Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arch Virol ; 149(6): 1083-94, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15168196

RESUMO

Herpesvirus Macaca arctoides (HVMA), an Epstein-Barr virus (EBV)-related herpesvirus of macaque origin, induces malignant lymphomas in rabbits. To get more insights into the oncogenesis of the EBV/HVMA infection the aim of the present study was to prove the in vitro transforming ability of HVMA for rabbit lymphocytes as well as human umbilical cord blood lymphocytes. As a result, B-cell transformation could be demonstrated after infection with HVMA in all mononuclear cell samples of 20 rabbits. The transformation was evaluated microscopically and confirmed by the expression of EBV-related nuclear antigens. The transforming activity led to the establishment of permanent rabbit lymphoblastoid cell lines cultured up to more than 90 passages. The cell lines contained EBV-like HVMA-DNA. Interestingly, the transformed rabbit lymphocytes showed chromosomal abnormalities with a subtetraploid karyotype. The low extent of lytic cycle-dependent expression of virus capsid antigen in the established cell lines increased after treatment with the inducing agents iododeoxyuridine and mitomycin C. In contrast, no transformation could be induced after exposure of human umbilical cord blood lymphocytes to HVMA. The permanent rabbit lymphoblastoid cell lines provide a model for further studies on the role of EBV/HVMA in oncogenesis of lymphomas. In addition, it might be suitable for testing potential antiviral compounds in vitro.


Assuntos
Transformação Celular Viral , Herpesvirus Humano 4/patogenicidade , Linfócitos/virologia , Animais , Antígenos Virais/análise , Linfócitos B/virologia , Proteínas do Capsídeo/análise , Linhagem Celular , Núcleo Celular/imunologia , Aberrações Cromossômicas , DNA Viral/análise , Imunofluorescência , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/imunologia , Humanos , Linfócitos/metabolismo , Linfócitos/patologia , Macaca/virologia , Reação em Cadeia da Polimerase , Coelhos , Fatores de Tempo
2.
Mutagenesis ; 19(1): 27-33, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14681310

RESUMO

Due to the very limited transduction capacity of hitherto available vectors, the success of gene therapy of tumours by means of suicide genes has so far essentially depended on the transfer of toxic drug metabolites from transduced (metabolizing) cells to adjacent non-transduced cells via gap junctions (bystander effect). Most experimental systems for the detection of a bystander effect yield net data of cell losses and cannot differentiate between killed transduced versus killed bystander cells. Here we report on metabolic cooperation in vitro between CHO cells stably transfected with the thymidine kinase gene of herpes simplex virus type-1 (HSVtk) (metabolizing cells) and Swiss albino 3T3 cells (bystander cells). Both cell lines are readily distinguishable by single cell and colony morphology and by their chromosomal constitution. While 3T3 cells cultured alone were refractory to the antiviral drug ganciclovir (GCV), co-culture with CHO-HSVtk(+) cells led to a dramatic reduction in plating efficiency as well as to a 4-fold increase in sister chromatid exchange rates induced by very low GCV concentrations in the 3T3 bystander cells. The modulator of gap junctional cooperation, all-trans retinoic acid, caused a strong augmentation of the bystander effect, while 18alpha-glycyrrhetinic acid, an inhibitor of gap junctional communication, drastically diminished the toxicity of GCV in the bystander cells. Whereas CHO-HSVtk(+) cells showed a distinct immunoreactivity for connexin43 in the cell membranes, 3T3 cells were almost negative. The co-culture system described here allows unequivocal distinction between metabolizing and bystander cells. In this way, mechanistic aspects of the transfer of genotoxic/cytotoxic metabolites to cells, which per se are unable to form them, become accessible to investigation.


Assuntos
Efeito Espectador , Análise Citogenética/métodos , Ganciclovir/metabolismo , Troca de Cromátide Irmã/efeitos dos fármacos , Timidina Quinase/genética , Células 3T3 , Animais , Células CHO , Conexina 43/efeitos dos fármacos , Conexina 43/metabolismo , Cricetinae , Ganciclovir/farmacologia , Junções Comunicantes/efeitos dos fármacos , Terapia Genética/métodos , Ácido Glicirretínico/análogos & derivados , Ácido Glicirretínico/farmacologia , Herpesvirus Humano 1/genética , Camundongos , Transfecção
3.
Neoplasma ; 50(3): 165-71, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12937848

