Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Publication year range
1.
Nucleic Acids Res ; 49(3): 1581-1596, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33434265

ABSTRACT

DNA gyrase, a type II topoisomerase found predominantly in bacteria, is the target for a variety of 'poisons', namely natural product toxins (e.g. albicidin, microcin B17) and clinically important synthetic molecules (e.g. fluoroquinolones). Resistance to both groups can be mediated by pentapeptide repeat proteins (PRPs). Despite long-term studies, the mechanism of action of these protective PRPs is not known. We show that a PRP, QnrB1 provides specific protection against fluoroquinolones, which strictly requires ATP hydrolysis by gyrase. QnrB1 binds to the GyrB protein and stimulates ATPase activity of the isolated N-terminal ATPase domain of GyrB (GyrB43). We probed the QnrB1 binding site using site-specific incorporation of a photoreactive amino acid and mapped the crosslinks to the GyrB43 protein. We propose a model in which QnrB1 binding allosterically promotes dissociation of the fluoroquinolone molecule from the cleavage complex.


Subject(s)
Bacterial Proteins/metabolism , DNA Gyrase/metabolism , Topoisomerase II Inhibitors/toxicity , Adenosine Triphosphate/metabolism , Bacteriocins/toxicity , Ciprofloxacin/toxicity , DNA/metabolism , Escherichia coli/enzymology , Hydrolysis , Organic Chemicals/toxicity , Xanthomonas
2.
Biochem Biophys Res Commun ; 561: 7-13, 2021 07 05.
Article in English | MEDLINE | ID: mdl-33992835

ABSTRACT

Doxorubicin (DOX) is a broad-spectrum antineoplastic drug; however, its serious cardiotoxic side effects in inflammatory responses limit its use in clinical applications. Dopamine D1 receptor (DRD1), a G protein-coupled receptor, is crucial for the development and function of the nervous system; additionally, it also play a role in immune regulation. However, the specific role of DRD1 in DOX-induced cardiac inflammation has not yet been clarified. Here, we discovered that DRD1 expression was induced by DOX treatment in H9C2 cardiomyocytes. DRD1 activation by A-68930, a DRD1-specific agonist, decreased DOX-induced nucleotide-binding domain-like receptor protein 3 (NLRP3) expression, caspase-1 activation, and IL-1ß maturation in H9C2 cells. Expression of the cytokines IL-1ß and IL-18 in the supernatants was also inhibited by A-68930 treatment. DRD1 knockdown, using siRNA, abolished the effects of A-68930 on the DOX-induced NLRP3 inflammasome. Furthermore, we found that DRD1 signaling downregulated the NLRP3 inflammasome in H9C2 cells through cyclic adenosine monophosphate (cAMP). Moreover, application of A-68930 to activate DRD1 reduced cardiac injury and fibrosis in a DOX-treated mouse model by suppressing the NLRP3 inflammasome in the heart. These findings indicate that DRD1 signaling may protect against DOX-induced cardiac injury by inhibiting the NLRP3 inflammasome-mediated inflammation.


Subject(s)
Cardiotoxicity/prevention & control , Chromans/pharmacology , Doxorubicin/toxicity , Inflammasomes/antagonists & inhibitors , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Receptors, Dopamine D1/agonists , Animals , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Cells, Cultured , Cytokines/metabolism , Dopamine Agonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Receptors, Dopamine D1/metabolism , Signal Transduction , Topoisomerase II Inhibitors/toxicity
3.
Bioorg Med Chem Lett ; 49: 128274, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34303812

ABSTRACT

Two series of (hetero)arylamino-naphthoquinones and benzo-fused carbazolequinones were considered for study with the rationale that related structural motifs are present in numerous drugs, clinical trial agents, natural products and hTopoIIα inhibitors. Total 42 compounds were synthesized by reactions including dehydrogenative CN and Pd-catalyzed CC bond forming transformations. These compounds were screened against numerous cancer cells including highly metastatic one (MCF-7, MDA-MB-231, H-357 and HEK293T), and normal cells (MCF 10A). Some of the active compounds were evaluated for clonogenic cell survival and apoptotic effects in cancer cells (DAPI nuclear staining, Comet assay, Annexin-V-FITC/PI dual staining, flow cytometry, and western blot analysis with relevant proteins). All compounds were tested for hTopoIIα inhibitory activity. The investigated series compounds showed important properties like significant apoptotic antiproliferation in cancer cells with cell cycle arrest at S-phase and downregulation of NF- κß signaling cascade, relatively less cytotoxicity to normal cells, and hTopoIIα inhibition with more efficiency compared to an anticancer drug etoposide.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , DNA Topoisomerases, Type II/metabolism , Naphthoquinones/pharmacology , Poly-ADP-Ribose Binding Proteins/metabolism , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Carbazoles/chemical synthesis , Carbazoles/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Naphthoquinones/chemical synthesis , Naphthoquinones/toxicity , S Phase Cell Cycle Checkpoints/drug effects , Signal Transduction/drug effects , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/toxicity
4.
Bioorg Chem ; 114: 105042, 2021 09.
Article in English | MEDLINE | ID: mdl-34120024

ABSTRACT

S. aureus resistant to methicillin (MRSA) is one of the most-concerned multidrug resistant bacteria, due to its role in life-threatening infections. There is an urgent need to develop new antibiotics against MRSA. In this study, we firstly compiled a data set of 2,3-diaminoquinoxalines by chemical synthesis and antibacterial screening against S. aureus, and then performed cheminformatics modeling and virtual screening. The compound with the Specs ID of AG-205/33156020 was discovered as a new antibacterial agent, and was further identified as a Gyrase B (GyrB) inhibitor. In light of the common features, we hypothesized that the 6c as the representative of 2,3-diaminoquinoxalines also inhibited GyrB and eventually proved it. Via molecular docking and molecular dynamics simulations, we identified binding modes of AG-205/33156020 and 6c to the ATPase domain of GyrB. Importantly, these GyrB inhibitors inhibited the MRSA strains and showed selectivity to HepG2 and HUVEC. Taken together, this research work provides an effective ligand-based computational workflow for scaffold hopping in anti-MRSA drug discovery, and discovers two new GyrB inhibitors that are worthy of further development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Quinoxalines/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/toxicity , DNA Gyrase/metabolism , Drug Evaluation, Preclinical , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Ligands , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Quinoxalines/chemical synthesis , Quinoxalines/metabolism , Quinoxalines/toxicity , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/metabolism , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/toxicity
5.
Arch Toxicol ; 95(12): 3803-3813, 2021 12.
Article in English | MEDLINE | ID: mdl-34609522

ABSTRACT

The comet assay is widely used in basic research, genotoxicity testing, and human biomonitoring. However, interpretation of the comet assay data might benefit from a better understanding of the future fate of a cell with DNA damage. DNA damage is in principle repairable, or if extensive, can lead to cell death. Here, we have correlated the maximally induced DNA damage with three test substances in TK6 cells with the survival of the cells. For this, we selected hydrogen peroxide (H2O2) as an oxidizing agent, methyl methanesulfonate (MMS) as an alkylating agent and etoposide as a topoisomerase II inhibitor. We measured cell viability, cell proliferation, apoptosis, and micronucleus frequency on the following day, in the same cell culture, which had been analyzed in the comet assay. After treatment, a concentration dependent increase in DNA damage and in the percentage of non-vital and apoptotic cells was found for each substance. Values greater than 20-30% DNA in tail caused the death of more than 50% of the cells, with etoposide causing slightly more cell death than H2O2 or MMS. Despite that, cells seemed to repair of at least some DNA damage within few hours after substance removal. Overall, the reduction of DNA damage over time is due to both DNA repair and death of heavily damaged cells. We recommend that in experiments with induction of DNA damage of more than 20% DNA in tail, survival data for the cells are provided.


Subject(s)
Apoptosis/drug effects , Cell Survival/drug effects , Comet Assay , DNA Damage/drug effects , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/toxicity , Cell Line , Cell Proliferation/drug effects , DNA Repair/drug effects , Dose-Response Relationship, Drug , Etoposide/administration & dosage , Etoposide/toxicity , Humans , Hydrogen Peroxide/administration & dosage , Hydrogen Peroxide/toxicity , Methyl Methanesulfonate/administration & dosage , Methyl Methanesulfonate/toxicity , Oxidants/administration & dosage , Oxidants/toxicity , Time Factors , Topoisomerase II Inhibitors/administration & dosage , Topoisomerase II Inhibitors/toxicity
6.
J Appl Toxicol ; 41(1): 175-189, 2021 01.
Article in English | MEDLINE | ID: mdl-32969520

ABSTRACT

Heterocycles have long been the focus of intensive study in attempts to develop novel therapeutic compounds, and acridine, a polynuclear nitrogen molecule containing a heterocycle, has attracted a considerable amount of scientific attention. Acridine derivatives have been studied in detail and have been found to possess multitarget properties, which inhibit topoisomerase enzymes that regulate topological changes in DNA and interfere with the essential biological function of DNA. This article describes some recent advancements in the field of new 9-substituted acridine heterocyclic agents and describes both the structure and the structure-activity relationship of the most promising molecules. The article will also present the IC50 values of the novel derivatives against various human cancer cell lines. The mini review also investigates the topoisomerase inhibition and antibacterial and antimalarial activity of these polycyclic aromatic derivatives.


