Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 286
Filtrar
1.
Biochem Biophys Res Commun ; 594: 57-62, 2022 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-35074586

RESUMO

DNA-damaging agents, such as radiation and chemotherapy, are common in cancer treatment, but the dosing has proven to be challenging, leading to severe side effects in some patients. Hence, to be able to personalize DNA-damaging chemotherapy, it is important to develop fast and reliable methods to measure the resulting DNA damage in patient cells. Here, we demonstrate how single DNA molecule imaging using fluorescence microscopy can quantify DNA-damage caused by the topoisomerase II (TopoII) poison etoposide. The assay uses an enzyme cocktail consisting of base excision repair (BER) enzymes to repair the DNA damage caused by etoposide and label the sites using a DNA polymerase and fluorescently labeled nucleotides. Using this DNA-damage detection assay we find a large variation in etoposide induced DNA-damage after in vitro treatment of blood cells from healthy individuals. We furthermore used the TopoII inhibitor ICRF-193 to show that the etoposide-induced damage in DNA was TopoII dependent. We discuss how our results support a potential future use of the assay for personalized dosing of chemotherapy.


Assuntos
Dano ao DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA de Cadeia Simples/efeitos dos fármacos , DNA de Cadeia Simples/genética , Dicetopiperazinas/farmacologia , Etoposídeo/farmacologia , Imagem Individual de Molécula , Antineoplásicos Fitogênicos/farmacologia , DNA/efeitos dos fármacos , Reparo do DNA , Relação Dose-Resposta a Droga , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Microscopia de Fluorescência , Inibidores da Topoisomerase II/farmacologia
2.
Genes Cells ; 25(11): 718-729, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32939879

RESUMO

Ring finger protein 43 (RNF43) is an E3 ubiquitin ligase which is well-known for its role in negative regulation of the Wnt-signaling pathway. However, the function in DNA double-strand break repairs has not been investigated. In this study, we used a lymphoblast cell line, DT40, and mouse embryonic fibroblast as cellular models to study DNA double-strand break (DSB) repairs. For this purpose, we created RNF43 knockout, RNF43-/- DT40 cell line to investigate DSB repairs. We found that deletion of RNF43 does not interfere with cell proliferation. However, after exposure to various types of DNA-damaging agents, RNF43-/- cells become more sensitive to topoisomerase II inhibitors, etoposide, and ICRF193, than wild type cells. Our results also showed that depletion of RNF43 results in apoptosis upon etoposide-mediated DNA damage. The delay in resolution of γH2AX and 53BP1 foci formation after etoposide treatment, as well as epistasis analysis with DNAPKcs, suggested that RNF43 might participate in DNA repair of etoposide-induced DSB via non-homologous end joining. Disturbed γH2AX foci formation in MEFs following pulse etoposide treatment supported the notion that RNF43 also functions DNA repair in mammalian cells. These findings propose two possible functions of RNF43, either participating in NHEJ or removing the blockage of 5' topo II adducts from DSB ends.


Assuntos
Reparo do DNA/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Etoposídeo/efeitos adversos , Etoposídeo/farmacologia , Técnicas de Inativação de Genes/métodos , Camundongos , Proteínas Oncogênicas/genética , Recombinação Genética/efeitos dos fármacos , Inibidores da Topoisomerase II/farmacologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/fisiologia
3.
Arch Toxicol ; 95(12): 3787-3802, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34635930

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , DNA Topoisomerases Tipo I/efeitos dos fármacos , Flavonas/intoxicação , Ciclo Celular/efeitos dos fármacos , Neoplasias do Colo/enzimologia , DNA Topoisomerases Tipo II/efeitos dos fármacos , Relação Dose-Resposta a Droga , Flavonas/administração & dosagem , Células HT29 , Humanos , Concentração Inibidora 50 , Proteínas de Ligação a Poli-ADP-Ribose/efeitos dos fármacos , Fatores de Tempo
4.
Nucleic Acids Res ; 47(12): 6172-6183, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31106359

RESUMO

Topoisomerase II (Top2) is an essential enzyme that decatenates DNA via a transient Top2-DNA covalent intermediate. This intermediate can be stabilized by a class of drugs termed Top2 poisons, resulting in massive DNA damage. Thus, Top2 activity is a double-edged sword that needs to be carefully controlled to maintain genome stability. We show that Uls1, an adenosine triphosphate (ATP)-dependent chromatin remodelling (Snf2) enzyme, can alter Top2 chromatin binding and prevent Top2 poisoning in yeast. Deletion mutants of ULS1 are hypersensitive to the Top2 poison acriflavine (ACF), activating the DNA damage checkpoint. We map Uls1's Top2 interaction domain and show that this, together with its ATPase activity, is essential for Uls1 function. By performing ChIP-seq, we show that ACF leads to a general increase in Top2 binding across the genome. We map Uls1 binding sites and identify tRNA genes as key regions where Uls1 associates after ACF treatment. Importantly, the presence of Uls1 at these sites prevents ACF-dependent Top2 accumulation. Our data reveal the effect of Top2 poisons on the global Top2 binding landscape and highlights the role of Uls1 in antagonizing Top2 function. Remodelling Top2 binding is thus an important new means by which Snf2 enzymes promote genome stability.


Assuntos
DNA Helicases/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Acriflavina/toxicidade , DNA Helicases/química , DNA Helicases/genética , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Fúngico/metabolismo , Deleção de Genes , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
5.
Bioorg Med Chem Lett ; 29(15): 1881-1885, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31182315

RESUMO

A number of natural products with medicinal properties increase DNA cleavage mediated by type II topoisomerases. In an effort to identify additional natural compounds that affect the activity of human type II topoisomerases, a blind screen of a library of 341 Mediterranean plant extracts was conducted. Extracts from Nuphar lutea, the yellow water lily, were identified in this screen. N. lutea has been used in traditional medicine by a variety of indigenous populations. The active compound in N. lutea, 6,6'-dihydroxythiobinupharidine, was found to enhance DNA cleavage mediated by human topoisomerase IIα and IIß âˆ¼8-fold and ∼3-fold, respectively. Mechanistic studies with topoisomerase IIα indicate that 6,6'-dihydroxythiobinupharidine is a "covalent poison" that acts by adducting the enzyme outside of the DNA cleavage-ligation active site and requires the N-terminal domain of the protein for its activity. Results suggest that some of the medicinal properties of N. lutea may result from the interactions between 6,6'-dihydroxythiobinupharidine and the human type II enzymes.


Assuntos
Alcaloides/efeitos adversos , DNA Topoisomerases Tipo II/efeitos dos fármacos , Extratos Vegetais/efeitos adversos , Humanos , Venenos
6.
Bioorg Med Chem Lett ; 29(23): 126714, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31635931

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo I/efeitos dos fármacos , Metilaminas/síntese química , Simulação de Acoplamento Molecular/métodos , Humanos , Metilaminas/química , Estrutura Molecular , Relação Estrutura-Atividade
7.
Bioorg Chem ; 92: 103109, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31521987

RESUMO

New series of fused pyrazolopyridines were prepared and assessed for antimicrobial, antiquorum-sensing and antitumor activities. Antimicrobial evaluation toward selected Gram-positive bacteria, Gram-negative bacteria and fungi indicated that 5-phenylpyrazolopyridotriazinone 4a has good and broad-spectrum antimicrobial activity. In addition, 5-(4-chlorophenyl)pyrazolopyridotriazinone 4b and 5-(4-(dimethylamino)phenyl)pyrazolopyridotriazinone 4c exhibited good activity against the selected Gram-positive bacteria and A. fumigatus, whereas 5-amino-4-phenylpyrazolopyridopyrimidine 6a demonstrated good activity against B. cereus and P. aeruginosa. Furthermore, 6-amino-5-imino-4-phenylpyrazolopyridopyrimidine 7a and 6-amino-4-(4-chlorophenyl)-5-iminopyrazolopyridopyrimidine 7b demonstrated promising activity against the tested Gram-negative bacteria and fungi, and moderate activity against Gram-positive bacteria. Antiquorum-sensing screening over C. violaceum illustrated that 4a, 6a and 7a-c have strong activity. In vitro antiproliferative assessment of the new derivatives against HepG2, HCT-116 and MCF-7 cancer cells revealed that 7a is the most active analog against all tested cell lines. Likewise, 3,7-dimethyl-4-phenylpyrazolopyridopyrimidinone 2a and 6-amino-4-(4-chlorophenyl)-5-iminopyrazolopyridopyrimidine 7b manifested strong activity against all examined cell lines. In vivo antitumor testing of 2a, 7a and 7b against EAC cells in mice indicated that 7a has the highest activity. Cytotoxicity toward WI38 and WISH normal cells was also assessed and results assured that all of the investigated analogs have lower cytotoxicity than doxorubicin. DNA-binding affinity and topoisomerase IIß inhibitory activity were evaluated, and results revealed that 5b, 7a and 7b bind strongly to DNA; in addition, 2a, 4a, 7a and 7b manifested higher topoisomerase IIß inhibitory activity than that of doxorubicin. Analogs 5b, 7a and 7b were docked into topoisomerase IIß, and results indicated that 7a and 7b have the highest binding affinity toward topoisomerase IIß. In silico simulation studies referred that most of the new analogs comply with the optimum needs for good oral absorption. Also, computational carcinogenicity evaluation was predicted.


Assuntos
Anti-Infecciosos/síntese química , Antineoplásicos/síntese química , Pirazóis/síntese química , Piridinas/síntese química , Animais , Anti-Infecciosos/farmacologia , Antineoplásicos/farmacologia , Bactérias/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Simulação por Computador , DNA/química , DNA Topoisomerases Tipo II/efeitos dos fármacos , Proteínas de Ligação a DNA/química , Avaliação Pré-Clínica de Medicamentos , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Pirazóis/farmacologia , Piridinas/farmacologia , Relação Estrutura-Atividade
8.
Nucleic Acids Res ; 45(10): 5995-6010, 2017 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-28472494

RESUMO

Topoisomerase (topo) IIα and IIß maintain genome stability and are targets for anti-tumor drugs. In this study, we demonstrate that the decatenation checkpoint is regulated, not only by topo IIα, as previously reported, but also by topo IIß. The decatenation checkpoint is most efficient when both isoforms are present. Regulation of this checkpoint and sensitivity to topo II-targeted drugs is influenced by the C-terminal domain (CTD) of the topo II isoforms and by a conserved non-catalytic tyrosine, Y640 in topo IIα and Y656 in topo IIß. Deletion of most of the CTD of topo IIα, while preserving the nuclear localization signal (NLS), enhances the decatenation checkpoint and sensitivity to topo II-targeted drugs. In contrast, deletion of most of the CTD of topo IIß, while preserving the NLS, and mutation of Y640 in topo IIα and Y656 in topo IIß inhibits these activities. Structural studies suggest that the differential impact of the CTD on topo IIα and topo IIß function may be due to differences in CTD charge distribution and differential alignment of the CTD with reference to transport DNA. Together these results suggest that topo IIα and topo IIß cooperate to maintain genome stability, which may be distinctly modulated by their CTDs.


Assuntos
Antígenos de Neoplasias/química , Pontos de Checagem do Ciclo Celular/fisiologia , Instabilidade Cromossômica/fisiologia , DNA Topoisomerases Tipo II/química , Proteínas de Ligação a DNA/química , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/efeitos dos fármacos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/fisiologia , Linhagem Celular , Dano ao DNA , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/genética , DNA Topoisomerases Tipo II/fisiologia , DNA Complementar/genética , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Resistencia a Medicamentos Antineoplásicos , Fibroblastos , Células HL-60 , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Domínios Proteicos , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Inibidores da Topoisomerase II/farmacologia
9.
Arch Pharm (Weinheim) ; 351(6): e1800023, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29737542

RESUMO

The design and synthesis of dihydropyrazolo[1,5-c]quinazolines (1a-h) as human topoisomerase II (TopoII) catalytic inhibitors are reported. The compounds were investigated for their antiproliferative activity against the C6 rat glial cell line. Two compounds, 1b and 1h, were found to be potent cytotoxic agents against glioma cells and exerted selective TopoII inhibitory activity. Furthermore, the compounds induced alterations in reactive oxygen species levels as measured by DCFDA assay and were found to induce cell cycle arrest at the G1 phase at lower concentrations and profound apoptosis at higher concentrations. The interaction of selected investigational molecules with TopoII was further corroborated by molecular modeling.


Assuntos
Antineoplásicos/farmacologia , Glioma/tratamento farmacológico , Quinazolinas/farmacologia , Inibidores da Topoisomerase II/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Relação Dose-Resposta a Droga , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Glioma/enzimologia , Humanos , Modelos Moleculares , Quinazolinas/síntese química , Quinazolinas/química , Ratos , Espécies Reativas de Oxigênio/metabolismo , Inibidores da Topoisomerase II/síntese química , Inibidores da Topoisomerase II/química
10.
Arch Toxicol ; 91(4): 2007-2016, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27682608

RESUMO

Type II DNA-topoisomerases (topo II) play a crucial role in the maintenance of DNA topology. Previously, fungi of the Alternaria genus were found to produce mycotoxins that target human topo II. These results implied the question why a fungus should produce secondary metabolites that target a human enzyme. In the current work, the homology between human topo II and its bacterial equivalent, gyrase, served as basis to study a potential dual inhibition of both enzymes by mycotoxins. A total of 15 secondary metabolites produced by fungi of the genera Alternaria and Fusarium were assessed for their impact on topo II of human and bacterial origin in the decatenation and the supercoiling assay, respectively. In line with the theory of dual topo II inhibition, six of the tested Alternaria mycotoxins were active against both enzymes, the dibenzo-α-pyrones alternariol (AOH) and alternariol monomethyl ether (AME), as well as the perylene-quinones altertoxin I (ATX I) and II (ATX II), alterperylenol (ALP) and stemphyltoxin III (STTX III). The Alternaria metabolites altersetin (ALN), macrosporin (MAC), altenusine (ALS) and pyrenophorol (PYR) impaired the function of human topo II, but did not show any effect on gyrase. The potency to inhibit topo II activity declined in the row STTX III (initial inhibitory concentration 10 µM) > AOH (25 µM) = AME (25 µM) = ALS (25 µM) = ATX II (25 µM) > ALN (50 µM) = ATX I (50 µM) > ALP (75 µM) = PYR (75 µM) > MAC (150 µM). Inhibition of gyrase activity was most pronounced for AOH and AME (initial inhibitory concentration 10 µM) followed by ATX II (25 µM) > ATX I = ALP = STTX III (50 µM). In contrast, none of the investigated Fusarium mycotoxins deoxynivalenol (DON), fumonisin B1, fusarin C and moniliformin, as well as the Alternaria metabolite tentoxin, had any impact on the activity of neither human nor bacterial topo II.


Assuntos
Alternaria/metabolismo , Fusarium/metabolismo , Micotoxinas/farmacologia , Inibidores da Topoisomerase II/farmacologia , DNA Girase/efeitos dos fármacos , DNA Girase/metabolismo , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Relação Dose-Resposta a Droga , Humanos , Micotoxinas/administração & dosagem , Micotoxinas/isolamento & purificação , Metabolismo Secundário , Inibidores da Topoisomerase II/administração & dosagem , Inibidores da Topoisomerase II/isolamento & purificação
11.
J Biol Chem ; 290(33): 20313-24, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26055707

RESUMO

Historically, type II topoisomerases have yielded clinically useful drugs for the treatment of bacterial infections and cancer, but the corresponding enzymes from malaria parasites remain understudied. This is due to the general challenges of producing malaria proteins in functional forms in heterologous expression systems. Here, we express full-length Plasmodium falciparum topoisomerase II (PfTopoII) in a wheat germ cell-free transcription-translation system. Functional activity of soluble PfTopoII from the translation lysates was confirmed through both a plasmid relaxation and a DNA decatenation activity that was dependent on magnesium and ATP. To facilitate future drug discovery, a convenient and sensitive fluorescence assay was established to follow DNA decatenation, and a stable, truncated PfTopoII was engineered for high level enzyme production. PfTopoII was purified using a DNA affinity column. Existing TopoII inhibitors previously developed for other non-malaria indications inhibited PfTopoII, as well as malaria parasites in culture at submicromolar concentrations. Even before optimization, inhibitors of bacterial gyrase, GSK299423, ciprofloxacin, and etoposide exhibited 15-, 57-, and 3-fold selectivity for the malarial enzyme over human TopoII. Finally, it was possible to use the purified PfTopoII to dissect the different modes by which these varying classes of TopoII inhibitors could trap partially processed DNA. The present biochemical advancements will allow high throughput chemical screening of compound libraries and lead optimization to develop new lines of antimalarials.


Assuntos
DNA Topoisomerases Tipo II/metabolismo , Plasmodium falciparum/enzimologia , Inibidores da Topoisomerase II/farmacologia , Sequência de Aminoácidos , Animais , Sistema Livre de Células , DNA Topoisomerases Tipo II/química , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/isolamento & purificação , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Triticum/genética
12.
Cytometry A ; 89(9): 852-60, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27517472

RESUMO

Topoisomerase II (Top2) is an important target for anticancer therapy. A variety of drugs that poison Top2, including several epipodophyllotoxins, anthracyclines, and anthracenediones, are widely used in the clinic for both hematologic and solid tumors. The poisoning of Top2 involves the formation of a reaction intermediate Top2-DNA, termed Top2 cleavage complex (Top2cc), which is persistent in the presence of the drug and involves a 5' end of DNA covalently bound to a tyrosine from the enzyme through a phosphodiester group. Drug-induced Top2cc leads to Top2 linked-DNA breaks which are the major responsible for their cytotoxicity. While biochemical detection is very laborious, quantification of drug-induced Top2cc by immunofluorescence-based microscopy techniques is time consuming and requires extensive image segmentation for the analysis of a small population of cells. Here, we developed a flow cytometry-based method for the analysis of drug-induced Top2cc. This method allows a rapid analysis of a high number of cells in their cell cycle phase context. Moreover, it can be applied to almost any human cell type, including clinical samples. The methodology is useful for a high-throughput analysis of drugs that poison Top2, allowing not just the discrimination of the Top2 isoform that is targeted but also to track its removal. © 2016 International Society for Advancement of Cytometry.


Assuntos
DNA Topoisomerases Tipo II/isolamento & purificação , Proteínas de Ligação a DNA/isolamento & purificação , Citometria de Fluxo/métodos , Neoplasias/tratamento farmacológico , Inibidores da Topoisomerase II/química , Antraquinonas/química , Antraquinonas/isolamento & purificação , Antraquinonas/uso terapêutico , Dano ao DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/química , DNA Topoisomerases Tipo II/efeitos dos fármacos , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Ensaios de Triagem em Larga Escala , Humanos , Substâncias Macromoleculares/química , Substâncias Macromoleculares/isolamento & purificação , Inibidores da Topoisomerase II/isolamento & purificação , Inibidores da Topoisomerase II/uso terapêutico
13.
Molecules ; 20(4): 6254-72, 2015 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-25859782

RESUMO

We have synthesized and examined the antibacterial activity, toxicity and affinity towards bacterial type II topoisomerases of a series of 1,2,4-triazole-ciprofloxacin hybrids. A number of these compounds displayed enhanced activity against Gram-positive and Gram-negative bacteria when compared to ciprofloxacin. The toxic concentrations of the obtained derivatives, evaluated on HEK-293 cells using MTT assay, were much higher than concentrations required to produce antibacterial effect. Finally, the results of enzymatic studies showed that the analyzed compounds demonstrated other preferences as regards primary and secondary molecular targets than ciprofloxacin.


Assuntos
Antibacterianos/farmacologia , Ciprofloxacina/farmacologia , Inibidores da Topoisomerase II/farmacologia , Triazóis/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Ciprofloxacina/síntese química , Ciprofloxacina/química , DNA Topoisomerases Tipo II/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Células HEK293 , Humanos , Relação Estrutura-Atividade , Inibidores da Topoisomerase II/síntese química , Inibidores da Topoisomerase II/química , Triazóis/síntese química , Triazóis/química
14.
Mar Drugs ; 12(9): 4833-50, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25244109

RESUMO

Recently, we characterized neoamphimedine (neo) as an ATP-competitive inhibitor of the ATPase domain of human Topoisomerase IIα. Thus far, neo is the only pyridoacridine with this mechanism of action. One limiting factor in the development of neo as a therapeutic agent has been access to sufficient amounts of material for biological testing. Although there are two reported syntheses of neo, both require 12 steps with low overall yields (≤6%). In this article, we report an improved total synthesis of neo achieved in 10 steps with a 25% overall yield. In addition, we report an expanded cytotoxicity study using a panel of human cancer cell lines, including: breast, colorectal, lung, and leukemia. Neo displays potent cytotoxicity (nM IC50 values) in all, with significant potency against colorectal cancer (lowest IC50 = 6 nM). We show that neo is cytotoxic not cytostatic, and that neo exerts cytotoxicity by inducing G2-M cell cycle arrest and apoptosis.


Assuntos
Acridinas/síntese química , Acridinas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Inibidores da Topoisomerase II/síntese química , Inibidores da Topoisomerase II/farmacologia , Fosfatase Ácida/metabolismo , Antígenos de Neoplasias/efeitos dos fármacos , Antígenos de Neoplasias/metabolismo , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Fase G2/efeitos dos fármacos , Humanos , Modelos Moleculares , Rodaminas/química
15.
FEBS Open Bio ; 14(6): 1001-1010, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38531625

RESUMO

Myeloperoxidase (MPO) is found almost exclusively in granulocytes and immature myeloid cells. It plays a key role in the innate immune system, catalysing the formation of reactive oxygen species that are important in anti-microbial action, but MPO also oxidatively transforms the topoisomerase II (TOP2) poison etoposide to chemical forms that have elevated DNA damaging properties. TOP2 poisons such as etoposide are widely used anti-cancer drugs, but they are linked to cases of secondary acute myeloid leukaemias through a mechanism that involves DNA damage and presumably erroneous repair leading to leukaemogenic chromosome translocations. This leads to the possibility that myeloperoxidase inhibitors could reduce the rate of therapy-related leukaemia by protecting haematopoietic cells from TOP2 poison-mediated genotoxic damage while preserving the anti-cancer efficacy of the treatment. We show here that myeloperoxidase inhibition reduces etoposide-induced TOP2B-DNA covalent complexes and resulting DNA double-strand break formation in primary ex vivo expanded CD34+ progenitor cells and unfractionated bone marrow mononuclear cells. Since MPO inhibitors are currently being developed as anti-inflammatory agents this raises the possibility that repurposing of these potential new drugs could provide a means of suppressing secondary acute myeloid leukaemias associated with therapies containing TOP2 poisons.


Assuntos
Dano ao DNA , DNA Topoisomerases Tipo II , Etoposídeo , Peroxidase , Proteínas de Ligação a Poli-ADP-Ribose , Humanos , Antineoplásicos/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Etoposídeo/farmacologia , Etoposídeo/toxicidade , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Peroxidase/antagonistas & inibidores , Peroxidase/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/antagonistas & inibidores , Inibidores da Topoisomerase II/farmacologia
16.
J Biol Chem ; 287(46): 38590-9, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23012366

RESUMO

To investigate the potency of the topoisomerase II (topo II) poisons doxorubicin and etoposide to stimulate the DNA damage response (DDR), S139 phosphorylation of histone H2AX (γH2AX) was analyzed using rat cardiomyoblast cells (H9c2). Etoposide caused a dose-dependent increase in the γH2AX level as shown by Western blotting. By contrast, the doxorubicin response was bell-shaped with high doses failing to increase H2AX phosphorylation. Identical results were obtained by immunohistochemical analysis of γH2AX focus formation, comet assay-based DNA strand break analysis, and measuring the formation of the topo II-DNA cleavable complex. At low dose, doxorubicin activated ataxia telangiectasia mutated (ATM) but not ATM and Rad3-related (ATR). Both the lipid-lowering drug lovastatin and the Rac1-specific inhibitor NSC23766 attenuated doxorubicin- and etoposide-stimulated H2AX phosphorylation, induction of DNA strand breaks, and topo II-DNA complex formation. Lovastatin and NSC23766 acted in an additive manner. They did not attenuate doxorubicin-induced increase in p-ATM and p-Chk2 levels. DDR stimulated by topo II poisons was partially blocked by inhibition of type I p21-associated kinases. DDR evoked by the topoisomerase I poison topotecan remained unaffected by lovastatin. The data show that the mechanisms involved in DDR stimulated by topo II poisons are agent-specific with anthracyclines lacking DDR-stimulating activity at high doses. Pharmacological inhibition of Rac1 signaling counteracts doxorubicin- and etoposide-stimulated DDR by disabling the formation of the topo II-DNA cleavable complex. Based on the data we suggest that Rac1-regulated mechanisms are required for DNA damage induction and subsequent activation of the DDR following treatment with topo II but not topo I poisons.


Assuntos
Dano ao DNA , DNA Topoisomerases Tipo II/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Antineoplásicos/farmacologia , Morte Celular , Linhagem Celular , DNA Topoisomerases Tipo I/metabolismo , DNA Topoisomerases Tipo II/efeitos dos fármacos , Histonas/química , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Imuno-Histoquímica/métodos , Neoplasias/tratamento farmacológico , Venenos/química , Ratos , Transdução de Sinais , Inibidores da Topoisomerase II/farmacologia , Proteínas rac1 de Ligação ao GTP/química , Proteínas rho de Ligação ao GTP/metabolismo
17.
Antimicrob Agents Chemother ; 56(2): 893-902, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22106228

RESUMO

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.


Assuntos
Antígenos de Neoplasias/efeitos dos fármacos , Antivirais/farmacologia , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo I/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Herpesvirus Humano 8/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Camptotecina/farmacologia , Linhagem Celular , Replicação do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo I/genética , DNA Topoisomerases Tipo I/metabolismo , DNA Topoisomerases Tipo II/genética , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Inativação Gênica , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Humanos , Novobiocina/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Vírion/metabolismo
18.
Biol Reprod ; 86(3): 96, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22190700

RESUMO

Doxorubicin (DXR) is a frontline chemotherapy agent implicated in unintended ovarian failure in female cancer survivors. The fertility preservation techniques currently available for cancer patients are often time and cost prohibitive and do not necessarily preserve endocrine function. There are no drug-based ovary protection therapies clinically available. This study provides the first investigation using dexrazoxane (Dexra) to limit DXR insult in ovarian tissue. In KK-15 granulosa cells, a 3-h DXR treatment increased double-strand (ds) DNA breaks 40%-50%, as quantified by the neutral comet assay, and dose-dependent cytotoxicity. Dexra exhibited low toxicity in KK-15 cells, inducing no DNA damage and less than 20% cell loss. Cotreating KK-15 cells with Dexra prevented acute DXR-induced dsDNA damage. Similarly, Dexra attenuated the DXR-induced 40%-65% increase in dsDNA breaks in primary murine granulosa cells and cells from in vitro cultured murine ovaries. DXR can cause DNA damage either through a topoisomerase II-mediated pathway, based on DXR intercalation into DNA, or through oxidative stress. Cotreating KK-15 cells with 2 µM Dexra was sufficient to prevent DXR-induced, but not H(2)O(2)-induced, DNA damage. These data indicated the protective effects are likely due to Dexra's inhibition of topoisomerase II catalytic activity. This putative protective agent attenuated downstream cellular responses to DXR, preventing H2AFX activation in KK-15 cells and increasing viability as demonstrated by increasing the DXR lethal dose in KK-15 cells 5- to 8-fold (LD(20)) and primary murine granulosa cells 1.5- to 2-fold (LD(50)). These data demonstrate Dexra protects ovarian cells from DXR insult and suggest that it is a promising tool to limit DXR ovarian toxicity in vivo.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Doxorrubicina/efeitos adversos , Infertilidade Feminina/prevenção & controle , Ovário/efeitos dos fármacos , Razoxano/farmacologia , Animais , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/fisiologia , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Feminino , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Histonas/antagonistas & inibidores , Histonas/fisiologia , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Endogâmicos , Modelos Animais , Ovário/citologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia
19.
Cancer Cell ; 5(3): 275-85, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15050919

RESUMO

Chronic myelogeneous leukemia (CML) is a two-stage disease associated with expression of the BCR/ABL tyrosine kinase protein. However, whether BCR/ABL expression directly causes blast crisis, and if so by what mechanism, is unknown. We have found that BCR/ABL translocates from the cytoplasm to the nucleus after genotoxic stress. Furthermore, BCR/ABL increases DNA double-strand damage after etoposide treatment and leads to a defect in an intra-S phase checkpoint, causing a radioresistant DNA synthesis (RDS) phenotype. In the nucleus, BCR/ABL associates with the ataxia-telangiectasia and rad 3-related protein (ATR) and disrupts ATR-dependent signal transduction. Overexpression of ATR in a BCR/ABL-expressing cell line corrects the DNA damage phenotype. These results demonstrate a nuclear role for BCR/ABL in altering the cellular response to DNA damage.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Proteínas de Fusão bcr-abl/metabolismo , Proteínas Serina-Treonina Quinases , Fase S/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Células Cultivadas , Citoplasma/metabolismo , Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , DNA Topoisomerases Tipo II/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/fisiopatologia , Transporte Proteico/fisiologia , Transdução de Sinais/fisiologia
20.
Nat Commun ; 13(1): 842, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149673

RESUMO

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.


Assuntos
Produtos Biológicos/farmacologia , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo I/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Metagenoma , Produtos Biológicos/química , Produtos Biológicos/isolamento & purificação , Vias Biossintéticas/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Biologia Computacional , DNA Topoisomerases Tipo I/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/isolamento & purificação , Humanos , Metagenoma/genética , Metagenômica , Família Multigênica , Solo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA