Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Natl Cancer Inst ; 81(22): 1732-5, 1989 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-2553992

RESUMEN

Topoisomerase inhibitors comprise an important group of agents that is used in cancer treatment. Because the development of resistance to cancer chemotherapeutic agents represents a major limitation of cancer chemotherapy, we investigated the mechanism of resistance by murine P388 leukemia to camptothecin (topoisomerase I inhibitor) or amsacrine (topoisomerase II inhibitor). The resistant cells contained reduced levels of topoisomerase activity and messenger RNA. The topoisomerase gene of these cells was rearranged (only in one allele) and hypermethylated. These topoisomerase gene alterations probably resulted in reduced transcription and, thus, enzyme production, which was correlated with resistance to the topoisomerase inhibitor.


Asunto(s)
Amsacrina/farmacología , Camptotecina/farmacología , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo I/genética , Reordenamiento Génico , Leucemia P388/enzimología , Leucemia Experimental/enzimología , Alelos , Animales , ADN-Topoisomerasas de Tipo I/análisis , ADN-Topoisomerasas de Tipo II/análisis , Resistencia a Medicamentos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Metilación , Metiltransferasas/metabolismo , Ratones , ARN Mensajero/análisis , Inhibidores de Topoisomerasa I , Inhibidores de Topoisomerasa II , Transcripción Genética/efectos de los fármacos
2.
Cancer Res ; 50(9): 2636-40, 1990 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-2158396

RESUMEN

A combination of tumor necrosis factor (TNF) and the topoisomerase I inhibitor, camptothecin, or the topoisomerase II inhibitors, teniposide and amsacrine, produced dose-dependent synergistic cytotoxicity against the murine L929 fibrosarcoma cells. Similar synergy was not observed with a combination of TNF and bleomycin. To define the role of TNF in the augmentation of tumor cell killing by topoisomerase I or II inhibitors, the effect of TNF on the production of enzyme-linked DNA strand breaks induced in cells by topoisomerase inhibitors was investigated. L929 cells incubated for 1 h with the topoisomerase inhibitors contained protein-linked strand breaks. In contrast, TNF alone did not induce DNA strand breakage. However, when cells were incubated simultaneously with TNF and camptothecin, amsacrine, Adriamycin, actinomycin D, teniposide, or etoposide, increased numbers of strand breaks were produced. Preincubation of the cells with TNF for 30 min or 3 h before the addition of camptothecin or etoposide resulted in no more strand breaks than that observed in cells incubated with the drugs alone. TNF treatment of L929 cells produced a rapid and transient increase in specific activity of extractable topoisomerases I and II. These increases were maximum at 2-5 min of TNF treatment and by 30 min the activities of extractable enzymes were equal to or less than those detected in extracts from untreated cell controls. The transient nature of the increase in extractable topoisomerase activity may explain the kinetics and significance of the order of addition of TNF and inhibitors for maximal synergistic activity. These data are consistent also with a role for topoisomerase-linked DNA lesions in the TNF-mediated potentiation of killing of L929 cells by topoisomerase inhibitors.


Asunto(s)
Daño del ADN , Inhibidores de Topoisomerasa I , Inhibidores de Topoisomerasa II , Factor de Necrosis Tumoral alfa/farmacología , Amsacrina/farmacología , Animales , Camptotecina/farmacología , Supervivencia Celular/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/análisis , ADN-Topoisomerasas de Tipo II/análisis , Sinergismo Farmacológico , Ratones , Células Tumorales Cultivadas
3.
Cancer Res ; 51(21): 5813-6, 1991 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-1657371

RESUMEN

Topotecan (SK&F 104864), a water-soluble analogue of the topoisomerase I inhibitor camptothecin, is currently in Phase II clinical trial for solid tumors. We have characterized topotecan in terms of its effect upon gamma-radiation-induced cell killing. In colony formation experiments, subtoxic concentrations of topotecan (2 microM) potentiated radiation-induced killing of exponentially growing Chinese hamster ovary or P388 murine leukemia cultured cells. Survival curve shoulders were reduced; the slopes of the exponential portions of the curves were decreased to a small extent. D37 and D10 (radiation dose resulting in 37 and 10% survival of colony-forming ability) values were reduced by approximately 60 and 50%, respectively, in the case of Chinese hamster ovary cells. In P388 cells, topotecan reduced D37 by 35 to 40% and D10 by 20 to 25%. Potentiation of radiation-induced cell killing by topotecan was absolutely dependent upon the presence of the topoisomerase I inhibitor during the first few (less than 30) min after irradiation. Association of topoisomerase I with this effect was confirmed in studies of Chinese hamster ovary cells previously made resistant to camptothecin (and cross-resistant to topotecan), resulting in decreased cellular content of topoisomerase I. These cells were found to be 2- to 3-fold hypersensitive to gamma-radiation-induced killing. P388 camptothecin-resistant cells were further sensitized to the lethal effects of ionizing radiation by nontoxic treatment with the topoisomerase II inhibitor novobiocin, consistent with increased dependence of topoisomerase I-deficient cells upon topoisomerase II.


Asunto(s)
Antineoplásicos/farmacología , Camptotecina/análogos & derivados , Supervivencia Celular/efectos de la radiación , Inhibidores de Topoisomerasa I , Animales , Células CHO , Camptotecina/farmacología , Supervivencia Celular/efectos de los fármacos , Cricetinae , Relación Dosis-Respuesta en la Radiación , Ensayos de Selección de Medicamentos Antitumorales , Rayos gamma , Leucemia P388 , Ratones , Topotecan , Células Tumorales Cultivadas
4.
Cancer Res ; 49(10): 2578-83, 1989 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-2540903

RESUMEN

Merbarone has previously been shown to have antitumor activity of unknown mechanism in P388 and L1210 tumor models (A. D. Brewer et al., Biochem. Pharmacol., 34:2047-2050, 1985) and is currently undergoing Phase I clinical trials. Here we report that merbarone is an inhibitor of topoisomerase II. Merbarone inhibited purified mammalian topoisomerase II with a 50% inhibitory concentration of 20 microM, as assessed by ATP-dependent unknotting of P4 phage DNA or relaxation of supercoiled pBR322 plasmid. In contrast to the type II enzyme, inhibition of catalytic activity of topoisomerase I required about 10-fold higher concentrations of merbarone, with a 50% inhibitory concentration of approximately 200 microM. Unlike epipodophyllotoxin analogues and certain DNA intercalative agents which stabilize the topoisomerase II-DNA "cleavable complex," merbarone did not cause detectable topoisomerase II-induced DNA cleavage. Furthermore, merbarone inhibited the production by amsacrine or teniposide of topoisomerase II-associated DNA strand breaks; under identical conditions novobiocin did not decrease these breaks, setting merbarone apart from a novobiocin-like class of topoisomerase II inhibitor. In L1210 cells, merbarone produced only small numbers of protein-associated DNA strand breaks, and only at very high concentrations. Merbarone reduced in a concentration-dependent manner the number of amsacrine- or teniposide-stimulated protein-associated DNA strand breaks in L1210 cells or their isolated nuclei. The data suggest that merbarone represents a novel type of topoisomerase II inhibitor.


Asunto(s)
Antineoplásicos/farmacología , Tiobarbitúricos/farmacología , Inhibidores de Topoisomerasa II , Amsacrina/farmacología , Animales , Camptotecina/farmacología , ADN/metabolismo , Daño del ADN , Leucemia Experimental/enzimología , Ratones , Tenipósido/farmacología , Inhibidores de Topoisomerasa I
5.
Cancer Res ; 44(3): 961-6, 1984 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-6692418

RESUMEN

Two DNA repair assays were used to group human cells. (a) The first assay, survival of N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)-treated adenovirus infecting cellular monolayers, was previously used to define the Mer phenotype of the strain. Strains that supported the growth of MNNG-treated viruses as well as did human fibroblasts were "Mer+"; those that gave rise to clearly less virus survival were "Mer-." (b) The second assay, data from which are presented in this paper, was that of post-MNNG colony-forming ability, and defined the Rem phenotype of the strain. Strains having post-MNNG colony-forming ability like that of human fibroblasts were "Rem+"; more sensitive strains were "Rem-". In all, 22 human cell strains were analyzed for their post-MNNG colony-forming ability. The most resistant strains (eight Mer+ Rem+ strains) had an average inactivation slope of 0.32 "lethal hit"/microM and were those fully able to repair O6-methylguanine (O6mGua) produced in their DNA by a 5 microM dose of MNNG. The most sensitive strains (9 Mer- Rem- strains) had an average inactivation slope of 7.0 "lethal hits"/microM, and were strains that failed to repair O6mGua. Five strains of intermediate sensitivity (Mer+ Rem-) had an average inactivation slope of 0.93 "lethal hit"/microM and were able to repair some labeled O6mGua produced by a 5 microM dose of labeled MNNG, but they repaired significantly less labeled O6mGua if pretreated with unlabeled MNNG. Representative strains from each group were treated with MNNG and assayed for ability: (a) to perform DNA repair synthesis (and DNA repair replication); (b) to support the growth of MNNG-treated adenoviruses; and (c) to restore control levels of tertiary structure to their DNA as assayed by nucleoid sedimentation. The results support the hypothesis that a lesion (both produced by agents that produce O6mGua and repaired by cell strains that repair O6mGua, but not by those that do not) is a lesion lethal to Mer- Rem- strains. This lesion may also initiate induction of excess DNA repair synthesis, the relaxed conformation of nucleoids, the reduced ability to repair MNNG-treated adenovirus, and sister chromatid exchanges as well.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Metilnitronitrosoguanidina/toxicidad , Neoplasias/fisiopatología , Adenoviridae/genética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Transformación Celular Viral , Humanos , Neoplasias/genética , Fenotipo
6.
Cancer Res ; 44(6): 2467-74, 1984 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-6722789

RESUMEN

In order to investigate the mechanisms of cellular damage by alkylating agents, human fibroblasts and tumor cell strains having different sensitivities to killing by N-methyl-N'-nitro-N-nitrosoguanidine [and different abilities to repair O6-methylguanine ( O6mGua ) in their DNA] were treated with other alkylating agents. Methyl methanesulfonate, 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), 1-(2-hydroxyethyl)-3-(2-chloroethyl)-3-nitrosourea, and N-ethyl-N'-nitro-N-nitrosoguanidine gave rise to sensitivity differences, but the differences were less than those observed with N-methyl-N'-nitro-N-nitrosoguanidine. After treatment with UV light, the strains showed similar survival. The data show that the DNA repair mechanism(s) responsible for the differential survival of the strains after N-methyl-N'-nitro-N-nitrosoguanidine treatment probably play(s) a role in repairing DNA damage produced by methyl methanesulfonate, N-ethyl-N'-nitro-N-nitrosoguanidine, BCNU, and 1-(2-hydroxyethyl)-3-(2-chloroethyl)-3-nitrosourea but not that produced by UV. Furthermore, the results support the idea that a breakdown product of BCNU, that does not cause damage repairable by O6mGua repair mechanisms, contributes to the lethal effects due to BCNU-produced DNA-damage that is repairable by O6mGua repair mechanisms. The survival data, along with nucleoid sedimentation and adenovirus host-cell reactivation data, are consistent with the hypothesis that the lesion(s) lethal to tumor cells defective in O6mGua DNA repair are lesions in which DNA oxygen atoms are alkylated.


Asunto(s)
Alquilantes/toxicidad , Reparación del ADN , Guanina/análogos & derivados , Compuestos de Nitrosourea/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Femenino , Guanina/metabolismo , Humanos , Cinética , Neoplasias/fisiopatología , Relación Estructura-Actividad , Rayos Ultravioleta
7.
Cancer Res ; 50(10): 2901-8, 1990 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-2159365

RESUMEN

The activity of topoisomerase II and the cellular content of the 170kD and 180kD forms of the enzyme were studied as functions of transformation and growth state by using normal and ras-transformed NIH-3T3 cells. Total topoisomerase II activity, as measured by the unknotting of P4 DNA, was higher in ras-transformed than in normal cells in similar growth states, and was higher in exponentially growing than in plateau cells for both cell lines. Total topoisomerase II levels, as measured by immunoblotting, showed a similar dependence on transformation and growth state. The relative amounts of the 170kD and 180kD forms of the enzyme varied as a function of transformation and growth state. The proportion of 170kD topoisomerase II was higher in ras-transformed than in untransformed cells and depended much less on growth state in the ras-transformed cells. The topoisomerase II activity in extracts of ras-transformed cells was more sensitive to inhibition by teniposide and merbarone, drugs which selectively inhibit the 170kD form of topoisomerase II. The ras-transformed cells were also more sensitive to the cytotoxic effects of these drugs. An increase in the relative cellular content of 170kD topoisomerase II is characteristic of ras-transformed 3T3 cells, and the levels of this form of the enzyme appear to be less dependent on proliferation state than in untransformed cells. The susceptibility of certain tumors to killing by topoisomerase II-directed drugs may be due to a higher proportion of 170kD enzyme as well as a higher level of total topoisomerase II activity.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Genes ras , Animales , Northern Blotting , Western Blotting , División Celular , Línea Celular , Núcleo Celular/enzimología , Daño del ADN , ADN-Topoisomerasas de Tipo II/clasificación , Ratones , Peso Molecular , Novobiocina/farmacología , ARN Mensajero/genética , Tenipósido/farmacología , Tiobarbitúricos/farmacología , Inhibidores de Topoisomerasa II
8.
Cancer Res ; 46(2): 611-6, 1986 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3000581

RESUMEN

DNA intercalating drugs and the epipodophyllotoxins etoposide and teniposide interfere with the action of mammalian DNA topoisomerase II by trapping an intermediate complex of the enzyme covalently linked to the 5'-termini of DNA breaks. This effect can be observed in intact cells by alkaline elution measurement of protein-associated DNA strand breaks. To assess the cytotoxic role of this effect, we have studied a subline of DC3F Chinese hamster lung cells selected for resistance to the intercalating agent 9-hydroxyellipticine. This subline (DC3F/9-OHE) was cross-resistant to other intercalators as well as to etoposide. Resistance to Adriamycin was associated with reduced uptake. However, resistance to 4'-(9-acridinylamino)methanesulfon-m-aniside and 2-methyl-9-hydroxyellipticinium was observed in the absence of changes in drug uptake, suggesting a second mode of resistance. DC3F/9-OHE cells formed fewer protein-associated DNA strand breaks in response to 4'-(9-acridinylamino)methanesulfon-m-aniside, 2-methyl-9-hydroxyellipticinium, or etoposide than did the sensitive parental cells. The same was true for isolated nuclei from these cells, which is consistent with a mode of resistance unrelated to drug uptake through the plasma membrane. These data suggest that resistance to DNA topoisomerase II inhibitors exhibited by DC3F/9-OHE cells is due in part to a modification of topoisomerase II activity.


Asunto(s)
Alcaloides/toxicidad , Aminoacridinas/toxicidad , Reparación del ADN , ADN/metabolismo , Elipticinas/toxicidad , Podofilotoxina/toxicidad , Inhibidores de Topoisomerasa II , Amsacrina , Animales , Transporte Biológico , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cricetinae , Cricetulus , Resistencia a Medicamentos , Pulmón , Proteínas/metabolismo
9.
Cancer Res ; 46(11): 5821-6, 1986 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3019538

RESUMEN

Ditercalinium, a 7H-pyridocarbazole dimer (bisintercalator) belongs to a new class of antineoplastic intercalating agents. To investigate its mechanism of cytotoxicity, the effects of ditercalinium on DNA were assessed using normal (L1210) and drug-resistant (L1210/PyDi1) mouse leukemia cells. Alkaline elution assays demonstrated that ditercalinium produced no DNA strand breaks, DNA-protein cross-links, or DNA-DNA cross-links, eliminating these effects as cytotoxic lesions. This result sets ditercalinium apart from other intercalating agents with respect to its interaction with DNA. Nucleoids (histone-depleted chromatin) from ditercalinium-treated L1210 cells were considerably more compact than those from untreated cells, as determined by sedimentation in neutral sucrose gradients. In contrast, nucleoids from ditercalinium-treated L1210/PyDi1 (resistant) cells were similar in compactness to those from control cells. Thus, ditercalinium altered chromatin structure in vivo. The effect of the bisintercalator on purified DNA topoisomerase II, an intracellular target of monointercalators, was measured in vitro. Ditercalinium (5 X 10(-7) M) completely inhibited both the formation of covalent complexes between this enzyme and simian virus 40 DNA and the enzyme-induced DNA cleavage. In addition, ditercalinium induced DNA catenation in the presence of topoisomerase II and adenosine triphosphate. Thus, the cytotoxicity of ditercalinium may derive from a mechanism that, although involving topoisomerase II, is manifested by condensation of DNA rather than by the induction of protein-associated DNA strand breaks.


Asunto(s)
Carbazoles/farmacología , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Sustancias Intercalantes/farmacología , Inhibidores de Topoisomerasa II , Amsacrina/farmacología , Animales , Reactivos de Enlaces Cruzados/farmacología , Etopósido/farmacología , Leucemia L1210/enzimología , Ratones , Conformación de Ácido Nucleico/efectos de los fármacos , Relación Estructura-Actividad
10.
Cancer Res ; 47(7): 1793-8, 1987 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-3028614

RESUMEN

Results of filter elution assays of lesions produced in the DNA of cultured L1210 cells by the antineoplastic alkaloid camptothecin support the notion that topoisomerase I is an intracellular target of this drug. One to 10 microM camptothecin induced DNA single-strand, but not double-strand, breaks when incubated with intact cells or with their isolated nuclei. Approximately one half of the strand breakage was protein concealed, as judged by filter elution. Camptothecin-induced, protein-concealed DNA strand breaks disappeared rapidly after drug removal. DNA-protein cross-links were generated by camptothecin with frequencies approximately equal to those of protein-concealed DNA strand breaks. It is likely that camptothecin can inhibit topoisomerase I in intact cells in a manner similar to that in which other antineoplastic agents such as amsacrine or teniposide inhibit topoisomerase II. DNA-breaking lesions other than those resulting from trapped topoisomerase I-DNA complexes may also be generated by camptothecin. The yields of DNA strand breaks induced by camptothecin, amsacrine, or teniposide were approximately doubled when cells were incubated for 16 h with 3-aminobenzamide, an inhibitor of poly(ADP ribosylation) of proteins, prior to 1-h exposure to the antineoplastic compounds. 3-Aminobenzamide also enhanced the cytotoxic action of camptothecin, amsacrine, and teniposide. These results suggest that protein-concealed strand breaks can be lethal lesions and that intracellular topoisomerase I and II activity may be regulated coordinately through poly(ADP ribosylation).


Asunto(s)
Camptotecina/toxicidad , Leucemia L1210/patología , Inhibidores de Topoisomerasa I , Animales , Daño del ADN , ADN de Neoplasias/aislamiento & purificación , ADN de Neoplasias/efectos de la radiación , Leucemia L1210/enzimología , Ratones , Proteínas de Neoplasias/efectos de la radiación
11.
Cancer Res ; 43(12 Pt 1): 5718-24, 1983 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-6416668

RESUMEN

The free radical scavengers, dimethyl sulfoxide (Me2SO) and thiourea, were used to assess the role of free radicals in the production of intercalator-induced DNA breaks and cytotoxicity in mouse leukemia L1210 cells. Both agents decreased X-ray break production, and this decrease was comparable in magnitude to the degree of inhibition of X-ray-induced cell killing. By contrast, Me2SO increased the DNA breaks produced by the intercalators, Adriamycin, 5-iminodaunorubicin, and 4'-(9-acridinylamino)methanesulfon-m-anisidide. This was not due to an enhancement of Adriamycin or 4'-(9-acridinylamino)methanesulfon-m-anisidide uptake by Me2SO. Strand break production by intercalators was decreased by thiourea. This was not due to an inactivation of the intercalators or to a decrease of Adriamycin or 4'-(9-acridinylamino)methanesulfon-m-anisidide uptake by thiourea. Experiments using nucleoid sedimentation to assess the DNA linking number and domain size from cells treated with Me2SO and thiourea indicated that these chemicals alter chromatin structure in a fashion which may account for effects on intercalator-induced DNA scission. The alterations in intercalator-induced DNA scission were not accompanied by corresponding alterations in cytotoxicity, thus dissociating intercalator-induced strand break production from lethality and the mechanism of X-ray break production.


Asunto(s)
ADN de Neoplasias/genética , ADN de Cadena Simple/genética , Dimetilsulfóxido/farmacología , Sustancias Intercalantes/toxicidad , Leucemia L1210/fisiopatología , Tiourea/farmacología , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Radicales Libres , Ratones , Relación Estructura-Actividad
12.
Cancer Res ; 46(11): 5486-93, 1986 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3756897

RESUMEN

We have previously reported the cytotoxicity and antitumor activity of bis(diphenylphosphino)ethane (DPPE) and a variety of its transition metal complexes. During studies of the chemistry of a gold complex of this group [(AuCl)2(DPPE)], it was observed that this complex readily underwent ring closure on reaction with DPPE to form the tetrahedral complex [Au(DPPE)2]+. Various counterion forms (e.g., Cl-) of this cation were isolated and were found to exhibit a remarkably high stability in solution. Evaluation of [Au(DPPE)2]Cl in mice bearing i.p. P388 leukemia demonstrated that the compound produced an average of 87% increase in life span at its maximally tolerated dose (2-3 mumol/kg/day for 5 days). Activity was also seen in i.p. M5076 reticulum cell sarcoma (60% increase in life span) and s.c. mammary adenocarcinoma 16/c. Modest activity was evident in i.p. B16 melanoma and L1210 leukemia. A subline of P388 leukemia resistant to cisplatin was not cross-resistant to [Au(DPPE)2]Cl. In addition, combination therapy of [Au(DPPE)2]Cl and cisplatin against i.p. P388 demonstrated an advantage over single-agent therapy. In vitro studies of [Au(DPPE)2]Cl showed that the compound: is cytotoxic to tumor cell lines; is only minimally inhibited in its cytotoxic activity by the presence of serum; produces DNA protein cross-links and DNA strand breaks in cells; and inhibits macromolecular synthesis with a preferential inhibitory effect on protein synthesis relative to DNA and RNA synthesis. 31P nuclear magnetic resonance spectroscopy indicated that the compound is stable in the presence of serum proteins, thiols, or disulfides and that it reacts with Cu(II) resulting in the formation of a Cu(I)DPPE complex. The results of these in vivo and in vitro experiments suggest that the contrasting pharmacological profile of [Au(DPPE)2]Cl with respect to other gold(I) phosphine complexes may be related to both the kinetic stability of the complex and its stability in the presence of thiols.


Asunto(s)
Antineoplásicos , Oro/uso terapéutico , Leucemia P388/tratamiento farmacológico , Leucemia Experimental/tratamiento farmacológico , Compuestos Organometálicos , Compuestos Organofosforados/uso terapéutico , Sarcoma Experimental/tratamiento farmacológico , Animales , Supervivencia Celular/efectos de los fármacos , Fenómenos Químicos , Química , Cisplatino/administración & dosificación , Cobre , Sulfato de Cobre , ADN/efectos de los fármacos , Leucemia L1210/tratamiento farmacológico , Espectroscopía de Resonancia Magnética , Melanoma Experimental/tratamiento farmacológico , Ratones , Ácidos Nucleicos/biosíntesis , Compuestos Orgánicos de Oro , Biosíntesis de Proteínas
13.
Oncogene ; 18(28): 4047-54, 1999 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10435585

RESUMEN

In response to DNA damage and replication blocks, cells activate pathways that arrest the cell cycle and induce the transcription of genes that facilitate repair. In mammals, ATM (ataxia telangiectasia mutated) kinase together with other checkpoint kinases are important components in this response. We have cloned the rat and human homologs of Saccharomyces cerevisiae Rad 53 and Schizosaccharomyces pombe Cds1, called checkpoint kinase 2 (chk2). Complementation studies suggest that Chk2 can partially replace the function of the defective checkpoint kinase in the Cds1 deficient yeast strain. Chk2 was phosphorylated and activated in response to DNA damage in an ATM dependent manner. Its activation in response to replication blocks by hydroxyurea (HU) treatment, however, was independent of ATM. Using mass spectrometry, we found that, similar to Chk1, Chk2 can phosphorylate serine 216 in Cdc25C, a site known to be involved in negative regulation of Cdc25C. These results suggest that Chk2 is a downstream effector of the ATM-dependent DNA damage checkpoint pathway. Activation of Chk2 might not only delay mitotic entry, but also increase the capacity of cultured cells to survive after treatment with gamma-radiation or with the topoisomerase-I inhibitor topotecan.


Asunto(s)
Daño del ADN , Reparación del ADN/genética , Proteínas Quinasas , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas/fisiología , ras-GRF1 , Alquilantes/farmacología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Quinasa de Punto de Control 2 , Clonación Molecular , ADN Complementario/genética , ADN de Hongos/efectos de los fármacos , ADN de Hongos/genética , ADN de Hongos/efectos de la radiación , Proteínas de Unión al ADN , Inhibidores Enzimáticos/farmacología , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/fisiología , Rayos gamma , Prueba de Complementación Genética , Humanos , Hidroxiurea/farmacología , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/efectos de la radiación , Schizosaccharomyces/efectos de los fármacos , Schizosaccharomyces/genética , Schizosaccharomyces/efectos de la radiación , Proteínas de Schizosaccharomyces pombe , Transducción de Señal , Especificidad de la Especie , Inhibidores de Topoisomerasa I , Topotecan/farmacología , Proteínas Supresoras de Tumor
14.
Biochim Biophys Acta ; 563(2): 293-305, 1979 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-465491

RESUMEN

Chinese hamster ovary cells labelled with [14C]thymidine were made permeable, incubated with various concentrations of the intercalating dye ethidium bromide, and centrifuged through neutral sucrose gradients. The gradient profiles of these cells were qualitatively similar to those obtained by centrifuging DNA from untreated, lysed permeable cells through gradients containing ethidium bromide. The sedimentation distance of DNA had a biphasic dependence on the concentration of ethidium bromide, suggesting that the dye altered the amount of DNA supercoiling in situ. The effect of ethidium bromide intercalation on incorporation of [3H]dTMP into acid-precipitable material in an in vitro DNA synthesis mixture was measured. The incorporation of [3H]dTMP was unaffected by less than 1 microgram/ml of ethidium bromide, enhanced up to two-fold by 1--10 microgram/ml, and inhibited by concentrations greater than 10 micrograms/ml. Alkaline sucrose gradient analysis revealed a higher percentage of small DNA fragments (6--20 S) in the cells treated with 2 micrograms/ml ethidium bromide than in control cells. These fragments attained parental size within the same time as the fragments in control cells. In cells treated with 2 micrograms/ml ethidium bromide, a significant fraction of newly synthesized DNA resulted from new starts, whereas in untreated cells practically none of the newly synthesized DNA resulted from new starts. These results suggest that relaxation of DNA supercoiled structures ahead of the replication fork generates spurious initiations of DNA synthesis and that in intact cells the rate of chain elongation is limited by supercoiled regions ahead of the growing point.


Asunto(s)
Replicación del ADN/efectos de los fármacos , ADN Superhelicoidal/metabolismo , ADN/biosíntesis , Etidio/farmacología , Animales , Línea Celular , Cricetinae , Femenino , Cinética , Ovario , Timidina Monofosfato/metabolismo
15.
Biochim Biophys Acta ; 563(2): 306-12, 1979 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-465492

RESUMEN

Novobiocin, an inhibitor of gyrase-induced DNA supercoiling and DNA replication in prokaryotes, inhibited the incorporation of DNA precursors into DNA in both intact and permeable Chinese hamster ovary cells; much higher concentrations were required for permeable cells, in which no new replicons were initiated. Nucleoids were prepared from cells that were incubated for 60 min with 200 micrograms/ml novobiocin, made permeable, and incubated with 0--50 micrograms/ml ethidium bromide. Sedimentation of the nucleoids in neutral sucrose gradients suggested that the number of supercoils in the average nucleoid had been reduced by prior incubation with novobiocin. In intact cells, novobiocin is required inside the cell for continued inhibition of DNA synthesis, suggesting that it does not act directly on the DNA. Alkaline sucrose gradient profiles of DNA synthesized in the presence of novobiocin in intact cells indicated that the drug inhibited replicon initiation while having little if any effect on chain elongation. These data are consistent with the idea that an activity similar to the bacterial gyrase generates supercoils in mammalian DNA and produces the proper conformation for the initiation of DNA replication.


Asunto(s)
Replicación del ADN/efectos de los fármacos , ADN Superhelicoidal/metabolismo , ADN/biosíntesis , Novobiocina/farmacología , Animales , Línea Celular , Cricetinae , Etidio/farmacología , Femenino , Cinética , Ovario
16.
Biochim Biophys Acta ; 653(2): 248-58, 1981 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-6261824

RESUMEN

Novobiocin and nalidixic acid, inhibitors of the bacterial enzyme DNA gyrase, inhibit DNA, RNA and protein synthesis in several human and rodent cell lines. The sensitivity of DNA synthesis (both replicative and repair) to inhibition by novobiocin and nalidixic acid is greater than that of protein synthesis. Novobiocin inhibits RNA synthesis about half as effectively as it does DNA synthesis, whereas nalidixic acid inhibits both equally well. Replicative DNA synthesis, as measured by incorporation of [3H]thymidine, is blocked by novobiocin in a number of cell strains; the inhibition is reversible with respect to both DNA synthesis and cell killing, and continues for as long as 20--30 h if the cells are kept in novobiocin-containing growth medium. Both novobiocin and nalidixic acid inhibit repair DNA synthesis (measured by BND-cellulose chromatography) induced by ultraviolet light or N-methyl-N'-nitro-N-nitrosoguanidine (but not that induced by methyl methanesulfonate) at lower concentration (as low as 5 micrograms/ml) than those required to inhibit replicative DNA synthesis (50 micrograms/ml or greater). Neither novobiocin nor nalidixic acid alone induces DNA repair synthesis. Incubation of ultraviolet-irradiated cells with 10--100 micrograms/ml novobiocin results in little, if any, further reduction of colony-forming ability (beyond that caused by the ultraviolet irradiation). Novobiocin at sufficiently low concentrations (200 micrograms/ml) apparently generates a quiescent state (in terms of cellular DNA metabolism) from which recovery is possible. Under more drastic conditions of time in contact with cells and concentration, however, novobiocin itself induces mammalian cell killing.


Asunto(s)
Reparación del ADN , Replicación del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/metabolismo , ADN Superhelicoidal/metabolismo , ADN/biosíntesis , Ácido Nalidíxico/farmacología , Novobiocina/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Humanos , Cinética , Metilnitronitrosoguanidina/farmacología , Xerodermia Pigmentosa
17.
Biochim Biophys Acta ; 395(1): 48-55, 1975 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-166695

RESUMEN

Three mammalian cell lines (WI-38, SV40-transformed WI-38 and Chinese hamster ovary) were exposed to high doses of 137-Cs gamma rays and their DNA analysed, following various periods of postirradiation incubation, for products of the 5,6-dihydroxy-dihydrothymine type. Within fifteen minutes of incubation at 37 degrees C 70 to 90 percent of these radiation products were removed from acid-precipitable material in all three cell lines. The amount of DNA degradation induced by radiation varied from approximately one percent in WI-38 cells to 15 percent in SV40-transformed WI-38 cells. Comparison of DNA degradation with the amount of thymine radiation product removed indicates that a selective gamma ray-induced excision repair capability exists in mammalian cells. Because of its more rapid kinetics, gamma ray excision repair is probably a distinct process as compared with ultraviolet-induced pyrimidine dimer excision.


Asunto(s)
ADN/efectos de la radiación , Efectos de la Radiación , Timina/efectos de la radiación , Animales , Línea Celular , Transformación Celular Neoplásica , Cricetinae , ADN/metabolismo , Femenino , Pulmón/embriología , Ovario , Virus 40 de los Simios , Timina/metabolismo , Factores de Tiempo
18.
FEBS Lett ; 276(1-2): 17-20, 1990 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-2176162

RESUMEN

Cultured rat mesangial cells contain high affinity endothelin (ET) receptors at high densities. Exposure of these cells to ET resulted in a transient activation of topoisomerase I extractable activity, which reached its maximum value at approximately 2 min and returned to basal value after approximately 10 min of treatment. The activation of this enzyme was dependent upon the concentration of ET added. Incubation of the cells with pertussis toxin inhibited ET-induced increases in topoisomerase I activity in a concentration-dependent manner, suggesting that involvement of pertussis toxin-sensitive GTP-binding protein in ET-mediated action. Endothelin had no detectable effect upon extractable topoisomerase II activity.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , Endotelinas/farmacología , Mesangio Glomerular/enzimología , Corteza Renal/enzimología , Toxina del Pertussis , Factores de Virulencia de Bordetella/farmacología , Animales , Células Cultivadas , ADN-Topoisomerasas de Tipo I/aislamiento & purificación , Activación Enzimática , Mesangio Glomerular/efectos de los fármacos , Cinética , Ratas , Ratas Endogámicas , Receptores de Superficie Celular/fisiología , Receptores de Endotelina , Inhibidores de Topoisomerasa I
19.
J Med Chem ; 32(3): 715-20, 1989 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2537428

RESUMEN

Several camptothecin derivatives containing a modified hydroxy lactone ring have been synthesized and evaluated for inhibition of topoisomerase I and cytotoxicity to mammalian cells. Each of the groups of the hydroxy lactone moiety, the carbonyl oxygen, the ring lactone oxygen, and the 20-hydroxy group, were shown to be critical for enzyme inhibition. For example the lactol, lactam, thiolactone, and 20-deoxy derivatives did not stabilize the covalent DNA-topoisomerase I complex. With a few exceptions, those compounds that did not inhibit topoisomerase I were not cytotoxic to mammalian cells. Two cytotoxic derivatives that did not inhibit topoisomerase I were shown to produce non-protein-associated DNA single-strand breaks and are likely to have a different mechanism of action. One of these compounds was tested for antitumor activity and was found to be inactive. The present findings, as well as other reports that the hydroxy lactone ring of camptothecin is critical for antitumor activity in vivo, correlate with the structure-activity relationships at the level of topoisomerase I and support the hypothesis that antitumor activity is related to inhibition of this target enzyme.


Asunto(s)
Antineoplásicos Fitogénicos/síntesis química , Camptotecina/análogos & derivados , Inhibidores de Topoisomerasa I , Animales , Antineoplásicos Fitogénicos/farmacología , Camptotecina/síntesis química , Camptotecina/farmacología , Fenómenos Químicos , Química , Ensayos de Selección de Medicamentos Antitumorales , Leucemia L1210/tratamiento farmacológico , Ratones , Relación Estructura-Actividad
20.
Org Lett ; 3(25): 4047-9, 2001 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-11735581

RESUMEN

[structure: see text] Bioassay-guided fractionation of the plant Acacia aulacocarpa, guided by a bioassay for Tie2 tyrosine kinase activity, yielded the novel triterpene 3,21-dioxo-olean-18-en-oic acid (1) as the first naturally occurring non-protein inhibitor of Tie2 kinase. The structure of 1 was assigned by analysis of spectral data. In addition to its activity as an inhibitor of Tie2 kinase, compound 1 also shows modest activity against a variety of cultured mammalian cells.


Asunto(s)
Acacia/química , Inhibidores Enzimáticos/química , Ácido Oleanólico/química , Extractos Vegetales/química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Triterpenos/química , Animales , Células Cultivadas , Inhibidores Enzimáticos/aislamiento & purificación , Inhibidores Enzimáticos/farmacología , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Estructura Molecular , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/aislamiento & purificación , Ácido Oleanólico/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor TIE-2 , Triterpenos/aislamiento & purificación , Triterpenos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA