Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 409(1): 9-18, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11099695

RESUMO

Cryptolepine and neocryptolepine are two indoloquinoline derivatives isolated from the roots of the african plant Cryptolepis sanguinolenta. These two alkaloids, which only differ by the respective orientation of their indole and quinoline rings, display potent cytotoxic activities against tumour cells and present antibacterial and antiparasitic properties. Our previous molecular studies indicated that these two natural products intercalate into DNA and interfere with the catalytic activity of human topoisomerase II. Here we have extended the study of their mechanism of action at the cellular level. Murine and human leukemia cells were used to evaluate the cytotoxicity of the drugs and their effects on the cell cycle were measured by flow cytometry. Cryptolepine, and to a lesser extent neocryptolepine, provoke a massive accumulation of P388 murine leukemia cells in the G2/M phase. With HL-60 human leukemia cells, the treatment with cryptolepine leads to the appearance of a hypo-diploid DNA content peak (sub-G1) characteristic of the apoptotic cell population. With both P388 and HL-60 cells, cryptolepine proved about four times more toxic than its isomer. But the use of the HL-60/MX2 cell line resistant to the anticancer drug mitoxantrone suggests that topoisomerase II may not represent the essential cellular target for the alkaloids, which are both only two times less toxic to the resistant HL-60/MX2 cells compared to the parental cells. The capacity of the drugs to induce apoptosis of HL-60 human leukemia cells was examined by complementary biochemical techniques. Western blotting analysis revealed that cryptolepine, but not neocryptolepine, induces cleavage of poly(ADP-ribose) polymerase but both alkaloids induce the release of cytochrome c from the mitochondria. The cleavage of poly(ADP-ribose) polymerase observed with cryptolepine correlates with the appearance of a marked sub-G1 peak in the cell cycle experiments. The proteolytic activity of Asp-Glu-Val-Asp- or Ile-Glu-Thr-Asp-caspases was found to be enhanced much more strongly with cryptolepine than with its isomer, as expected from their different cytotoxic potential. Despite the activation of the caspase cascade, we did not detect internucleosomal cleavage of DNA in the HL-60 cells treated with the alkaloids. Altogether, the results shed light on the mechanism of action of these two plant alkaloids.


Assuntos
Alcaloides/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Indóis , Quinolinas , Animais , Caspase 3 , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Células HL-60 , Humanos , Alcaloides Indólicos , Leucemia P388/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Mitoxantrona/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo
2.
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
3.
Eur J Pharm Sci ; 10(2): 125-31, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10727878

RESUMO

Leukemia cells (HL-60 and P388) treated with the topoisomerase I inhibitor camptothecin (CPT) undergo rapid apoptosis as judged from internucleosomal degradation of genomic DNA, morphological changes and flow cytometry analysis. The intracellular free calcium concentration is not affected by the treatment with a high dose of CPT. In contrast, fluorescence measurements of cells loaded with the pH indicator BCECF-AM indicate that the intracellular pH decreases significantly. Incubation of the leukemia cells with a high drug concentration for 5 h or with lower drug concentrations for 15 h results in a pronounced intracellular acidification. Measurements with the whole cell population show a decrease of 0.3-0.4 pH units. The extent of the acidic shift is proportional to the drug concentration and the period of incubation. No such effects were observed with P388CPT5 cells resistant to CPT. The results support the hypothesis that apoptosis induced in leukemia cells by CPT is associated with decreased intracellular pH. Modification of intracellular pH by topoisomerase inhibitors is viewed as an essential event responsible for the induction and/or propagation of apoptosis. The role of CPT-induced cellular acidification in the mechanism of action of the drug is discussed.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Camptotecina/farmacologia , Animais , Cálcio/metabolismo , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Eletroforese em Gel de Ágar , Inibidores Enzimáticos/farmacologia , Células HL-60 , Humanos , Concentração de Íons de Hidrogênio , Leucemia P388/tratamento farmacológico , Leucemia P388/metabolismo , Inibidores da Topoisomerase II
4.
Biochemistry ; 38(47): 15556-63, 1999 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-10569939

RESUMO

Homocamptothecin (hCPT) contains a seven-membered beta-hydroxylactone in place of the conventional six-membered alpha-hydroxylactone ring found in camptothecin and its tumor active analogues, including topotecan and irinotecan. The homologation of the lactone E-ring reinforces the stability of the lactone, thus reducing considerably its conversion into a carboxylate form which is inactive. We have recently shown that hCPT is much more active than the parent compound against a variety of tumor cells in vitro and in xenograft models, suggesting that a highly reactive lactone is not essential for topoisomerase I-mediated anticancer activity [Lesueur-Ginot et al. (1999) Cancer Res. 59, 2939-2943]. In the present study, we provide further evidence that hCPT has superior topoisomerase I inhibition capacities to CPT. In particular, we show that replacement of the camptothecin lactone E-ring with a homologous seven-membered lactone ring changes the sequence-specificity of the drug-induced DNA cleavage by topoisomerase I. Both CPT and hCPT stimulate the cleavage by topoisomerase I at T( downward arrow)G sites, but in addition, hCPT stabilizes cleavage at specific sites containing the sequence AAC( downward arrow)G. At low drug concentrations, the cleavage at the T( downward arrow)G sites and at the hCPT-specific C( downward arrow)G sites is more pronounced and more stable with hCPT than with CPT. The in vitro data were confirmed in cells. Higher levels of protein-DNA complexes were detected in P388 leukemia cells treated with hCPT than those treated with CPT. Immunoblotting experiments revealed that endogenous topoisomerase I was efficiently trapped onto DNA by hCPT in cells. Finally, the use of a leukemia cell line resistant to CPT provided evidence that topoisomerase I is involved in the cytotoxicity of hCPT. Altogether, the results show that the beta-hydroxylactone ring of hCPT plays an important and positive role in the poisoning of topoisomerase I. An explanation is proposed to account for such remarkable changes in the sequence specificity of topoisomerase I cleavage consequent to the modification of the lactone. The study sheds new light on the importance of the lactone ring of camptothecins for the stabilization of topoisomerase I-DNA complexes.


Assuntos
Camptotecina/análogos & derivados , DNA Topoisomerases Tipo I/química , DNA Super-Helicoidal/química , Inibidores Enzimáticos/química , Inibidores da Topoisomerase I , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Camptotecina/química , Camptotecina/farmacologia , Camptotecina/toxicidade , Sobrevivência Celular/efeitos dos fármacos , DNA Super-Helicoidal/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/toxicidade , Leucemia P388 , Camundongos , Relação Estrutura-Atividade , Células Tumorais Cultivadas
5.
Biochem Pharmacol ; 58(8): 1307-12, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10487533

RESUMO

Lucanthone is an antitumour drug used as an adjuvant in radiation therapy. The drug intercalates into DNA and inhibits topoisomerase II. An indazole analogue of lucanthone (IA-5) was examined for its ability to modulate topoisomerase II-DNA cleavable complex formation in vitro. The drug contains a methylbenzothiopyranoindazole chromophore instead of the methyl-thioxanthenone nucleus of lucanthone. Using a radiolabelled linear plasmid DNA as a substrate, both lucanthone and the indazole analogue were shown to promote the cleavage of DNA by human topoisomerase II. Sequencing experiments with different restriction fragments indicated that the indazole drug promoted DNA cleavage primarily at sites having a C on the 3' side of the cleaved bond (-1 position). By contrast, in the same sequencing methodology lucanthone exerted a much weaker effect on topoisomerase II. The sequence selectivity of IA-5 is reminiscent of that of the anticancer drug mitoxantrone and its anthrapyrazole analogue losoxantrone, which is structurally close to IA-5. Binding to DNA and topoisomerase II inhibition are two distinct processes contributing separately to the cytotoxic activity of the indazole drug.


Assuntos
DNA Topoisomerases Tipo II/metabolismo , DNA/efeitos dos fármacos , Indazóis/farmacologia , Lucantona/farmacologia , DNA/metabolismo , DNA Topoisomerases Tipo II/efeitos dos fármacos , Humanos , Indazóis/química , Lucantona/análogos & derivados , Estrutura Molecular
6.
Bioorg Med Chem Lett ; 9(14): 2025-30, 1999 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-10450974

RESUMO

Indolequinone derivatives of the antitumour antibiotic BE 10988 were synthesized and evaluated for their cytotoxicity and action mechanism. The quinone system is essential to biological activity and the thiazole ring plays a major role in the poisoning of topoisomerase II.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Tiazóis/química , Inibidores da Topoisomerase II , Animais , Divisão Celular/efeitos dos fármacos , DNA/metabolismo , Inibidores Enzimáticos/metabolismo , Indóis/química , Concentração Inibidora 50 , Leucemia L1210/tratamento farmacológico , Camundongos , Quinonas/química , Solubilidade , Relação Estrutura-Atividade
7.
Biochemistry ; 38(24): 7719-26, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10387011

RESUMO

Cryptolepine, matadine, and serpentine are three indoloquinoline alkaloids isolated from the roots of African plants: Cryptolepis sanguinolenta, Strychnos gossweileri, and Rauwolfia serpentina, respectively. For a long time, these alkaloids have been used in African folk medicine in the form of plant extracts for the treatment of multiple diseases, in particular as antimalarial drugs. To date, the molecular basis for their diverse biological effects remains poorly understood. To elucidate their mechanism of action, we studied their interaction with DNA and their effects on topoisomerase II. The strength and mode of binding to DNA of the three alkaloids were investigated by spectroscopy. The alkaloids bind tightly to DNA and behave as typical intercalating agents. All three compounds stabilize the topoisomerase II-DNA covalent complex and stimulate the cutting of DNA by topoisomerase II. The poisoning effect is more pronounced with cryptolepine than with matadine and serpentine, but none of the drugs exhibit a preference for cutting at a specific base. Cryptolepine which binds 10-fold more tightly to DNA than the two related alkaloids proves to be much more cytotoxic toward B16 melanoma cells than matadine and serpentine. The cellular consequences of the inhibition of topoisomerase II by cryptolepine were investigated using the HL60 leukemia cell line. The flow cytometry analysis shows that the drug alters the cell cycle distribution, but no sign of drug-induced apoptosis was detected when evaluating the internucleosomal fragmentation of DNA in cells. Cryptolepine-treated cells probably die via necrosis rather than via apoptosis. The results provide evidence that DNA and topoisomerase II are the primary targets of cryptolepine, matadine, and serpentine.


Assuntos
Alcaloides/farmacologia , DNA Topoisomerases Tipo II/química , DNA/química , DNA/efeitos dos fármacos , Indóis , Substâncias Intercalantes/farmacologia , Quinolinas , Alcaloides/química , Alcaloides/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Sítios de Ligação/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , DNA/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Eletroquímica , Inibidores Enzimáticos/farmacologia , Polarização de Fluorescência , Células HL-60 , Humanos , Alcaloides Indólicos , Substâncias Intercalantes/química , Substâncias Intercalantes/metabolismo , Alcaloides de Triptamina e Secologanina/química , Alcaloides de Triptamina e Secologanina/metabolismo , Alcaloides de Triptamina e Secologanina/farmacologia , Análise Espectral , Inibidores da Topoisomerase II
8.
Cancer Res ; 59(12): 2853-60, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10383146

RESUMO

The DNA-intercalating antitumor drug NB-506 is a potent topoisomerase poison currently undergoing phase I/II clinical trials. It contains a planar indolocarbazole chromophore substituted with a glucose residue. Up until now, it was thought that intercalation of the drug into DNA was essential for the stabilization of topoisomerase I-DNA covalent complexes. But, in the present study, we show that a regio-isomeric form of NB-506 has lost its capacity to intercalate into DNA, but remains an extremely potent topoisomerase I poison. The new analogue contains two hydroxyl groups at positions 2,10 instead of positions 1,11 in NB-506. The relocation of the two OH groups reduces considerably the strength of binding to DNA and prevents the drug from intercalating into the DNA double helix. However, the topoisomerase I inhibition capacity of the new analogue remains very high. The two drug isomers are equally potent at maintaining the integrity of the topoisomerase I-DNA covalent complexes, but stimulate cleavage at different sites on DNA. NB-506 stabilizes topoisomerase I preferentially at sites having a pyrimidine (T or C) and a G on the 5' and 3' sides of the cleaved bond, respectively. The 2,10-isomer induces topoisomerase I-mediated cleavage only at TG sites and, thus, behaves exactly as the reference topoisomerase I poison camptothecin. Finally, cytotoxicity measurements performed with a panel of murine and human cancer cell lines reveal that the newly designed drug is considerably (up to 100-fold) more toxic to tumor cells than the parent drug NB-506. We conclude that the DNA-binding and topoisomerase I poisoning activities of NB-506 can be viewed as two separate mechanisms.


Assuntos
Antineoplásicos/farmacologia , Carbazóis/farmacologia , DNA/efeitos dos fármacos , Glucosídeos/farmacologia , Inibidores da Topoisomerase I , Animais , Antineoplásicos/química , Carbazóis/química , DNA/metabolismo , Pegada de DNA , Desenho de Fármacos , Glucosídeos/química , Humanos , Concentração Inibidora 50 , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacologia , Camundongos , Estereoisomerismo , Células Tumorais Cultivadas
9.
Cancer Res ; 59(12): 2939-43, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10383158

RESUMO

Homocamptothecin (hCPT) is a semisynthetic analogue of camptothecin (CPT) with a seven-membered beta-hydroxylactone resulting from the insertion of a methylene spacer between the alcohol moiety and the carboxyl function of the naturally occurring six-membered alpha-hydroxylactone of CPT. This E-ring modification provides a less reactive lactone with enhanced stability and decreased protein binding in human plasma. Biological testing against CPT revealed that, instead of being detrimental, the modified lactone of hCPT has a positive impact on topoisomerase I (Topo I) poisoning properties. In vitro tests showed hCPT to fully conserve the ability to stabilize Topo I-DNA cleavage complexes and to stimulate a higher level of DNA cleavage than CPT. A similar trend toward improvement was also observed in antiproliferative assays with human tumor cell lines (including cells overexpressing P-glycoprotein). In two distinct in vivo models, using L1210 murine leukemia or human colon carcinoma HT29, hCPT was found to be more efficacious than CPT. The slow, but irreversible, hydrolysis of hCPT, instead of the fast equilibrium of CPT, may account for its good in vivo activity. Overall, these results provide evidence that a highly reactive lactone is not a requisite for the Topo I-mediated antitumor activity of CPT analogues, and that hCPT is an interesting pharmacological tool with improved solution behavior as well as a promising new template for the preparation of more efficacious Topo I poisons.


Assuntos
Antineoplásicos/uso terapêutico , Camptotecina/análogos & derivados , Lactonas/metabolismo , Inibidores da Topoisomerase I , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Camptotecina/química , Camptotecina/farmacologia , Divisão Celular/efeitos dos fármacos , DNA/efeitos dos fármacos , DNA/metabolismo , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais , Estabilidade de Medicamentos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/uso terapêutico , Células HT29 , Humanos , Células K562 , Leucemia L1210/tratamento farmacológico , Leucemia Experimental/tratamento farmacológico , Transplante de Neoplasias , Transplante Heterólogo , Células Tumorais Cultivadas
10.
Anticancer Drug Des ; 14(1): 47-60, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10363027

RESUMO

Four diphenylfuran derivatives possessing different dicationic terminal side chains were used to investigate sequence-specific binding to DNA and poisoning of human topoisomerase II. Footprinting experiments with a range of DNA substrates attest that all four drugs bind selectively to AT-rich sequences in DNA. However, the quantitative analysis of the footprinting profiles reveals significant differences in terms of AT-selectivity according to the nature of the basic side chains. Furimidazoline (DB60) shows a reduced capacity to interact selectively with A.T tetrads compared with furamidine (DB75) and the 3-pentyl-substituted diamidine analogue DB226. DB244, for which the two amidine ends are substituted with a cyclopentyl group, exhibits the most pronounced AT specificity. It binds tightly to sites composed of at least four adjacent AT base pairs, such as 5'-TAAT, AATT and TTTT. At low concentrations (< 2 microM) DB60 is also capable of forming stable complexes with AT sites but at higher concentrations the binding becomes totally non-specific due to additional intercalation of drug molecules into GC-rich sequences. Nevertheless, DB60 is the only drug is the series which stabilizes DNA-topoisomerase II covalent complexes. This compound effectively promotes DNA cleavage by topoisomerase II whereas DB75, DB226 and DB244 have practically no effect. The topoisomerase II poisoning activity of DB60 correlates with its ability to intercalate into GC sites in DNA whereas the three other diphenylfurans essentially behave as typical AT-selective minor groove binders. The study suggests that the antimicrobial activity of the diphenylfurans, which are active against the Pneumocystis carinii pathogen (PCP), depends essentially on their capacity to recognize AT-rich DNA sequences rather than their ability to interfere with topoisomerase II. In contrast, the cytotoxicity of drugs like DB60 would be connected with the formation of intercalation complexes and the stimulation of DNA cleavage by human topoisomerase II.


Assuntos
Antibacterianos/farmacologia , Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Furanos/farmacologia , Inibidores da Topoisomerase II , Antibacterianos/química , Antibacterianos/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Sítios de Ligação , Pegada de DNA , DNA Topoisomerases Tipo I/química , DNA Topoisomerases Tipo II/química , DNA Bacteriano/metabolismo , DNA Super-Helicoidal/química , Eletroforese em Gel de Poliacrilamida , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Escherichia coli/metabolismo , Furanos/química , Furanos/metabolismo , Humanos , Indicadores e Reagentes , Permanganato de Potássio
11.
Anticancer Res ; 19(6B): 5245-50, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10697543

RESUMO

Usambarensine is a plant alkaloid isolated from the roots of Strychnos usambarensis collected in Central Africa. This bis-indole compound displays potent antiamoebic activities and shows antigardial, antimalarial and cytotoxic effects. Usambarensine is highly toxic to B16 melanoma cells and inhibits the growth of leukemia and carcinoma cells. To date, the molecular basis for its diverse biological effects remains totally unknown. However, its capacity to inhibit nucleic acids synthesis in melanoma cells, on the one hand, and its structural analogy with DNA-binding pyridoindole plant alkaloids recently studied (cryptolepine and matadine), on the other hand, suggested that usambarensine could also bind to DNA. Consequently, we studied the strength and mode of binding to DNA of usambarensine by means of absorption, circular and linear dichroism. The results of the optical measurements indicate that the alkaloid effectively binds to DNA and behaves as a typical intercalating agent. Biochemical experiments indicated that, in contrast to cryptolepine and matadine, usambarensine does not interfere with the catalytic activity of topoisomerase II. Human HL60 leukemia cells were used to assess the cytotoxicity of the alkaloid and its effect on the cell cycle. Usambarensine treatment is associated with a loss of cells in the G1 phase accompanied with a large increase in the sub-G1 region which is characteristic of apoptotic cells. The DNA of usambarensine-treated cells was severely fragmented and the proteolytic activity of DEVD-caspases is enhanced. Usambarensine is thus characterized as DNA intercalator inducing apoptosis in leukemia cells.


Assuntos
Alcaloides/farmacologia , Indóis/farmacologia , Substâncias Intercalantes/farmacologia , Alcaloides/química , Alcaloides/metabolismo , Dicroísmo Circular , DNA/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Células HL-60 , Humanos , Indóis/química , Indóis/metabolismo , Substâncias Intercalantes/química , Substâncias Intercalantes/metabolismo , Plantas/química , Inibidores da Topoisomerase I
12.
Bull Cancer ; 85(3): 254-61, 1998 Mar.
Artigo em Francês | MEDLINE | ID: mdl-9752316

RESUMO

The treatment of cancer with alkylating drugs or topoisomerase II inhibitors can be responsible for the development of myelodysplastic syndromes and acute myelogenous leukemia. Alkylating agents such as melphalan and cisplatinum mainly produce damages at chromosomes 5 and 7 whereas topoisomerase II inhibitors-induced lesions essentially affect chromosomes 11 and 21. Rearrangements of the MLL gene at band 11q23 are frequently observed in human de novo myeloid and lymphoid leukemia as well as in leukemia or myelodysplasia secondary to therapy with drugs targetting topoisomerase II such as the epipodophyllotoxins. A relationship between the treatment with etoposide on teniposide and the development of translocations of the MLL gene has been clearly evidenced. The potential molecular basis of the chromosomal rearrangements implicating topoisomerase II and its inhibitors are discussed. The chemical structure of the inhibitors, their mechanism of action and the genes targetted by these drugs are presented. DNA cleavages induced directly by topoisomerase II inhibitors or by the drug induced apoptotic cellular response are responsible for nonrandom chromosomal aberrations and contribute to leukemogenesis.


Assuntos
Antineoplásicos/efeitos adversos , Aberrações Cromossômicas , Leucemia Mieloide/induzido quimicamente , Proteínas de Neoplasias/genética , Segunda Neoplasia Primária/induzido quimicamente , Leucemia-Linfoma Linfoblástico de Células Precursoras/induzido quimicamente , Inibidores da Topoisomerase II , Doença Aguda , Antineoplásicos/uso terapêutico , Cromossomos Humanos Par 11/efeitos dos fármacos , Cromossomos Humanos Par 21/efeitos dos fármacos , DNA de Neoplasias/efeitos dos fármacos , Rearranjo Gênico , Humanos , Leucemia Mieloide/genética , Segunda Neoplasia Primária/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Recombinantes de Fusão/genética , Translocação Genética
13.
Biochemistry ; 37(15): 5136-46, 1998 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-9548744

RESUMO

Cryptolepine hydrochloride is an indoloquinoline alkaloid isolated from the roots of Cryptolepis sanguinolenta. It is characterized by a multiplicity of host-mediated biological activities, including antibacterial, antiviral, and antimalarial properties. To date, the molecular basis for its diverse biological effects remains largely uncertain. Several lines of evidence strongly suggest that DNA might correspond to its principal cellular target. Consequently, we studied the strength and mode of binding to DNA of cryptolepine by means of absorption, fluorescence, circular, and linear dichroism, as well as by a relaxation assay using DNA topoisomerases. The results of various optical and gel electrophoresis techniques converge to reveal that the alkaloid binds tightly to DNA and behaves as a typical intercalating agent. In DNAase I footprinting experiments it was found that the drug interacts preferentially with GC-rich sequences and discriminates against homo-oligomeric runs of A and T. This study has also led to the discovery that cryptolepine is a potent topoisomerase II inhibitor and a promising antitumor agent. It stabilizes topoisomerase II-DNA covalent complexes and stimulates the cutting of DNA at a subset of preexisting topoisomerase II cleavage sites. Taking advantage of the fluorescence of the indoloquinoline chromophore, fluorescence microscopy was used to map cellular uptake of the drug. Cryptolepine easily crosses the cell membranes and accumulates selectively into the nuclei rather than in the cytoplasm of B16 melanoma cells. Quantitative analyses of DNA in cells after Feulgen reaction and image cytometry reveal that the drug blocks the cell cycle in G2/M phases. It is also shown that the alkaloid is more potent at inhibiting DNA synthesis rather than RNA and protein synthesis. Altogether, the results provide direct evidence that DNA is the primary target of cryptolepine and suggest that this alkaloid is a valid candidate for the development of tumor active compounds.


Assuntos
Alcaloides/farmacologia , Antineoplásicos/farmacologia , DNA de Neoplasias/efeitos dos fármacos , Indóis , Substâncias Intercalantes/farmacologia , Melanoma Experimental/metabolismo , Quinolinas , Inibidores da Topoisomerase II , Animais , Sequência de Bases , Pegada de DNA , Relação Dose-Resposta a Droga , Alcaloides Indólicos , Camundongos , Dados de Sequência Molecular , Desnaturação de Ácido Nucleico/efeitos dos fármacos , Análise Espectral
14.
Nucleic Acids Res ; 25(22): 4487-92, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9358156

RESUMO

The transactivation response region (TAR) RNA is an essential component in transcriptional regulation of the human immunodeficiency virus type-1 (HIV-1) genome. We have examined the interaction between TAR RNA and the bisbenzimidazole derivative Hoechst 33258. Previous studies have shown that this drug, which is well known as an AT-selective DNA minor groove binder, can also interact with GC-rich sequences in DNA as well as with RNA, possibly by intercalation. Absorption spectroscopy, circular dichroism and electric linear dichroism, as well as RNase A footprinting, were employed to compare binding of Hoechst 33258 to wild-type RNA and its analogue lacking the pyrimidine bulge. The uridine bulge, which is an important contributor to the structural stability of TAR, plays an essential role in drug binding. Deletion of the bulge destabilizes both free and drug-bound forms of TAR and markedly affects the orientation of the drug chromophore complexed with the RNA. According to the linear dichroism data, the bisbenzimidazole is oriented more or less perpendicular to the RNA helix axis. The data are compatible with a model in which the bisbenzimidazole chromophore is inserted into the existing cavity created by the pyrimidine bulge. The footprinting experiments, showing that the drug binds to a unique site opposite the unpaired uridine residues, also support this model. The binding of Hoechst 33258 to the sequence 5'-GCUCU, which delimits the cavity, reflects the greater accessibility of that region compared with other sites in the RNA molecule. The identification of a binding site for small molecules in TAR offers promising perspectives for developing drugs that would block the access of TAR RNA to proteins and therefore for the design of anti-HIV agents.


Assuntos
Bisbenzimidazol/metabolismo , Repetição Terminal Longa de HIV , HIV-1/genética , Conformação de Ácido Nucleico , RNA Viral/metabolismo , Absorção , Sítios de Ligação , Bisbenzimidazol/química , Dicroísmo Circular , Pegada de DNA , Calefação , Humanos , Desnaturação de Ácido Nucleico , Oligonucleotídeos/síntese química , Oligonucleotídeos/metabolismo , Pirimidinas/química , RNA Viral/química , Deleção de Sequência , Relação Estrutura-Atividade , Uridina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...