RESUMO

A new cell line, designated as Tuwei00, is described. It originated from an Epstein-Barr virus-positive skin tumor biopsy of a heart transplant recipient, whose numerous cutaneous neoplasms were treated with the antiviral drug cidofovir what caused at least transient remissions. The cell line was established in vitro and maintained for more than 70 passages. Cells of early passages were characterized by a slower growth, the inability to form colonies and a higher sensitivity to cidofovir. After overcoming a crisis, the cells grew faster, to a higher density and were able to form adherent colonies from single cells as well as colonies in soft agar. Chromosome analysis showed diploidy/hyperdiploidy at the earlier and hypodiploidy at the later passages. Sensitivity to cidofovir was distinctly higher in early passages of Tuwei00 cells than in later passages and was characterized by distinct decline of cell survival after long term cidofovir exposure. Established normal human keratinocytes, HaCaT cells, which were checked for comparison, showed a low cidofovir sensitivity similar to late passage Tuwei00 cells.


Assuntos
Antivirais/uso terapêutico , Citosina/análogos & derivados , Citosina/uso terapêutico , Transplante de Coração , Organofosfonatos , Compostos Organofosforados/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Idoso , Biópsia , Divisão Celular/efeitos dos fármacos , Aberrações Cromossômicas , Cidofovir , DNA Viral/metabolismo , Resistencia a Medicamentos Antineoplásicos , Herpesvirus Humano 4/isolamento & purificação , Humanos , Técnicas Imunoenzimáticas , Cariotipagem , Queratinócitos/efeitos dos fármacos , Queratinócitos/patologia , Masculino , Neoplasias Cutâneas/virologia , Células Tumorais Cultivadas
4.
Cancer Res ; 61(20): 7399-403, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11606369

RESUMO

The efficacy of suicide herpes simplex virus-1 thymidine kinase (HSVtk)/ganciclovir (GCV) gene therapy is often limited by intrinsic resistance of tumor cells. Here we show that repair of GCV incorporated in DNA is a factor involved in GCV resistance. A protective role of DNA repair in GCV-induced cell killing is supported by the following findings: (a) GCV-exposed Chinese hamster ovary-HSVtk cells exhibited both reduced repair of GCV and cloning efficiency in the presence of a specific polymerase beta (beta-pol) inhibitor, prunasin; (b) DNA beta-pol-deficient mouse fibroblasts were more sensitive to the cytotoxic, apoptosis-inducing, and genotoxic (DNA breakage and chromosomal aberration-inducing) effects of GCV as compared with wild-type and beta-pol-complemented cell lines; (c) methoxyamine, an inhibitor of beta-pol-dependent short-patch base excision repair, sensitized wild-type and complemented beta-pol cells to GCV, whereas it had no effect on the sensitivity of beta-pol-null cells to GCV. Because methoxyamine-mediated sensitization of beta-pol wild-type and beta-pol-complemented cells to GCV did not reach the level of null cells, we suggest that both beta-pol-dependent short- and long-patch base excision repair are involved in protection of cells to GCV. Some implications for HSVtk/GCV gene therapy are being discussed.


Assuntos
Dano ao DNA , DNA Polimerase beta/metabolismo , Ganciclovir/toxicidade , Animais , Células CHO , Cricetinae , DNA/metabolismo , DNA Polimerase beta/antagonistas & inibidores , DNA Polimerase beta/deficiência , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Ganciclovir/metabolismo , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Hidroxilaminas/farmacologia , Camundongos , Camundongos Knockout , Nitrilas/farmacologia , Timidina Quinase/genética , Timidina Quinase/metabolismo , Transfecção
5.
Antiviral Res ; 49(2): 55-74, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11248359

RESUMO

The available informations on the genotoxic effects in experimental systems of the antiherpesvirus nucleosides aciclovir, penciclovir, ganciclovir, brivudine and cidofovir as well as of the antiretrovirals zidovudine (AZT), lamivudine, zalcitabine (ddC), didanosine and stavudine are reviewed. Furthermore, data on carcinogenic activity of these drugs in laboratory rodents are compiled. Most nucleoside analogue antivirals induce chromosomal aberrations but are inactive in gene mutation assays. Carcinogenicity findings in mice and rats are variable but clearly positive for AZT and ddC. The possible mechanisms by which these agents may cause damage in the genetic information are still largely hypothetical, and experimental findings do not permit relevant extrapolations to the situation in man. There is no conclusive evidence that any of the drugs caused tumours in humans. The use of nucleoside analogues in antiviral therapy remains a pragmatic option that seems justified by risk/benefit assessment.


Assuntos
Antivirais/efeitos adversos , Carcinógenos , Mutagênicos , Nucleosídeos/efeitos adversos , Animais , Antivirais/química , Antivirais/toxicidade , Testes de Carcinogenicidade , Aberrações Cromossômicas , Humanos , Camundongos , Testes de Mutagenicidade , Nucleosídeos/química , Nucleosídeos/toxicidade
6.
Ophthalmic Res ; 32(2-3): 118-25, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10754445

RESUMO

Povidone-iodine (PVP-I) is a broad-spectrum microbicide with in vitro activity against bacteria, viruses, fungi and protozoans. A 5% solution of PVP-I proved to be highly effective in ophthalmic surgery for the prophylaxis of endophthalmitis. For the antiseptic treatment of eye infections a novel application form of PVP-I has been developed by using a PVP-I liposome complex which demonstrated an excellent antimicrobial efficacy. In this study it could be shown that the novel liposomal formulations containing 2.5 or 5% PVP-I were as active as the aqueous solution against herpes simplex virus type 1, adenovirus type 8, coxsackievirus A9 and Chlamydia trachomatis in cell culture referring to equal PVP-I concentrations. Long-term cytotoxicity experiments demonstrated a moderate cytotoxicity for both formulations with a better tolerability of the liposomal PVP-I formulation compared with the aqueous solution. There is no evidence for a genotoxic activity of these agents.


Assuntos
Adenovírus Humanos/efeitos dos fármacos , Anti-Infecciosos Locais/farmacologia , Chlamydia trachomatis/efeitos dos fármacos , Enterovirus/efeitos dos fármacos , Herpesvirus Humano 1/efeitos dos fármacos , Soluções Oftálmicas/farmacologia , Povidona-Iodo/farmacologia , Animais , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Humanos , Lipossomos , Masculino , Coelhos , Células Vero
7.
Cancer Gene Ther ; 7(1): 107-17, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10678363

RESUMO

We studied the genotoxic and apoptosis-inducing properties of ganciclovir (GCV) and penciclovir (PCV) using Chinese hamster ovary cells stably transfected with the thymidine kinase (tk) gene of herpes simplex virus-1 (HSV-1). Cells expressing HSVtk were 300 and 100 times more sensitive than their isogenic HSVtk- counterparts to the cytotoxic effects of GCV and PCV, respectively. Using radiolabeled drugs, GCV was found to be incorporated into the genomic DNA much more effectively than PCV. GCV was highly potent in inducing chromosomal aberrations compared with PCV, which provoked less sister chromatid exchanges and chromosomal changes using equimolar or equitoxic doses. For both agents, apoptosis was shown to be the major route of cell killing. Time course experiments revealed that neither genotoxicity nor apoptosis were induced within the cell cycle exposed to the drug; they are late events provoked in the following cell cycle(s). This indicates that the incorporation/exposure step of GCV or PCV into DNA is not decisive for triggering genotoxicity and apoptosis, but that events occurring subsequently, presumably during replication of a DNA containing the nucleotide analogs, are of major importance. Because PCV, unlike GCV, induced highly effectively apoptosis without exerting much genotoxicity, the use of PCV as a relatively safe alternative drug for suicide gene therapy of malignant diseases is recommended.


Assuntos
Aciclovir/análogos & derivados , Apoptose , Ganciclovir/farmacologia , Terapia Genética , Timidina Quinase/genética , Aciclovir/metabolismo , Aciclovir/farmacologia , Animais , Células CHO , Ciclo Celular/efeitos dos fármacos , Cricetinae , DNA/metabolismo , Replicação do DNA/efeitos dos fármacos , Ganciclovir/metabolismo , Guanina , Herpesvirus Humano 1/enzimologia , Testes de Mutagenicidade , Necrose , Troca de Cromátide Irmã/efeitos dos fármacos , Timidina Quinase/metabolismo , Transfecção
8.
Mutagenesis ; 15(2): 177-84, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10719044

RESUMO

The three anti-herpes nucleoside analogues ganciclovir, penciclovir and aciclovir were investigated as to their recombinogenic [sister chromatid exchange (SCE) inducing] and clastogenic activity in CHO cells expressing the thymidine kinase gene of HSV-1, which is a precondition of therapeutic activity of these drugs. The compounds were applied for the duration of one cell cycle and cytogenetic end-points were measured between 0 and 42 h after exposure. Although the nucleoside analogues are quite similar with respect to chemical structure, they differ basically in their genotoxic potency, aberration types induced as well as the time course of chromosomal damage. Aciclovir induced SCEs and chromosomal aberrations immediately after exposure but only in a concentration range much higher than that reached in blood plasma during anti-herpes therapy. The direct genotoxic activity is explained by the obligate chain terminating property of aciclovir upon incorporation into genomic DNA. On the other hand, genotoxic damage caused by ganciclovir and penciclovir is of the delayed type requiring at least one post-exposure cell cycle for its expression. Unlike aciclovir, ganciclovir is an extremely potent inducer of SCEs and chromosome breaks and translocations at concentrations far below those impairing the proliferative activity and triggering apoptosis of the target cells (as shown by our previous investigation). Penciclovir is essentially devoid of genotoxic activity. It induces SCEs only at cytotoxic/apoptotic concentrations, is only weakly clastogenic and induces premature chromosome condensation which appears to result from uncoupling of karyokinesis and cytokinesis. The genotoxic activity of ganciclovir is explained as due to repair processes triggered in the second post-exposure replication cycle at the sites of nucleoside analogue incorporation into genomic DNA. The findings have considerable implications with respect to the use of ganciclovir or other antiviral drugs in suicide gene therapy of malignant diseases.


Assuntos
Aciclovir/análogos & derivados , Aciclovir/toxicidade , Antivirais/toxicidade , Ganciclovir/toxicidade , Herpesvirus Humano 1/metabolismo , Timidina Quinase/metabolismo , Animais , Células CHO , Cromossomos/efeitos dos fármacos , Cricetinae , Relação Dose-Resposta a Droga , Guanina , Mutagênicos/toxicidade , Troca de Cromátide Irmã/efeitos dos fármacos , Timidina Quinase/genética , Fatores de Tempo
9.
Antiviral Res ; 31(1-2): 105-13, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8793014

RESUMO

The antiherpes virostatics acyclovir (ACV), valaciclovir (VACV), penciclovir (PCV), famciclovir (FCV) and ganciclovir (GCV), which belong to the group of purine acyclic nucleoside analogues, were tested for clastogenic and sister chromatid exchange (SCE)-inducing activity in Chinese hamster V79-E cells upon chronic application with and without a recovery period. ACV induced borderline effects in both cytogenetic assays, a dose-dependent reduction of the mitotic index and an increasing cell cycle delay. With VACV and PCV only a decrease of the mitotic index and an increase of cell cycle delay were observed. FCV was negative with respect to the four parameters studied, presumably due to the incapacity of the target cells of metabolizing FCV to PCV. GCV was a very potent genotoxin in both assays. It induced a statistically significant SCE response even in the range of the cytomegalovirus IC50 of < 10 microM. By variation of the experimental protocol it was shown that SCEs are induced in the second cell cycle following exposure to GCV but not in the first one. It is assumed that the drugs under study are metabolized to their respective triphosphates and then inhibit DNA replication as detected by decreasing mitotic index and increasing cell cycle delay. In the case of GCV it is suggested that GCV-TP is incorporated into the target cell DNA and that chromosomal aberrations and SCEs are secondary lesions due to repair processes at the substituted template.


Assuntos
2-Aminopurina/análogos & derivados , Aciclovir/análogos & derivados , Ganciclovir/toxicidade , Mutagênicos/toxicidade , Valina/análogos & derivados , 2-Aminopurina/toxicidade , Aciclovir/toxicidade , Animais , Células CHO , Cricetinae , Famciclovir , Guanina , Testes de Mutagenicidade , Valaciclovir , Valina/toxicidade
10.
Int J Cancer ; 65(4): 506-12, 1996 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-8621235

RESUMO

The DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) removes alkyl groups from the O6 position of guanine in DNA and thus may protect cells against genotoxic effects of agents inducing this lesion. To analyze quantitatively the level of protection mediated by MGMT against antineoplastic drugs, we determined the cytotoxic and recombinogenic (sister-chromatid exchange inducing) effects of various chemotherapeutic agents in a pair of isogenic Chinese hamster cell lines deficient and proficient for MGMT, generated upon transfection with human MGMT cDNA. Furthermore, we compared the responses of the human cell lines HeLa MR (MGMT deficient) and HeLa S3 (MGMT proficient) to the various agents. It is shown that: (1) MGMT proficient cells are resistant in cell killing to the methylating drug streptozotocin and all the chloroethylating nitrosoureas tested. There was a marked agent specificity in protection. The level of resistance provoked by MGMT increased in the order BCNU < CCNU < ACNU < HeCNU < streptozotocin. (2) MGMT did not protect cells against killing induced by chlorambucil, cisplatin, melphalan, activated cyclophosphamide (malosfamide) and activated ifosfamide (4-hydroperoxy-ifosfamide). (3) MGMT caused protection against the recombinogenic effect of all nitrosoureas tested. The lowest level of protection was again observed for BCNU, followed by CCNU, ACNU < HeCNU < streptozotocin. (4) MGMT proficient cells did not exhibit resistance in SCE induction towards cyclophosphamide (activated by microsomes), 4-hydroperoxy-ifosfamide, mafosfamide, chlorambucil and melphalan. Some protection was afforded, however, against cisplatin (and transplatin). This effect was abolished by pretreatment of cells with O6-benzylguanine, which depletes MGMT, indicating that some lesion(s) induced by cisplatin giving rise to SCEs can be repaired by MGMT. Taken together, these results indicate that streptozotocin, HeCNU and ACNU are more selective than CCNU and BCNU in killing MGMT deficient cells, and that in the cases of cyclophosphamide, ifosfamide, chlorambucil, cisplatin and melphalan MGMT is not involved in mediating cytotoxic drug resistance.


Assuntos
Antineoplásicos/farmacologia , Metiltransferases/fisiologia , Troca de Cromátide Irmã/efeitos dos fármacos , Animais , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , O(6)-Metilguanina-DNA Metiltransferase
11.
Cell Biol Toxicol ; 7(2): 145-65, 1991 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1889006

RESUMO

A series of seven 1-aryl-3.3-dialkyltriazenes, including 1-phenyl-3.3-dimethyltriazene (DMPT), 1-phenyl-3.3-di-(trideuteromethyl)-triazene (DMPT-ds), 1-p-methylphenyl-3.3-dimethyltriazene (DMpMPT), 1-p-nitrophenyl-3.3-dimethyltriazene (DMpNPT), 1-phenyl-3.3-diethyltriazene (DEPT), 1-phenyl-3.3-di-n-propyltriazene (DnPrPT) and 1-phenyl-3.3-diisopropyltriazene (DiPrPT) and 1.3-diphenyl-3-methyltriazene (DPMT), was synthesized and characterized by UV/VIS, IR and 1H-NMR spectroscopy. Chemical half-life was determined in phosphate buffer at 37 degrees using UV/VIS spectroscopy. With the exception of DMpNPT, which was stable, the triazenes underwent pH-dependent hydrolytic decomposition (acid catalysis). By means of UV/VIS spectra, TLC and HPLC, phenol, aniline and secondary azocoupling products were identified after complete hydrolytic cleavage of the parent compounds. Pathways of spontaneous hydrolysis are proposed and discussed. Genotoxic activity of the triazenes was assayed by measurement of sister chromatid exchanges (SCE) in V79-E cells without and with rat liver S9 mix as an exogenous metabolizing system. In the direct SCE assay (without S9 mix), all triazenes except DMpNPT exerted a toxic action (cell cycle delay) in a narrow concentration range between no effect and overt cytotoxicity. This non-specific toxicity depended on the pH of the incubation system and was inversely proportional to chemical half-life. The toxicity of these agents is most likely due to the arenediazonium cation which is a relatively stable intermediate. In a sublethal concentration range most triazeness induced significant increases of SCE rates. These are interpreted as an indirect consequence of cytotoxicity. Upon metabolic activation, the compounds were genotoxic in a dose-dependent fashion. Their SCE-inducing capacity depended on the nature of the alkylating species generated, i.e., the alkyldiazonium cation, and on chemical stability. Surprisingly, no deuterium isotope effect was observed in DMPT-d6. The order of genotoxic activity among the aryldialkyltriazenes was DMpNPT much greater than DMPT = DMPT-ds greater than DMpMPT much greater than DEPT greater than DnPrPT greater than or equal to DiPrPT. DPMT was a marginal SCE inducer but very toxic upon metabolic activation. As monooxygenation of DPMT, like spontaneous hydrolysis, should generate a phenyldiazonium cation, the results suggest that arylation of DNA causes a very low SCE induction, if any.


Assuntos
Ciclo Celular/efeitos dos fármacos , Troca de Cromátide Irmã , Triazenos/toxicidade , Animais , Biotransformação , Linhagem Celular , Cricetinae , Cricetulus , Meia-Vida , Hidrólise , Espectroscopia de Ressonância Magnética , Masculino , Microssomos Hepáticos/metabolismo , Ratos , Espectrofotometria Infravermelho , Espectrofotometria Ultravioleta , Relação Estrutura-Atividade , Triazenos/análise , Triazenos/química
12.
Arch Geschwulstforsch ; 60(3): 179-86, 1990.
Artigo em Alemão | MEDLINE | ID: mdl-2369280

RESUMO

The pre- and postnatal administration of DEMNU induces a high frequency of tumors when applied via the intravenous route, and the latency periods show a dose dependence (table I). Tumors of the brain, spinal cord and cranial nerves clearly predominate. Furthermore, a large number of neoplasms of kidney, heart and soft tissue was observed (table II). As DEMNU is per se a very stable compound, it is suggested that this agent is metabolized by monooxygenases. 3-Ethyl-1-methyl-1-nitrosourea should be formed as an intermediate product via this pathway, which is relatively stable and might explain the mainly neurotropic carcinogenicity of DEMNU. Species differences in the carcinogenicity of trialkyl-nitrosoureas and the mode of metabolic activation are discussed.


Assuntos
Neoplasias Experimentais/induzido quimicamente , Compostos de Nitrosoureia/toxicidade , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Injeções Intravenosas , Masculino , Compostos de Metilureia/administração & dosagem , Compostos de Metilureia/toxicidade , Neoplasias Experimentais/patologia , Compostos de Nitrosoureia/administração & dosagem , Gravidez , Ratos , Ratos Endogâmicos , Fatores de Tempo
13.
Carcinogenesis ; 10(10): 1787-91, 1989 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2791198

RESUMO

Trisubstituted nitrosoureas are very stable in aqueous systems. But they are potent genotoxins in Chinese hamster V79-E cells, if no exogenous metabolizing system is added, and the mechanism of their genotoxic and carcinogenic activity has been largely unknown. This investigation shows that the sister-chromatid-exchange (SCE)-inducing capacity of 1,3-dimethyl-3-phenyl-1-nitrosourea (DMPNU) is eliminated by adding diisopropylflurophosphate (DFP) or porcine liver carboxylesterase to the incubation system. These effects are caused by two different mechanisms: (i) DFP inhibits endogenous amidases existing in V79-E cells, thus preventing the intracellular decomposition, which means an activation; and (ii) exogenous carboxylesterase cleaves DMPNU extracellularly, and the genotoxic decomposition product is obviously too short-lived to reach a critical intracellular target. A second trisubstituted nitrosourea, 3,3-diethyl-1-methyl-1-nitrosourea (DEMNU), which is mainly activated by monooxygenases, but in the absence of an exogenous metabolizing system also induces SCEs in V79-E cells, was studied in the same way. It was found that the 'direct' genotoxicity of DEMNU may be inhibited by DFP as well, but carboxylesterase decomposes this trialklynitrosourea with a much lower efficiency than DMPNU suggesting a low substrate affinity. The SCE-inducing capacity of both compounds is strongly influenced by the presence of calf serum in the culture medium. The nature of the serum factor is still unknown. Pathways for the amidase catalysis of DMPNU and for the activation of DEMNU by monooxygenases and amidases are proposed and discussed with respect to the topical or systemic carcinogenicity of these agents.


Assuntos
Compostos de Nitrosoureia/farmacologia , Serina Endopeptidases/metabolismo , Troca de Cromátide Irmã/efeitos dos fármacos , Animais , Biotransformação , Hidrolases de Éster Carboxílico/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Cricetinae , Cricetulus , Isoflurofato/farmacologia , Pulmão , Compostos de Nitrosoureia/metabolismo
14.
Cell Biol Toxicol ; 4(2): 241-57, 1988 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3233533

RESUMO

The mechanism of cytogenetic genotoxicity (clastogenicity, induction, cell cycle delay) of 10(-3) M glutathione in V79-E cells, as described by Thust and Bach (1985), was studied in detail by using different treatment conditions. It was found that 1-cystine is the essential cofactor in the incubation system. Catalase, but not superoxide dismutase, abolished the genotoxic effect, and the iron chelator desferoxamine, as well as the hydroxyl radical scavenger mannitol, diminished the activity. It is suggested that glutathione, in combination with V79-E cells and cystine, forms a hydrogen peroxide-generating system which provokes the adverse effects. Glutathione as well as 1-cysteine and 2-mercaptopropionylglycine, which were checked for comparison, show a "paradoxic genotoxicity," i.e., at 10(-2) M the effects return almost to the level of controls. Concentration dependence and other criteria of cytogenetic genotoxicity observed with glutathione show obvious similarities to those of other oxidatively acting agents and reveal striking differences to the cytogenetic effects of "typical" genotoxins.


Assuntos
Glutationa/toxicidade , Animais , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Aberrações Cromossômicas/efeitos dos fármacos , Relação Dose-Resposta a Droga , Cobaias , Testes de Mutagenicidade , Ratos
15.
Carcinogenesis ; 8(2): 237-40, 1987 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-3802407

RESUMO

1,3-Dimethyl-3-phenyl-1-nitrosourea (DMPNU) is a very potent local carcinogen in rats and induces a 100% frequency of forestomach carcinomas when applied i.g. in two different dosages (10 applications of 0.3 or 0.03 mmol/kg body wt, respectively, at 14-day intervals), but it is inactive upon i.v. administration (10 applications of 0.03 mmol/kg body wt at 14-day intervals). By means of the direct sister chromatid exchange (SCE) assay in V79-E cells in the presence of rat blood, serum or plasma, respectively, as well as by a 'host-mediated' SCE assay (in which the agent was given i.v. to rats, and blood taken from the animal was checked for the recovery of genotoxic activity in cell cultures), we tried to elucidate the unexpected lack of carcinogenic activity of i.v. DMPNU. The direct SCE assay revealed a drastic reduction of DMPNU genotoxicity by rat blood, serum or plasma, respectively, which is abolished by the esterase inhibitor diisopropylfluorophosphate. In the 'host-mediated' SCE assay a genotoxic activity of DMPNU was only recoverable after a very high i.v. dose and when the blood added to the cell cultures had been taken from the rat heart within 1 min after DMPNU administration in vivo. 1-Methyl-1-nitrosourea (MNU) and 1-methyl-3-phenyl-1-nitrosourea (MPNU) were used as positive controls in these experiments and also gave a lower response than theoretically expected, but the relative loss of activity with the latter compounds was much lower than with DMPNU. It is assumed that an esterase in rat blood effectively decomposes this trisubstituted nitrosourea. Problems of the novel 'host-mediated' SCE assay are discussed.


Assuntos
Carcinoma de Células Escamosas/induzido quimicamente , Metilnitrosoureia/análogos & derivados , Troca de Cromátide Irmã/efeitos dos fármacos , Neoplasias Gástricas/induzido quimicamente , Animais , Células Cultivadas , Cricetinae , Feminino , Inativação Metabólica , Injeções Intravenosas , Intubação Gastrointestinal , Masculino , Metilnitrosoureia/administração & dosagem , Metilnitrosoureia/metabolismo , Metilnitrosoureia/toxicidade , Testes de Mutagenicidade , Ratos
16.
Cell Biol Toxicol ; 1(3): 123-31, 1985 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3916979

RESUMO

Glutathione (GSH) dissolved in Eagle's MEM and added to cultures of V79-E cells in concentrations between 2.5 X 10(-4) and 10(-3) moles/l for 1 h induces a dose-dependent cell cycle delay, sister chromatid exchanges and clastogenic damage. 7-8% of the metaphases showed endoreduplication at a recovery phase of 25 and 30 h after treatment with 10(-3) moles/l GSH. Higher concentrations were lethal. The highest tolerated dose corresponds to the intracellular GSH level in V79-E cells. In the same range of concentrations, glutathione disulfide was inactive. Endoreduplication induction by GSH is G2-phase specific and endoreduplication metaphases show a reduced occurrence of single SCEs when extrapolated to the diploid complement. The adverse effects of GSH are independent of the presence of serum in the culture fluid but completely abolished when the treatment is performed in Hank's solution instead of MEM. The mechanism of genotoxicity of exogenous GSH is discussed but, at present, no pertinent explanation can be given.


Assuntos
Replicação do DNA/efeitos dos fármacos , Glutationa/farmacologia , Mutagênicos , Troca de Cromátide Irmã/efeitos dos fármacos , Animais , Linhagem Celular , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga
17.
Carcinogenesis ; 6(6): 873-6, 1985 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-4006073

RESUMO

The genotoxicity of 1-methyl-3-phenyl-1-nitrosourea (MPNU), 1-methyl-3-(p-chlorophenyl)-1-nitrosourea (C1-MPNU), 1-ethyl-3-phenyl-1-nitrosourea (EPNU), 1,3-dimethyl-3-phenyl-1-nitrosourea (DMPNU) and their derivatives substituted by deuterium in different positions was studied using sister chromatid exchange (SCE) induction in Chinese hamster V79-E cells. Deuterium substitution in the 1-methyl group of MPNU (MPNU-d3) and C1-MPNU (C1-MPNU-d3) diminished the SCE-inducing capacity by 20-30% and by 30-40% in DMPNU (DMPNU-d3B). There was no altered SCE activity detected when the phenyl group of MPNU (MPNU-d5) or the 3-methyl group of DMPNU (DMPNU-d3A) was deuterium labeled. No isotope effect was detected in deuterated EPNU derivatives, presumably due to the instability of these compounds. It is surmised that the easier delocalization of the positive charge in the deuterated alkyl diazonium ion causes a diminished reactivity and therefore influences the type and amount of DNA alkylation. Furthermore, the experiments with DMPNU and its derivatives revealed that, in contrast to mono- and disubstituted nitrosoureas, the biological activities of these very stable trisubstituted nitrosoureas are strongly influenced by a serum factor in the culture fluid.


Assuntos
Deutério/farmacologia , Compostos de Nitrosoureia/toxicidade , Troca de Cromátide Irmã/efeitos dos fármacos , Animais , Linhagem Celular , Cricetinae , Cricetulus , Etilnitrosoureia/toxicidade , Técnicas In Vitro , Metilnitrosoureia/toxicidade
18.
Mutat Res ; 126(3): 259-64, 1984 May.
Artigo em Inglês | MEDLINE | ID: mdl-6717462

RESUMO

The genotoxic activity of 3,3-diethyl-1-methyl-1-nitrosourea ( DEMNU ), 1,3-dimethyl-3-phenyl-1-nitrosourea ( DMPNU ) and 1-chloroethyl-3-methyl-3-phenyl-1-nitrosourea ( CEMPNU ) was studied in the SCE assay in V79-E cells in vitro. These compounds are very stable in aqueous solutions, but are directly acting genotoxins . The SCE rates increase linearly with the length of the incubation period. This direct activity is presumably due to an intracellular catalytic decomposition. Whereas the SCE-inducing effect of DMPNU and CEMPNU is not influenced by addition of S9 mix, that of DEMNU is strongly potentiated by rat and Syrian hamster S9 mix. This DEMNU activation is an NADPH-dependent enzymatic reaction and is inducible by phenobarbital. The absence of a direct mutagenic effect of DEMNU in the Ames test, as reported by other authors, is probably caused by a striking insensitivity to tri-substituted nitrosoureas of the Salmonella assay. This assumption was substantiated by long-term application of very low DMPNU doses to V79-E. Long-term simultaneous treatment with DMPNU and bromodeoxyuridine (BUdR) significantly diminished the rate of SCE induction.


Assuntos
Troca Genética/efeitos dos fármacos , Compostos de Nitrosoureia/toxicidade , Troca de Cromátide Irmã/efeitos dos fármacos , Animais , Biotransformação , Bromodesoxiuridina/farmacologia , Ciclo Celular , Células Cultivadas , Cricetinae , Relação Dose-Resposta a Droga , Testes de Mutagenicidade , Relação Estrutura-Atividade
19.
Mutat Res ; 139(4): 207-10, 1984 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-6325903

RESUMO

Benzo[alpha]pyrene BaP), freely dissolved or incorporated in liposomes prepared from egg yolk lecithin, was checked for SCE induction in Chinese hamster V79-E and rat liver RL-19 cells in vitro. SCE induction in V79-E was observed only when freely dissolved BaP was added together with S9 mix. RL-19 cells were per se highly sensitive to SCE induction by BaP either freely dissolved or incorporated in liposomes. It is suggested that the incorporation of genotoxins in liposomes is a practicable method for the application, in mammalian genotoxicity assays, of agents which are barely soluble or completely insoluble in water, provided no exogenous metabolizing system is required.


Assuntos
Benzopirenos/administração & dosagem , Troca Genética/efeitos dos fármacos , Lipossomos/administração & dosagem , Mutagênicos/administração & dosagem , Troca de Cromátide Irmã/efeitos dos fármacos , Animais , Benzo(a)pireno , Células Cultivadas , Cricetinae , Testes de Mutagenicidade , Solubilidade
20.
Zentralbl Allg Pathol ; 129(4): 365-70, 1984.
Artigo em Alemão | MEDLINE | ID: mdl-6395564

RESUMO

A survey is given about problems related to cheking for genotoxicity of environmental chemicals. Cytogenetic effects, induction of structural chromosome aberrations (clastogenicity) and sister chromatid exchanges (SCEs), are very sensitive indicators of genetic damage in eukaryotic cells. Problems and methods of the clastogenicity and SCE assay in mammalian cells in vitro are described. These bioassays are correlation tests and do not allow a definitive prediction of carcinogenic activity of chemicals, but they are a valuable tool for the evaluation of genetic/carcinogenic risks. Problems of extrapolation to man are discussed.


Assuntos
Carcinógenos , Avaliação Pré-Clínica de Medicamentos , Testes de Mutagenicidade/métodos , Animais , Células Cultivadas , Aberrações Cromossômicas , Mamíferos , Troca de Cromátide Irmã
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...