Subject(s)
Acridines/chemistry , Acridines/pharmacology , Acridines/toxicity , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/toxicity , Tumor Cells, Cultured/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Antimalarials/chemistry , Antimalarials/pharmacology , Antimalarials/toxicity , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Humans , Structure-Activity Relationship
7.
Int Heart J ; 62(5): 1112-1123, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34544967

ABSTRACT

Doxorubicin (DOX) is a widely used anticancer drug, but its cardiotoxicity largely limits its clinical utilization. Circular RNA spindle and kinetochore-associated protein 3 (circ-SKA3) were found to be differentially expressed in heart failure patients. In this study, we investigated the role and mechanism of circ-SKA3 in DOX-induced cardiotoxicity.The quantitative real-time polymerase chain reaction and western blot assays were applied to measure the expression of circ-SKA3, microRNA (miR) -1303, and toll-like receptor 4 (TLR4). The viability and apoptosis of AC16 cells were analyzed using cell counting kit-8, flow cytometry, and western blot assays. The interaction between miR-1303 and circ-SKA3 or TLR4 was verified using dual-luciferase reporter and RNA immunoprecipitation assays. Exosomes were collected from culture media by the use of commercial kits and then qualified by transmission electron microscopy.The expression of circ-SKA3 and TLR4 was increased, whereas miR-1303 expression was decreased in DOX-treated AC16 cells. DOX treatment promoted cell apoptosis and inhibited cell viability in AC16 cells in vitro, which was partially reversed by circ-SKA3 knockdown, TLR4 silencing, or miR-1303 overexpression. Mechanistically, circ-SKA3 served as a sponge for miR-1303 to upregulate TLR4, which was confirmed to be a target of miR-1303. Additionally, circ-SKA3 contributed to DOX-induced cardiotoxicity through the miR-1303/TLR4 axis. Further studies suggested that circ-SKA3 was overexpressed in exosomes extracted from DOX-mediated AC16 cells, which could be internalized by surrounding untreated AC16 cells.Circ-SKA3 enhanced DOX-induced toxicity in AC16 cells through the miR-1303/TLR4 axis. Extracellular circ-SKA3 was packaged into exosomes, and exosomal circ-SKA3 could function as a mediator in intercellular communication between AC16 cells.


Subject(s)
Cell Cycle Proteins/genetics , Doxorubicin/toxicity , Microtubule-Associated Proteins/genetics , Myocytes, Cardiac/drug effects , Topoisomerase II Inhibitors/toxicity , Apoptosis/drug effects , Cardiotoxicity/genetics , Cell Cycle Proteins/drug effects , Cell Survival/drug effects , Exosomes/genetics , Heart Failure/genetics , Humans , MicroRNAs/genetics , Microscopy, Electron, Transmission/methods , Microtubule-Associated Proteins/drug effects , Myocytes, Cardiac/pathology , RNA, Circular/genetics , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/genetics , Transfection/methods , Up-Regulation
8.
Mol Cell Biochem ; 475(1-2): 119-126, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32754875

ABSTRACT

Doxorubicin (Dox) is a widely neoplasm chemotherapeutic drug with high incidences of cardiotoxicity. Prodigiosin (PG), a red bacterial pigment from Serratia marcescens, has been demonstrated to potentiate Dox's cytotoxicity against oral squamous cell carcinoma cells through elevating Dox influx and identified as a Dox enhancer via PG-induced autophagy; however, toxicity of normal cell remains unclear. This study is conducted to evaluate putative cytotoxicity features of PG/Dox synergism in the liver, kidney, and heart cells and further elucidate whether PG augmented Dox's effect via modulating Dox metabolism in normal cells. Murine hepatocytes FL83B, cardio-myoblast h9c2, and human kidney epithelial cells HK-2 were sequentially treated with PG and Dox by measuring cell viability, cell death characteristics, oxidative stress, Dox flux, and Dox metabolism. PG could slightly significant increase Dox cytotoxicity in all tested normal cells whose toxic alteration was less than that of oral squamous carcinoma cells. The augmentation of Dox cytotoxicity might be attributed to the increase of Dox-mediated ROS accumulation that might cause slight reduction of Dox influx and reduction of Dox metabolism. It was noteworthy to notice that sustained cytotoxicity appeared in normal cells after PG and Dox were removed. Taken together, moderately metabolic reduction of Dox might be ascribed to the mechanism of increase Dox cytotoxicity in PG-induced normal cells; nevertheless, the determination of PG/Dox dose with sustained cytotoxicity in normal cells needs to be comprehensively considered.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Doxorubicin/pharmacology , Neoplasms/drug therapy , Prodigiosin/pharmacology , Animals , Anti-Bacterial Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cell Line , Cell Survival/drug effects , Doxorubicin/administration & dosage , Drug Synergism , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Prodigiosin/adverse effects , Topoisomerase II Inhibitors/metabolism , Topoisomerase II Inhibitors/toxicity
9.
Nutr Cancer ; 72(2): 252-259, 2020.
Article in English | MEDLINE | ID: mdl-31184509

ABSTRACT

The purpose of this study was to investigate the effects of in vivo creatine monohydrate (Cr) supplementation on doxorubicin (Dox)-induced muscle dysfunction. Male rats were fed a diet supplemented with 3% Cr or a standard chow for 2 wk. After 2 wk of feeding, animals received Dox or saline as a placebo. Five days post-injection, grip strength was measured, and muscle fatigue was analyzed ex vivo. When compared with controls, a significantly lower grip strength was observed with Dox treatment, but no significant handgrip difference was observed with Cr feeding prior to Dox treatment when compared to controls. In the isolated muscle fatigue experiments, solei (primarily type I muscle) from controls produced significantly less force than baseline at 60 s and solei from Dox treated rats produced significantly less force than baseline at 30 s; however, Cr feeding prior to Dox produced significantly less force than baseline at 60 s. In the primarily type II EDL, a decline in force production from baseline was observed at 50 s in controls and Cr + Dox and at 20 s in standard chow + Dox. Cr attenuated the increase in fatigue that accompanies Dox treatment suggesting that Cr supplementation may have use in managing Dox myotoxicity.


Subject(s)
Creatine/pharmacology , Dietary Supplements , Doxorubicin/toxicity , Hand Strength/physiology , Muscle Fatigue/drug effects , Muscle Strength/physiology , Animals , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Topoisomerase II Inhibitors/toxicity
10.
Chem Res Toxicol ; 32(10): 2135-2143, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31512855

ABSTRACT

Topoisomerase II is a nuclear enzyme involved in the maintenance of DNA and is an effective anticancer drug target. However, several clinical topoisomerase II-targeted agents display significant off-target toxicities and adverse events. Thus, it is important to continue characterizing compounds with activity against topoisomerase II. We previously analyzed α-(N)-heterocyclic thiosemicarbazone copper(II) complexes against human topoisomerase IIα (TOP2A), but humans also express topoisomerase IIß (TOP2B), which has distinct functional roles. Therefore, we examined two α-(N)-heterocyclic thiosemicarbazone copper [Cu(II)] complexes for activity against TOP2B in a purified system. The Cu(II) complexes, Cu(APY-ETSC)Cl and Cu(BZP-ETSC)Cl, were examined using plasmid DNA cleavage, supercoiled DNA relaxation, enzyme inactivation, protein cross-linking, DNA ligation, and ATP hydrolysis assays with TOP2B to determine whether these compounds act similarly against both enzymes. Both of the Cu(II) thiosemicarbazone (Cu-TSC) complexes we tested disrupted the function of TOP2B in a way similar to the effect on TOP2A. In particular, TOP2B DNA cleavage activity is increased in the presence of these compounds, while the relaxation and ATPase activities are inhibited. Further, both Cu-TSCs stabilize the N-terminal DNA clamp of TOP2A and TOP2B and rapidly inactivate TOP2B when the compounds are present before DNA. Our data provide evidence that the Cu-TSC complexes we tested utilize a similar mechanism against both isoforms of the enzyme. This mechanism may involve interaction with the ATPase domain of TOP2A and TOP2B outside of the ATP binding pocket. Additionally, these data support a model of TOP2 function where the ATPase domain communicates with the DNA cleavage/ligation domain.


Subject(s)
Organometallic Compounds/pharmacology , Organometallic Compounds/toxicity , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/toxicity , Copper/chemistry , Copper/pharmacology , DNA Cleavage/drug effects , DNA Topoisomerases, Type II/metabolism , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Humans , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry
11.
Curr Microbiol ; 76(3): 312-319, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30603963

ABSTRACT

The ability of fermentates of two potential probiotic strains, Bacillus amyloliquefaciens B-1895 and Bacillus subtilis KATMIRA1933, to lower the SOS response in bacteria was evaluated using Escherichia coli-based Lux biosensors (pRecA-lux) and the tested bacilli fermentates obtained through solid-state fermentation. The SOS response was stimulated by the addition of ciprofloxacine. Preparations of both Bacillus fermentates demonstrated SOS-inhibitory activity (up to 54.21%). The strain КATMIRA1933 was characterized by higher SOS-inhibitory activity. The active components of the fermentates were stable against heating, proteinase, and RNase action.


Subject(s)
Antimutagenic Agents/pharmacology , Bacillus amyloliquefaciens/metabolism , Bacillus subtilis/metabolism , Probiotics/pharmacology , SOS Response, Genetics/drug effects , Antimutagenic Agents/metabolism , Bacillus/metabolism , Biosensing Techniques , Ciprofloxacin/toxicity , Escherichia coli/drug effects , Escherichia coli/genetics , Fermentation , Probiotics/metabolism , Topoisomerase II Inhibitors/toxicity
12.
Int J Cancer ; 140(4): 864-876, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27813122

ABSTRACT

Quercetin (Que) is an abundant flavonoid in the human diet and high-concentration food supplement with reported pro- and anti-carcinogenic activities. Topoisomerase II (TopoII) inhibition and subsequent DNA damage induction by Que was implicated in the mixed lineage leukemia gene (MLL) rearrangements that can induce infant and adult leukemias. This notion raised concerns regarding possible genotoxicities of Que in hematopoietic stem and progenitor cells (HSPCs). However, molecular targets mediating Que effects on DNA repair relevant to MLL translocations have not been defined. In this study we describe novel and potentially genotoxic Que activities in suppressing non-homologous end joining and homologous recombination pathways downstream of MLL cleavage. Using pharmacological dissection of DNA-PK, ATM and PI3K signalling we defined PI3K inhibition by Que with a concomitant decrease in the abundance of key DNA repair genes to be responsible for DNA repair inhibition. Evidence for the downstream TopoII-independent mutagenic potential of Que was obtained by documenting further increased frequencies of MLL rearrangements in human HSPCs concomitantly treated with Etoposide and Que versus single treatments. Importantly, by engaging a tissue engineered placental barrier, we have established the extent of Que transplacental transfer and hence provided the evidence for Que reaching fetal HSPCs. Thus, Que exhibits genotoxic effects in human HSPCs via different mechanisms when applied continuously and at high concentrations. In light of the demonstrated Que transfer to the fetal compartment our findings are key to understanding the mechanisms underlying infant leukemia and provide molecular markers for the development of safety values.


Subject(s)
Cell Transformation, Neoplastic/drug effects , DNA Damage , DNA Repair/drug effects , DNA Topoisomerases, Type II/physiology , Hematopoietic Stem Cells/drug effects , Histone-Lysine N-Methyltransferase/genetics , Leukemia/chemically induced , Myeloid-Lymphoid Leukemia Protein/genetics , Phosphoinositide-3 Kinase Inhibitors , Quercetin/toxicity , Signal Transduction/drug effects , Topoisomerase II Inhibitors/toxicity , Adult , Ascorbic Acid/pharmacology , Cell Culture Techniques , Cells, Cultured , Colony-Forming Units Assay , Dose-Response Relationship, Drug , Etoposide/pharmacology , Female , Genistein/pharmacology , Histones/analysis , Humans , Infant , Leukemia/genetics , Maternal-Fetal Exchange , Phosphatidylinositol 3-Kinases/physiology , Pregnancy
13.
Genes Cells ; 21(3): 226-40, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26891792

ABSTRACT

Doxorubicin is a widely used chemotherapeutic agent, but its utility is limited by cellular resistance and off-target effects. To understand the molecular mechanisms regulating chemotherapeutic responses to doxorubicin, we previously carried out a genomewide search of doxorubicin-resistance genes in Schizosaccharomyces pombe fission yeast and showed that these genes are organized into networks that counteract doxorubicin cytotoxicity. Here, we describe the identification of a subgroup of doxorubicin-resistance genes that, when disrupted, leads to reduced tolerance to exogenous calcium. Unexpectedly, we observed a suppressive effect of calcium on doxorubicin cytotoxicity, where concurrent calcium and doxorubicin treatment resulted in significantly higher cell survival compared with cells treated with doxorubicin alone. Conversely, inhibitors of voltage-gated calcium channels enhanced doxorubicin cytotoxicity in the mutants. Consistent with these observations in fission yeast, calcium also suppressed doxorubicin cytotoxicity in human breast cancer cells. Further epistasis analyses in yeast showed that this suppression of doxorubicin toxicity by calcium was synergistically dependent on Rav1 and Vph2, two regulators of vacuolar-ATPase assembly; this suggests potential modulation of the calcium-doxorubicin interaction by fluctuating proton concentrations within the cellular environment. Thus, the modulatory effects of drugs or diet on calcium concentrations should be considered in doxorubicin treatment regimes.


Subject(s)
Calcium/pharmacology , Doxorubicin/toxicity , Topoisomerase II Inhibitors/toxicity , Calcium Channel Blockers/pharmacology , Cell Survival , Drug Resistance, Fungal/genetics , Genes, Fungal , Humans , MCF-7 Cells , Schizosaccharomyces/drug effects , Schizosaccharomyces/metabolism
14.
Arch Toxicol ; 91(8): 2763-2780, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28536863

ABSTRACT

Infant leukaemia (<1 year old) is a rare disease of an in utero origin at an early phase of foetal development. Rearrangements of the mixed-lineage leukaemia (MLL) gene producing abnormal fusion proteins are the most frequent genetic/molecular findings in infant B cell-acute lymphoblastic leukaemia. In small epidemiological studies, mother/foetus exposures to some chemicals including pesticides have been associated with infant leukaemia; however, the strength of evidence and power of these studies are weak at best. Experimental in vitro or in vivo models do not sufficiently recapitulate the human disease and regulatory toxicology studies are unlikely to capture this kind of hazard. Here, we develop an adverse outcome pathway (AOP) based substantially on an analogous disease-secondary acute leukaemia caused by the topoisomerase II (topo II) poison etoposide-and on cellular and animal models. The hallmark of the AOP is the formation of MLL gene rearrangements via topo II poisoning, leading to fusion genes and ultimately acute leukaemia by global (epi)genetic dysregulation. The AOP condenses molecular, pathological, regulatory and clinical knowledge in a pragmatic, transparent and weight of evidence-based framework. This facilitates the interpretation and integration of epidemiological studies in the process of risk assessment by defining the biologically plausible causative mechanism(s). The AOP identified important gaps in the knowledge relevant to aetiology and risk assessment, including the specific embryonic target cell during the short and spatially restricted period of susceptibility, and the role of (epi)genetic features modifying the initiation and progression of the disease. Furthermore, the suggested AOP informs on a potential Integrated Approach to Testing and Assessment to address the risk caused by environmental chemicals in the future.


Subject(s)
Adverse Outcome Pathways , Pesticides/toxicity , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Animals , Environmental Exposure , Etoposide/toxicity , Gene Rearrangement , Histone-Lysine N-Methyltransferase/genetics , Humans , Infant , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Risk Assessment/methods , Topoisomerase II Inhibitors/toxicity
15.
Environ Toxicol ; 32(6): 1836-1843, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28206703

ABSTRACT

Etoposide, a topoisomerase II inhibitor, has been widely used as a clinical anticancer drug to treat diverse cancer patients. Since not only rapidly dividing cancer cells but also the cells of normal human tissues and every living organism in environmental ecosystems have topoisomerases, it is crucial to study the toxicity of etoposide in other organisms in addition to cancer cells. In this study, we evaluated the toxicity of etoposide in both a soil nematode, Caenorhabditis elegans, and 3T3-L1 normal murine cells. Etoposide significantly retarded the growth, egg laying, and hatching in C. elegans. Etoposide also affected the reproductive gonad tissue, decreased the number of germ cells and induced abnormally enlarged nuclei in C. elegans. In addition, etoposide inhibited 3T3-L1 cell proliferation, with IC50 values of 37.8 ± 7.3 and 9.8 ± 1.8 µM after 24 and 48 hours of treatment, respectively, via the induction of cell cycle arrest at the G2/M phase and apoptotic cell death. Etoposide also induced nuclear enlargement in 3T3-L1 normal murine cells. The reproductive toxicity and abnormal nuclear morphological changes seemed to correlate with the adverse effects of etoposide. We suggest that these experimental platforms, i.e., the toxicological evaluation of both nematodes and 3T3-L1 cells, may be useful to study the mechanisms underlying the side effects of chemicals, including topoisomerase inhibitors.


Subject(s)
Antineoplastic Agents/toxicity , Caenorhabditis elegans/drug effects , Cell Nucleus/drug effects , Etoposide/toxicity , Topoisomerase II Inhibitors/toxicity , 3T3-L1 Cells , Animals , Apoptosis/drug effects , Cell Culture Techniques , Cell Cycle Checkpoints/drug effects , Cell Nucleus/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Mice , Microscopy, Fluorescence , Reproduction/drug effects
16.
Bioorg Med Chem Lett ; 26(9): 2159-63, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27036521

ABSTRACT

A simple and efficient method for the selective synthesis of 2-pyrdones from 4H-pyrans using iodine as catalyst and ethanol as solvent was developed. The present method is equally effective for both aromatic and hetero aromatic ring containing 4H-pyrans. The compatibility with various functional groups, mild reaction conditions, high yields and application of inexpensive, readily and easily available iodine as catalyst and formation of 2-pyridones as major products are the advantages of the present procedure. In vitro antiproliferative activity of the final synthesized compounds was evaluated with four different human cancer cell lines (Lung adenocarcinoma-A549, Hepatocarcinoma-HepG2, Breast carcinoma-MCF-7 and Ovarian carcinoma-SKOV3) and normal human lung fibroblast cell line (MRC-5). Compounds 2b showed better inhibition against MCF-7, HepG2 and A549 cell lines (IC50 8.00 ± 0.11, 11.93 ± 0.01 and 15.85 ± 0.04 µM, respectively) as compared with doxorubicin and also 2e showed moderate inhibition against MCF-7, HepG2 (IC50 9.32 ± 0.21 and 20.22 ± 0.01 µM, respectively, cell lines, respectively) as compared with doxorubicin. As many clinically used antiproliferative agents induce apoptosis in cancer cells hence, the 2-pyridone analogues were also tested for their ability to induce apoptosis in MCF-7 cells using the caspases-3 and -9 assays.


Subject(s)
Antineoplastic Agents/chemical synthesis , Iodine/chemistry , Pyridones/chemical synthesis , Topoisomerase II Inhibitors/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Catalysis , Cell Line, Tumor , DNA Gyrase/metabolism , Drug Screening Assays, Antitumor , Humans , Pyridones/pharmacology , Pyridones/toxicity , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/toxicity
17.
Bioorg Med Chem ; 24(1): 42-52, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26678175

ABSTRACT

Recently numerous non-fluoroquinolone-based bacterial type II topoisomerase inhibitors from both the GyrA and GyrB classes have been reported as antibacterial agents. Inhibitors of the GyrA class include aminopiperidine-based novel bacterial type II topoisomerase inhibitors (NBTIs). However, inhibition of the cardiac ion channel remains a serious liability for the aminopiperidine based NBTIs. In this paper we replaced central aminopiperidine linker with piperazine moiety and tested for its biological activity. We developed a series of twenty four compounds with a piperazine linker 1-(2-(piperazin-1-yl)ethyl)-1,5-naphthyridin-2(1H)-one, by following a multistep protocol. Among them compound 4-(2-(7-methoxy-2-oxo-1,5-naphthyridin-1(2H)-yl)ethyl)-N-(4-nitrophenyl)piperazine-1-carboxamide (11) was the most promising inhibitor with Mycobacterium tuberculosis (MTB) DNA gyrase enzyme supercoiling IC50 of 0.29±0.22µM, with a good MTB MIC of 3.45µM. These kind of compounds retains good potency and showed reduced cardiotoxicity compared to aminopiperidines.


Subject(s)
Antitubercular Agents/pharmacology , Cardiotoxicity/drug therapy , Mycobacterium tuberculosis/enzymology , Naphthyridines/pharmacology , Piperazines/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/toxicity , Atrioventricular Block/drug therapy , DNA Gyrase/metabolism , Enzyme Assays , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Heart Rate/drug effects , Naphthyridines/chemical synthesis , Naphthyridines/toxicity , Novobiocin/pharmacology , Piperazines/chemical synthesis , Piperazines/toxicity , Terfenadine/pharmacology , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/toxicity , Zebrafish , Zebrafish Proteins/antagonists & inhibitors
18.
Biol Pharm Bull ; 39(4): 631-5, 2016.
Article in English | MEDLINE | ID: mdl-26754253

ABSTRACT

Derris scandens (ROXB.) BENTH. (Fabaceae) is used as an alternative treatment for cancer in Thai traditional medicine. Investigation of the topoisomerase II (Top2) poison of compounds isolated from this plant may reveal new drug leads for the treatment of cancer. Bioassay-guided isolation was performed on an extract of D. scandens stems using a yeast cell-based assay. A yeast strain expressing the top2-1 temperature-sensitive mutant was used to assay Top2 activity. At the permissive temperature of 25°C, yeast cells were highly sensitive to Top2 poison agents. At the semi-permissive temperature of 30°C, where enzyme activity was present but greatly diminished, cells displayed only marginal sensitivity. The bioassay-guided fractionation of the extract led to the isolation of two known isoflavones: 5,7,4'-trihydroxy-6,8-diprenylisoflavone (1) and lupalbigenin (2). These two compounds also displayed cytotoxicity against three different cancer cell lines, KB, MCF-7 and NCI-H187. In conclusion, Top2 poison agents from D. scandens are reported for the first time, substantiating the use of D. scandens in Thai traditional medicine for cancer treatment.


Subject(s)
Antineoplastic Agents , Derris , Isoflavones , Topoisomerase II Inhibitors , Animals , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/toxicity , Biological Assay , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Humans , Isoflavones/isolation & purification , Isoflavones/toxicity , Plant Stems , Saccharomyces cerevisiae/genetics , Topoisomerase II Inhibitors/isolation & purification , Topoisomerase II Inhibitors/toxicity , Vero Cells
19.
Bioorg Med Chem Lett ; 25(22): 5067-71, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26483137

ABSTRACT

The first total synthesis of a C5-curcumin-2-hexadecynoic acid (C5-Curc-2-HDA, 6) conjugate was successfully performed. Through a three-step synthetic route, conjugate 6 was obtained in 13% overall yield and tested for antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA) strains. Our results revealed that 6 was active against eight MRSA strains at MICs that range between 31.3 and 62.5 µg/mL. It was found that the presence of 2-hexadecynoic acid (2-HDA, 4) in conjugate 6 increased 4-8-fold its antibacterial activity against MRSA strains supporting our hypothesis that the chemical connection of 4 to C5-curcumin (2) increases the antibacterial activity of 2 against Gram-positive bacteria. Combinational index (CIn) values that range between 1.6 and 2.3 were obtained when eight MRSA strains were treated with an equimolar mixture of 2 and 4. These results demonstrated that an antagonistic effect is taking place. Finally, it was investigated whether conjugate 6 can affect the replication process of S. aureus, since this compound inhibited the supercoiling activity of the S. aureus DNA gyrase at minimum inhibitory concentrations (MIC) of 250 µg/mL (IC50=100.2±13.9 µg/mL). Moreover, it was observed that the presence of 4 in conjugate 6 improves the anti-topoisomerase activity of 2 towards S. aureus DNA gyrase, which is in agreement with results obtained from antibacterial susceptibility tests involving MRSA strains.


Subject(s)
Alkynes/pharmacology , Anti-Bacterial Agents/pharmacology , Curcumin/analogs & derivatives , DNA, Superhelical/chemistry , DNA, Superhelical/pharmacology , Fatty Acids, Unsaturated/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Alkynes/chemistry , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/toxicity , Chlorocebus aethiops , Curcumin/chemical synthesis , Curcumin/pharmacology , Curcumin/toxicity , DNA Gyrase/chemistry , Escherichia coli/drug effects , Fatty Acids, Unsaturated/chemistry , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Staphylococcus aureus/enzymology , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/toxicity , Vero Cells
20.
Bioorg Med Chem ; 23(19): 6404-17, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26456623

ABSTRACT

Three series of novel 3,5-bis(arylmethylene)-1-(N-(ortho-substituted aryl)maleamoyl)-4-piperidones, designed as simplified analogs of curcumin with maleic diamide tether, were synthesized and bioevaluated. These compounds displayed potent cytotoxicity towards human Molt 4/C8 and CEM T-lymphocytes as well as murine L1210 leukemic cells. In contrast, the related N-arylmaleamic acids possessed little or no cytotoxicity in these three screens. Design of these compounds was based on molecular modeling studies performed on a related series of molecule in a previous study. Representative title compounds were found to be significantly potent in inhibiting the activity of topoisomerase II alpha indicating the possible mode of action of these compounds. These compounds were also potent antioxidants in vitro and attenuated the AAPH triggered peroxyl radical production in human fibroblasts. Various members of these series were also well tolerated in both in vitro and in vivo toxicity analysis.


Subject(s)
Curcumin/chemistry , DNA-Binding Proteins/antagonists & inhibitors , Piperidones/chemistry , Topoisomerase II Inhibitors/chemistry , Animals , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line , Cell Survival/drug effects , Curcumin/chemical synthesis , Curcumin/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Drug Design , Drug Screening Assays, Antitumor , Humans , Mice , Quantitative Structure-Activity Relationship , Reactive Oxygen Species/metabolism , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL