.8, 0.4, and 2.6 µM, respectively. Mechanism studies show that 1n inhibits hemozoin polymerization and decreases the mitochondrial membrane potential but also inhibits P. falciparum DNA gyrase. 1n not only combines different mechanisms of action but also exhibits a high therapeutic index (CC50/IC50 = 675), high selectivity, and a notable drug-like profile.
Subject(s)
Antimalarials/pharmacology , Ascomycota/chemistry , Isocoumarins/pharmacology , Animals , Anthozoa/microbiology , Antimalarials/chemical synthesis , Aquatic Organisms/chemistry , China , Chlorocebus aethiops , DNA Gyrase , Hemeproteins , Isocoumarins/chemical synthesis , Membrane Potential, Mitochondrial/drug effects , Molecular Structure , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Topoisomerase II Inhibitors/pharmacology , Vero CellsABSTRACT
From a medical point of view lot of existing antibiotics became unusable because microbial gained strong antibiotic resistance. The combination of two compounds in one core may lead to kill such type of pathogens. Herein, we developed pyranopyrazole derivatives comprising benzoxazole moiety by green approach strategy and studied their antimicrobial performance on four bacteria and two fungi. As a result, most of the compounds delivered reliable toxicity to kill the pathogens. In those,6aexhibited considerable activity against the microbial pathogens. Moreover,compounds 6d, 6l,and6nshowed prominent antibacterial activity. In addition, molecular docking studies of docked compounds revealed the strong bonding interaction with DNA-Gyrase and were docked into the intercalation location of DNA of the DNA-gyrase complex. The molecule bounded to the DNA stabilized by the H bonds, hydrophobic interactions, and π-π interaction. In addition, the linked 5-chlorobenazoxazole structure stabilized by the DT-8 and DG2009 of the F chain with pi-pi interactions. From the computer-aided results, it was observed that compound6a demonstrated maximum docking score -10.0 kcal/mole towards DNA-gyrase. Overall, this investigation suggested that these biologically active compounds can be utilized as leads for preclinical studies with the goal of developing newer antimicrobial drugs.
Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Anti-Infective Agents/chemical synthesis , Bacteria/drug effects , Bacteria/enzymology , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Benzoxazoles/chemical synthesis , DNA Gyrase/metabolism , Drug Design , Drug Resistance, Bacterial , Drug Resistance, Fungal , Fungi/drug effects , Fungi/enzymology , Green Chemistry Technology , Humans , Molecular Docking Simulation , Mycoses/drug therapy , Mycoses/microbiology , Pyrazoles/chemical synthesis , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacologyABSTRACT
Twelve 2-(quinolin-4-ylmethylene) hydrazinecarbothioamide derivatives were synthetized and their biological properties were investigated, among which, the ability to interact with DNA and BSA through UV-Vis absorption, fluorescence, Circular Dichroism, molecular docking and relative viscosity, antiproliferative activity against MCF-7 and T-47D mammary tumor cells and RAW-264.7 macrophages and inhibitory capacity of the enzyme topoisomerase IIα. In the binding study with DNA and BSA, all the compounds displayed affinity for interaction with both biomolecules, especially JF-92 (p-ethyl-substituted), with binding constant of 1.62â¯×â¯106 and 1.43â¯×â¯105, respectively, and DNA binding mode by intercalation. The IC50 values were obtained between 0.81 and 1.48⯵M and topoisomerase inhibition results in 10⯵M. Thus, we conclude that the reduction of the acridine to quinoline ring did not disrupt the antitumor action and that substitution patterns are important for biomolecule interaction affinity as they demonstrate the potential of these compounds for anticancer therapy.
Subject(s)
Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , Quinolines/pharmacology , Thiosemicarbazones/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Macromolecular Substances/chemical synthesis , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Models, Molecular , Molecular Structure , Quinolines/chemical synthesis , Quinolines/chemistry , RAW 264.7 Cells , Structure-Activity Relationship , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/chemistry , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , ViscosityABSTRACT
Common approaches to antibiotic discovery include small-molecule screens for growth inhibition in target pathogens and screens for inhibitors of purified enzymes. These approaches have a shared intent of seeking to directly target a vital Achilles heel in a pathogen of interest. Here, we report the first screen against a sporulation pathway in a non-pathogenic bacterium as a means of discovering novel antibiotics-this effort has resulted in two important discoveries. First, we show that the sporulation program of Streptomyces venezuelae is exquisitely sensitive to numerous forms of DNA damage. Second, we have identified a DNA gyrase inhibitor. This molecule, EN-7, is active against pathogenic species that are resistant to ciprofloxacin and other clinically important antibiotics. We suggest that this strategy could be applied to other morphogenetic pathways in prokaryotes or eukaryotes as a means of identifying novel chemical matter having scientific and clinical utility.
Subject(s)
DNA Gyrase/drug effects , Drug Discovery/methods , Topoisomerase II Inhibitors/isolation & purification , Anti-Bacterial Agents/pharmacology , DNA Gyrase/metabolism , Microbial Sensitivity Tests , Spores, Bacterial/drug effects , Streptomyces/drug effects , Streptomyces/metabolism , Topoisomerase II Inhibitors/metabolism , Topoisomerase II Inhibitors/pharmacologyABSTRACT
BACKGROUND: Considering the need for the development of new antitumor drugs, associated with the great antitumor potential of thiophene and thiosemicarbazonic derivatives, in this work we promote molecular hybridization approach to synthesize new compounds with increased anticancer activity. OBJECTIVE: Investigate the antitumor activity and their likely mechanisms of action of a series of N-substituted 2-(5-nitro-thiophene)-thiosemicarbazone derivatives. METHODS: Methods were performed in vitro (cytotoxicity, cell cycle progression, morphological analysis, mitochondrial membrane potential evaluation and topoisomerase assay), spectroscopic (DNA interaction studies), and in silico studies (docking and molecular modelling). RESULTS: Most of the compounds presented significant inhibitory activity; the NCIH-292 cell line was the most resistant, and the HL-60 cell line was the most sensitive. The most promising compound was LNN-05 with IC50 values ranging from 0.5 to 1.9 µg.mL-1. The in vitro studies revealed that LNN-05 was able to depolarize (dose-dependently) the mitochondrial membrane, induceG1 phase cell cycle arrest noticeably, promote morphological cell changes associated with apoptosis in chronic human myelocytic leukaemia (K-562) cells, and presented no topoisomerase II inhibition. Spectroscopic UV-vis and molecular fluorescence studies showed that LNN compounds interact with ctDNA forming supramolecular complexes. Intercalation between nitrogenous bases was revealed through KI quenching and competitive ethidium bromide assays. Docking and Molecular Dynamics suggested that 5-nitro-thiophene-thiosemicarbazone compounds interact against the larger DNA groove, and corroborating the spectroscopic results, may assume an intercalating interaction mode. CONCLUSION: Our findings highlight 5-nitro-thiophene-thiosemicarbazone derivatives, especially LNN-05, as a promising new class of compounds for further studies to provide new anticancer therapies.
Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA, Neoplasm/drug effects , Nitro Compounds/pharmacology , Thiophenes/pharmacology , Thiosemicarbazones/pharmacology , Topoisomerase II Inhibitors/pharmacology , Adult , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Proliferation/drug effects , DNA Topoisomerases, Type II/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Nitro Compounds/chemical synthesis , Nitro Compounds/chemistry , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/chemistry , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , Tumor Cells, CulturedABSTRACT
Cells are daily submitted to high levels of DNA lesions that trigger complex pathways and cellular responses by cell cycle arrest, apoptosis, alterations in transcriptional response, and the onset of DNA repair. Members of the NIMA-related kinase (NEK) family have been related to DNA damage response and repair and the first insight about NEK5 in this context is related to its role in centrosome separation resulting in defects in chromosome integrity. Here we investigate the potential correlation between NEK5 and the DNA damage repair index. The effect of NEK5 in double-strand breaks caused by etoposide was accessed by alkaline comet assay and revealed that NEK5-silenced cells are more sensitive to etoposide treatment. Topoisomerase IIß (TOPIIß) is a target of etoposide that leads to the production of DNA breaks. We demonstrate that NEK5 interacts with TOPIIß, and the dynamics of this interaction is evaluated by proximity ligation assay. The complex NEK5/TOPIIß is formed immediately after etoposide treatment. Taken together, the results of our study reveal that NEK5 depletion increases DNA damage and impairs proper DNA damage response, pointing out NEK5 as a potential kinase contributor to genomic stability.
Subject(s)
DNA Damage/genetics , DNA Repair/genetics , DNA Topoisomerases, Type II/metabolism , Etoposide/pharmacology , NIMA-Related Kinases/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA/drug effects , DNA/genetics , HEK293 Cells , Humans , NIMA-Related Kinases/genetics , RNA Interference , RNA, Small Interfering/geneticsABSTRACT
Due to their unique and versatile biochemical properties, ruthenium-based compounds have emerged as promising anticancer agents. Previous studies showed that three ruthenium(II) compounds: [Ru(pySH)(bipy)(dppb)]PF6 (1), [Ru(HSpym)(bipy)(dppb)]PF6 (2) and Ru[(SpymMe2)(bipy)(dppb)]PF6 (3) presented anticancer properties higher than doxorubicin and cisplatin and acted as human topoisomerase IB (Topo I) inhibitors. Here, we focused our studies on in vitro intestinal permeability and anticancer mechanisms of these three complexes. Caco-2 permeation studies showed that 1 did not permeate the monolayer of intestinal cells, suggesting a lack of absorption on oral administration, while 2 and 3 permeated the cells after 60 and 120 min, respectively. Complexes 2 and 3 fully inhibited Topo II relaxation activity at 125 µM. In previously studies, 3 was the most potent inhibitor of Topo I, here, we concluded that it is a dual topoisomerase inhibitor. Moreover, it presented selectivity to cancer cells when evaluated by clonogenic assay. Thus, 3 was selected to gene expression assay front MDA-MB-231 cells from triple-negative breast cancer (TNBC), which represents the highly aggressive subgroup of breast cancers with poor prognosis. The analyses revealed changes of 27 out of 84 sought target genes. PARP1 and PARP2 were 5.29 and 1.83 times down-regulated after treatment with 3, respectively. PARPs have been attractive antitumor drug targets, considering PARP inhibition could suppress DNA damage repair and sensitize tumor cells to DNA damage agents. Recent advances in DNA repair studies have shown that an approach that causes cell lethality using synthetic PARP-inhibiting drugs has produced promising results in TNBC.
Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Ruthenium/pharmacology , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Caco-2 Cells , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemistry , DNA Topoisomerases, Type II/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Ruthenium/chemistry , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemistryABSTRACT
Nine new spiroacridine derivatives were synthetized by introducing cyano-N-acylhydrazone group between the acridine and phenyl-substituted rings followed by spontaneous cyclization. The new compounds were assayed for their DNA binding properties, human topoisomerase IIα inhibition and bovine serum albumin (BSA) interaction. Besides, docking analysis were performed in order to better understanding the biomolecule-compounds interactions. All compounds interacted with BSA which was demonstrated by the fluorescence suppression constant of 104â¯M-1. Compounds with chloro and NO2 substituents at that para-position on phenyl ring demonstrated the best results for BSA interaction. DNA binding constant determined by UV-vis data demonstrated high values for AMTAC-11 and AMTAC-14, 1.1â¯×â¯108â¯M-1 and 4.8â¯×â¯106â¯M-1, respectively, and all others presented constant values of 105â¯M-1. AMTAC-06 with chloro at para-position on phenyl ring presented a topoisomerase II inhibition of 84.34% in comparison to the positive controls used. Docking studies indicated that AMTAC-06 is able to intercalate the DNA base pairs at topoisomerase IIα active site, preventing DNA connection after break, in a process known as poisoning. Topoisomerase enzyme inhibition result was correlated to BSA interaction profile, since AMTAC-06 showed the best results in both analysis. The findings obtained here proved that methoxy or chloro substitution on phenyl ring at para-position is fundamental for in vitro activity of new spiroacridine derivatives, and indicates that AMTAC-06 is a promising entity and should serve as a lead compound in the development of new DNA and protein binders, as well as human topoisomerase II inhibitors.
Subject(s)
Acridines/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA/chemistry , Serum Albumin, Bovine/chemistry , Topoisomerase II Inhibitors/pharmacology , Acridines/chemical synthesis , Acridines/chemistry , Animals , Cattle , Dose-Response Relationship, Drug , Fluorescence , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistryABSTRACT
Naphthoquinones and 1,2,3-triazoles are structural pharmacophore that is known to impart several cancer cells. This work shows a synthetic methodology to obtain hybrid molecules involving naphthoquinone and triazol scaffold as multiple ligands. A simple and efficient synthetic route was used to prepare a series of sixteen compounds being eight 2-(1-aryl-1H-1,2,3-triazol-4-yl)-2,3-dihydronaphtho[1,2â¯b]furan-4,5-diones and eight 2-(1-aryl-1H-1,2,3-triazol-4-yl)-2,3-dihydronaphtho[2,3-b]furan-4,9-diones. These compounds were tested in MDA-MB231, Caco-2 and Calu-3 human cancer cells, and among them 7a was the most selective compound on Caco-2â¯cells, the most sensitized cell line in this study. In silico study suggest that the blockage of topoisomerase I and IIα may be one of the mechanisms of action responsible for the cytotoxic effect of 7a in Caco-2â¯cells.
Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Triazoles/chemistry , Triazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Caco-2 Cells , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , Humans , Models, Molecular , Naphthoquinones/chemical synthesis , Neoplasms/drug therapy , Neoplasms/metabolism , Structure-Activity Relationship , Topoisomerase I Inhibitors/chemical synthesis , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology , Triazoles/chemical synthesisABSTRACT
Anthracyclines, e.g., doxorubicin (DOX), and anthracenediones, e.g., mitoxantrone (MTX), are drugs used in the chemotherapy of several cancer types, including solid and non-solid malignancies such as breast cancer, leukemia, lymphomas, and sarcomas. Although they are effective in tumor therapy, treatment with these two drugs may lead to side effects such as arrhythmia and heart failure. At the same clinically equivalent dose, MTX causes slightly reduced cardiotoxicity compared with DOX. These drugs interact with iron to generate reactive oxygen species (ROS), target topoisomerase 2 (Top2), and impair mitochondria. These are some of the mechanisms through which these drugs induce late cardiomyopathy. In this review, we compare the cardiotoxicities of these two chemotherapeutic drugs, DOX and MTX. As described here, even though they share similarities in their modes of toxicant action, DOX and MTX seem to differ in a key aspect. DOX is a more redox-interfering drug, while MTX induces energy imbalance. In addition, DOX toxicity can be explained by underlying mechanisms that include targeting of Top2 beta, mitochondrial impairment, and increases in ROS generation. These modes of action have not yet been demonstrated for MTX, and this knowledge gap needs to be filled.
Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Heart Diseases/chemically induced , Mitoxantrone/toxicity , Myocytes, Cardiac/drug effects , Signal Transduction/drug effects , Animals , Antigens, Neoplasm/metabolism , Cardiotoxicity , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/prevention & control , Humans , Iron/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidation-Reduction , Oxidative Stress/drug effects , Poly-ADP-Ribose Binding Proteins , Reactive Oxygen Species/metabolism , Topoisomerase II Inhibitors/pharmacologyABSTRACT
Mitoxantrone (MXT) is an anticancer drug structurally related to anthracyclines, such as doxorubicin (DOX). Here we report that cells deficient in nucleotide excision repair (NER) are very sensitive to MXT. However, cells deficient in each of the NER sub-pathways - transcription coupled repair (deficient in CSB protein) and global genome repair (deficient in XPC protein) - demonstrate a difference in sensitivity from each other and also show different responses in cell cycle profile, DNA synthesis and topo II DNA complex formation upon MXT treatment. XPC-deficient cells are slightly more resistant than CSB-deficient cells, and in the same way as MRC5 NER-proficient cells, show G2/M arrest, normal DNA synthesis rate and a pattern of formation of complexes similar to proficient cells, whereas CSB-deficient cells show accumulation in S phase, reduced DNA synthesis and a more intense signal of topo II DNA complexes, indicating that they remain longer in these cells. Complementation of CSB mutant cells with CSB rescue MXT-induced sensitivity and also a decrease in the signal intensity of the complexes, suggest that resolution of these lesions would take place. Taken together, our results indicate that NER proteins are implicated in the response to MXT and that CSB protein has a key role in processing MXT-induced topo II DNA complexes.
Subject(s)
Antineoplastic Agents/pharmacology , DNA Repair/drug effects , Mitoxantrone/pharmacology , Topoisomerase II Inhibitors/pharmacology , DNA/biosynthesis , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Damage , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA Topoisomerases, Type II/metabolism , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Models, Molecular , Nucleic Acid Conformation , Poly-ADP-Ribose Binding ProteinsABSTRACT
In cancer treatment, chemotherapy is one of the main strategies used. The knowledge of the cellular and molecular characteristics of tumors allows the use of more specific drugs, making the removal of tumors more efficient. Among the drugs of choice in these treatments, topoisomerase inhibitors are widely used against different types of tumors. Topoisomerases are enzymes responsible for maintaining the structure of DNA, altering its topological state temporarily during the processes of replication and transcription, in order to avoid supercoiling and entanglements at the double helix. The DNA damage formed as a result of topoisomerase inhibition can be repaired by DNA repair mechanisms. Thus, DNA repair pathways can modulate the effectiveness of chemotherapy. Homologous recombination (HR) and non-homologous end joining (NHEJ) are the main pathways involved in the removal of double strand breaks (DSBs); while nucleotide excision repair (NER) is mainly characterized by the removal of lesions that lead to significant structural distortions in the DNA double helix. Evidence has shown that DSBs are the main type of damage resulting from the inhibition of the DNA topoisomerase II enzyme, and therefore the involvement of HR and NHEJ pathways in the repair process is well established. However, some topoisomerase II inhibitors induce other types of lesions, like DNA adducts, interstrand crosslinks and reactive oxygen species, and studies have shown that other DNA repair pathways might be participating in removing injury induced by these drugs. This review aims to correlate the involvement of proteins from different DNA repair pathways in response to these drugs, with an emphasis on NER.