Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 641
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 599(7883): 120-124, 2021 11.
Article in English | MEDLINE | ID: mdl-34646011

ABSTRACT

Antibiotics are used to fight pathogens but also target commensal bacteria, disturbing the composition of gut microbiota and causing dysbiosis and disease1. Despite this well-known collateral damage, the activity spectrum of different antibiotic classes on gut bacteria remains poorly characterized. Here we characterize further 144 antibiotics from a previous screen of more than 1,000 drugs on 38 representative human gut microbiome species2. Antibiotic classes exhibited distinct inhibition spectra, including generation dependence for quinolones and phylogeny independence for ß-lactams. Macrolides and tetracyclines, both prototypic bacteriostatic protein synthesis inhibitors, inhibited nearly all commensals tested but also killed several species. Killed bacteria were more readily eliminated from in vitro communities than those inhibited. This species-specific killing activity challenges the long-standing distinction between bactericidal and bacteriostatic antibiotic classes and provides a possible explanation for the strong effect of macrolides on animal3-5 and human6,7 gut microbiomes. To mitigate this collateral damage of macrolides and tetracyclines, we screened for drugs that specifically antagonized the antibiotic activity against abundant Bacteroides species but not against relevant pathogens. Such antidotes selectively protected Bacteroides species from erythromycin treatment in human-stool-derived communities and gnotobiotic mice. These findings illluminate the activity spectra of antibiotics in commensal bacteria and suggest strategies to circumvent their adverse effects on the gut microbiota.


Subject(s)
Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Gastrointestinal Microbiome/drug effects , Animals , Anti-Bacterial Agents/classification , Bacteria/classification , Bacteria, Anaerobic/drug effects , Bacteroides/drug effects , Clostridioides difficile/drug effects , Dicumarol/pharmacology , Erythromycin/pharmacology , Feces/microbiology , Female , Germ-Free Life , Humans , Macrolides/pharmacology , Male , Mice , Microbiota/drug effects , Symbiosis/drug effects , Tetracyclines/pharmacology
2.
Bioorg Chem ; 129: 106191, 2022 12.
Article in English | MEDLINE | ID: mdl-36270169

ABSTRACT

PPARγ full agonists, thiazolidinediones (TZDs), have been known as a class of most effective drugs for the treatment of type 2 diabetes mellitus (T2DM). However, recently their therapeutic benefits have been compromised by several undesirable side effects. In this study, a host-based repurposing strategy and in combination with comprehensive biological evaluations were synergistically employed to seek for potent PPARγ ligands, which led to the identification of an anti-thrombotic drug, dicoumarol (Dic), as the novel and safer selectively PPARγ modulator (SPPARγM) with advantages over current TZD drugs. The results in vitro showed that Dic had a potent binding affinity and weakly agonistic activity for PPARγ and its downstream key genes. Moreover, in diabetic model, it significantly reduced blood glucose without leading to the weight gain of both body and main organ tissues. Further mechanistic investigations revealed that Dic possessed such desired pharmacological properties mainly through effectively inhibiting the phosphorylation of PPARγ-Ser273 and selectively regulating the expressions of insulin-sensitive and resistance genes. Finally, the docking studies on the analysis of the potent binding mode of Dic with PPARγ revealed a remarkable difference on interaction region compared with other developed PPARγ agonists, which not only gave a proof of concept for the abovementioned mechanism but also provided the molecular basis for the discrimination of Dic from other PPARγ ligands, especially TZD drugs. Taken together, our findings suggested that Dic could serve as a new and promising candidate with good therapeutic index for treating T2DM, especially for those T2DM patients with thrombosis.


Subject(s)
Anticoagulants , Diabetes Mellitus, Type 2 , Dicumarol , Hypoglycemic Agents , PPAR gamma , Thrombosis , Humans , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Dicumarol/chemistry , Dicumarol/pharmacology , Dicumarol/therapeutic use , Hypoglycemic Agents/chemistry , Ligands , PPAR gamma/agonists , Thiazolidinediones/adverse effects , Thiazolidinediones/pharmacology , Thrombosis/drug therapy , Thrombosis/etiology , Anticoagulants/chemistry , Anticoagulants/pharmacology
3.
Molecules ; 27(7)2022 Mar 31.
Article in English | MEDLINE | ID: mdl-35408682

ABSTRACT

A new dicoumarin, jusan coumarin, (1), has been isolated from Artemisia glauca aerial parts. The chemical structure of jusan coumarin was estimated, by 1D, 2D NMR as well as HR-Ms spectroscopic methods, to be 7-hydroxy-6-methoxy-3-[(2-oxo-2H-chromen-6-yl)oxy]-2H-chromen-2-one. As the first time to be introduced in nature, its potential against SARS-CoV-2 has been estimated using various in silico methods. Molecular similarity and fingerprints experiments have been utilized for 1 against nine co-crystallized ligands of COVID-19 vital proteins. The results declared a great similarity between Jusan Coumarin and X77, the ligand of COVID-19 main protease (PDB ID: 6W63), Mpro. To authenticate the obtained outputs, a DFT experiment was achieved to confirm the similarity of X77 and 1. Consequently, 1 was docked against Mpro. The results clarified that 1 bonded in a correct way inside Mpro active site, with a binding energy of -18.45 kcal/mol. Furthermore, the ADMET and toxicity profiles of 1 were evaluated and showed the safety of 1 and its likeness to be a drug. Finally, to confirm the binding and understand the thermodynamic characters between 1 and Mpro, several molecular dynamics (MD) simulations studies have been administered. Additionally, the known coumarin derivative, 7-isopentenyloxycoumarin (2), has been isolated as well as ß-sitosterol (3).


Subject(s)
Artemisia , Coronavirus 3C Proteases , Coumarins , Protease Inhibitors , SARS-CoV-2 , Artemisia/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Coumarins/chemistry , Coumarins/pharmacology , Dicumarol/chemistry , Dicumarol/pharmacology , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology
4.
Neurochem Res ; 46(1): 88-99, 2021 Jan.
Article in English | MEDLINE | ID: mdl-31902045

ABSTRACT

The reduction of water-soluble tetrazolium salts (WSTs) is frequently used to determine the metabolic integrity and the viability of cultured cells. Recently, we have reported that the electron cycler menadione can efficiently connect intracellular oxidation reactions in cultured astrocytes with the extracellular reduction of WST1 and that this menadione cycling reaction involves an enzyme. The enzymatic reaction involved in the menadione-dependent WST1 reduction was found strongly enriched in the cytosolic fraction of cultured astrocytes and is able to efficiently use both NADH and NADPH as electron donors. In addition, the reaction was highly sensitive towards dicoumarol with Kic values in the low nanomolar range, suggesting that the NAD(P)H:quinone oxidoreductase 1 (NQO1) catalyzes the menadione-dependent WST1 reduction in astrocytes. Also, in intact astrocytes, dicoumarol inhibited the menadione-dependent WST1 reduction in a concentration-dependent manner with half-maximal inhibition observed at around 50 nM. Moreover, the menadione-dependent WST1 reduction by viable astrocytes was strongly affected by the availability of glucose. In the absence of glucose only residual WST1 reduction was observed, while a concentration-dependent increase in WST1 reduction was found during a 30 min incubation with maximal WST1 reduction already determined in the presence of 0.5 mM glucose. Mannose could fully replace glucose as substrate for astrocytic WST1 reduction, while other hexoses, lactate and the mitochondrial substrate ß-hydroxybutyrate failed to provide electrons for the cell-dependent WST1 reduction. These results demonstrate that the menadione-mediated WST1 reduction involves cytosolic NQO1 activity and that this process is strongly affected by the availability of glucose as metabolic substrate.


Subject(s)
Astrocytes/metabolism , Glucose/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Tetrazolium Salts/metabolism , Vitamin K 3/metabolism , Animals , Animals, Newborn , Cells, Cultured , Dicumarol/pharmacology , Enzyme Inhibitors/pharmacology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Oxidation-Reduction , Rats, Wistar , Tetrazolium Salts/chemistry
5.
Biochem Biophys Res Commun ; 524(4): 1003-1009, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32063361

ABSTRACT

Colon cancer is one of the leading causes of cancer-related deaths and its five-year survival rate remains low in locally advanced or metastatic stages of colon cancer. Overexpression of high mobility group protein AT-hook2 (HMGA2) is associated with cancer progression, metastasis, and poor prognosis in many malignancies. Oxidative stress regulates cellular mechanisms and provides an environment that favors the cancer cells to survive and progress, yet, at the same time, oxidative stress can also be utilized as a cancer-damaging strategy. The molecular regulatory roles of HMGA2 in oxidative stress and their involvement in cancer progression are largely unknown. In this study, we investigated the involvement of HMGA2 in regulation of oxidative stress responses by luciferase reporter assays. Moreover, we utilized dicoumarol (DIC), a derivative of coumarin which has been suggested to be involved in oxidation regulation with anticancer effects, and demonstrated that DIC could induce apoptosis and inhibit cell migration of HMGA2 overexpressing colon cancer cells. Further investigation also evidenced that DIC can enhance the cancer inhibition effect of 5-FU in colony formation assays. Taken together, our data revealed novel insights into the molecular mechanisms underlying HMGA2 and highlighted the possibility of targeting the cellular antioxidant system for treating patients and preventing from cancer progression in HMGA2 overexpressing colon cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Dicumarol/pharmacology , HMGA2 Protein/metabolism , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Fluorouracil/pharmacology , Humans , Oxidative Stress/drug effects
6.
Toxicol Appl Pharmacol ; 404: 115180, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32739527

ABSTRACT

Numerous studies conducted in the past have reported deaths in the human population due to cardiovascular diseases (CVD) on exposure to air particulate matter (APM). BP-1,6-quinone (BP-1,6-Q) is one of the significant components of APM. However, the mechanism(s) by which it can exert its toxicity in endothelial cells is not yet completely understood. NAD(P)H: quinone oxidoreductase-1 (NQO1) is expressed highly in myocardium and vasculature tissues of the heart and plays a vital role in maintaining vascular homeostasis. This study, demonstrated that BP-1,6-Q diminishes NQO1 enzyme activity in a dose-dependent manner in human EA.hy926 endothelial cells. The decrease in the NQO1 enzyme causes potentiation in BP-1,6-Q-mediated toxicity in EA.hy926 endothelial cells. The enhancement of NQO1 in endothelial cells showed cytoprotection against BP-1,6-Q-induced cellular toxicity, lipid, and protein damage suggesting an essential role of NQO1 in cytoprotection against BP-1,6-Q toxicity. Using various biochemical assays and genetic approaches, results from this study further demonstrated that NQO1 also plays a crucial role in BP-1,6-Q-induced production of reactive oxygen species (ROS). These findings will contribute to elucidating BP-1,6-Q mediated toxicity and its role in the development of atherosclerosis.


Subject(s)
Benzopyrenes/toxicity , Endothelial Cells/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Reactive Oxygen Species/metabolism , Benzopyrenes/chemistry , Cell Line , Cell Survival/drug effects , Dicumarol/pharmacology , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/metabolism , Molecular Structure , NAD(P)H Dehydrogenase (Quinone)/genetics
7.
Neurochem Res ; 45(10): 2442-2455, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32789798

ABSTRACT

ß-lapachone (ß-lap) is reduced in tumor cells by the enzyme NAD(P)H: quinone acceptor oxidoreductase 1 (NQO1) to a labile hydroquinone which spontaneously reoxidises to ß-lap, thereby generating reactive oxygen species (ROS) and oxidative stress. To test for the consequences of an acute exposure of brain cells to ß-lap, cultured primary rat astrocytes were incubated with ß-lap for up to 4 h. The presence of ß-lap in concentrations of up to 10 µM had no detectable adverse consequences, while higher concentrations of ß-lap compromised the cell viability and the metabolism of astrocytes in a concentration- and time-dependent manner with half-maximal effects observed for around 15 µM ß-lap after a 4 h incubation. Exposure of astrocytes to ß-lap caused already within 5 min a severe increase in the cellular production of ROS as well as a rapid oxidation of glutathione (GSH) to glutathione disulfide (GSSG). The transient cellular accumulation of GSSG was followed by GSSG export. The ß-lap-induced ROS production and GSSG accumulation were completely prevented in the presence of the NQO1 inhibitor dicoumarol. In addition, application of dicoumarol to ß-lap-exposed astrocytes caused rapid regeneration of the normal high cellular GSH to GSSG ratio. These results demonstrate that application of ß-lap to cultured astrocytes causes acute oxidative stress that depends on the activity of NQO1. The sequential application of ß-lap and dicoumarol to rapidly induce and terminate oxidative stress, respectively, is a suitable experimental paradigm to study consequences of a defined period of acute oxidative stress in NQO1-expressing cells.


Subject(s)
Astrocytes/drug effects , Dicumarol/pharmacology , Enzyme Inhibitors/pharmacology , Naphthoquinones/adverse effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Cell Survival/drug effects , Glutathione/chemistry , Glutathione/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Oxidation-Reduction , Rats, Wistar , Reactive Oxygen Species/metabolism
8.
Pharmacol Res ; 160: 105193, 2020 10.
Article in English | MEDLINE | ID: mdl-32911072

ABSTRACT

Dicoumarol is an oral anticoagulant agent prescribed in clinical for decades. It is a natural hydroxycoumarin discovered from the spoilage of Melilotus officinalis (L.) Pall and is originally discovered as a rodenticide. Due to its structural similarity to that of vitamin K, it significantly inhibits vitamin K epoxide reductase and acts as a vitamin K antagonist. Dicoumarol is mainly used as an anticoagulant to prevent thrombogenesis and to cure vascular thrombosis. Other biological activities besides anticoagulants such as anticancer, antimicrobial, antiviral, etc., have also been documented. The side effects of dicoumarol raise safety concerns for clinical application. In this review, the physicochemical property, the pharmacological activities, the side effects, and the pharmacokinetics of dicoumarol were summarized, aiming to provide a whole picture of the "old" anticoagulant.


Subject(s)
Anticoagulants/pharmacology , Dicumarol/pharmacology , Animals , Anticoagulants/therapeutic use , Dicumarol/chemistry , Dicumarol/therapeutic use , Humans , Melilotus/chemistry , Vitamin K/antagonists & inhibitors , Vitamin K Epoxide Reductases/antagonists & inhibitors
9.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33228195

ABSTRACT

Derivatives of tirapazamine and other heteroaromatic N-oxides (ArN→O) exhibit tumoricidal, antibacterial, and antiprotozoal activities, which are typically attributed to bioreductive activation and free radical generation. In this work, we aimed to clarify the role of NAD(P)H:quinone oxidoreductase (NQO1) in ArN→O aerobic cytotoxicity. We synthesized 9 representatives of ArN→O with uncharacterized redox properties and examined their single-electron reduction by rat NADPH:cytochrome P-450 reductase (P-450R) and Plasmodium falciparum ferredoxin:NADP+ oxidoreductase (PfFNR), and by rat NQO1. NQO1 catalyzed both redox cycling and the formation of stable reduction products of ArN→O. The reactivity of ArN→O in NQO1-catalyzed reactions did not correlate with the geometric average of their activity towards P-450R- and PfFNR, which was taken for the parameter of their redox cycling efficacy. The cytotoxicity of compounds in murine hepatoma MH22a cells was decreased by antioxidants and the inhibitor of NQO1, dicoumarol. The multiparameter regression analysis of the data of this and a previous study (DOI: 10.3390/ijms20184602) shows that the cytotoxicity of ArN→O (n = 18) in MH22a and human colon carcinoma HCT-116 cells increases with the geometric average of their reactivity towards P-450R and PfFNR, and with their reactivity towards NQO1. These data demonstrate that NQO1 is a potentially important target of action of heteroaromatic N-oxides.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Antiprotozoal Agents/pharmacology , Cyclic N-Oxides/pharmacology , Ferredoxin-NADP Reductase/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NADPH-Ferrihemoprotein Reductase/antagonists & inhibitors , Aerobiosis , Animals , Anti-Bacterial Agents/chemical synthesis , Antioxidants/chemical synthesis , Antiprotozoal Agents/chemical synthesis , Cell Line, Tumor , Cell Survival/drug effects , Cyclic N-Oxides/chemical synthesis , Dicumarol/pharmacology , Enzyme Assays , Enzyme Inhibitors/pharmacology , Ferredoxin-NADP Reductase/chemistry , Ferredoxin-NADP Reductase/metabolism , HCT116 Cells , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Kinetics , Mice , NAD(P)H Dehydrogenase (Quinone)/chemistry , NAD(P)H Dehydrogenase (Quinone)/metabolism , NADPH-Ferrihemoprotein Reductase/chemistry , NADPH-Ferrihemoprotein Reductase/metabolism , Oxidation-Reduction , Plasmodium falciparum/chemistry , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Rats , Tirapazamine/chemistry , Tirapazamine/pharmacology
10.
Angew Chem Int Ed Engl ; 59(47): 21143-21150, 2020 11 16.
Article in English | MEDLINE | ID: mdl-32729980

ABSTRACT

Unpredictable in vivo therapeutic feedback of hydroxyl radical (. OH) efficiency is the major bottleneck of chemodynamic therapy. Herein, we describe novel Fenton-based nanotheranostics NQ-Cy@Fe&GOD for spatio-temporally reporting intratumor . OH-mediated treatment, which innovatively unites dual-channel near-infrared (NIR) fluorescence and magnetic resonance imaging (MRI) signals. Specifically, MRI signal traces the dose distribution of Fenton-based iron oxide nanoparticles (IONPs) with high-spatial resolution, meanwhile timely fluorescence signal quantifies . OH-mediated therapeutic response with high spatio-temporal resolution. NQ-Cy@Fe&GOD can successfully monitor the intracellular release of IONPs and . OH-induced NQO1 enzyme in living cells and tumor-bearing mice, which makes a breakthrough in conquering the inherent unpredictable obstacles on spatio-temporally reporting chemodynamic therapy, so as to manipulate dose-dependent therapeutic process.


Subject(s)
Antineoplastic Agents/pharmacology , Hydrogen Peroxide/pharmacology , Hydroxyl Radical/pharmacology , Iron/pharmacology , Magnetic Iron Oxide Nanoparticles/chemistry , Magnetic Resonance Imaging , Optical Imaging , A549 Cells , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dicumarol/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Hydrogen Peroxide/chemical synthesis , Hydrogen Peroxide/chemistry , Hydroxyl Radical/chemistry , Infrared Rays , Iron/chemistry , Mice , Mice, Nude , Molecular Structure , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism
11.
Chembiochem ; 20(22): 2841-2849, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31165578

ABSTRACT

NAD(P)H quinone oxidoreductase-1 (NQO1) is a homodimeric protein that acts as a detoxifying enzyme or as a chaperone protein. Dicourmarol interacts with NQO1 at the NAD(P)H binding site and can both inhibit enzyme activity and modulate the interaction of NQO1 with other proteins. We show that the binding of dicoumarol and related compounds to NQO1 generates negative cooperativity between the monomers. This does not occur in the presence of the reducing cofactor, NAD(P)H, alone. Alteration of Gly150 (but not Gly149 or Gly174) abolished the dicoumarol-induced negative cooperativity. Analysis of the dynamics of NQO1 with the Gaussian network model indicates a high degree of collective motion by monomers and domains within NQO1. Ligand binding is predicted to alter NQO1 dynamics both proximal to the ligand binding site and remotely, close to the second binding site. Thus, drug-induced modulation of protein motion might contribute to the biological effects of putative inhibitors of NQO1.


Subject(s)
Allosteric Regulation/drug effects , Dicumarol/pharmacology , Enzyme Inhibitors/pharmacology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Amino Acid Substitution , Catalytic Domain , Cell Line, Tumor , Dicumarol/metabolism , Enzyme Inhibitors/metabolism , Humans , Ligands , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Protein Binding , Tumor Suppressor Protein p53/metabolism
12.
Neurochem Res ; 44(2): 333-346, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30443714

ABSTRACT

Dicoumarol is frequently used as inhibitor of the detoxifying enzyme NAD(P)H:quinone acceptor oxidoreductase 1 (NQO1). In order to test whether dicoumarol may also affect the cellular glutathione (GSH) metabolism, we have exposed cultured primary astrocytes to dicoumarol and investigated potential effects of this compound on the cell viability as well as on the cellular and extracellular contents of GSH and its metabolites. Incubation of astrocytes with dicoumarol in concentrations of up to 100 µM did not acutely compromise cell viability nor was any GSH consumption or GSH oxidation to glutathione disulfide (GSSG) observed. However, unexpectedly dicoumarol inhibited the cellular multidrug resistance protein (Mrp) 1-dependent export of GSH in a time- and concentration-dependent manner with half-maximal effects observed at low micromolar concentrations of dicoumarol. Inhibition of GSH export by dicoumarol was not additive to that observed for the known Mrp1 inhibitor MK571. In addition, dicoumarol inhibited also the Mrp1-mediated export of GSSG during menadione-induced oxidative stress and the export of the GSH-bimane-conjugate (GS-B) that had been generated in the cells after exposure to monochlorobimane. Half-maximal inhibition of the export of Mrp1 substrates was observed at dicoumarol concentrations of around 4 µM (GSH and GSSG) and 30 µM (GS-B). These data demonstrate that dicoumarol strongly affects the GSH metabolism of viable cultured astrocytes by inhibiting Mrp1-mediated export processes and identifies for the first time Mrp1 as additional cellular target of dicoumarol.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Astrocytes/drug effects , Cell Survival/drug effects , Dicumarol/pharmacology , Propionates/pharmacology , Quinolines/pharmacology , ATP Binding Cassette Transporter, Subfamily B/drug effects , Animals , Animals, Newborn , Astrocytes/metabolism , Cells, Cultured , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Rats, Wistar
13.
Bioorg Chem ; 80: 741-752, 2018 10.
Article in English | MEDLINE | ID: mdl-30077781

ABSTRACT

Dicoumarol derivatives were synthesized in the InCl3 catalyzed pseudo three-component reactions of 4-hydroxycoumarin with aromatic aldehydes in excellent yields. The reactions were performed in water under microwave irradiation. All synthesized compounds were characterized using NMR, IR, and UV-Vis spectroscopy, as well as with TD-DFT. Obtained dicoumarols were subjected to evaluation of their in vitro lipid peroxidation and soybean lipoxygenase inhibition activities. It was shown that five of ten examined compounds (3e, 3h, 3b, 3d, 3f) possess significant potential of antilipid peroxidation (84-97%), and that compounds 3b, 3e, 3h provided the highest soybean lipoxygenase (LOX-Ib) inhibition (IC50 = 52.5 µM) and 3i somewhat lower activity (IC50 = 55.5 µM). The bioactive conformations of the best LOX-Ib inhibitors were obtained by means of molecular docking and molecular dynamics. It was shown that, within the bioactive conformations interior to LOX-Ib active site, the most active compounds form the pyramidal structure made of two 4-hydroxycoumarin cores and a central phenyl substituent. This form serves as a spatial barrier which prevents LOX-Ib Fe2+/Fe3+ ion activity to generate the coordinative bond with the C13 hydroxyl group of the α-linoleate. It is worth pointing out that the most active compounds 3b, 3e, 3h and 3i can be candidates for further examination of their in vitro and in vivo anti-inflammatory activity and that molecular modeling study results provide possibility to screen bioactive conformations and elucidate the mechanism of dicoumarols anti-LOX activity.


Subject(s)
Dicumarol/analogs & derivatives , Dicumarol/pharmacology , Lipoxygenase Inhibitors/chemistry , Lipoxygenase Inhibitors/pharmacology , Dicumarol/chemical synthesis , Drug Design , Green Chemistry Technology , Lipid Peroxidation/drug effects , Lipoxygenase/metabolism , Lipoxygenase Inhibitors/chemical synthesis , Molecular Docking Simulation , Glycine max/enzymology , Structure-Activity Relationship
14.
Article in English | MEDLINE | ID: mdl-28807907

ABSTRACT

De novo pyrimidine biosynthesis is a key metabolic pathway involved in multiple biosynthetic processes. Here, we identified an original series of 3-(1H-indol-3-yl)-2,3-dihydro-4H-furo[3,2-c]chromen-4-one derivatives as a new class of pyrimidine biosynthesis inhibitors formed by two edge-fused polycyclic moieties. We show that identified compounds exhibit broad-spectrum antiviral activity and immunostimulatory properties, in line with recent reports linking de novo pyrimidine biosynthesis with innate defense mechanisms against viruses. Most importantly, we establish that pyrimidine deprivation can amplify the production of both type I and type III interferons by cells stimulated with retinoic acid-inducible gene 1 (RIG-I) ligands. Altogether, our results further expand the current panel of pyrimidine biosynthesis inhibitors and illustrate how the production of antiviral interferons is tightly coupled to this metabolic pathway. Functional and structural similarities between this new chemical series and dicoumarol, which was reported before to inhibit pyrimidine biosynthesis at the dihydroorotate dehydrogenase (DHODH) step, are discussed.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya virus/immunology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Interferon Type I/biosynthesis , Interferons/biosynthesis , Measles virus/immunology , Pyrimidines/biosynthesis , Antiviral Agents/chemistry , Cell Line , Chromones/chemistry , Dicumarol/pharmacology , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Immunity, Innate/immunology , Indoles/chemistry , Interferon Type I/immunology , Interferons/immunology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Structure-Activity Relationship , Interferon Lambda
15.
Biochem Biophys Res Commun ; 483(1): 680-686, 2017 01 29.
Article in English | MEDLINE | ID: mdl-27986568

ABSTRACT

Deoxynyboquinone (DNQ), a potent novel quinone-based antineoplastic agent, selectively kills solid cancers with overexpressed cytosolic NAD(P)H:quinone oxidoreductase-1 (NQO1) via excessive ROS production. A genetically encoded redox-sensitive probe was used to monitor intraorganellar glutathione redox potentials (EGSH) as a direct indicator of cellular oxidative stress following chemotherapeutic administration. Beta-lapachone (ß-lap) and DNQ-induced spatiotemporal redox responses were monitored in human lung A549 and pancreatic MIA-PaCa-2 adenocarcinoma cells incubated with or without dicumarol and ES936, potent NQO1 inhibitors. Immediate oxidation of EGSH in both the cytosol and mitochondrial matrix was observed in response to DNQ and ß-lap. The DNQ-induced cytosolic oxidation was fully prevented with NQO1 inhibition, whereas mitochondrial oxidation in A549 was NQO1-independent in contrast to MIA-PaCa-2 cells. However, at pharmacologic concentrations of ß-lap both quinone-based substrates directly oxidized the redox probe, a possible sign of off-target reactivity with cellular thiols. Together, these data provide new evidence that DNQ's direct and discerning NQO1 substrate specificity underlies its pharmacologic potency, while ß-lap elicits off-target responses at its effective doses.


Subject(s)
Antineoplastic Agents/pharmacology , Glutathione/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress/drug effects , Quinones/pharmacology , Biosensing Techniques , Cell Line, Tumor , Cytosol/drug effects , Cytosol/metabolism , Dicumarol/pharmacology , Fluorescent Dyes/analysis , Glutaredoxins/analysis , Glutaredoxins/genetics , Glutathione/analysis , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Indolequinones/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Imaging , Molecular Probes/genetics , Molecular Targeted Therapy , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Naphthoquinones/metabolism , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Substrate Specificity
16.
Bioorg Med Chem Lett ; 27(5): 1325-1328, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28159415

ABSTRACT

A series of novel ROS inducers were designed by merging the fragments of piperlongumine and dicoumarol. Most of these derivatives showed potent in vitro activity against three cancer cell lines and good selectivity towards normal lung cells. The most potent and selective compound 3e was proven to exhibit obvious ROS elevation and excellent in vivo antitumor activity with suppressed tumor growth by 48.46% at the dose of 5mg/kg. Supported by these investigation, these findings encourage further investigation around this interesting antitumor chemotype.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Dicumarol/chemistry , Dioxolanes/chemistry , Reactive Oxygen Species , A549 Cells , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dicumarol/chemical synthesis , Dicumarol/pharmacology , Dioxolanes/chemical synthesis , Dioxolanes/pharmacology , Drug Screening Assays, Antitumor , Heterografts , Humans , Inhibitory Concentration 50 , Mice , Molecular Structure , Reactive Oxygen Species/metabolism
17.
Mol Hum Reprod ; 22(1): 57-67, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26612783

ABSTRACT

STUDY HYPOTHESIS: Dicoumarol (DC) has potential for use as a gonad-safe anticancer agent. STUDY FINDING: DC altered cell proliferation, decreased viability and increased apoptosis in Vero and MCF-7 cell lines but did not show any toxic effect on mouse ovarian tissues and developing oocytes in vitro and in vivo. WHAT IS KNOWN ALREADY: DC suppresses cell proliferation and increases apoptosis in various cancer cells such as breast, urogenital and melanoma. DC has also been reported to alter the anticancer effects of several chemotherapeutics, including cisplatin, gemcitabine and doxorubicin in prostate, liver and uroepithelial cancer cells, respectively. STUDY DESIGN, SAMPLES/MATERIALS, METHODS: Vero (African green monkey kidney epithelial cells) and MCF-7 (human cancerous breast epithelial cells) cell lines and mouse granulosa cells isolated from 21-day-old female BALB/c mice (n = 21) were used to assess the effects of DC (10, 50, 100 and 200 µm) for 24 and 48 h on cell proliferation, viability and apoptotic cell death. In vivo experiments were performed with a single i.p. injection of 32 mg/kg DC in 21-day-old female BALB/c mice (n = 12). Following 48 h, animals were sacrificed by cervical dislocation and histological sections of isolated ovaries were evaluated for apoptosis. Viability assays were based on the trypan blue dye exclusion method and an automated cell counter device was used. Terminal deoxynucleotidyltransferase-mediated dUTP nick-end labelling (TUNEL) and Annexin-V immunofluorescence were assessed by 3D confocal microscopy to address apoptotic cell death. We also assessed whether DC inhibits cell proliferation and viability through NQO1 [NAD(P)H Quinone Oxidoreductase 1], an intracellular inhibitor of reactive oxygen species (ROS). The meiotic spindle and chromosomes were studied in mouse oocytes by α-ß-tubulin and 7-aminoactinomycine D (7-AAD) immunostaining in vitro and in vivo. MAIN RESULTS AND THE ROLE OF CHANCE: DC does not block oocyte maturation and no significant alteration was noted in meiotic spindle or chromosome morphology in metaphase-II (M-II) stage oocytes following DC treatment in vitro or in vivo. In contrast, exposure to DC for 24 h suppressed cell proliferation (P = 0.026 at 200 µm), decreased viability (P = 0.002 at 200 µm) and increased apoptosis (P = 0.048 at 100 µm) in Vero and MCF-7 cell lines, compared with controls. These changes were not related to intracellular NQO1 levels. Mouse granulosa cells were unaffected by 50 or 100 µm DC treatment for 24 and 48 h in vitro. DC treatment in vivo did not alter the number of primordial follicles or the ratio of apoptosis in primordial, primary and secondary follicles, as well as in antral follicles, compared with the controls. LIMITATIONS, REASONS FOR CAUTION: DC was tested for ovarian toxicity only in isolated mouse oocytes/ovaries and healthy BALB/c mice. No cancer formation was used as an in vivo test model. The possibility that DC may potentiate ovarian toxicity when combined with traditional chemotherapeutic agents, such as mitomycin-C, cisplatin, gemcitabine and doxorubicin, must be taken into account, as DC is known to alter their effects in some cancer cells. WIDER IMPLICATIONS OF THE FINDINGS: The present study evaluated, for the first time, the effect of DC on ovarian tissue. The results suggested that DC is not toxic to ovarian tissues and developing oocytes; therefore, DC should be assessed further as a potential anticancer agent when female fertility preservation is a concern. LARGE SCALE DATA: N/A. STUDY FUNDING AND COMPETING INTERESTS: This work includes data from dissertation thesis entitled 'Effects of dicoumarol on mitotic and meiotic cells as an anticancer agent' by DA, 2014 and was partly supported by The National Scientific and Technological Research Council of Turkey (SBAG-109S415) to AC, OC and SO. The authors confirm that this article content presents no conflicts of interest.


Subject(s)
Antineoplastic Agents/pharmacology , Dicumarol/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Chlorocebus aethiops , Dicumarol/administration & dosage , Dicumarol/toxicity , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Granulosa Cells/drug effects , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mitotic Index , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/physiology , Oocytes/drug effects , Organ Sparing Treatments , Ovary/drug effects , Spindle Apparatus/drug effects , Spindle Apparatus/ultrastructure , Vero Cells
18.
Bioorg Med Chem Lett ; 26(2): 512-517, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26681508

ABSTRACT

Naphtho[1',2':4,5]imidazo[1,2-a]pyridine-5,6-diones (NPDOs), a new type of N-heterocycle-fused o-quinones, have been synthesized. They have been found to be efficient electron-accepting substrates of NADPH-dependent single-electron-transferring P-450R and two-electron transferring NQO1, generating reactive oxygen species (ROS) with a concomitant decrease in NADPH, which is consistent with redox-cycling. The reactivity of NPDOs toward P-450R (in terms of kcat/Km) varied in the range of 10(6)-10(7)M(-1)s(-1), while their reduction by NQO1 proceeded much faster, approaching the diffusion control limit (kcat/Km∼10(8)-10(9)M(-1)s(-1)). NPDOs exhibited relatively high cytotoxic activity against human lung carcinoma (A-549) and breast tumor (MCF-7) cell lines (LC50=0.1-8.3µM), while promyelocytic leukemia cells (HL-60) were less sensitive to NPDOs (LC50⩾10µM). 3-Nitro-substituted NPDO (11) revealed the highest potency against both A-549 and MCF-7 cell lines, with LC50 of 0.12±0.03µM and 0.28±0.08µM, respectively. Dicoumarol partly suppressed the activity of the compounds against A-594 and MCF-7 cell lines, suggesting that their cytotoxic action might be partially influenced by NQO1-mediated bioreductive activation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Naphthoquinones/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cytochrome P-450 Enzyme System/metabolism , Dicumarol/pharmacology , Escherichia coli/drug effects , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacology , Naphthoquinones/chemical synthesis , Naphthoquinones/chemistry , Oxidation-Reduction , Pyridines/chemical synthesis , Pyridines/chemistry , Pyridines/pharmacology , Salmonella enterica/drug effects , Staphylococcus aureus/drug effects
19.
BMC Cancer ; 15: 553, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26219569

ABSTRACT

BACKGROUND: Because anaplastic lymphoma kinase (ALK) is dependent on Hsp90 for protein stability, Hsp90 inhibitors are effective in controlling growth of lung cancer cells with ALK rearrangement. We investigated the mechanism of acquired resistance to 17-(Dimethylaminoethylamino)-17-demethoxygeldanamycin (17-DMAG), a geldanamycin analogue Hsp90 inhibitor, in H3122 and H2228 non-small cell lung cancer cell lines with ALK rearrangement. METHODS: Resistant cell lines (H3122/DR-1, H3122/DR-2 and H2228/DR) were established by repeated exposure to increasing concentrations of 17-DMAG. Mechanisms for resistance by either NAD(P)H/quinone oxidoreductase 1 (NQO1), previously known as a factor related to 17-DMAG resistance, or P-glycoprotein (P-gp; ABCB1/MDR1) were queried using RT-PCR, western blot analysis, chemical inhibitors, the MTT cell proliferation/survival assay, and cellular efflux of rhodamine 123. RESULTS: The resistant cells showed no cross-resistance to AUY922 or ALK inhibitors, suggesting that ALK dependency persists in cells with acquired resistance to 17-DMAG. Although expression of NQO1 was decreased in H3122/DR-1 and H3122/DR-2, NQO1 inhibition by dicumarol did not affect the response of parental cells (H2228 and H3122) to 17-DMAG. Interestingly, all resistant cells showed the induction of P-gp at the protein and RNA levels, which was associated with an increased efflux of the P-gp substrate rhodamine 123 (Rho123). Transfection with siRNA directed against P-gp or treatment with verapamil, an inhibitor of P-gp, restored the sensitivity to the drug in all cells with acquired resistance to 17-DMAG. Furthermore, we also observed that the growth-inhibitory effect of 17-DMAG was decreased in A549/PR and H460/PR cells generated to over-express P-gp by long-term exposure to paclitaxel, and these cells recovered their sensitivity to 17-DMAG through the inhibition of P-gp. CONCLUSION: P-gp over-expression is a possible mechanism of acquired resistance to 17-DMAG in cells with ALK rearrangement.


Subject(s)
Benzoquinones/pharmacology , Drug Resistance, Neoplasm , Lactams, Macrocyclic/pharmacology , Lung Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dicumarol/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Paclitaxel/pharmacology
20.
Exp Cell Res ; 323(1): 144-154, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24462458

ABSTRACT

In the study, we investigated the effect of dicoumarol, an anti-coagulant agent with the inhibitory activity of NAD(P)H quinone oxidoreductase 1 (NQO1), on TRAIL-induced apoptosis in renal cancer cell. Combined treatment with dicoumarol and TRAIL significantly induced apoptosis in various human renal carcinoma cells including Caki, ACHN, and A498, but not in normal human skin fibroblasts (HSF) and mouse kidney cells (TMCK-1). When we elucidated the relevance of NQO1 in dicoumarol plus TRAIL-mediated apoptosis, both ES936 (a NQO1 inhibitor) and knockdown of NQO1 with siRNA had no effect on TRAIL-mediated apoptosis, suggesting that the stimulating effect of dicoumarol on TRAIL-mediated apoptosis is independent of NQO1 activity. We found that dicoumarol transcriptionally down-regulated Bcl-2 expression via inhibition of NF-κB and CREB activity, whereas it down-regulated Mcl-1 and c-FLIP expression at the post-translational level. Overexpression of Bcl-2, Mcl-1, or c-FLIP overcame the dicoumarol plus TRAIL-induced apoptosis, indicating that down-regualtion of these anti-apoptotic proteins may critically contribute to the sensitizing effect of dicoumarol on TRAIL-mediated apoptosis.


Subject(s)
Apoptosis/physiology , Carcinoma, Renal Cell/metabolism , Dicumarol/pharmacology , Enzyme Inhibitors/pharmacology , Kidney Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/biosynthesis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CREB-Binding Protein/antagonists & inhibitors , Cell Line, Tumor , Down-Regulation , Humans , Indolequinones/pharmacology , Mice , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transcription Factor CHOP/genetics , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL