Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-26774667

RESUMO

This work seeks to provide users with guidance on cell culture, treatment, processing and analytical conditions for achieving optimal performance of the in vitro micronucleus assay using the In Vitro MicroFlow(®) method. Experimental data are provided to support the advice described. The information provided covers specific topics or issues that are identified as critical to the methodology and thus is meant to work with instruction manuals, published papers and other references, and not as a replacement for these documents. The content is divided into several sections. Cell culture and treatment describes conditions for routine maintenance of cells as well as treatment with test articles. Preparation and processing of samples details steps found to be critical in execution of the procedure. Instrument parameters and analysis covers set-up of the flow cytometer and evaluation of the samples. General assay considerations and interpretation of results describes examination of data in terms of assay validity, viability and genotoxicity assessment. The goal is to educate users and enable them to design, conduct and interpret flow cytometric in vitro micronucleus (MN) studies. Readers should obtain an understanding of specific cell culture practices, options for assay formatting and execution and the information required to successfully integrate and validate the in vitro MN assay into their existing safety program.


Assuntos
Citometria de Fluxo/métodos , Testes para Micronúcleos/métodos , Animais , Apoptose/efeitos dos fármacos , Azidas/toxicidade , Técnicas de Cultura de Células , Células Cultivadas , Cricetinae , Dano ao DNA/efeitos dos fármacos , Necrose/induzido quimicamente , Necrose/patologia , Timidina/toxicidade
3.
Artigo em Inglês | MEDLINE | ID: mdl-26340763

RESUMO

Sample dilution is one major challenge in dried blood spot (DBS) bioanalysis. To resolve this issue, we applied a no-dilution strategy for DBS analysis by using a calibration curve with very wide linear range. We developed an LC-MS/MS DBS assay with a linear range of 5 orders of magnitude (50-5000,000ng/mL) for BMS-986001, an HIV drug under development, by simultaneously monitoring two selective reaction monitoring transitions of different intensity. The assay was validated and successfully applied to the analysis of DBS samples collected in a toxicology study in rats dosed with BMS-986001. All samples were analyzed without any dilution. We also compared the concentration data generated from the DBS method and a validated plasma assay for the same study. The two sets of data agreed well with each other, demonstrating the validity of this strategy for DBS analysis. This approach provides an effective and convenient way to eliminate complicated dilution for DBS and other sample collection techniques.


Assuntos
Fármacos Anti-HIV/sangue , Cromatografia Líquida/métodos , Espectrometria de Massas em Tandem/métodos , Timidina/análogos & derivados , Animais , Fármacos Anti-HIV/toxicidade , Feminino , Limite de Detecção , Gravidez , Ratos , Timidina/sangue , Timidina/toxicidade
4.
Chem Res Toxicol ; 26(11): 1730-8, 2013 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-24134187

RESUMO

Causal links exist between smoking cigarettes and cancer development. Some genotoxic agents in cigarette smoke are capable of alkylating nucleobases in DNA, and higher levels of ethylated DNA lesions were observed in smokers than in nonsmokers. In this study, we examined comprehensively how the regioisomeric O(2)-, N3-, and O(4)-ethylthymidine (O(2)-, N3-, and O(4)-EtdT, respectively) perturb DNA replication mediated by purified human DNA polymerases (hPols) η, κ, and ι, yeast DNA polymerase ζ (yPol ζ), and the exonuclease-free Klenow fragment (Kf(-)) of Escherichia coli DNA polymerase I. Our results showed that hPol η and Kf(-) could bypass all three lesions and generate full-length replication products, whereas hPol ι stalled after inserting a single nucleotide opposite the lesions. Bypass conducted by hPol κ and yPol ζ differed markedly among the three lesions. Consistent with its known ability to efficiently bypass the minor groove N(2)-substituted 2'-deoxyguanosine lesions, hPol κ was able to bypass O(2)-EtdT, though it experienced great difficulty in bypassing N3-EtdT and O(4)-EtdT. yPol ζ was only modestly blocked by O(4)-EtdT, but the polymerase was strongly hindered by O(2)-EtdT and N3-EtdT. LC-MS/MS analysis of the replication products revealed that DNA synthesis opposite O(4)-EtdT was highly error-prone, with dGMP being preferentially inserted, while the presence of O(2)-EtdT and N3-EtdT in template DNA directed substantial frequencies of misincorporation of dGMP and, for hPol ι and Kf(-), dTMP. Thus, our results suggested that O(2)-EtdT and N3-EtdT may also contribute to the AT → TA and AT → GC mutations observed in cells and tissues of animals exposed to ethylating agents.


Assuntos
Replicação do DNA/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/metabolismo , Timidina/toxicidade , Sequência de Bases , Cromatografia Líquida de Alta Pressão , DNA/metabolismo , DNA Polimerase I/metabolismo , Primers do DNA/química , Primers do DNA/metabolismo , DNA Bacteriano/biossíntese , Escherichia coli/enzimologia , Humanos , Saccharomyces cerevisiae/enzimologia , Estereoisomerismo , Espectrometria de Massas em Tandem , Timidina/análogos & derivados , Timidina/análise , DNA Polimerase iota
5.
Nucleic Acids Res ; 40(13): 6016-25, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22447450

RESUMO

Deamination of cytosine (C), 5-methylcytosine (mC) and 5-hydroxymethylcytosine (hmC) occurs spontaneously in mammalian DNA with several hundred deaminations occurring in each cell every day. The resulting potentially mutagenic mispairs of uracil (U), thymine (T) or 5-hydroxymethyluracil (hmU) with guanine (G) are substrates for repair by various DNA glycosylases. Here, we show that targeted inactivation of the mouse Smug1 DNA glycosylase gene is sufficient to ablate nearly all hmU-DNA excision activity as judged by assay of tissue extracts from knockout mice as well as by the resistance of their embryo fibroblasts to 5-hydroxymethyldeoxyuridine toxicity. Inactivation of Smug1 when combined with inactivation of the Ung uracil-DNA glycosylase gene leads to a loss of nearly all detectable uracil excision activity. Thus, SMUG1 is the dominant glycosylase responsible for hmU-excision in mice as well as the major UNG-backup for U-excision. Both Smug1-knockout and Smug1/Ung-double knockout mice breed normally and remain apparently healthy beyond 1 year of age. However, combined deficiency in SMUG1 and UNG exacerbates the cancer predisposition of Msh2(-/-) mice suggesting that when both base excision and mismatch repair pathways are defective, the mutagenic effects of spontaneous cytosine deamination are sufficient to increase cancer incidence but do not preclude mouse development.


Assuntos
Reparo do DNA , Pentoxil (Uracila)/análogos & derivados , Uracila-DNA Glicosidase/genética , Uracila-DNA Glicosidase/metabolismo , Animais , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Fluoruracila/metabolismo , Marcação de Genes , Predisposição Genética para Doença , Longevidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 2 Homóloga a MutS/genética , Neoplasias Experimentais/genética , Pentoxil (Uracila)/metabolismo , Timidina/análogos & derivados , Timidina/toxicidade , beta-Galactosidase/genética
6.
Mol Biosyst ; 7(11): 3075-86, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21897988

RESUMO

Novel thymidine- or uridine-based nucleolipids, containing one hydrophilic oligo(ethylene glycol) chain and one or two oleic acid residues (called ToThy, HoThy and DoHu), have been synthesized with the aim to develop bio-compatible nanocarriers for drug delivery and/or produce pro-drugs. Microstructural characterization of their aggregates has been determined in pure water and in pseudo-physiological conditions through DLS and SANS experiments. In all cases stable vesicles, with mean hydrodynamic radii ranging between 120 nm and 250 nm have been revealed. Biological validation of the nucleolipidic nanocarriers was ensured by evaluation of their toxicological profiles, performed by administration of the nanoaggregates to a panel of different cell lines. ToThy exhibited a weak cytotoxicity and, at high concentration, some ability to interfere with cell viability and/or proliferation. In contrast, DoHu and HoThy exhibited no toxicological relevance, behaving similarly to POPC-based liposomes, widely used for systemic drug delivery. Taken together, these results show nucleolipid-based nanocarriers as finely tunable, multi-functional self-assembling materials of interest for the in vivo transport of biomolecules or drugs.


Assuntos
Portadores de Fármacos/síntese química , Nanocápsulas/química , Ácidos Oleicos/síntese química , Timidina/análogos & derivados , Uridina/análogos & derivados , Células 3T3-L1 , Animais , Células CACO-2 , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Portadores de Fármacos/química , Portadores de Fármacos/toxicidade , Sistemas de Liberação de Medicamentos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Ácidos Oleicos/química , Ácidos Oleicos/toxicidade , Pró-Fármacos/síntese química , Pró-Fármacos/química , Ratos , Timidina/síntese química , Timidina/química , Timidina/toxicidade , Uridina/síntese química , Uridina/química , Uridina/toxicidade
7.
Nucleic Acids Res ; 39(19): 8430-44, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21745813

RESUMO

Cytotoxicity of 5-fluorouracil (FU) and 5-fluoro-2'-deoxyuridine (FdUrd) due to DNA fragmentation during DNA repair has been proposed as an alternative to effects from thymidylate synthase (TS) inhibition or RNA incorporation. The goal of the present study was to investigate the relative contribution of the proposed mechanisms for cytotoxicity of 5-fluoropyrimidines. We demonstrate that in human cancer cells, base excision repair (BER) initiated by the uracil-DNA glycosylase UNG is the major route for FU-DNA repair in vitro and in vivo. SMUG1, TDG and MBD4 contributed modestly in vitro and not detectably in vivo. Contribution from mismatch repair was limited to FU:G contexts at best. Surprisingly, knockdown of individual uracil-DNA glycosylases or MSH2 did not affect sensitivity to FU or FdUrd. Inhibitors of common steps of BER or DNA damage signalling affected sensitivity to FdUrd and HmdUrd, but not to FU. In support of predominantly RNA-mediated cytotoxicity, FU-treated cells accumulated ~3000- to 15 000-fold more FU in RNA than in DNA. Moreover, FU-cytotoxicity was partially reversed by ribonucleosides, but not deoxyribonucleosides and FU displayed modest TS-inhibition compared to FdUrd. In conclusion, UNG-initiated BER is the major route for FU-DNA repair, but cytotoxicity of FU is predominantly RNA-mediated, while DNA-mediated effects are limited to FdUrd.


Assuntos
Reparo do DNA , Fluoruracila/metabolismo , Uracila-DNA Glicosidase/metabolismo , Animais , Ciclo Celular , Linhagem Celular Tumoral , DNA/química , DNA/metabolismo , Dano ao DNA , Endodesoxirribonucleases/genética , Floxuridina/metabolismo , Floxuridina/toxicidade , Fluoruracila/toxicidade , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Proteína 2 Homóloga a MutS/genética , RNA/metabolismo , Timidina/análogos & derivados , Timidina/metabolismo , Timidina/toxicidade , Timina DNA Glicosilase/genética , Timina DNA Glicosilase/metabolismo , Uracila-DNA Glicosidase/genética , Uridina/análogos & derivados , Uridina/metabolismo , Uridina/toxicidade
8.
Br J Cancer ; 104(12): 1869-76, 2011 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-21610703

RESUMO

BACKGROUND: Thiothymidine (S(4)TdR) can be incorporated into DNA and sensitise cells to DNA damage and cell death following exposure to UVA light. Studies were performed to determine if the combination of S(4)TdR and UVA could be an effective treatment for bladder cancer. METHODS: Uptake and incorporation of S(4)TdR was determined in rat and human bladder tumour cell lines. Measures of DNA crosslinking and apoptosis were also performed. In vivo activity of the combination of S(4)TdR and UVA was investigated in an orthotopic model of bladder cancer in rats. RESULTS: Thiothymidine (200 µM) replaced up to 0.63% of thymidine in rat and tumour bladder cancer cells. The combination of S(4)TdR (10-200 µM) and UVA (1-5 kJ m(-2)) caused apoptosis and cell death at doses that were not toxic alone. Addition of raltitrexed (Astra Zeneca, Alderley Edge, Cheshire, UK) increased the incorporation of S(4)TdR into DNA (up to 20-fold at IC(5)) and further sensitised cells to UVA. Cytotoxic effect was associated with crosslinking of DNA, at least partially to protein. Intravenous administration of S(4)TdR, in combination with UVA delivered directly to the bladder, resulted in an antitumour effect in three of five animals treated. CONCLUSION: These data indicate that the combination of S(4)TdR and UVA has potential as a treatment for bladder cancer, and give some insight into the mechanism of action. Further work is necessary to optimise the delivery of the two components.


Assuntos
Radiossensibilizantes/uso terapêutico , Timidina/análogos & derivados , Terapia Ultravioleta , Neoplasias da Bexiga Urinária/terapia , Animais , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Dano ao DNA , Feminino , Humanos , Quinazolinas/farmacologia , Ratos , Ratos Endogâmicos F344 , Tiofenos/farmacologia , Timidina/metabolismo , Timidina/uso terapêutico , Timidina/toxicidade , Neoplasias da Bexiga Urinária/patologia
9.
Biochemistry ; 48(33): 7833-41, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19618962

RESUMO

Tandem lesions are comprised of two contiguously damaged nucleotides. Tandem lesions make up the major family of reaction products generated from a pyrimidine nucleobase radical, which are formed in large amounts by ionizing radiation. One of these tandem lesions contains a thymidine glycol lesion flanked on its 5'-side by 2-deoxyribonolactone (LTg). The replication of this tandem lesion was investigated in Escherichia coli using single-stranded genomes. LTg is a much more potent replication block than thymidine glycol and is bypassed only under SOS-induced conditions. The adjacent thymidine glycol does not significantly affect nucleotide incorporation opposite 2-deoxyribonolactone in wild-type cells. In contrast, the misinsertion frequency opposite thymidine glycol, which is negligible in the absence of 2-deoxyribonolactone, increases to 10% in wild-type cells when LTg is flanked by a 3'-dG. Experiments in which the flanking nucleotides are varied and in cells lacking one of the SOS-induced bypass polymerases indicate that the mutations are due to a mechanism in which the primer misaligns prior to bypassing the lesion, which allows for an additional nucleotide to be incorporated across from the 3'-flanking nucleotide. Subsequent realignment and extension results in the observed mutations. DNA polymerases II and IV are responsible for misalignment induced mutations and compete with DNA polymerase V which reads through the tandem lesion. These experiments reveal that incorporation of the thymidine glycol into a tandem lesion indirectly induces increases in mutations by blocking replication, which enables the misalignment-realignment mechanism to compete with direct bypass by DNA polymerase V.


Assuntos
Dano ao DNA , DNA Bacteriano/efeitos dos fármacos , Repetições de Dinucleotídeos/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Mutagênese , Timidina/análogos & derivados , Regiões 5' não Traduzidas/efeitos dos fármacos , Regiões 5' não Traduzidas/genética , Bacteriófago M13/genética , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , DNA Bacteriano/síntese química , Repetições de Dinucleotídeos/genética , Genoma Bacteriano/efeitos dos fármacos , Radical Hidroxila/toxicidade , Resposta SOS em Genética/efeitos dos fármacos , Deleção de Sequência/efeitos dos fármacos , Açúcares Ácidos/toxicidade , Timidina/toxicidade
10.
Exp Cell Res ; 315(15): 2539-43, 2009 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-19481540

RESUMO

The nucleoside analogs 9-beta-D-arabinofuranosylguanine (araG) and 1-beta-d-arabinofuranosylthymine (araT) are substrates of mitochondrial nucleoside kinases and have previously been shown to be predominantly incorporated into mtDNA of cells, but the pharmacological importance of their accumulation in mtDNA is not known. Here, we examined the role of mtDNA in the response to araG, araT and other anti-cancer and anti-viral agents in a MOLT-4 wild-type (wt) T-lymphoblastoid cell line and its petite mutant MOLT-4 rho(0) cells (lacking mtDNA). The mRNA levels and activities of deoxyguanosine kinase (dGK), deoxycytidine kinase (dCK), thymidine kinase 1 (TK1) and thymidine kinase 2 (TK2) were determined in the two cell lines. Compared to that in the MOLT-4 wt cells the mRNA level of the constitutively expressed TK2 was higher (p<0.01) in the rho(0) cells, whereas the TK1 mRNA level was lower (p<0.05). The enzyme activity of the S-phase restricted TK1 was also lower (p<0.05) in the MOLT-4 rho(0) cells, whereas the activities of dGK, dCK and TK2 were similar in MOLT-4 wt and rho(0) cell lines. The sensitivities to different cytotoxic nucleoside analogs were determined and compared between the two cell lines. Interestingly, we found that the acute cytotoxicity of araG, araT and other anti-viral and anti-cancer agents is independent of the presence of mtDNA in MOLT-4 T-lymphoblastoid cells.


Assuntos
Antineoplásicos/toxicidade , Arabinonucleosídeos/toxicidade , DNA Mitocondrial/metabolismo , Timidina/análogos & derivados , Animais , Linhagem Celular Tumoral/efeitos dos fármacos , DNA Mitocondrial/genética , Desoxicitidina Quinase/genética , Desoxicitidina Quinase/metabolismo , Humanos , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Timidina/toxicidade , Timidina Quinase/genética , Timidina Quinase/metabolismo
11.
Antiviral Res ; 76(2): 124-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17658623

RESUMO

N-Methanocarbathymidine [(N)-MCT] is a newly identified inhibitor of orthopoxvirus replication in cell culture and in mice. Limited published animal studies indicated the compound is effective by intraperitoneal (i.p.) route at 10-100 mg/(kg day). More extensive studies using different treatment regimens in intranasally infected mice were conducted in order to further explore the potential of this compound compared to cidofovir in treating vaccinia virus infections. (N)-MCT was given twice a day for 7 days, whereas cidofovir was administered once a day for 2 days, each starting 24h after virus exposure for most experiments. (N)-MCT was not toxic up to 1000 mg/(kg day) by the i.p. treatment route. Oral and i.p. treatment regimens with (N)-MCT were directly compared during a vaccinia virus (IHD strain) infection, indicating that the nucleoside has good oral bioavailability in mice. Treatments by i.p. route with (N)-MCT (100 mg/(kg day)) reduced lung, nasal, and brain virus titers during an IHD virus infection, but not nearly to the same extent as i.p. cidofovir (100 mg/(kg day)). Treatment with both compounds decreased liver, spleen, and kidney virus titers, as well as reduced lung consolidation scores and lung weights. Onset of treatment could be delayed by 2 days with (N)-MCT and by 3 days with cidofovir, providing significant survival benefit during the IHD virus infection. Against a vaccinia virus (WR strain) infection in mice, i.p. (N)-MCT treatment prevented death at 500 mg/(kg day), which was comparable in activity to i.p. cidofovir (100 mg/(kg day)). Significant reductions in tissue virus titers occurred with both treatment regimens. (N)-MCT could be further pursued for its potential to treat orthopoxvirus infections in humans.


Assuntos
Antivirais/uso terapêutico , Timidina/análogos & derivados , Vaccinia virus/efeitos dos fármacos , Vacínia/tratamento farmacológico , Administração Oral , Animais , Cidofovir , Citosina/administração & dosagem , Citosina/análogos & derivados , Citosina/uso terapêutico , Injeções Intraperitoneais , Camundongos , Organofosfonatos/administração & dosagem , Organofosfonatos/uso terapêutico , Análise de Sobrevida , Timidina/administração & dosagem , Timidina/uso terapêutico , Timidina/toxicidade , Ensaio de Placa Viral
12.
Nucleic Acids Res ; 35(12): 4103-13, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17567611

RESUMO

The Cockayne syndrome B (CSB) protein--defective in a majority of patients suffering from the rare autosomal disorder CS--is a member of the SWI2/SNF2 family with roles in DNA repair and transcription. We demonstrate herein that purified recombinant CSB and the major human apurinic/apyrimidinic (AP) endonuclease, APE1, physically and functionally interact. CSB stimulates the AP site incision activity of APE1 on normal (i.e. fully paired) and bubble AP-DNA substrates, with the latter being more pronounced (up to 6-fold). This activation is ATP-independent, and specific for the human CSB and full-length APE1 protein, as no CSB-dependent stimulation was observed with Escherichia coli endonuclease IV or an N-terminal truncated APE1 fragment. CSB and APE1 were also found in a common protein complex in human cell extracts, and recombinant CSB, when added back to CSB-deficient whole cell extracts, resulted in increased total AP site incision capacity. Moreover, human fibroblasts defective in CSB were found to be hypersensitive to both methyl methanesulfonate (MMS) and 5-hydroxymethyl-2'-deoxyuridine, agents that introduce base excision repair (BER) DNA substrates/intermediates.


Assuntos
DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Linhagem Celular Transformada , DNA Helicases/fisiologia , Enzimas Reparadoras do DNA/fisiologia , Genoma Humano , Humanos , Metanossulfonato de Metila/toxicidade , Proteínas de Ligação a Poli-ADP-Ribose , Timidina/análogos & derivados , Timidina/toxicidade
13.
Radiats Biol Radioecol ; 47(1): 108-16, 2007.
Artigo em Russo | MEDLINE | ID: mdl-17388002

RESUMO

We found that low doses (0.12-0.46Gy) of (methyl-) 3H-thymidine incorporated into human endothelial cells induce the accumulation cells in G2-phase of the cell cycle. Temperate doses of (1-6 Gy) gamma-rays 137Cs were less effective in the G2-block estimated by flow cytometry analysis of DNA content. Furthermore, the induced the high level of the chromosome aberrations (bridges and fragments in anaphases). 1Gy of gamma-ray 137Cs and 0.005 Gy of beta-rays induced the same per cent of the aberrant anaphases. Apparently, that the damages of the cellular hereditary structures are responsible for the blocking of the cellular proliferation in G2-phase. We suggest, that the disposition 3H-thymidine into radiosensitive target (DNA) defines the high cytotoxic of the beta-rays.


Assuntos
DNA/efeitos da radiação , Células Endoteliais/efeitos da radiação , Fase G2/efeitos da radiação , Raios gama , Partículas beta , Proliferação de Células/efeitos da radiação , Radioisótopos de Césio/toxicidade , Instabilidade Cromossômica , Aberrações Cromossômicas , DNA/química , DNA/metabolismo , Dano ao DNA , Humanos , Timidina/análise , Timidina/metabolismo , Timidina/toxicidade , Trítio
14.
J Virol ; 81(9): 4900-3, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17301142

RESUMO

The retroviral mutation rate is susceptible to a number of variables, including the balance between intracellular deoxynucleoside triphosphate (dNTP) pools. While this follows from tissue culture studies, the issue has never been addressed directly in vivo. To explore this question in a tractable experimental system, we analyzed the impact of thymidine treatment on the synthesis of gypsy retroelement cDNA from Drosophila melanogaster during development through to hatching. The mutation frequency was enhanced approximately 16-fold over the levels seen in the experimental background. Due to the lack of proofreading, these gypsy elements represent hypervariable loci within the Drosophila genome, suggesting that dNTP pool imbalances in vivo are mutagenic.


Assuntos
Drosophila melanogaster/genética , Mutagênese/efeitos dos fármacos , Retroelementos/genética , Timidina/toxicidade , Sequência de Aminoácidos , Animais , Sequência de Bases , Códon/genética , Drosophila melanogaster/crescimento & desenvolvimento , Larva/efeitos dos fármacos , Larva/crescimento & desenvolvimento , Dados de Sequência Molecular , Óvulo/efeitos dos fármacos
15.
J Mol Biol ; 328(3): 521-35, 2003 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-12706714

RESUMO

The RAD51 protein, a eukaryotic homologue of the Escherichia coli RecA protein, plays an important role in the repair of DNA double-strand breaks (DSBs) by homologous recombination (HR) in mammalian cells. Recent findings suggest that HR may be important in repair following replication arrest in mammalian cells. Here, we have investigated the role of RAD51 in the repair of different types of damage induced during DNA replication with etoposide, hydroxyurea or thymidine. We show that etoposide induces DSBs at newly replicated DNA more frequently than gamma-rays, and that these DSBs are different from those induced by hydroxyurea. No DSB was found following treatment with thymidine. Although these compounds appear to induce different DNA lesions during DNA replication, we show that a cell line overexpressing RAD51 is resistant to all of them, indicating that RAD51 is involved in repair of a wide range of DNA lesions during DNA replication. We observe fewer etoposide-induced DSBs in RAD51-overexpressing cells and that HR repair of etoposide-induced DSBs is faster. Finally, we show that induced long-tract HR in the hprt gene is suppressed in RAD51-overexpressing cells, although global HR appears not to be suppressed. This suggests that overexpression of RAD51 prevents long-tract HR occurring during DNA replication. We discuss our results in light of recent models suggested for HR at stalled replication forks.


Assuntos
Dano ao DNA , Reparo do DNA , Replicação do DNA , Proteínas de Ligação a DNA/fisiologia , Animais , Apoptose , Linhagem Celular , Cricetinae , Relação Dose-Resposta a Droga , Etoposídeo/toxicidade , Hidroxiureia/toxicidade , Hipoxantina Fosforribosiltransferase/genética , Cinética , Modelos Genéticos , Rad51 Recombinase , Recombinação Genética , Timidina/toxicidade
16.
Carcinogenesis ; 22(1): 179-86, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11159757

RESUMO

We have demonstrated previously that the toxicity of 5-hydroxymethyl-2'-deoxyuridine (hmdUrd) to Chinese hamster fibroblasts (V79 cells) results from enzymatic removal of large numbers of hydroxymethyluracil residues from the DNA backbone [Boorstein,R. et al. (1992) Mol. Cell. Biol., 12, 5536-5540]. Here we report that a significant portion of the hmdUrd-induced cell death that is dependent on DNA base excision repair in V79 cells is apoptosis. Incubation of V79 cells with pharmacologically relevant concentrations of hmdUrd resulted in the characteristic changes of apoptosis as measured by gel electrophoresis, flow cytometry and phase contrast microscopy. However, hmdUrd did not induce apoptosis in V79mut1 cells, which are deficient in DNA base excision repair of 5-hydroxymethyluracil (hmUra). Apoptosis was not prevented by addition of 3-aminobenzamide, which inhibits synthesis of poly(ADP-ribose) from NAD, indicating that apoptosis was not the direct consequence of NAD depletion. Pulsed field gel electrophoresis indicated that hmdUrd treatment resulted in high molecular weight (2.2-4.5 Mb) DNA double-strand breaks prior to formation of internucleosomal ladders in V79 cells. Simultaneous measurement of DNA strand breaks with bromodeoxyuridine/terminal deoxynucleotidyl transferase-fluorescein isothiocyanate labeling and of cell cycle distribution indicated that cells with DNA strand breaks accumulated in late S/G(2) and that hmdUrd-treated cells underwent apoptosis after arrest in late S/G(2) phase. Our results indicate that excessive DNA base excision repair results in the generation of high molecular weight DNA double-strand breaks and eventually leads to apoptosis in V79 cells. Thus, delayed apoptosis following DNA damage can be a consequence of excessive DNA repair activity. Immunochemical analysis showed that both V79 and V79mut1 cells contained mutant p53, indicating that apoptosis induced by DNA base excision repair can be independent of p53.


Assuntos
Apoptose/fisiologia , Reparo do DNA/fisiologia , DNA/fisiologia , Pentoxil (Uracila)/metabolismo , Timidina/análogos & derivados , Proteína Supressora de Tumor p53/genética , Animais , Apoptose/efeitos dos fármacos , Bromodesoxiuridina , Ciclo Celular/fisiologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , DNA/efeitos dos fármacos , DNA/metabolismo , Dano ao DNA , DNA Nucleotidilexotransferase , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Fluoresceína-5-Isotiocianato , Fase G2/efeitos dos fármacos , Genes p53 , Células HL-60 , Humanos , Pulmão/citologia , Camundongos , Mutação , Pentoxil (Uracila)/análogos & derivados , Fosfatidilserinas/metabolismo , Timidina/toxicidade , Proteína Supressora de Tumor p53/biossíntese
17.
Mutat Res ; 374(2): 287-95, 1997 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-9100852

RESUMO

V79mut1 cells are resistant to the toxic effects of 5-hydroxymethyl-2'-deoxyuridine (hmdUrd) and are deficient in the DNA repair enzyme hydroxymethyluracil-DNA glycosylase (hmUDG). We have therefore proposed that the toxicity of hmdUrd results from the repair of the lesion from DNA. In order to clarify the biological role of hmUDG, we have determined whether the repair-deficient cells showed resistance or sensitivity to the toxic or mutagenic effects of other DNA-damaging agents. Cells were exposed to hmdUrd, ionizing or ultraviolet radiation, to the alkylating agent MNNG, and to oxidative stress produced by hypoxanthine/xanthine oxidase, glucose/glucose oxidase, nitric oxide donor SNAP, or to H2O2. The V79mut1 cells did not show increased mutagenesis in response to hmdUrd. Relative to the V79 parent cells, the V79mut1 cells were not markedly altered in sensitivity to oxidizing agents and ionizing radiation (which produce hmdUra in DNA). The repair-deficient cells wee equally sensitive as the parent V79 cells to DNA damage induced by ultraviolet radiation or by MNNG. No significant differences were seen between the parent and the repair-deficient cells in terms of synthesis of poly(ADP-ribose) in response to damage or in their sensitization to 3-aminobenzamide. Thus, the loss of the 5-hydroxymethyluracil (hmUra)-DNA glycosylase activity in mammalian cells in culture confers no obvious deleterious effect on cell survival or mutagenicity in response to a wide range of DNA damage. These studies indicate that the major lesion known to be repaired by hmUra-DNA glycosylase, an hmUra residue replacing thymine, is produced in cells only in small quantities as the result of exposure to common DNA-damaging agents. These results raise the possibility that hmUra-DNA glycosylase may have evolved to respond to other lesions than hmUra residues formed from the oxidation of thymine.


Assuntos
Dano ao DNA , DNA Glicosilases , Mutagênicos/toxicidade , N-Glicosil Hidrolases/metabolismo , Timidina/análogos & derivados , Alquilantes/toxicidade , Animais , Benzamidas/toxicidade , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cricetinae , Reparo do DNA , Resistência a Medicamentos , Raios gama , Glicosilação , Mutagênese/genética , Testes de Mutagenicidade , Oxidantes/toxicidade , Poli Adenosina Difosfato Ribose/biossíntese , Timidina/toxicidade , Raios Ultravioleta
19.
Anticancer Res ; 15(3): 951-8, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7645985

RESUMO

3'-Aminocyanoborane-2', 3'-dideoxythymidine (VIIa) and 3'-aminocyanoborane-2', 3'-dideoxyuridine (VIIIb) were successfully synthesized. The thymidine derivative (VIIIa) was shown to be a potent cytotoxic agent in murine and selected human suspended and solid tumor cell lines. Compound VIIIa inhibited L-1210 leukemia DNA and RNA synthesis with the protein synthesis requiring a higher concentration of drug for inhibition within 60 min. The purine pathway appeared to be the major target of Compound VIIIa with inhibition of IMP dehydrogenase and dihydrofolate reductase activities. The compound affected metabolic enzyme activities in the pyrimidine pathway as well as the nucleoside kinase activities. The DNA molecule did not appear to be target of the 3'-aminocyanoborane-2', 3'-dideoxythymidine (VIIIa), in that there was no change in ct-DNA viscosity, thermal denaturation or absorption of nucleosides of DNA nor was there any L-1210 DNA strand scission or inhibition of L-1210 DNA topoisomerase II activity when compound VIIIa was incubated at 100 microM.


Assuntos
Antineoplásicos/toxicidade , Compostos de Boro/síntese química , Compostos de Boro/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Desoxiuridina/análogos & derivados , Timidina/análogos & derivados , Animais , Antineoplásicos/síntese química , Linhagem Celular , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , DNA de Neoplasias/antagonistas & inibidores , DNA de Neoplasias/biossíntese , DNA de Neoplasias/química , Desoxiuridina/síntese química , Desoxiuridina/toxicidade , Ensaios de Seleção de Medicamentos Antitumorais , Células HeLa , Humanos , Indicadores e Reagentes , Leucemia L1210 , Camundongos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Desnaturação de Ácido Nucleico , RNA Neoplásico/antagonistas & inibidores , RNA Neoplásico/biossíntese , Ratos , Relação Estrutura-Atividade , Timidina/síntese química , Timidina/toxicidade , Células Tumorais Cultivadas , Viscosidade
20.
Mutat Res ; 336(3): 293-305, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7739616

RESUMO

5-Hydroxymethyl-2'-deoxyuridine (hmdUrd) is incorporated into the DNA of V79 Chinese hamster cells as an analogue of thymidine. Incorporated residues are then recognized and excised by hmUra-DNA glycosylase (hmUDG). The removal of large numbers of hmUra residues and subsequent strand breakage is cytotoxic, as has been demonstrated by our finding that a mutant cell line, which is deficient in this enzyme, is resistant to hmdUrd (Boorstein et al., 1992a). In order to determine whether topoisomerase I plays a role in hmUDG initiated base excision repair, V79 cells and repair deficient V79mut1 cells were exposed to combinations of hmdUrd and the topoisomerase I inhibitors camptothecin (CPT), CPT-11, and beta-lapachone. Treatment of V79 cells with hmdUrd followed by non-toxic concentrations of camptothecin or CPT-11 showed significant enhancement of the baseline cytotoxicity of the hmdUrd alone. In contrast, camptothecin and CPT-11 had no effect in combination with hmdUrd in the V79mut1 cells. Non-toxic concentrations of beta-lapachone, which inhibits topoisomerase I by a different mechanism than camptothecin and CPT-11, produced no synergistic toxicity in V79 cells. Neither camptothecin nor CPT-11 inhibited removal of hmdUrd from hmdUrd treated cells, nor did they affect hmdUrd-induced poly(ADP-ribose) synthesis. Camptothecin did not alter the cell cycle distribution of either hmdUrd treated cells or untreated cells at concentrations sufficient to cause synergistic toxicity with hmdUrd. Results from our study indicate that the utility of topoisomerase I inhibitors may be enhanced by sensitizing cells with hmdUrd initiated repair activity which arrests cells in S-phase and produces DNA lesions that are further converted into lethal damage by camptothecin.


Assuntos
Antineoplásicos/farmacologia , Camptotecina/toxicidade , Reparo do DNA/fisiologia , Timidina/análogos & derivados , Inibidores da Topoisomerase I , Animais , Antibióticos Antineoplásicos/toxicidade , Camptotecina/análogos & derivados , Ciclo Celular/efeitos dos fármacos , Linhagem Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Reparo do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Irinotecano , Pulmão/citologia , Naftoquinonas/toxicidade , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/biossíntese , Fase S , Timidina/metabolismo , Timidina/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...