Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
DNA Repair (Amst) ; 94: 102926, 2020 10.
Article in English | MEDLINE | ID: mdl-32674013

ABSTRACT

Topoisomerases play a pivotal role in ensuring DNA metabolisms during replication, transcription and chromosomal segregation. To manage DNA topology, topoisomerases generate break(s) in the DNA backbone by forming transient enzyme-DNA cleavage complexes (TOPcc) with phosphotyrosyl linkages between DNA ends and topoisomerase catalytic tyrosyl residues. Topoisomerases have been identified as the cellular targets of a variety of anti-cancer drugs (e.g. topotecan, irinotecan, etoposide and doxorubicin, and antibiotics (e.g. ciprofloxacin and levofloxacin). These drugs, as well as other exogenous and endogenous agents, convert the transient TOPcc into persistent TOPcc, which we refer to as topoisomerase DNA-protein crosslinks (TOP-DPC) that challenge genome integrity and lead to cell death if left unrepaired. Proteolysis of the bulky protein component of TOP-DPC (debulking) is a poorly understood repair process employed across eukaryotes. TOP-DPC proteolysis can be achieved either by the ubiquitin-proteasome pathway (UPP) or by non-proteasomal proteases, which are typified by the metalloprotease SPRTN/WSS1. Debulking of TOP-DPC exposes the phosphotyrosyl bonds, hence enables tyrosyl-DNA phosphodiesterases (TDP1 and TDP2) to access and cleave the bonds. In this review, we focus on current knowledge of the protease pathways for debulking TOP-DPC and highlighting recent advances in understanding the mechanisms regulating the proteolytic repair pathways. We also discuss the avenues that are being exploited to target the proteolytic repair pathways for improving the clinical outcome of topoisomerase inhibitors.


Subject(s)
DNA Adducts/metabolism , DNA Repair , Phosphoric Diester Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , DNA/chemistry , DNA/metabolism , DNA Adducts/chemistry , DNA Topoisomerases/chemistry , DNA Topoisomerases/drug effects , DNA Topoisomerases/metabolism , Eukaryota/genetics , Eukaryota/metabolism , Humans , Proteolysis , Topoisomerase Inhibitors/pharmacology
2.
Article in English | MEDLINE | ID: mdl-31678841

ABSTRACT

Leishmania is the aethiological agent responsible for the visceral leishmaniasis, a serious parasite-borne disease widely spread all over the World. The emergence of resistant strains makes classical treatments less effective; therefore, new and better drugs are necessary. Naphthoquinones are interesting compounds for which many pharmacological properties have been described, including leishmanicidal activity. This work shows the antileishmanial effect of two series of terpenyl-1,4-naphthoquinones (NQ) and 1,4-anthraquinones (AQ) obtained from natural terpenoids, such as myrcene and myrceocommunic acid. They were evaluated both in vitro and ex vivo against the transgenic iRFP-Leishmania infantum strain and also tested on liver HepG2 cells to determine their selectivity indexes. The results indicated that NQ derivatives showed better antileishmanial activity than AQ analogues, and among them, compounds with a diacetylated hydroquinone moiety provided better results than their corresponding quinones. Regarding the terpenic precursor, compounds obtained from the monoterpenoid myrcene displayed good antiparasitic efficiency and low cytotoxicity for mammalian cells, whereas those derived from the diterpenoid showed better antileishmanial activity without selectivity. In order to explore their mechanism of action, all the compounds have been tested as potential inhibitors of Leishmania type IB DNA topoisomerases, but only some compounds that displayed the quinone ring were able to inhibit the recombinant enzyme in vitro. This fact together with the docking studies performed on LTopIB suggested the existence of another mechanism of action, alternative or complementary to LTopIB inhibition. In silico druglikeness and ADME evaluation of the best leishmanicidal compounds has shown good predictable druggability.


Subject(s)
Antiprotozoal Agents/pharmacology , DNA Topoisomerases/drug effects , Leishmania infantum/drug effects , Leishmania infantum/enzymology , Quinones/pharmacology , Animals , Anthraquinones/pharmacology , Camptothecin/chemistry , Camptothecin/pharmacology , DNA Topoisomerases/chemistry , DNA Topoisomerases/genetics , Drug Resistance , Female , Hep G2 Cells/parasitology , Humans , Leishmaniasis, Visceral/drug therapy , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Naphthoquinones/pharmacology , Quinones/chemistry , Spleen/cytology , Topoisomerase Inhibitors/chemistry , Topoisomerase Inhibitors/pharmacology
3.
Indian J Med Res ; 149(2): 192-198, 2019 02.
Article in English | MEDLINE | ID: mdl-31219083

ABSTRACT

Background & objectives: Infection from fluoroquinolone-resistant extra-intestinal Escherichia coli is a global concern. In this study, isolation and characterization of fluoroquinolone-resistant extra-intestinal E. coli isolates obtained from hospital samples were undertaken to detect plasmid-mediated quinolone resistance (PMQR) genes. Methods: Forty three isolates of E. coli obtained from patients with extra-intestinal infections were subjected to antibiogram to detect fluoroquinolone resistance. The mechanism of fluoroquinolone resistance was determined by the detection of PMQR genes and mutations in quinolone resistance determining region (QRDR). Results: Of the 43 isolates, 36 were resistant to nalidixic acid (83.72%) and 28 to ciprofloxacin (65.11%). Eight E. coli isolates showed total resistance to both the antimicrobials without any minimum inhibitory concentration. The detection of PMQR genes with qnr primers showed the presence of qnrA in two, qnrB in six and qnrS in 21 isolates. The gene coding for quinolone efflux pump (qepA) was not detected in any of the isolates tested. The presence of some unexpressed PMQR genes in fluoroquinolone sensitive isolates was also observed. Interpretation & conclusions: The detection of silent PMQR genes as observed in the present study presents a risk of the transfer of the silent resistance genes to other microorganisms if present in conjugative plasmids, thus posing a therapeutic challenge to the physicians. Hence, frequent monitoring is to be done for all resistance determinants.


Subject(s)
Drug Resistance, Bacterial/genetics , Escherichia coli Infections/drug therapy , Escherichia coli/drug effects , Fluoroquinolones/adverse effects , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Ciprofloxacin/adverse effects , Ciprofloxacin/pharmacology , DNA Topoisomerases/drug effects , DNA Topoisomerases/genetics , Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Fluoroquinolones/therapeutic use , Humans , Plasmids/drug effects , Plasmids/genetics
4.
Eur J Med Chem ; 175: 269-286, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31096151

ABSTRACT

Organometallic complexes have widely been used for the treatment of various diseases viz., malaria, arthritis, syphilis, pernicious anemia, tuberculosis and particular in cancers. Recent decades have witnessed an upsurging interest in the application of organometallic compounds to treat various phenotypes of cancers with multiple etiologies. The unique and exceptional properties of organometallic compounds, intermediate between classical inorganic and organic materials provide new insight in the progress of inorganic medicinal chemistry. Herein, we have selectively focused on various organometallic sandwich and half-sandwich complexes of ruthenium (Ru), titanium (Ti), gold (Au) and iron (Fe) exhibiting promising activity towards a panel of cancer cell lines and resistant cancer cell lines. These complexes exhibit novel mechanisms of drug action through incorporation of outer-sphere recognition of molecular targets and controlled activation features based on ligand substitution along with monometallic and heterometallic redox processes. Furthermore, they are usually found to be uncharged or neutral possessing metals in a low oxidation state, exhibit kinetic stability, relative lipophilicity and are amenable to a host of various chemical transformations. This review mainly sheds light on the successful advancement of organometallic complexes as anticancer drug aspirants in relation to their versatile structural chemistry and innovative mechanisms of action targeting nucleic acids, several enzymes viz; thioredoxin reductases (Thrx), EGFR, transferrin, cathepsin B, topoisomerases etc.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Organometallic Compounds/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cathepsin B/drug effects , Cell Line , Cell Line, Tumor , DNA Topoisomerases/drug effects , ErbB Receptors/drug effects , Gold/chemistry , Humans , Neoplasms/pathology , Nucleic Acids/drug effects , Organometallic Compounds/chemistry , Organometallic Compounds/therapeutic use , Ruthenium/chemistry , Thioredoxin-Disulfide Reductase/drug effects , Titanium/chemistry , Transferrin/drug effects
5.
Int J Oncol ; 47(5): 1854-62, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26398524

ABSTRACT

Iron depletion and stimulation of iron-dependent free radical damage is a rapidly developing field for chelation therapy, but the iron mobilization from ferritin by chelators has received less attention. In this study, the di-2-pyridylketone 2-pyridine carboxylic acid hydrazone (DPPCAH) and its copper complex was prepared and characterized by NMR and MS spectra. The proliferation inhibition assay showed that both DPPCAH and its copper complex exhibited selectively proliferation inhibition for HepG2 (IC50, 4.6 ± 0.2 µM for DPPACH and 1.3 ± 0.2 µM for its copper complex), but less inhibition for HCT-116 cell line (IC50, >100 µM for DPPACH and 7.8 ± 0.4 µM for its copper complex). The mechanistic studies revealed that DPPACH could remove iron from ferritin in a oxygen-catalytic manner, and contributed to redox activity of labile iron pool (LIP), that is less reported for the chelators that possess significant biological activity. The reactive oxygen species (ROS) generation and DNA cleavage assay in vitro and in vivo showed that both DPPACH-Fe(II) and DPPACH-Cu were redox-active species, indicating that ROS may mediate their antitumor activity. Further study revealed that both DPPACH and its copper complex displayed certain degree of inhibition of type II topoisomerase (Top) which contributed to their antitumor activity. Thus, the mechanism that iron mobilization by DPPACH from ferritin contributed to LIP was proposed, and both DPPACH and its copper complex were involved in ROS generation and Top II inhibition for their antitumor activities.


Subject(s)
Cell Proliferation/drug effects , Hydrazones/administration & dosage , Liver Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Catalysis , Copper/chemistry , DNA Topoisomerases/drug effects , Hep G2 Cells , Humans , Hydrazones/chemistry , Iron/chemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Oxygen/chemistry , Topoisomerase Inhibitors/administration & dosage , Topoisomerase Inhibitors/chemistry
6.
Phytochemistry ; 116: 221-229, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25986863

ABSTRACT

Nine lanostane-type triterpenoids were isolated from branches and leaves of Abies faxoniana, along with 10 known compounds. Two were isolated as inseparable mixtures of epimers at C-23 of the γ-lactone ring that had a lactol structure. The structures of the nine compounds were established by spectroscopic analysis and circular dichroism (CD) data. The absolute configurations at the stereogenic centres of two of the known compounds were confirmed by X-ray crystallography. One compound showed cytotoxic activities against HCT-116, MCF-7, and A549 cells with IC50 values of 8.9, 7.6, and 4.2µM, respectively. The isolated compounds were tested for their effects on human DNA topoisomerases I and II. One was found to be a selective inhibitor of human topo II activity with an IC50 value of 53.5µM, which was comparable to that of the topo II inhibitor etoposide (IC50=49.6µM).


Subject(s)
Abies/chemistry , DNA Topoisomerases/drug effects , Lanosterol/isolation & purification , Lanosterol/pharmacology , Topoisomerase II Inhibitors/isolation & purification , Topoisomerase II Inhibitors/pharmacology , Topoisomerase Inhibitors/isolation & purification , Topoisomerase Inhibitors/pharmacology , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Lanosterol/chemistry , Molecular Structure , Plant Components, Aerial/chemistry , Stereoisomerism , Topoisomerase II Inhibitors/chemistry , Topoisomerase Inhibitors/chemistry
7.
Tuberculosis (Edinb) ; 95 Suppl 1: S200-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25801335

ABSTRACT

Discovering new drugs to treat tuberculosis more efficiently and to overcome multidrug resistance is a world health priority. To find novel antitubercular agents several approaches have been used in various institutions worldwide, including target-based approaches against several validated mycobacterial enzymes and phenotypic screens. We screened more than 17,000 compounds from Vichem's Nested Chemical Library™ using an integrated strategy involving whole cell-based assays with Corynebacterium glutamicum and Mycobacterium tuberculosis, and target-based assays with protein kinases PknA, PknB and PknG as well as other targets such as PimA and bacterial topoisomerases simultaneously. With the help of the target-based approach we have found very potent hits inhibiting the selected target enzymes, but good minimal inhibitory concentrations (MIC) against M. tuberculosis were not achieved. Focussing on the whole cell-based approach several potent hits were found which displayed minimal inhibitory concentrations (MIC) against M. tuberculosis below 10 µM and were non-mutagenic, non-cytotoxic and the targets of some of the hits were also identified. The most active hits represented various scaffolds. Medicinal chemistry-based lead optimization was performed applying various strategies and, as a consequence, a series of novel potent compounds were synthesized. These efforts resulted in some effective potential antitubercular lead compounds which were confirmed in phenotypic assays.


Subject(s)
Antitubercular Agents/isolation & purification , Drug Design , Small Molecule Libraries , Tuberculosis, Multidrug-Resistant/drug therapy , DNA Gyrase/drug effects , DNA Topoisomerases/drug effects , Enzyme Assays , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Humans , Mannosyltransferases/antagonists & inhibitors , Microbial Sensitivity Tests , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/isolation & purification , Topoisomerase Inhibitors/chemistry , Topoisomerase Inhibitors/isolation & purification
8.
J Pharmacol Exp Ther ; 347(3): 607-14, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24049059

ABSTRACT

Nitric oxide (·NO) was originally identified as an innate cytotoxin. However, in tumors it can enhance resistance to chemotherapy and exacerbate cancer progression. Our previous studies indicated that (·NO/·NO-derived species react with etoposide (VP-16) in vitro and form products that show significantly reduced activity toward HL60 cells and lipopolysaccharide (LPS)-induced macrophages. Here, we further confirm the hypothesis that (÷)NO generation contributes to VP-16 resistance by examining interactions of ·NO with VP-16 in inducible nitric-oxide synthase (iNOS)-expressing human melanoma A375 cells. Inhibition of iNOS catalysis by N(6)-(1-iminoethyl)-L-lysine dihydrochloride (L-NIL) in human melanoma A375 cells reversed VP-16 resistance, leading to increased DNA damage and apoptosis. Furthermore, we found that coculturing A375 melanoma cells with LPS-induced macrophage RAW cells also significantly reduced VP-16 cytotoxicity and DNA damage in A375 cells. We also examined the interactions of (·)NO with another topoisomerase active drug, Adriamycin, in A375 cells. In contrast, to VP-16, (·)NO caused no significant modulation of cytotoxicity or Adriamycin-dependent apoptosis, suggesting that (⋅)NO does not interact with Adriamycin. Our studies support the hypothesis that (·)NO oxidative chemistry can detoxify VP-16 through direct nitrogen oxide radical attack. Our results provide insights into the pharmacology and anticancer mechanisms of VP-16 that may ultimately contribute to increased resistance, treatment failure, and induction of secondary leukemia in VP-16-treated patients.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents/pharmacology , DNA Topoisomerases/drug effects , Doxorubicin/pharmacology , Etoposide/pharmacology , Melanoma/drug therapy , Nitric Oxide/pharmacology , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Humans , Lysine/analogs & derivatives , Lysine/pharmacology , Macrophages/drug effects , Nitric Oxide Synthase/antagonists & inhibitors
9.
Yao Xue Xue Bao ; 47(8): 969-77, 2012 Aug.
Article in Chinese | MEDLINE | ID: mdl-23162891

ABSTRACT

The fluoroquinolones are the most widely used broad-spectrum antibiotics, accounting for 18% of global antibacterial market share. They can kill bacteria rapidly with variety of derivatives available. Different quinolones vary significantly in rate and spectrum of killing, oxygen requirement for metabolism and reliance upon protein synthesis. Further understanding the sophisticated mechanisms of action of this important antibiotic family based on the molecular genetic response of bacteria can facilitate the discovery of better quinolone derivatives. Factors such as SOS response, bacterial toxin-antitoxin system, programmed death, chromosome fragmentation and reactive oxygen have been implicated in the action to some extent. "Two steps characteristic" of quinolones killing is also emphasized, which might inspire future better quinolones modification.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria , Quinolones/pharmacology , Apoptosis/drug effects , Bacteria/drug effects , Bacteria/enzymology , Bacteria/genetics , Chromosomes, Bacterial/drug effects , DNA Cleavage/drug effects , DNA Gyrase/drug effects , DNA Replication/drug effects , DNA Topoisomerases/drug effects , Fluoroquinolones/pharmacology , Reactive Oxygen Species , SOS Response, Genetics/drug effects
10.
Planta Med ; 77(6): 586-97, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20945274

ABSTRACT

Trypanosoma brucei is the causative agent of human African trypanosomiasis (sleeping sickness) which is fatal if left untreated. This disease occurs in 36 African countries, south of the Sahara, where 60 million people are at risk of acquiring infection. The current chemotherapy relies on only four drugs, three of which were developed more than 60 years ago. These drugs have many limitations, ranging from oral inabsorption, acute toxicities, short duration of action and the emergence of trypanosomal resistance. Despite decades of use of most of the current trypanocides, little is known about their mode of action. That being said, African trypanosomes continue to be among the most extensively studied parasitic protists to date. Many of their intriguing biological features have been well documented and can be viewed as attractive targets for antitrypanosomal chemotherapy. A considerable number of natural products with diverse molecular structures have revealed antiparasitic potency in the laboratory and represent interesting lead compounds for the development of new and urgently needed antiparasitics. The major validated drug targets in T. brucei are discussed with particular emphasis on those known to be attacked by natural compounds.


Subject(s)
Biological Products/pharmacology , Trypanocidal Agents/therapeutic use , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy , Africa/epidemiology , Animals , Biological Products/chemistry , Biological Products/therapeutic use , Carbohydrate Metabolism/drug effects , DNA Topoisomerases/drug effects , DNA, Kinetoplast/drug effects , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Drug Resistance , Farnesyltranstransferase/drug effects , Humans , Oxidoreductases/drug effects , Protozoan Proteins/drug effects , Sulfhydryl Compounds/metabolism , Trypanocidal Agents/chemistry , Trypanocidal Agents/isolation & purification , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei brucei/pathogenicity , Trypanosomiasis, African/epidemiology , Tubulin/drug effects
11.
Arch Microbiol ; 192(11): 893-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20734190

ABSTRACT

Effects of different flavonoids on various bacterial strains have been extensively reported; however, the mechanism(s) of their action on bacterial cells remain largely elusive. In this study, the antibacterial mechanism of soybean isoflavone (SI) on Staphylococcus aureus is systematically investigated using 4'6-diamidino-2-phenylindole (DAPI) staining, pBR322DNA decatenation experiment mediated by topoisomerase and agarose gel electrophoresis for direct decatenation. The results of fluorescence microscopy and fluorescence spectrophotometer indicated that DAPI was integrated in Staphylococcus aureus. Additionally, the quantity of both DNA and RNA reduced to 66.47 and 60.18%, respectively, after treated with SI for 28 h. Effects of SI on topoisomerase I and II were also investigated. SI completely inhibited the pBR322DNA unwinding mediated by topoisomerase I and topoisomerase II at the concentration of 6.4 mg/ml and could denature the plasmid DNA at the concentration of 12.8 mg/ml. These results indicate that topoisomerase I and II are the most important targets by SI to restrain bacterial cell division.


Subject(s)
Anti-Bacterial Agents/pharmacology , Glycine max/chemistry , Isoflavones/pharmacology , Staphylococcus aureus/drug effects , Cell Division , DNA Topoisomerases/drug effects , DNA, Bacterial/biosynthesis , DNA, Bacterial/drug effects , Indoles/chemistry , Microbial Sensitivity Tests , Staphylococcus aureus/metabolism
12.
Eur J Pharmacol ; 638(1-3): 13-20, 2010 Jul 25.
Article in English | MEDLINE | ID: mdl-20406630

ABSTRACT

Chemopreventive non-steroidal anti-inflammatory drugs (NSAIDs) exhibit diverse pharmacological and biological activities mainly through their inhibitory effect on cyclooxygenase (COX). However, COX-independent mechanisms involving kinase inhibition have been proposed to explain certain therapeutic effects of NSAIDs. Here, we explored the potential relationship between chemopreventive NSAIDs and DNA damage responses induced by treatment with topoisomerase-targeting drugs. (1) Sodium salicylate, a non-COX-selective NSAID, was shown to reduce DNA damage-induced RPA and p53 phosphorylation. (2) The formation of enzyme cleavable complexes by topoisomerase-targeting drugs was not affected in the presence of sodium salicylate. (3) The attenuating effect of NSAIDs on the DNA damage responses is COX-2-independent, since COX-2-selective inhibitors failed to inhibit DNA damage-induced phosphorylation of replication protein A (RPA) and p53. (4) This COX-2-independent attenuating effect was mediated through interference of neither nuclear factor kappa B nor extracellular signal-regulated kinase pathways. (5) The activation of ataxia telangiectasia mutated (ATM) kinase and DNA-dependent protein kinase (DNA-PK), two key signal transducers upstream of RPA and p53, was found to be significantly reduced with sodium salicylate treatment. (6) Most importantly, sodium salicylate and other NSAIDs directly inhibited kinase activity of ATM and DNA-PK. The extent of inhibition on the kinase activity also correlated with the degree of attenuation on the DNA damage responses. (7) Unexpectedly, sodium salicylate showed a p53-independent protection effect on topoisomerase-mediated cell killing. Together, our study provides evidence that NSAIDs exhibit a novel COX-independent modulating activity of NSAIDs on the DNA damage responses and it is through inhibition of phosphoinositide 3-kinase-like kinases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , DNA Damage/drug effects , DNA Topoisomerases/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sodium Salicylate/pharmacology , Tumor Suppressor Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins , Camptothecin/pharmacology , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Drug Interactions , HCT116 Cells , Humans , NF-kappa B , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Replication Protein A/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
13.
Bioorg Med Chem Lett ; 14(11): 2857-62, 2004 Jun 07.
Article in English | MEDLINE | ID: mdl-15125947

ABSTRACT

In this study, we report the design, synthesis and structure-activity relationships of novel indazole derivatives as DNA gyrase inhibitors with Gram-positive antibacterial activity. Our results show that selected compounds from this series exhibit potent antibacterial activity against Gram-positive bacteria including multi-drug resistant strains that is methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus faecalis (VRE).


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Gram-Positive Bacteria/drug effects , Imidazoles/pharmacology , Topoisomerase II Inhibitors , Anti-Bacterial Agents/pharmacology , DNA Topoisomerases/drug effects , Drug Design , Drug Resistance, Multiple , Enterococcus faecalis/drug effects , Enterococcus faecalis/enzymology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Gram-Positive Bacteria/enzymology , Humans , Imidazoles/chemical synthesis , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Staphylococcaceae/drug effects , Staphylococcaceae/enzymology , Structure-Activity Relationship
14.
Bioorg Med Chem ; 12(10): 2597-601, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15110841

ABSTRACT

During screening for mammalian DNA polymerase inhibitors, we found and succeeded in isolating a potent inhibitor from a higher plant, Taxus cuspidate. The compound was unexpectedly determined to be taxinine, an intermediate of paclitaxel (taxol) metabolism. Taxinine was found to selectively inhibit DNA polymerase alpha (pol.alpha) and beta (pol.beta). We therefore, tested taxol and other derivatives and found that taxol itself had no such inhibitory effect, and only taxinine could inhibit both pol.alpha and beta. The other compounds used, one derivative, cephalomannine, and five intermediates synthesized chemically inhibited only the pol.alpha activity in vitro. None of the compounds, including taxinine, influenced the activities of the other DNA polymerases, which are reportedly targeted by many pol.beta inhibitors. With both pol.alpha and beta, all of the compounds tested noncompetitively inhibited with respect to both the DNA template-primer and the dNTP substrate.


Subject(s)
DNA Polymerase I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Paclitaxel/analogs & derivatives , Taxoids/pharmacology , Animals , Cell Line, Tumor , DNA Topoisomerases/drug effects , Drosophila/enzymology , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Nucleic Acid Synthesis Inhibitors/isolation & purification , Nucleic Acid Synthesis Inhibitors/pharmacology , Paclitaxel/pharmacology , Taxoids/isolation & purification , Taxus/chemistry
15.
J Biol Chem ; 279(18): 18641-7, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-14988409

ABSTRACT

Previous studies have suggested two possible roles for Rad9 in mammalian cells subjected to replication stress or DNA damage. One model suggests that a Rad9-containing clamp is loaded onto damaged DNA, where it participates in Chk1 activation and subsequent events that contribute to cell survival. The other model suggests that Rad9 translocates to mitochondria, where it triggers apoptosis by binding to and inhibiting Bcl-2 and Bcl-x(L). To further study the role of Rad9, parental and Rad9(-/-) murine embryonic stem (ES) cells were treated with camptothecin, etoposide, or cytarabine, all prototypic examples of three classes of widely used anticancer agents. All three agents induced Rad9 chromatin binding. Each of these agents also triggered S-phase checkpoint activation in parental ES cells, as indicated by a caffeine-inhibitable decrease in [3H]thymidine incorporation into DNA and Cdc25A down-regulation. Interestingly, the ability of cytarabine to activate the S-phase checkpoint was severely compromised in Rad9(-/-) cells, whereas activation of this checkpoint by camptothecin and etoposide was unaltered, suggesting that the action of cytarabine is readily distinguished from that of classical topoisomerase poisons. Nonetheless, Rad9 deletion sensitized ES cells to the cytotoxic effects of all three agents, as evidenced by enhanced apoptosis and diminished colony formation. Collectively, these results suggest that the predominant role of Rad9 in ES cells is to promote survival after replicative stress and topoisomerase-mediated DNA damage.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins/physiology , Protective Agents , Stem Cells/cytology , Topoisomerase Inhibitors , Animals , Camptothecin/pharmacology , Cell Cycle Proteins/genetics , Cytarabine/pharmacology , DNA Damage/drug effects , DNA Topoisomerases/drug effects , Embryo, Mammalian/cytology , Etoposide/pharmacology , Humans , Mice , Mice, Knockout , Stem Cells/drug effects , Transfection , Transgenes
16.
J Mol Neurosci ; 22(1-2): 19-31, 2004.
Article in English | MEDLINE | ID: mdl-14742907

ABSTRACT

Previously, we evidenced a B --> Z helical change in Alzheimer's brain genomic DNA, leading to a hypothesis that Alzheimer's disease (AD) etiological factors such as aluminum (Al), amyloid beta (Abeta) peptide, and Tau might play a role in modulating DNA topology. In the present study, we investigated the interaction of Al and Abeta with DNA. Our results show that Abeta(1-42) could induce a B --> Psi (Psi) conformational change in pUC 18 supercoiled DNA (scDNA), Abeta(1-16) caused an altered B-form, whereas Al induced a complex B-C-A mixed conformation. Ethidium bromide binding and agarose gel electrophoresis studies revealed that Al uncoiled the DNAto a fully relaxed form, whereas Abeta(1-42) and Abeta(1-16) effected a partial uncoiling and also showed differential sensitivity toward chloroquine-induced topoisomer separation. Our findings show for the first time that Abeta and Al modulate both helicity and superhelicity in scDNA. A new hypothetical model explaining the potential toxicity of Abeta and Al in terms of their DNA binding properties leading to DNA conformational alteration is proposed.


Subject(s)
Aluminum/chemistry , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , DNA, Superhelical/chemistry , Nucleic Acid Conformation/drug effects , Peptide Fragments/chemistry , Aged , Aluminum/metabolism , Aluminum/pharmacology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Apoptosis , Cell Nucleus/metabolism , Cell Nucleus/pathology , Chloroquine/pharmacology , DNA/chemistry , DNA/drug effects , DNA Topoisomerases/drug effects , DNA Topoisomerases/metabolism , DNA, Superhelical/drug effects , DNA, Z-Form/chemistry , DNA, Z-Form/drug effects , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Ethidium , Female , Gallium/chemistry , Gallium/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Male , Middle Aged , Peptide Fragments/metabolism , Peptide Fragments/pharmacology
17.
Proc Natl Acad Sci U S A ; 100(9): 5046-51, 2003 Apr 29.
Article in English | MEDLINE | ID: mdl-12704241

ABSTRACT

The cytotoxicity of several important antitumor drugs depends on formation of the covalent topoisomerase-DNA cleavage complex. However, cellular processes such as DNA replication are necessary to convert the cleavage complex into a cytotoxic lesion, but the molecular mechanism of this conversion and the precise nature of the cytotoxic lesion are unknown. Using a bacteriophage T4 model system, we have previously shown that antitumor drug-induced cleavage complexes block replication forks in vivo. In this report, we show that these blocked forks can be cleaved by T4 endonuclease VII to create overt DNA breaks. The accumulation of blocked forks increased in endonuclease VII-deficient infections, suggesting that endonuclease cleavage contributes to fork processing in vivo. Furthermore, purified endonuclease VII cleaved the blocked forks in vitro close to the branch points. These results suggest that an indirect pathway of branched-DNA cleavage contributes to the cytotoxicity of antitumor drugs that target DNA topoisomerases.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage , DNA Topoisomerases/drug effects , DNA/drug effects , Endodeoxyribonucleases/metabolism , Replication Origin , T-Phages/enzymology
18.
Antimicrob Agents Chemother ; 47(1): 274-82, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12499202

ABSTRACT

Gemifloxacin, a novel quinolone with potent activity against Staphylococcus aureus, was 8- to 16-fold more active against wild-type S. aureus than ciprofloxacin. The two- to fourfold increase in the MIC of gemifloxacin in genetically defined grlBA mutants and the twofold increase in a single gyrA mutant, supported by the low frequency of selection of resistant mutants at twice the MIC (7.4 x 10(-11) to 1.1 x 10(-10)), suggested similar targeting of the two enzymes by gemifloxacin. Dual mutations in both gyrase and topoisomerase IV caused a 64- to 128-fold increase in the MIC of gemifloxacin, similar to that seen with ciprofloxacin. Gemifloxacin also had similar activity in vitro against topoisomerase IV and gyrase purified from S. aureus (50% inhibitory concentrations of 0.25 and 0.31 micro g/ml, respectively). This activity was 10- to 20-fold higher than that of ciprofloxacin for topoisomerase IV and 33-fold higher than that for gyrase. In contrast to the in vitro findings, only topoisomerase IV mutants were selected in first-step mutants. Overexpression of the NorA efflux pump had a minimal effect on resistance to gemifloxacin, and a mutation in the promoter region of the gene for NorA was selected only in the sixth step of serial selection of mutants. Our data show that although gemifloxacin targets purified topoisomerase IV and gyrase similarly in vitro, topoisomerase IV is the preferred target in the bacteria. Selection of novel resistance mutations in grlA requires further expansion of quinolone-resistance-determining regions, and their study may provide increased insight into enzyme-quinolone interactions.


Subject(s)
Anti-Infective Agents/pharmacology , DNA Topoisomerases/drug effects , Drug Resistance, Bacterial/genetics , Fluoroquinolones , Microbial Sensitivity Tests , Naphthyridines/pharmacology , Staphylococcus aureus/enzymology , DNA Topoisomerases/classification , DNA Topoisomerases/genetics , Gemifloxacin , Mutation , Plasmids/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics
19.
Dev Growth Differ ; 44(4): 293-302, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12175364

ABSTRACT

To clarify the mechanisms of fish fertilization, the effects of inhibitors of DNA polymerase-alpha and DNA topoisomerases on nuclear behavior before and after fertilization were examined in eggs of the medaka, Oryzias latipes. Eggs underwent the fertilization process from sperm penetration to karyogamy of pronuclei, even when inseminated and incubated in the continuous presence of aphidicolin (DNA polymerase alpha inhibitor), camptothecin (DNA topoisomerase I inhibitor), etoposide, or beta-lapachone (DNA topoisomerase II inhibitor). However, continuous treatment with aphidicolin or camptothecin during fertilization inhibited the formation of sister chromosomes that were normally separated into blastomeres at the time of the subsequent cleavage. Sister chromosome formation appeared concomitantly with an increase in histone H1 kinase activity at the end of DNA synthesis, 30 min post insemination. However, non-activated eggs that were inseminated in saline containing anesthetic MS222 and aphidicolin had high levels of histone H1 kinase and MAP kinase activities, and transformation of the penetrated sperm nucleus to metaphase chromosomes occurred even in the presence of aphidicolin or camptothecin. The male chromosomes were normally separated into two anaphase chromosome masses upon egg activation. These results suggest that DNA polymerase alpha or DNA topoisomerase I, but not DNA topoisomerase II, may be required for the process by which the mitotic interphase nucleus transforms to separable metaphase chromosomes while the activity of MAP kinase is low, unlike the situation in meiotic division, during which MAP kinase activity is high and DNA replication is not required.


Subject(s)
Aphidicolin/pharmacology , Camptothecin/pharmacology , Chromosomes/drug effects , Fertilization/drug effects , Zygote/drug effects , Animals , DNA Polymerase I/antagonists & inhibitors , DNA Polymerase I/drug effects , DNA Topoisomerases/drug effects , Etoposide/pharmacology , Meiosis/drug effects , Mitogen-Activated Protein Kinases/drug effects , Mitosis/drug effects , Naphthoquinones/pharmacology , Oryzias , Protein Kinases/drug effects , Topoisomerase Inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...