Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
Chem Biol Interact ; 138(1): 59-75, 2001 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-11640915

RESUMO

Absorption, melting temperature and linear dichroism measurements were performed to investigate the interaction with DNA of a series of 16 tricyclic and tetracyclic compounds related to the antiviral agent B-220. The relative DNA affinity of the test compounds containing an indolo[2,3-b]quinoxaline, pyridopyrazino[2,3-b]indoles or pyrazino[2,3-b]indole planar chromophore varies significantly depending on the nature of the side chain grafted onto the indole nitrogen. Compounds with a dimethylaminoethyl chain strongly bind to DNA and exhibit a preference for GC-rich DNA sequences, as revealed by DNase I footprinting. Weaker DNA interactions were detected with those bearing a morpholinoethyl side chain. The incorporation of a 2,3-dihydroxypropyl side chain does not reinforce the DNA interaction compared with the unsubstituted analogues. Both the DNA relaxation assay and cytotoxicity study using two human leukemia cell lines sensitive (HL-60) or resistant (HL-60/MX2) to the antitumor drug mitoxantrone, indicate that topoisomerase II is not a privileged target for the test compounds which only weakly interfere with the catalytic activity of the DNA cleaving enzyme. Cytometry studies showed that the most cytotoxic compounds induce a massive accumulation of cells in the G2/M phase of the cell cycle. Collectively, the data show a relationship between DNA binding and cytotoxicity in the indolo[2,3-b]quinoxaline series.


Assuntos
DNA/química , Indóis/química , Quinoxalinas/química , Animais , Bovinos , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Pegada de DNA , Relação Dose-Resposta a Droga , Citometria de Fluxo , Células HL-60/efeitos dos fármacos , Humanos , Indóis/farmacologia , Substâncias Intercalantes/química , Quinoxalinas/farmacologia
2.
Biochemistry ; 40(15): 4663-71, 2001 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-11294633

RESUMO

The brominated anilinoquinazoline derivative PD153035 exhibits a very high affinity and selectivity for the epidermal growth factor receptor tyrosine kinase (EGF-R TK) and shows a remarkable cytotoxicity against several types of tumor cell lines. In contrast, its N-methyl derivative, designated EBE-A22, has no effect on EGF-R TK but maintains a high cytotoxic profile. The present study was performed to explore the possibility that PD153035 and its N-methyl analogue might interact with double-stranded DNA, which is a primary target for many conventional antitumor agents. We studied the strength and mode of binding to DNA of PD153035 and EBE-A22 by means of absorption, fluorescence, and circular and linear dichroism as well as by a relaxation assay using human DNA topoisomerases. The results of various optical and gel electrophoresis techniques converge to show that both drugs bind to DNA and behave as typical intercalating agents. In particular, EBE-A22 unwinds supercoiled plasmid, stabilizes duplex DNA against heat denaturation, and produces negative CD and ELD signals, as expected for an intercalating agent. Extensive DNase I footprinting experiments performed with a large range of DNA substrates show that EBE-A22, but not PD153035, interacts preferentially with GC-rich sequences and discriminates against homooligomeric runs of A and T which are often cut more readily by the enzyme in the presence of the drug compared to the control. Altogether, the results provide the first experimental evidence that DNA is a target of anilinoquinazoline derivatives and suggest that this N-methylated ring system is a valid candidate for the development of DNA-targeted cytotoxic compounds. The possible relevance of selective DNA binding to activity is considered. The unexpected GC-selective binding properties of EBE-A22 entreat further exploration into the use of N-methylanilinoquinazoline derivatives as tools for designing sequence-specific DNA binding ligands.


Assuntos
DNA/metabolismo , Inibidores Enzimáticos/metabolismo , Receptores ErbB/antagonistas & inibidores , Substâncias Intercalantes/metabolismo , Quinazolinas/metabolismo , Antineoplásicos/metabolismo , Sequência de Bases , Sítios de Ligação , Dicroísmo Circular , Pegada de DNA , DNA Super-Helicoidal/metabolismo , Densitometria , Eletroforese em Gel de Ágar , Humanos , Dados de Sequência Molecular , Desnaturação Proteica , Espectrometria de Fluorescência
3.
Chem Res Toxicol ; 14(1): 16-24, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11170504

RESUMO

Peroxisomicine A(1) (T-514) is a dimeric anthracenone first isolated from the plant Karwinskia humboldtiana. The compound presents a high and selective toxicity toward liver and skin cell cultures and is currently the subject of preclinical studies as an antitumor drug. To date, the molecular basis for its diverse biological effects remains poorly understood. To elucidate its mechanism of action, we studied its interaction with DNA and its effects on human DNA topoisomerases. Practically no interaction with DNA was detected. Peroxisomicine was found to inhibit topoisomerase II but not topoisomerase I. DNA relaxation and decatenation assays indicated that the drug interferes with the catalytic activity of topoisomerase II but does not stimulate DNA cleavage, in contrast to conventional topoisomerase poisons such as etoposide. Two human leukemia cell lines sensitive or resistant to mitoxantrone were used to assess the cytotoxicity of the toxin and its effect on the cell cycle. In both cases, peroxisomicine treatment was associated with a loss of cells from every phase of the cell cycle and was accompanied by a large increase in the sub-G1 region which is characteristic of apoptotic cells. The cell cycle changes were more pronounced with the sensitive HL-60 cells than with the resistant HL-60/MX2 cells (with reduced topoisomerase II activity), in agreement with the cytotoxicity measurements. Treatment of HL-60 cells with T-514 stimulated the cleavage of the poly(ADP-ribose) polymerase by intracellular proteases such as caspase-3. The cytometry and Western blot analyses reveal that peroxisomicine induces apoptosis in leukemia cells. In addition, we characterized a catabolite of peroxisomicine, named T-510R, in the form of a highly stable radical metabolite. The electron spin resonance and mass spectrometry data are consistent with the formation of an anionic semiquinonic radical. The oxidized product T-510R inhibits topoisomerase II with a reduced efficiency compared to the parent toxin and was found to be about 3-4 times less toxic to both the sensitive and resistant leukemia cell lines than T-514. Collectively, the results suggest that topoisomerase II inhibition plays a role in the cytotoxicity of the plant toxin peroxisomicine. Inhibition of topoisomerase II may serve as an inducing signal triggering the apoptotic cell death of leukemia cells exposed to the toxin. The dihydroxyanthracenone unit may represent a useful chemotype for the preparation of topoisomerase II-targeted anticancer agents.


Assuntos
Antracenos/metabolismo , Antracenos/toxicidade , Antineoplásicos Fitogênicos/metabolismo , Antineoplásicos Fitogênicos/toxicidade , Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/toxicidade , Inibidores da Topoisomerase II , Caspase 3 , Caspases/metabolismo , DNA de Neoplasias/metabolismo , DNA Super-Helicoidal/metabolismo , Resistencia a Medicamentos Antineoplásicos , Espectroscopia de Ressonância de Spin Eletrônica , Ativação Enzimática/efeitos dos fármacos , Radicais Livres/metabolismo , Radicais Livres/toxicidade , Células HL-60/citologia , Células HL-60/efeitos dos fármacos , Células HL-60/enzimologia , Humanos , Cinética , Mitoxantrona/farmacologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Inibidores da Topoisomerase I
4.
Anticancer Drug Des ; 16(1): 27-36, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11762642

RESUMO

We report the synthesis and pharmacological evaluation of a novel homocamptothecin (hCPT) derivative, 12-Cl-hCPT, which contains a seven-membered beta-hydroxylactone in place of the conventional six-membered alpha-hydroxylactone found in camptothecin (CPT) and bears a chloro substituent at position 12. The capacity of 12-Cl-hCPT to inhibit DNA topoisomerase I was compared with that of SN-38, the active metabolite of the clinically used antitumour prodrug CPT-11. In the DNA relaxation assay, 12-Cl-hCPT proved to be slightly more potent than SN-38 at stimulating the formation of nicked plasmid DNA molecules. A series of radiolabelled DNA restriction fragments were employed to identify and compare the position of the DNA cleavage sites induced by topoisomerase I in the presence of 12-Cl-hCPT and SN-38. These sequencing studies confirm that both 12-Cl-hCPT and SN-38 strongly promote DNA cleavage by topoisomerase I and reveal that the majority of the cleavage sites are located at the same nucleotide positions for the two drugs. However, a certain number of DNA cleavage sites were found to be specific to 12-Cl-hCPT. These sites, previously characterized with unsubstituted hCPT, generally correspond to 5'-CG sites whereas the sites common to the 12-Cl-hCPT and SN-38 essentially correspond to 5'-TG sites. We also quantified the formation of drug-induced protein-DNA complexes formed in HT29 human colon carcinoma cells. Trapping of endogenous proteins onto DNA was found to be much more efficient with 12-Cl-hCPT than with SN-38. These data provide a molecular basis to account for the enhanced antiproliferative activity of 12-Cl-hCPT compared with that of SN-38. Biological evaluation on a panel of sensitive and drug-resistant cell lines revealed 12-Cl-hCPT to be more cytotoxic to tumour cells than SN-38. 12-Cl-hCPT proved 14- and 23-fold more active than SN-38 toward the K562adr and T24anp multidrug-resistant cell lines, respectively. The marked topoisomerase I inhibitory properties of 12-Cl-hCPT coupled with its interesting antiproliferative activity, in particular against cancer cells presenting multidrug resistance phenotype with overexpression of P-glycoprotein, makes 12-Cl-hCPT a valid candidate for subsequent preclinical evaluation. Collectively, the data strengthen homocamptothecin as an extremely promising template to generate novel and potent antitumour agents.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos/farmacologia , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores da Topoisomerase I , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos/isolamento & purificação , Camptotecina/isolamento & purificação , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA de Neoplasias/genética , DNA de Neoplasias/isolamento & purificação , DNA Super-Helicoidal/efeitos dos fármacos , Doxorrubicina/farmacologia , Células HT29 , Humanos , Irinotecano , Plasmídeos/genética , Plasmídeos/isolamento & purificação
5.
Anticancer Drug Des ; 15(3): 191-201, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11049087

RESUMO

Cryptolepine and neocryptolepine are two indoloquinoline alkaloids isolated from the roots of the African plant Cryptolepis sanguinolenta. Both drugs have revealed antibacterial and antiparasitic activities and are strongly cytotoxic to tumour cells. We have recently shown that cryptolepine can intercalate into DNA and stimulates DNA cleavage by human topoisomerase II. In this study, we have investigated the mechanism of action and cytotoxicity of neocryptolepine, which differs from the parent isomer only by the orientation of the indole unit with respect to the quinoline moiety. The biochemical and physicochemical results presented here indicate that neocryptolepine also intercalates into DNA, preferentially at GC-rich sequences, but exhibits a reduced affinity for DNA compared with cryptolepine. The two alkaloids interfere with the catalytic activity of human topoisomerase II but the poisoning activity is slightly more pronounced with cryptolepine than with its isomer. The data provide a molecular basis to account for the reduced cytotoxicity of neocryptolepine compared with the parent drug.


Assuntos
Alcaloides/toxicidade , Antineoplásicos Fitogênicos/toxicidade , Indóis , Substâncias Intercalantes/toxicidade , Quinolinas , Inibidores da Topoisomerase II , Alcaloides/metabolismo , Animais , Antineoplásicos Fitogênicos/metabolismo , Bovinos , Dicroísmo Circular , DNA/efeitos dos fármacos , DNA/metabolismo , Dano ao DNA , Pegada de DNA , DNA Topoisomerases Tipo II/metabolismo , Humanos , Alcaloides Indólicos , Substâncias Intercalantes/metabolismo , Células KB , Melanoma Experimental/tratamento farmacológico , Camundongos , Camundongos Endogâmicos , Raízes de Plantas/química , Plantas Medicinais/química , Especificidade por Substrato
6.
Biochem Pharmacol ; 60(4): 527-37, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10874127

RESUMO

Ascididemin (ASC) is a pentacyclic DNA-intercalating agent isolated from the Mediterranean ascidian Cystodytes dellechiajei. This marine alkaloid exhibits marked cytotoxic activities against a range of tumor cells, but its mechanism of action remains poorly understood. We investigated the effects of ASC on DNA cleavage by human topoisomerases I and II. Relaxation assays using supercoiled DNA showed that ASC stimulated double-stranded cleavage of DNA by topoisomerase II, but exerted only a very weak effect on topoisomerase I. ASC is a conventional topoisomerase II poison that significantly promoted DNA cleavage, essentially at sites having a C on the 3' side of the cleaved bond (-1 position), as observed with etoposide. The stimulation of DNA cleavage by topoisomerase I in the presence of ASC was considerably weaker than that observed with camptothecin. Cytotoxicity measurements showed that ASC was even less toxic to P388 leukemia cells than to P388CPT5 cells resistant to camptothecin. In addition, the marine alkaloid was found to be equally toxic to HL-60 leukemia cells sensitive or resistant to mitoxantrone. It is therefore unlikely that topoisomerases are the main cellular targets for ASC. This alkaloid was found to strongly induce apoptosis in HL-60 and P388 leukemia cells. Cell cycle analysis showed that ASC treatment was associated with a loss of cells in the G1 phase accompanied with a large increase in the sub-G1 region. Cleavage experiments with poly(ADP-ribose) polymerase (PARP) revealed that caspase-3 was a mediator of the apoptotic pathway induced by ASC. The DNA of ASC-treated cells was severely fragmented. Collectively, these findings indicate that ASC is a potent inducer of apoptosis in leukemia cells.


Assuntos
Alcaloides/farmacologia , Apoptose , Fenantrolinas , Quinolinas , Inibidores da Topoisomerase II , Alcaloides/química , Animais , DNA Topoisomerases Tipo I/metabolismo , Células HL-60 , Humanos , Leucemia , Oceanos e Mares , Plâncton/química , Inibidores da Topoisomerase I , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA