Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Bioconjug Chem ; 34(1): 238-247, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36516871

ABSTRACT

As a counterpart to antibody-drug conjugates (ADCs), aptamer-drug conjugates (ApDCs) have been considered a promising strategy for targeted therapy due to the various benefits of aptamers. However, an aptamer merely serves as a targeting ligand in ApDCs, whereas the antibody enables the unexpected therapeutic efficacy of ADCs through antibody-dependent cellular cytotoxicity (ADCC). In this study, we developed a tumor-specific aptamer with an effector function and used it to confirm the feasibility of more potent ApDCs. First, we designed a nucleolin (NCL)-binding G-quadruplex (GQ) library based on the ability of NCL to bind to telomeric sequences. We then identified a bifunctional GQ aptamer (BGA) inhibiting the catalytic activity of topoisomerase 1 (TOP1) by forming an irreversible cleavage complex. Our BGA specifically targeted NCL-positive MCF-7 cells, exhibiting antiproliferative activity, and this suggested that tumor-specific therapeutic aptamers can be developed by using a biased library to screen aptamer candidates for functional targets. Finally, we utilized DM1, which has a synergistic interaction with TOP1 inhibitors, as a conjugated drug. BGA-DM1 exerted an anticancer effect 20-fold stronger than free DM1 and even 10-fold stronger than AS1411 (NCL aptamer)-DM1, highlighting our approach to develop synergistic ApDCs. Therefore, we anticipate that our library might be utilized for the identification of aptamers with effector functions. Furthermore, by employing such aptamers and appropriate drugs, synergistic ApDCs can be developed for targeted cancer therapy in a manner distinct from how ADCs exhibit additional therapeutic efficacy.


Subject(s)
Aptamers, Nucleotide , DNA Topoisomerases, Type I , RNA-Binding Proteins , Humans , Aptamers, Nucleotide/pharmacology , Aptamers, Nucleotide/metabolism , MCF-7 Cells , Phosphoproteins/metabolism , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , DNA Topoisomerases, Type I/drug effects , DNA Topoisomerases, Type I/metabolism , Drug Synergism , Nucleolin
2.
PLoS Genet ; 16(10): e1009085, 2020 10.
Article in English | MEDLINE | ID: mdl-33125364

ABSTRACT

DNA supercoiling is essential for all living cells because it controls all processes involving DNA. In bacteria, global DNA supercoiling results from the opposing activities of topoisomerase I, which relaxes DNA, and DNA gyrase, which compacts DNA. These enzymes are widely conserved, sharing >91% amino acid identity between the closely related species Escherichia coli and Salmonella enterica serovar Typhimurium. Why, then, do E. coli and Salmonella exhibit different DNA supercoiling when experiencing the same conditions? We now report that this surprising difference reflects disparate activation of their DNA gyrases by the polyamine spermidine and its precursor putrescine. In vitro, Salmonella DNA gyrase activity was sensitive to changes in putrescine concentration within the physiological range, whereas activity of the E. coli enzyme was not. In vivo, putrescine activated the Salmonella DNA gyrase and spermidine the E. coli enzyme. High extracellular Mg2+ decreased DNA supercoiling exclusively in Salmonella by reducing the putrescine concentration. Our results establish the basis for the differences in global DNA supercoiling between E. coli and Salmonella, define a signal transduction pathway regulating DNA supercoiling, and identify potential targets for antibacterial agents.


Subject(s)
DNA Gyrase/genetics , DNA Topoisomerases, Type I/genetics , DNA, Superhelical/genetics , Escherichia coli/genetics , Salmonella typhimurium/genetics , DNA Gyrase/drug effects , DNA Topoisomerases, Type I/drug effects , DNA, Superhelical/drug effects , Escherichia coli/drug effects , Escherichia coli/enzymology , Magnesium/pharmacology , Putrescine/pharmacology , Salmonella typhimurium/drug effects , Salmonella typhimurium/enzymology , Spermidine/biosynthesis
3.
Arch Toxicol ; 95(12): 3787-3802, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34635930

ABSTRACT

Nevadensin, an abundant polyphenol of basil, is reported to reduce alkenylbenzene DNA adduct formation. Furthermore, it has a wide spectrum of further pharmacological properties. The presented study focuses the impact of nevadensin on topoisomerases (TOPO) in vitro. Considering the DNA-intercalating properties of flavonoids, first, minor groove binding properties (IC50 = 31.63 µM), as well as DNA intercalation (IC50 = 296.91 µM) of nevadensin, was found. To determine potential in vitro effects on TOPO I and TOPO IIα, the relaxation and decatenation assay was performed in a concentration range of 1-500 µM nevadensin. A partial inhibition was detected for TOPO I at concentrations  ≥ 100 µM, whereas TOPO IIα activity is only inhibited at concentrations  ≥ 250 µM. To clarify the mode of action, the isolating in vivo complex of enzyme assay was carried out using human colon carcinoma HT29 cells. After 1 h of incubation, the amount of TOPO I linked to DNA was significantly increased by nevadensin (500 µM), why nevadensin was characterized as TOPO I poison. However, no effects on TOPO IIα were detected in the cellular test system. As a subsequent cellular response to TOPO I poisoning, a highly significant increase of DNA damage after 2 h and a decrease of cell viability after 48 h at the same concentration range were found. Furthermore, after 24 h of incubation a G2/M arrest was observed at concentrations ≥ 100 µM by flow cytometry. The analysis of cell death revealed that nevadensin induces the intrinsic apoptotic pathway via activation of caspase-9 and caspase-3. The results suggest that cell cycle disruption and apoptotic events play key roles in the cellular response to TOPO I poisoning caused by nevadensin in HT29 cells.


Subject(s)
Apoptosis/drug effects , DNA Damage/drug effects , DNA Topoisomerases, Type I/drug effects , Flavones/poisoning , Cell Cycle/drug effects , Colonic Neoplasms/enzymology , DNA Topoisomerases, Type II/drug effects , Dose-Response Relationship, Drug , Flavones/administration & dosage , HT29 Cells , Humans , Inhibitory Concentration 50 , Poly-ADP-Ribose Binding Proteins/drug effects , Time Factors
4.
Bioorg Chem ; 94: 103409, 2020 01.
Article in English | MEDLINE | ID: mdl-31732194

ABSTRACT

In the quest to ameliorate the camptothecin (CPT) downsides, we expedite to search for stable non-CPT analogues among 11 motifs of pyrazoloquinazolines reported. E-pharmacophore drug design approach helped filtering out pyrazolo[1,5-c]quinazolines as Topoisomerase I (TopoI) 'interfacial' inhibitors. Three compounds, 3c, 3e, and 3l were shown to be potent non-intercalating inhibitors of TopoI specifically and showed cancer cell-specific cytotoxicity in lung, breast and colon cancer cell lines. The compounds induced cell cycle arrest at S-phase, mitochondrial cell death pathway and modulated oxidative stress in cancer cells. Furthermore, a preliminary study was conducted to explore the feasibility of these compounds to be developed as dual TopoI-HDAC1 (histone deacetylase 1) inhibitors (4a) to combat resistance. Compound 4a was found to possess dual inhibitory capabilities in-vitro. Cytotoxic potential of 4a was found to be significantly higher than parent compound in 2D as well as 3D cancer cell models. Probable binding modes of 4a with TopoI and HDAC1 active sites were examined by molecular modelling.


Subject(s)
DNA Topoisomerases, Type I/drug effects , Enzyme Inhibitors/therapeutic use , Histone Deacetylases/drug effects , Quinazolines/therapeutic use , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Quinazolines/chemistry
5.
Bioorg Chem ; 103: 104162, 2020 10.
Article in English | MEDLINE | ID: mdl-32890988

ABSTRACT

In this work, 2'-alkoxymethyl substituted klavuzon derivatives were prepared starting from 2-methyl-1-naphthoic acid in eight steps. Anticancer potencies of the synthesized compounds were evaluated by performing MTT cell viability test over cancerous and healthy pancreatic cell lines, along with CRM1 inhibitory properties in HeLa cells by immunostaining and Topo I inhibition properties by supercoiled DNA relaxation assay. Their cytotoxic activities were also presented in hepatocellular carcinoma cells (HuH-7) derived 3D spheroids. Among the tested klavuzon derivatives, isobutoxymethyl substituted klavuzon showed the highest selectivity of cytotoxic activity against pancreatic cancer cell line. They showed potent Topo I inhibition while their CRM1 inhibitory properties somehow diminished compared to 4'-alkylsubstituted klavuzons. The most cytotoxic 2'-methoxymethyl derivative inhibited the growth of the spheroids derived from HuH-7 cell lines and PI staining exhibited time and concentration dependent cell death in 3D spheroids.


Subject(s)
DNA Topoisomerases, Type I/drug effects , Karyopherins/drug effects , Naphthalenes/chemistry , Naphthalenes/therapeutic use , Neoplasms/drug therapy , Pyrans/chemistry , Pyrans/therapeutic use , Receptors, Cytoplasmic and Nuclear/drug effects , Humans , Naphthalenes/pharmacology , Pyrans/pharmacology , Structure-Activity Relationship , Exportin 1 Protein
6.
Bioorg Med Chem Lett ; 29(23): 126714, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31635931

ABSTRACT

A series of novel N-phenylbenzamide-4-methylamine acridine derivatives were designed and synthesized based initially on the structure of amsacrine (m-AMSA). Molecular docking suggested that the representative compound 9a had affinity for binding DNA topoisomerase (Topo) II, which was comparable with that of m-AMSA, and furthermore that 9a could have preferential interactions with Topo I. After synthesis of 9a and analogues 9b-9f, these were all tested in vitro and the synthesized compounds displayed potent antiproliferative activity against three different cancer cell lines (K562, CCRF-CEM and U937). Among them, compounds 9b, 9c and 9d exhibiting the highest activity with IC50 value ranging from 0.82 to 0.91 µM against CCRF-CEM cells. In addition, 9b and 9d also showed high antiproliferative activity against U937 cells, with IC50 values of 0.33 and 0.23 µM, respectively. The pharmacological mechanistic studies of these compounds were evaluated by Topo I/II inhibition, western blot assay and cell apoptosis detection. In summary, 9b effectively inhibited the activity of Topo I/II and induced DNA damage in CCRF-CEM cells and, moreover, significantly induced cell apoptosis in a concentration-dependent manner. These observations provide new information and guidance for the structural optimization of more novel acridine derivatives.


Subject(s)
Apoptosis/drug effects , DNA Topoisomerases, Type II/drug effects , DNA Topoisomerases, Type I/drug effects , Methylamines/chemical synthesis , Molecular Docking Simulation/methods , Humans , Methylamines/chemistry , Molecular Structure , Structure-Activity Relationship
7.
Inorg Chem ; 58(10): 6804-6810, 2019 May 20.
Article in English | MEDLINE | ID: mdl-31046253

ABSTRACT

The substitution-inert polynuclear platinum complexes (SI-PPCs) are now recognized as a distinct subclass of platinum anticancer drugs with high DNA binding affinity. Here, we investigate the effects of SI-PPCs containing dangling amine groups in place of NH3 as ligands to increase the length of the molecule and therefore overall charge and its distribution. The results obtained with the aid of biophysical techniques, such as total intensity light scattering, gel electrophoresis, and atomic force microscopy, show that addition of dangling amine groups considerably augments the ability of SI-PPCs to condense/aggregate nucleic acids. Moreover, this enhanced capability of SI-PPCs correlates with their heightened efficiency to inhibit DNA-related enzymatic activities, such as those connected with DNA transcription, catalysis of DNA relaxation by DNA topoisomerase I, and DNA synthesis catalyzed by Taq DNA polymerase. Thus, the addition of the dangling amine groups resulting in structures of SI-PPCs, which differ so markedly from the derivatives of cisplatin used in the clinic, appears to contribute to the overall biological activity of these molecules.


Subject(s)
Amines/chemistry , Coordination Complexes/chemistry , DNA Topoisomerases, Type I/drug effects , DNA/chemistry , Platinum Compounds/chemistry , RNA/chemistry , Taq Polymerase/antagonists & inhibitors , Antineoplastic Agents/chemistry , Microscopy, Atomic Force , Topoisomerase I Inhibitors
8.
BMC Vet Res ; 15(1): 405, 2019 Nov 09.
Article in English | MEDLINE | ID: mdl-31706354

ABSTRACT

BACKGROUND: Canine leishmaniasis is a zoonotic disease caused by Leishmania infantum, being the dogs one of the major reservoirs of human visceral leishmaniasis. DNA topology is a consolidated target for drug discovery. In this regard, topoisomerase IB - one of the enzymes controlling DNA topology - has been poisoned by hundreds of compounds that increase DNA fragility and cell death. Aromathecins are novel molecules with a multiheterocyclic ring scaffold that have higher stability than camptothecins. RESULTS: Aromathecins showed strong activity against both forms of L. infantum parasites, free-living promastigotes and intra-macrophagic amastigotes harbored in ex vivo splenic explant cultures obtained from infected BALB/c mice. However, they prevented the relaxation activity of leishmanial topoisomerase IB weakly, which suggests that the inhibition of topoisomerase IB partially explains the antileishmanial effect of these compounds. The effect of aromathecins was also studied against a strain resistant to camptothecin, and results suggested that the trafficking of these compounds is not through the ABCG6 transporter. CONCLUSIONS: Aromathecins are promising novel compounds against canine leishmaniasis that can circumvent potential resistances based on drug efflux pumps.


Subject(s)
Antiprotozoal Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Leishmania infantum/drug effects , Topoisomerase I Inhibitors/pharmacology , Animals , Cell Culture Techniques , DNA Topoisomerases, Type I/drug effects , DNA Topoisomerases, Type I/metabolism , Female , Leishmania infantum/enzymology , Leishmania infantum/growth & development , Life Cycle Stages/drug effects , Mice, Inbred BALB C , Protozoan Proteins/antagonists & inhibitors , Spleen/parasitology
9.
Int J Mol Sci ; 20(21)2019 Oct 26.
Article in English | MEDLINE | ID: mdl-31717797

ABSTRACT

In the present study, a series of 4-acyloxy robustic acid derivatives were synthesized and characterized for evaluation of their anti-cancer activity. The structures of these derivatives were elucidated by mass spectra (MS) nuclear magnetic resonance spectra (NMR). The single-crystal X-ray diffraction structure of one of these compounds was obtained, for further validation of the target compound structures. The anticancer activities of the target products were evaluated against human leukemic cells HL-60, human non-small cell lung carcinoma cells A-549, human hepatic carcinoma cells SMMC-7721, human hepatocellular carcinoma cells HepG2, and human cervical carcinoma cells Hela. Three compounds among them exhibited potent in-vitro cytotoxicity and excellent DNA topoisomerase I inhibitory activity, even at 0.1 mM concentrations. The most noteworthy observation was the minor toxicity of two of these compounds to normal cells, with an activity similar to the positive control in cancerous cells. A Surflex-Dock docking study was performed to investigate the topoisomerase I activity of all compounds. Of all the other compounds, the most sensitive compound was selected for further investigation of its effect on apoptosis induction and cell cycle regulation in HL-60 cells. Our results suggest that the anticancer effects of these compounds can be attributed to their pharmacological effects on topoisomerase I, cell apoptosis, and cell cycle. These findings suggest that robustic acid derivatives could be used as potential antitumor drugs.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Isoflavones/chemistry , Pyranocoumarins/chemical synthesis , Pyranocoumarins/pharmacology , A549 Cells , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cells, Cultured , DNA Topoisomerases, Type I/drug effects , Dalbergia/chemistry , Drug Screening Assays, Antitumor , HL-60 Cells , HeLa Cells , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Isoflavones/pharmacology , Molecular Docking Simulation , Molecular Structure , Pyranocoumarins/chemistry , Pyranocoumarins/therapeutic use , Topoisomerase I Inhibitors/chemical synthesis , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology
10.
Article in English | MEDLINE | ID: mdl-28507113

ABSTRACT

Leishmaniasis is a disease found throughout the (sub)tropical parts of the world caused by protozoan parasites of the Leishmania genus. Despite the numerous problems associated with existing treatments, pharmaceutical companies continue to neglect the development of better ones. The high toxicity of current drugs combined with emerging resistance makes the discovery of new therapeutic alternatives urgent. We report here the evaluation of a binuclear cyclopalladated complex containing Pd(II) and N,N'-dimethylbenzylamine (Hdmba) against Leishmania amazonensis The compound [Pd(dmba)(µ-N3)]2 (CP2) inhibits promastigote growth (50% inhibitory concentration [IC50] = 13.2 ± 0.7 µM) and decreases the proliferation of intracellular amastigotes in in vitro incubated macrophages (IC50 = 10.2 ± 2.2 µM) without a cytotoxic effect when tested against peritoneal macrophages (50% cytotoxic concentration = 506.0 ± 10.7 µM). In addition, CP2 was also active against T. cruzi intracellular amastigotes (IC50 = 2.3 ± 0.5 µM, selective index = 225), an indication of its potential for use in Chagas disease therapy. In vivo assays using L. amazonensis-infected BALB/c showed an 80% reduction in parasite load compared to infected and nontreated animals. Also, compared to amphotericin B treatment, CP2 did not show any side effects, which was corroborated by the analysis of plasma levels of different hepatic and renal biomarkers. Furthermore, CP2 was able to inhibit Leishmania donovani topoisomerase 1B (Ldtopo1B), a potentially important target in this parasite. (This study has been registered at ClinicalTrials.gov under identifier NCT02169141.).


Subject(s)
Antiprotozoal Agents/therapeutic use , Benzylamines/therapeutic use , Leishmania mexicana/drug effects , Leishmaniasis, Cutaneous/drug therapy , Palladium/therapeutic use , Topoisomerase I Inhibitors/therapeutic use , Amphotericin B/therapeutic use , Animals , Antiprotozoal Agents/adverse effects , Benzylamines/chemistry , Catalytic Domain/drug effects , Cells, Cultured , DNA Topoisomerases, Type I/drug effects , Disease Models, Animal , Kidney Function Tests , Leishmania mexicana/growth & development , Liver Function Tests , Macrophages, Peritoneal/drug effects , Male , Mice , Mice, Inbred BALB C , Neglected Diseases/drug therapy , Neglected Diseases/parasitology , Palladium/chemistry , Parasite Load , Parasitic Sensitivity Tests
11.
J Nat Prod ; 79(11): 2961-2967, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27933897

ABSTRACT

Tyrosyl-DNA phosphodiesterase 1 (TDP1) is a repair enzyme for stalled DNA-topoisomerase 1 (Top1) cleavage complexes and other 3'-end DNA lesions. TDP1 is a perspective target for anticancer therapy based on Top1-poison-mediated DNA damage. Several novel usnic acid derivatives with an enamine moiety have been synthesized and tested as inhibitors of TDP1. The enamines of usnic acid showed IC50 values in the range of 0.16 to 2.0 µM. These compounds revealed moderate cytotoxicity against human tumor MCF-7 cells. These new compounds enhanced the cytotoxicity of the established Top1 poison camptothecin by an order of magnitude.


Subject(s)
Camptothecin/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/drug effects , Benzofurans/metabolism , Benzofurans/pharmacology , DNA Topoisomerases, Type I/drug effects , DNA Topoisomerases, Type I/metabolism , Humans , MCF-7 Cells/drug effects , Models, Molecular , Molecular Structure
12.
BMC Complement Altern Med ; 15: 165, 2015 Jun 06.
Article in English | MEDLINE | ID: mdl-26048712

ABSTRACT

BACKGROUND: 3beta,6beta,16beta-trihydroxylup-20(29)-ene is a lupane triterpene isolated from Combretum leprosum fruit. The lupane group has been extensively used in studies on anticancer effects; however, its possible activity against protozoa parasites is yet poorly known. The high toxicity of the compounds currently used in leishmaniasis chemotherapy stimulates the investigation of new molecules and drug targets for antileishmanial therapy. METHODS: The activity of 3beta,6beta,16beta-trihydroxylup-20(29)-ene was evaluated against Leishmania (L.) amazonensis by determining the cytotoxicity of the compound on murine peritoneal macrophages, as well as its effects on parasite survival inside host cells. To evaluate the effect of this compound on intracellular amastigotes, cultures of infected macrophages were treated for 24, 48 and 96 h and the percentage of infected macrophages and the number of intracellular parasites was scored using light microscopy. RESULTS: Lupane showed significant activity against the intracellular amastigotes of L. (L.) amazonensis. The treatment with 109 µM for 96 h reduced in 80 % the survival index of parasites in BALB/c peritoneal macrophages. At this concentration, the triterpene caused no cytotoxic effects against mouse peritoneal macrophages. Ultrastructural analyses of L. (L.) amazonensis intracellular amastigotes showed that lupane induced some morphological changes in parasites, such as cytosolic vacuolization, lipid body formation and mitochondrial swelling. Bioinformatic analyses through molecular docking suggest that this lupane has high-affinity binding with DNA topoisomerase. CONCLUSION: Taken together, our results have showed that the lupane triterpene from C. leprosum interferes with L. (L.) amazonensis amastigote replication and survival inside vertebrate host cells and bioinformatics analyses strongly indicate that this molecule may be a potential inhibitor of topoisomerase IB. Moreover, this study opens major prospects for the development of novel chemotherapeutic agents with leishmanicidal activity.


Subject(s)
Combretum/chemistry , Leishmania mexicana/drug effects , Leishmaniasis/parasitology , Macrophages, Peritoneal/parasitology , Plant Extracts/pharmacology , Triterpenes/pharmacology , Animals , Cytoplasm/parasitology , DNA Topoisomerases, Type I/drug effects , Female , Fruit/chemistry , In Vitro Techniques , Leishmaniasis/drug therapy , Mice , Mice, Inbred BALB C , Phytotherapy , Plant Extracts/chemistry , Triterpenes/isolation & purification
13.
J Nat Prod ; 77(6): 1494-504, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24937209

ABSTRACT

Two new (1 and 2) and four known arylnaphthalene lignan lactones (3-6) were isolated from different plant parts of Phyllanthus poilanei collected in Vietnam, with two further known analogues (7 and 8) being prepared from phyllanthusmin C (4). The structures of the new compounds were determined by interpretation of their spectroscopic data and by chemical methods, and the structure of phyllanthusmin D (1) was confirmed by single-crystal X-ray diffraction analysis. Several of these arylnaphthalene lignan lactones were cytotoxic toward HT-29 human colon cancer cells, with compounds 1 and 7-O-[(2,3,4-tri-O-acetyl)-α-L-arabinopyranosyl)]diphyllin (7) found to be the most potent, exhibiting IC50 values of 170 and 110 nM, respectively. Compound 1 showed activity when tested in an in vivo hollow fiber assay using HT-29 cells implanted in immunodeficient NCr nu/nu mice. Mechanistic studies showed that this compound mediated its cytotoxic effects by inducing tumor cell apoptosis through activation of caspase-3, but it did not inhibit DNA topoisomerase IIα activity.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Lactones/isolation & purification , Lactones/pharmacology , Lignans/isolation & purification , Lignans/pharmacology , Naphthalenes/isolation & purification , Naphthalenes/pharmacology , Phyllanthus/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Benzodioxoles/chemistry , Caspase 3/drug effects , DNA Topoisomerases, Type I/drug effects , DNA Topoisomerases, Type I/metabolism , Drug Screening Assays, Antitumor , Glycosides/chemistry , HT29 Cells , Humans , Lactones/chemistry , Lignans/chemistry , Mice , Molecular Structure , Naphthalenes/chemistry , Nuclear Magnetic Resonance, Biomolecular , Vietnam
14.
Mar Drugs ; 12(2): 779-98, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24473175

ABSTRACT

Lamellarin D (LamD) is a marine alkaloid with broad spectrum antitumor activities. Multiple intracellular targets of LamD, which affect cancer cell growth and induce apoptosis, have been identified. These include nuclear topoisomerase I, relevant kinases (such as cyclin-dependent kinase 2) and the mitochondrial electron transport chain. While we have previously demonstrated that LamD at micromolar range deploys strong cytotoxicity by inducing mitochondrial apoptosis, mechanisms of its cytostatic effect have not yet been characterized. Here, we demonstrated that induction of cellular senescence (depicted by cell cycle arrest in G2 associated with ß-galactosidase activity) is a common response to subtoxic concentrations of LamD. Cellular senescence is observed in a large panel of cancer cells following in vitro or in vivo exposure to LamD. The onset of cellular senescence is dependent on the presence of intact topoisomerase I since topoisomerase I-mutated cells are resistant to senescence induced by LamD. LamD-induced senescence occurs without important loss of telomere integrity. Instead, incubation with LamD results in the production of intracellular reactive oxygen species (ROS), which are critical for senescence as demonstrated by the inhibitory effect of antioxidants. In addition, cancer cells lacking mitochondrial DNA also exhibit cellular senescence upon LamD exposure indicating that LamD can trigger senescence, unlike apoptosis, in the absence of functional mitochondria. Overall, our results identify senescence-associated growth arrest as a powerful effect of LamD and add compelling evidence for the pharmacological interest of lamellarins as potential anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Cellular Senescence/drug effects , Coumarins/pharmacology , DNA Topoisomerases, Type I/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Isoquinolines/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , DNA, Mitochondrial/metabolism , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Mice, SCID , Mitochondria/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Telomere/metabolism , Topoisomerase Inhibitors/pharmacology
15.
Postepy Hig Med Dosw (Online) ; 67: 130-42, 2013 Mar 04.
Article in Polish | MEDLINE | ID: mdl-23475489

ABSTRACT

The bacterial chromosome is composed of topologically independent domains, whose spatial organization is controlled by enzymes called topoisomerases. Topology maintenance is crucial in many important cellular processes such as replication, transcription and recombination. Moreover, the role of chromosome topology in adaptation of bacteria to environmental changes and, in the case of pathogenic strains, in their virulence was described. In recent years higher numbers of pathogenic strains resistant to antibiotic treatment have been noticed. In this paper we present the current state of knowledge about the structure and cellular functions of bacterial topoisomerases IA. In particular, we discuss the potential use of these enzymes as new targets for antibacterial compounds. 


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/enzymology , DNA Topoisomerases, Type I/drug effects , DNA Topoisomerases, Type I/metabolism , Bacteria/pathogenicity , Chromosomes/metabolism , DNA Topoisomerases, Type I/chemistry , Drug Resistance, Bacterial/drug effects , Gene Expression Regulation, Bacterial/drug effects , Molecular Structure , Virulence/drug effects
16.
Antimicrob Agents Chemother ; 56(2): 893-902, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22106228

ABSTRACT

The lytic DNA replication of Kaposi's sarcoma-associated herpesvirus (KSHV) initiates at an origin (ori-Lyt) and requires trans-acting elements, both viral and cellular. We recently demonstrated that several host cellular proteins, including topoisomerases I and II (Topo I and II), are involved in KSHV lytic DNA replication (Y. Wang, H. Li, Q. Tang, G. G. Maul, and Y. Yuan. J. Virol. 82: 2867-2882, 2008). To assess the importance of these topoisomerases in viral lytic replication, shRNA-mediated gene silencing was used. Depletion of Topo I and II severely inhibited viral lytic DNA replication as well as virion production, suggesting essential roles of these cellular proteins in viral DNA replication. The discovery of Topo I and II as enzymes indispensable for KSHV DNA replication raises a possibility that these cellular proteins could be new targets of therapeutic approaches to halt KSHV replication and treat KSHV-associated diseases. In this report, we examined one Topo I inhibitor and several Topo II inhibitors (inclusive of Topo II poison and catalytic inhibitors) as potential therapeutic agents for blocking KSHV replication. The Topo II catalytic inhibitors in general exhibited marked inhibition on KSHV replication and minimal cytotoxicity. In particular, novobiocin, with the best selectivity index (SI = 31.62) among the inhibitors tested in this study, is effective in inhibiting KSHV DNA replication and virion production but shows little adverse effect on cell proliferation and cycle progression in its therapeutic concentration, suggesting its potential to become an effective and safe drug for the treatment of human diseases associated with KSHV infection.


Subject(s)
Antigens, Neoplasm/drug effects , Antiviral Agents/pharmacology , DNA Topoisomerases, Type II/drug effects , DNA Topoisomerases, Type I/drug effects , DNA-Binding Proteins/drug effects , Enzyme Inhibitors/pharmacology , Herpesvirus 8, Human/drug effects , Virus Replication/drug effects , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Camptothecin/pharmacology , Cell Line , DNA Replication/drug effects , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Silencing , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Humans , Novobiocin/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Virion/metabolism
17.
Nat Commun ; 13(1): 842, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35149673

ABSTRACT

In natural product discovery programs, the power of synthetic chemistry is often leveraged for the total synthesis and diversification of characterized metabolites. The synthesis of structures that are bioinformatically predicted to arise from uncharacterized biosynthetic gene clusters (BGCs) provides a means for synthetic chemistry to enter this process at an early stage. The recent identification of non-ribosomal peptides (NRPs) containing multiple ρ-aminobenzoic acids (PABAs) led us to search soil metagenomes for BGCs that polymerize PABA. Here, we use PABA-specific adenylation-domain sequences to guide the cloning of the lap BGC directly from soil. This BGC was predicted to encode a unique N-acylated PABA and thiazole containing structure. Chemical synthesis of this structure gave lapcin, a dual topoisomerase I/II inhibitor with nM to pM IC50s against diverse cancer cell lines. The discovery of lapcin highlights the power of coupling metagenomics, bioinformatics and total chemical synthesis to unlock the biosynthetic potential contained in even complex uncharacterized BGCs.


Subject(s)
Biological Products/pharmacology , DNA Topoisomerases, Type II/drug effects , DNA Topoisomerases, Type I/drug effects , Enzyme Inhibitors/pharmacology , Metagenome , Biological Products/chemistry , Biological Products/isolation & purification , Biosynthetic Pathways/genetics , Cell Line , Cell Survival/drug effects , Computational Biology , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Humans , Metagenome/genetics , Metagenomics , Multigene Family , Soil
18.
Arch Biochem Biophys ; 516(2): 108-12, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22033340

ABSTRACT

A gold(III) compound [Au(C^N^C)(IMe)]CF(3)SO(3) (Gold III) has been reported to have anticancer properties as it is able to reduce topoisomerase IB activity in vitro and suppress tumor growth in nude mice model. Here we have investigated the mechanism of inhibition of human topoisomerase IB activity by this compound, analyzing the various steps of the catalytic cycle. DNA supercoiled relaxation and the cleavage reaction are inhibited, but Gold III does not perturb the religation reaction, in contrast to what has been observed for camptothecin. Pre-incubation of enzyme with the inhibitor before adding DNA substrate increases the inhibitory effect. In addition, when Gold III is preincubated with the enzyme it prevents the stabilization of the cleavable complex by camptothecin. The analysis of the DNA-topoisomerase binding reaction indicates that the compound acts as a topoisomerase I inhibitor by preventing the enzyme-DNA interaction.


Subject(s)
DNA Topoisomerases, Type I/drug effects , Organogold Compounds/pharmacology , Topoisomerase I Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalysis , DNA/metabolism , DNA Topoisomerases, Type I/metabolism , Electrophoretic Mobility Shift Assay , Humans , Kinetics , Mice , Organogold Compounds/chemistry , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Substrate Specificity , Topoisomerase I Inhibitors/chemistry
19.
Bioorg Med Chem ; 19(5): 1790-801, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21315610

ABSTRACT

New acridine derivatives bearing two symmetrical imidazolidinone rings, 3,6-bis((1-alkyl-5-oxo-imidazolidin-2-yliden)imino)acridine hydrochlorides 6a-6e (alkyl=ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl), have been prepared and their interactions with calf thymus DNA and selected cell lines were studied. The DNA-binding of 6a-6e to ctDNA was examined by UV-vis, fluorescence, and CD spectroscopy. The binding constants determined by UV-vis spectroscopy were found in the range 1.9×10(5)-7.1×10(5) M(-1). An electrophoretic separation proved that ligands 6a-6e inhibited topoisomerase I in 40 µM concentration although only those with longer alkyl chains were able to penetrate the membranes and efficiently suppress the cell proliferation. The highest activity in cytotoxic tests was found for 3,6-bis((1-n-hexyl-5-oxo-imidazolidin-2-yliden)imino)acridine hydrochloride (6e) with IC(50)=2.12 µM (HL 60) and 5.28 µM (L1210) after 72 h incubation. Molecular dynamics simulations and calculations of solvent-accessible surface areas (SASAs) were used to explore the intercalation mechanism. MD simulations favor stacking between adjacent C:G base pairs from the minor groove side. MD and SASAs calculations indicate that the decrease of K with alkyl extension is due to negative entropic change upon binding.


Subject(s)
Acridines/chemical synthesis , DNA Topoisomerases, Type I/drug effects , Imidazolidines/chemical synthesis , Acridines/chemistry , Acridines/pharmacology , Animals , Cell Line, Tumor , Circular Dichroism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Imidazolidines/chemistry , Imidazolidines/pharmacology , Mice , Models, Molecular , Molecular Dynamics Simulation , Molecular Structure , Thermodynamics
20.
Bioorg Med Chem ; 19(16): 4971-84, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21783369

ABSTRACT

A novel 5-oxa-6a,8-diazaindeno[2,1-b]phenanthren-7-one scaffold was designed and synthesized as an active analogue of the cytotoxic marine alkaloid Lamellarin D. The design was based on molecular modeling of the site of interaction of Lamellarin D with DNA-topoisomerase I cleavable complex, whereas the synthesis capitalized on a simple Friedel-Crafts cyclization of indole to a ß-carbolinone nucleus. The product exhibited topoisomerase I poisoning activity and submicromolar cytotoxicity on human non-small cell lung cancer H460 cell line.


Subject(s)
Alkaloids/chemical synthesis , Antineoplastic Agents/chemical synthesis , Coumarins/chemistry , DNA Topoisomerases, Type I/drug effects , Heterocyclic Compounds, 4 or More Rings/chemistry , Isoquinolines/chemistry , Topoisomerase I Inhibitors/chemical synthesis , Alkaloids/chemistry , Alkaloids/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Coumarins/pharmacology , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Isoquinolines/pharmacology , Models, Molecular , Mollusca , Oceans and Seas , Simian virus 40/drug effects , Simian virus 40/genetics , Structure-Activity Relationship , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL