Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 21(18)2020 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-32927807

RESUMEN

BrdU (bromodeoxyuridine) and EdU (ethynyldeoxyuridine) have been largely utilized as the means of monitoring DNA replication and cellular division. Although BrdU induces gene and chromosomal mutations and induces sensitization to photons, EdU's effects have not been extensively studied yet. Therefore, we investigated EdU's potential cytotoxic and mutagenic effects and its related underlying mechanisms when administered to Chinese hamster ovary (CHO) wild type and DNA repair-deficient cells. EdU treatment displayed a higher cytotoxicity and genotoxicity than BrdU treatment. Cells with defective homologous recombination repair displayed a greater growth delay and severe inhibition of clonogenicity with EdU compared to wild type and other DNA repair-deficient cells. Inductions of sister chromatid exchange and hypoxanthine phosphorybosyl transferase (HPRT) mutation were observed in EdU-incorporated cells as well. Interestingly, on the other hand, EdU did not induce sensitization to photons to the same degree as BrdU. Our results demonstrate that elevated concentrations (similar to manufacturers suggested concentration; >5-10 µM) of EdU treatment were toxic to the cell cultures, particularly in cells with a defect in homologous recombination repair. Therefore, EdU should be administered with additional precautions.


Asunto(s)
Desoxiuridina/análogos & derivados , Células A549 , Animales , Bromodesoxiuridina , Células CHO , Cricetulus , Reparación del ADN , Desoxiuridina/toxicidad , Genes BRCA2 , Humanos , Pruebas de Mutagenicidad
2.
J Biol Chem ; 294(31): 11863-11875, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31201270

RESUMEN

Only a small portion of human immunodeficiency virus type 1 (HIV-1) particles entering the host cell results in productive infection, emphasizing the importance of identifying the functional virus population. Because integration of viral DNA (vDNA) is required for productive infection, efficient vDNA detection is crucial. Here, we use click chemistry to label viruses with integrase coupled to eGFP (HIVIN-eGFP) and visualize vDNA. Because click labeling with 5-ethynyl-2'-deoxyuridine is hampered by intense background staining of the host nucleus, we opted for developing HIV-1 reverse transcriptase (RT)-specific 2'-deoxynucleoside analogs that contain a clickable triple bond. We synthesized seven propargylated 2'-deoxynucleosides and tested them for lack of cytotoxicity and viral replication inhibition, RT-specific primer extension and incorporation kinetics in vitro, and the capacity to stain HIV-1 DNA. The triphosphate of analog A5 was specifically incorporated by HIV-1 RT, but no vDNA staining was detected during infection. Analog A3 was incorporated in vitro by HIV-1 RT and human DNA polymerase γ and did enable specific HIV-1 DNA labeling. Additionally, A3 supported mitochondria-specific DNA labeling, in line with the in vitro findings. After obtaining proof-of-principle of RT-specific DNA labeling reported here, further chemical refinement is necessary to develop even more efficient HIV-1 DNA labels without background staining of the nucleus or mitochondria.


Asunto(s)
Química Clic , Desoxiuridina/análogos & derivados , Transcriptasa Inversa del VIH/metabolismo , VIH-1/fisiología , Replicación Viral , Alquinos/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cartilla de ADN/metabolismo , Desoxiuridina/metabolismo , Desoxiuridina/toxicidad , Transcriptasa Inversa del VIH/antagonistas & inhibidores , VIH-1/genética , Humanos , Cinética , Microscopía Confocal , ARN Viral/química , ARN Viral/metabolismo
3.
Methods Mol Biol ; 1999: 307-318, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31127587

RESUMEN

Understanding the molecular dynamics of DNA replication in vivo has been a formidable challenge requiring the development of advanced technologies. Over the past 50 years or so, studies involving DNA autoradiography in bacterial cells have led to sophisticated DNA tract analyses in human cells to characterize replication dynamics at the single-molecule level. Our own lab has used DNA fiber analysis to characterize replication in helicase-deficient human cells. This work led us to propose a model in which the human DNA helicase RECQ1 acts as a governor of the single-stranded DNA binding protein RPA and regulates its bioavailability for DNA synthesis. We have also used the DNA fiber approach to investigate the interactive role of DDX11 helicase with a replication fork protection protein (Timeless) in human cells when they are under pharmacologically induced stress. In this methods chapter, we present a step-by-step protocol for the single-molecule DNA fiber assay. We describe experimental designs to study replication stress and staining patterns from pulse-chase labeling experiments to address the dynamics of replication forks in stressed cells.


Asunto(s)
Daño del ADN/genética , Replicación del ADN/genética , Imagen Individual de Molécula/métodos , Proteínas de Ciclo Celular/metabolismo , ARN Helicasas DEAD-box/metabolismo , Daño del ADN/efectos de los fármacos , ADN Helicasas/metabolismo , Replicación del ADN/efectos de los fármacos , ADN de Cadena Simple/metabolismo , Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidad , Células HeLa , Humanos , Idoxuridina/análogos & derivados , Idoxuridina/toxicidad , Péptidos y Proteínas de Señalización Intracelular/metabolismo , RecQ Helicasas/metabolismo , Proteína de Replicación A/metabolismo
4.
Int J Mol Sci ; 19(4)2018 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-29614057

RESUMEN

Replication errors that are caused by mutagens are critical for living cells. The aim of the study was to analyze the distribution of a DNA replication pattern on chromosomes of the H. vulgare 'Start' variety using pulse 5-ethynyl-2'-deoxyuridine (EdU) labeling, as well as its relationship to the DNA damage that is induced by mutagenic treatment with maleic hydrazide (MH) and γ ray. To the best of our knowledge, this is the first example of a study of the effects of mutagens on the DNA replication pattern in chromosomes, as well as the first to use EdU labeling for these purposes. The duration of the cell cycle of the Hordeum vulgare 'Start' variety was estimated for the first time, as well as the influence of MH and γ ray on it. The distribution of the signals of DNA replication along the chromosomes revealed relationships between DNA replication, the chromatin structure, and DNA damage. MH has a stronger impact on replication than γ ray. Application of EdU seems to be promising for precise analyses of cell cycle disturbances in the future, especially in plant species with small genomes.


Asunto(s)
Cromosomas de las Plantas/genética , Replicación del ADN/efectos de los fármacos , Hordeum/genética , Mutágenos/toxicidad , Cromosomas de las Plantas/efectos de los fármacos , Cromosomas de las Plantas/efectos de la radiación , Replicación del ADN/efectos de la radiación , Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidad , Rayos gamma/efectos adversos , Hordeum/efectos de los fármacos , Hordeum/efectos de la radiación
5.
Cereb Cortex ; 28(5): 1846-1856, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351579

RESUMEN

During differentiation, neurons progressively restrict their fate repressing the expression of specific genes. Here we describe the involvement in such developmental steps of the methyl-CpG binding protein 2 (MeCP2), an epigenetic factor that participates to chromatin folding and transcriptional regulation. We previously reported that, due to transcriptional impairments, the maturation of Mecp2 null neurons is delayed. To evaluate whether this could stem from altered progenitors proliferation and differentiation, we investigated whether lack of Mecp2 affects these features both in vitro and in vivo. We show that in Mecp2 null embryonic cortexes the expression of genes defining the identity of proliferating neuroprogenitors is enriched and that their permanence in the G1 phase is prolonged. Moreover, the number of cells transitioning from a stage of maturation to a more mature one is increased in Mecp2 null embryonic cortices, in line with the central role of G1 for cell identity refinement. We thus suggest that, possibly due to the lack of proper transcriptional control normally exerted by Mecp2, fate refinement is impaired in developing null cells. We propose that the maturation delay affecting the developing Mecp2 null cortex originates, at least in part, from deranged mechanisms of cell fate refinement.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Proteína 2 de Unión a Metil-CpG/deficiencia , Neuronas/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bromodesoxiuridina , Células Cultivadas , Ciclina D1/metabolismo , Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidad , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Femenino , Factores de Crecimiento de Fibroblastos/farmacología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Embarazo , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Mensajero
6.
Bioorg Med Chem ; 24(18): 4272-4280, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27460697

RESUMEN

A common method of evaluating cellular proliferation is to label DNA with chemical probes. 5-Ethynyl-2'-deoxyuridine (EdU) is a widely utilized chemical probe for labeling DNA, and upon incorporation, EdU treatment of cells is followed by a reaction with a small molecule fluorescent azide to allow detection. The limitations when using EdU include cytotoxicity and a reliance on nucleoside active transport mechanisms for entry into cells. Here we have developed six novel EdU pro-labels that consist of EdU modified with variable lipophilic acyl ester moieties. This pro-label:chemical probe relationship parallels the prodrug:drug relationship that is employed widely in medicinal chemistry. EdU and EdU pro-labels were evaluated for their labeling efficacy and cytotoxicity. Several EdU pro-label analogues incorporate into DNA at a similar level to EdU, suggesting that nucleoside transporters can be bypassed by the pro-labels. These EdU pro-labels also had reduced toxicity compared to EdU.


Asunto(s)
Desoxiuridina/análogos & derivados , Desoxiuridina/farmacología , Colorantes Fluorescentes/farmacología , Sondas Moleculares/farmacología , Línea Celular Tumoral , Proliferación Celular , ADN/genética , Desoxiuridina/síntesis química , Desoxiuridina/toxicidad , Ésteres/síntesis química , Ésteres/farmacología , Ésteres/toxicidad , Fluorescencia , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/toxicidad , Células HEK293 , Humanos , Sondas Moleculares/síntesis química , Sondas Moleculares/genética , Sondas Moleculares/toxicidad , Relación Estructura-Actividad
7.
PLoS One ; 10(2): e0117459, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25671308

RESUMEN

2'-Deoxy-5-ethynyluridine (EdU) has been previously shown to be a cell poison whose toxicity depends on the particular cell line. The reason is not known. Our data indicates that different efficiency of EdU incorporation plays an important role. The EdU-mediated toxicity was elevated by the inhibition of 2'-deoxythymidine 5'-monophosphate synthesis. EdU incorporation resulted in abnormalities of the cell cycle including the slowdown of the S phase and a decrease in DNA synthesis. The slowdown but not the cessation of the first cell division after EdU administration was observed in all of the tested cell lines. In HeLa cells, a 10 µM EdU concentration led to the cell death in the 100% of cells probably due to the activation of an intra S phase checkpoint in the subsequent S phase. Our data also indicates that this EdU concentration induces interstrand DNA crosslinks in HeLa cells. We suppose that these crosslinks are the primary DNA damage resulting in cell death. According to our results, the EdU-mediated toxicity is further increased by the inhibition of thymidylate synthase by EdU itself at its higher concentrations.


Asunto(s)
Citotoxinas/toxicidad , Daño del ADN , Desoxiuridina/análogos & derivados , Inhibidores Enzimáticos/toxicidad , Timidilato Sintasa/antagonistas & inhibidores , Línea Celular , Proliferación Celular/efectos de los fármacos , Citotoxinas/metabolismo , ADN/biosíntesis , ADN/genética , ADN/metabolismo , Replicación del ADN/efectos de los fármacos , Desoxiuridina/metabolismo , Desoxiuridina/toxicidad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/metabolismo , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Fase S/efectos de los fármacos , Tetrahidrofolatos/biosíntesis , Timidina/metabolismo , Timidina/farmacología , Timidina Monofosfato/metabolismo
8.
Angew Chem Int Ed Engl ; 53(39): 10541-4, 2014 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-25145319

RESUMEN

An aptamer specifically binding the interleukin-6 receptor and intrinsically comprising multiple units of the nucleoside analogue 5-fluoro-2'-deoxyuridine can exert a cytostatic effect direcly on certain cells presenting the receptor. Thus the modified aptamer fulfils the requirements for active drug targeting in an unprecedented manner. It can easily be synthesized in a single enzymatic step and it binds to a cell surface receptor that is conveyed into the lysosome. Upon degradation of the aptamer by intracellular nucleases the active drug is released within the targeted cells exclusively. In this way the aptamer acts as a prodrug meeting two major prerequisites of a drug delivery system: specific cell targeting and the controlled release of the drug triggered by an endogenous stimulus.


Asunto(s)
Aptámeros de Nucleótidos/química , Desoxiuridina/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Aptámeros de Nucleótidos/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Desoxiuridina/química , Desoxiuridina/uso terapéutico , Desoxiuridina/toxicidad , Portadores de Fármacos/química , G-Cuádruplex , Ratones , Neoplasias/tratamiento farmacológico , Profármacos/química , Profármacos/uso terapéutico , Profármacos/toxicidad , Unión Proteica , Receptores de Interleucina-6/química , Receptores de Interleucina-6/metabolismo
9.
Nucleic Acids Res ; 41(8): 4650-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23449220

RESUMEN

Therapeutic small interfering RNAs (siRNAs) are composed of chemically modified nucleotides, which enhance RNA stability and increase affinity in Watson-Crick base pairing. However, the precise fate of such modified nucleotides once the siRNA is degraded within the cell is unknown. Previously, we demonstrated that deoxythymidine release from degraded siRNAs reversed the cytotoxicity of thymidylate synthase (TS)-targeted siRNAs and other TS inhibitor compounds. We hypothesized that siRNAs could be designed with specific nucleoside analogues that, once released, would enhance siRNA cytotoxicity. TS-targeted siRNAs were designed that contained 5-fluoro-2'-deoxyuridine (FdU) moieties at various locations within the siRNA. After transfection, these siRNAs suppressed TS protein and messenger RNA expression with different efficiencies depending on the location of the FdU modification. FdU was rapidly released from the siRNA as evidenced by formation of the covalent inhibitory ternary complex formed between TS protein and the FdU metabolite, FdUMP. These modified siRNAs exhibited 10-100-fold greater cytotoxicity and induced multiple DNA damage repair and apoptotic pathways when compared with control siRNAs. The strategy of designing siRNA molecules that incorporate cytotoxic nucleosides represents a potentially novel drug development approach for the treatment of cancer and other human diseases.


Asunto(s)
Desoxiuridina/análogos & derivados , ARN Interferente Pequeño/química , ARN Interferente Pequeño/toxicidad , Apoptosis , Línea Celular Tumoral , Desoxiuridina/toxicidad , Humanos , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo , Transfección
10.
ChemMedChem ; 7(10): 1825-39, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22952012

RESUMEN

Apurinic/apyrimidinic (AP) endonuclease 1 (APE1) is a multifaceted protein with an essential role in the base excision repair (BER) pathway. Its implication in tumor development, progression, and resistance has been confirmed in multiple cancers, making it a viable target for intensive investigation. In this work, we designed and synthesized different classes of small-molecule inhibitors of the catalytic endonuclease function of APE1 that contain a 3-carbamoylbenzoic acid scaffold. Further structural modifications were made with the aim of increasing the activity and cytotoxicity of these inhibitors. Several of our compounds were shown to inhibit the catalytic endonuclease function of APE1 with potencies in the low-micromolar range in vitro, and therefore represent novel classes of APE1 inhibitors worthy of further development.


Asunto(s)
Ácido Benzoico/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , Diseño de Fármacos , Compuestos de Metilurea/química , Ácido Benzoico/síntesis química , Ácido Benzoico/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidad , Humanos
11.
Anal Biochem ; 417(1): 112-21, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21683679

RESUMEN

The labeling of newly synthesized DNA in cells to identify cell proliferation is an important experimental technique. The most accurate methods incorporate [(3)H]thymidine or 5-bromo-2'-deoxyruidine (BrdU) into dividing cells during S phase, which is subsequently detected by autoradiography or immunohistochemistry, directly measuring the newly synthesized DNA. Recently, a novel method was developed to detect DNA synthesis in proliferating cells based on a novel thymidine analog, 5-ethynyl-2'-deoxyuridine (EdU). EdU is incorporated into DNA and subsequently detected with a fluorescent azide via "click" chemistry. This novel technique is highly sensitive and does not require DNA denaturation. However, it was also found that EdU exhibits time-dependent inhibition effects on cell growth. Therefore, here we report a novel deoxycytidine analog, 5-ethynyl-2'-deoxycytidine (EdC), that can be used to detect DNA synthesis in vitro and in vivo at a similar sensitivity level compared with EdU. Furthermore, the EdC-induced cytotoxicity is much less than that of EdU when combined with thymidine. This will be a potential application for the long-term detection of proliferating cells.


Asunto(s)
ADN/metabolismo , Desoxiuridina/análogos & derivados , Coloración y Etiquetado/métodos , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN/biosíntesis , Desoxiuridina/química , Desoxiuridina/metabolismo , Desoxiuridina/toxicidad , Citometría de Flujo , Humanos , Ratones , Especificidad de Órganos/efectos de los fármacos , Timidina/metabolismo , Factores de Tiempo
12.
ChemMedChem ; 6(3): 457-64, 2011 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-21280228

RESUMEN

Recent evidence has shown that the gemcitabine metabolite, dFdU, is pharmacologically active. Though less potent, dFdU has a longer half-life and could potentiate or antagonize the activity of gemcitabine. Hence, studies were undertaken to evaluate the combined effects. Following chemical synthesis, an improved purification procedure for dFdU was developed (80 % yield; >99 % purity). Zebrafish phenotype-based embryo screens revealed no acute toxicity after gemcitabine or dFdU treatment. Only gemcitabine affected zebrafish development in a dose-dependent manner. Synergy or antagonism for the combination was not observed. Antitumor effects for dFdU were dose dependent. Antagonism was tumor cell-line dependent and did not depend on formation of the intracellular active metabolite of gemcitabine, suggesting that the drug-metabolite interaction occurs later. These studies highlight a platform for testing the pharmacologic activity for anticancer agent and metabolite combinations. Such analyses are expected to provide insight into the beneficial or harmful effect(s) of metabolites towards parent drug activity.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Desoxicitidina/análogos & derivados , Floxuridina/análogos & derivados , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Línea Celular Tumoral , Técnicas Químicas Combinatorias , Desoxicitidina/química , Desoxicitidina/metabolismo , Desoxicitidina/toxicidad , Desoxiuridina/química , Desoxiuridina/toxicidad , Embrión no Mamífero , Desarrollo Embrionario/efectos de los fármacos , Floxuridina/química , Floxuridina/toxicidad , Humanos , Ensayo de Tumor de Célula Madre , Pez Cebra/embriología , Gemcitabina
13.
Bioorg Med Chem Lett ; 21(4): 1151-4, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21251826

RESUMEN

This Letter describes the novel radiosensitizing agents based on nucleoside base modification. In addition to the known 5-phenylselenide derivative, 5-methylselenide modified thymidine, which has a van der Waals radius smaller than the phenyl group, was newly synthesized. The similar monomer activity of 5-methylselenide derivative under oxidation condition was confirmed by NMR experiments. The cytotoxicity tests and radiosensitizing experiments of both compounds were carried out using the H460 lung cancer cell line. Both the 5-phenylselenide and the 5-methylselenide derivatives showed a relatively low toxicity to the cells. However, in combination with γ-radiolysis, both exerted good radiosensitizing effects to the lung cancer cell lines in vitro. This result confirms that 5-methylselenide modified thymidine could be a useful candidate as a potential radiosensitizing agent in vivo.


Asunto(s)
Desoxiuridina/análogos & derivados , Compuestos de Organoselenio/química , Fármacos Sensibilizantes a Radiaciones/química , Línea Celular Tumoral , Desoxiuridina/síntesis química , Desoxiuridina/química , Desoxiuridina/toxicidad , Rayos gamma , Humanos , Espectroscopía de Resonancia Magnética , Compuestos de Organoselenio/síntesis química , Compuestos de Organoselenio/toxicidad , Oxidación-Reducción , Fármacos Sensibilizantes a Radiaciones/síntesis química , Fármacos Sensibilizantes a Radiaciones/toxicidad
14.
Cytometry A ; 75(6): 535-46, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19235202

RESUMEN

Using the nucleoside analogue EdU (5-ethynyl-2'-deoxyuridine) for thymidine substitution instead of BrdU (5-bromo-2'-deoxyuridine) in cell proliferation assays has recently been proposed. However, the effect of EdU on cell viability, DNA synthesis, and cell cycle progression and consequently its usability for dynamic cell proliferation analysis in vitro has not been explored. We compared the effect of EdU and BrdU incorporation into SK-BR-3 and BT474 breast cancer cells and the impact on cell cycle kinetics, cell viability, and DNA damage. We found that EdU can be used not only for pulse but also for continuous cell labeling and henceforth in high resolution EdU/Hoechst quenching assays. BrdU and EdU proliferation assays based on click chemistry revealed comparable results. However, cell viability of SK-BR-3 breast cancer cells was highly affected by long term exposure to EdU. Both SK-BR-3 as well as BT474 cells show cell cycle arrests upon long term EdU treatment whereas only SK-BR-3 cells were driven into necrotic cell death by long term exposure to EdU. In contrast BT474 cells appeared essentially unharmed by EdU treatment in terms of viability. Consequently using EdU enables highly sensitive and quantitative detection of proliferating cells and facilitates even continuous cell cycle assessment. Nevertheless, potential cellular susceptibility needs to be individually evaluated.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Desoxiuridina/análogos & derivados , Citometría de Flujo , Apoptosis , Bromodesoxiuridina/toxicidad , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclina B/efectos de los fármacos , Ciclina B/metabolismo , Ciclina B1 , ADN/metabolismo , Desoxiuridina/toxicidad , Femenino , Histonas/efectos de los fármacos , Histonas/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología
15.
Mutat Res ; 568(2): 195-209, 2004 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-15542107

RESUMEN

5-(2-chloroethyl)-2'-deoxyuridine (CEDU) had been developed for the treatment of herpes simplex infections. In the Salmonella reverse mutation test, the compound was found to be mutagenic in strains TA1535 and TA102 at very high concentrations (> or =2500 micro g/plate), both with and without S9-mix. The mutagenic potential of CEDU was further investigated in vivo and in vitro. It did not induce DNA repair in rat hepatocyte primary cultures, and was negative in the micronucleus test in V79 cells and in the comet assay in human leukocytes. In vivo, CEDU was negative in the bone marrow micronucleus test in CD1 mice. The mouse spot test provided a clearly positive result. Treatment of mice on day 9 of pregnancy with 2000 mg/kg resulted in 5.9% of the F1 animals having genetically relevant spots, whereas the corresponding vehicle control group had a spot rate of 1.9%. Since these data clearly identified CEDU as an inducer of gene mutations in vivo, this potential was further investigated in lacZ transgenic Muta Mouse. Six female animals were treated daily on five consecutive days with 2000 mg/kg/day and sacrificed, after a treatment-free sampling time, 14 days later. The data showed a clear increase in the mutant frequency in the bone marrow, the lung and in the spleen. CEDU is an exception in the group of nucleoside analogues, because it was found to be a strong gene mutagen and, in contrast to the other compounds of this group investigated so far, had no considerable clastogenic effects.


Asunto(s)
Antivirales/toxicidad , Reparación del ADN/genética , Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidad , Mutación/genética , Animales , Ensayo Cometa , Desoxiuridina/química , Femenino , Masculino , Ratones , Pruebas de Micronúcleos , Embarazo , Ratas , Salmonella , Pigmentación de la Piel/efectos de los fármacos
16.
Mutat Res ; 568(2): 211-20, 2004 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-15542108

RESUMEN

5-(2-chloroethyl)-2'-deoxyuridine (CEDU) is a pyrimidine nucleoside analogue formerly in development for the treatment of herpes simplex virus infections. The compound proved clearly mutagenic in the mouse spot test and exhibited weak activity in the Salmonella reverse mutation test, which led to the termination of the compound's development. In another study, CEDU, administered orally to beta-galactosidase (lacZ) transgenic mice (Muta Mouse) for five days, induced a clear increase in lacZ mutant frequencies in spleen, lung, and bone marrow. In the present follow-up study, we analyzed 32 of those lacZ mutants isolated from the bone marrow of the Muta Mouse animals of the experiments mentioned above, in order to obtain further information on the type of mutations induced by CEDU. CEDU induced a pronounced increase in A:T to G:C transitions. The distribution of A:T to G:C transitions was clearly non-random, showing a bias towards T to C substitutions in the coding DNA strand and a preference to occur in the sequence motif 5'-(G or C)-T-G-3'. Our data support the hypothesis that CEDU, after being phosphorylated, is incorporated into cellular DNA in place of thymidine, which leads to mispairing with guanosine during subsequent DNA replication. As a result, the compound is thought to exert its mutagenicity by inducing mismatches leading to T to C transitions. Our findings point towards a mode of mutagenic action of CEDU that differs fundamentally from that of other antiviral antinucleosides whose clastogenic and recombinogenic activities prevail.


Asunto(s)
Antivirales/toxicidad , Reparación del ADN/efectos de los fármacos , Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidad , Operón Lac/genética , Mutación/genética , Animales , Antivirales/química , Composición de Base , Secuencia de Bases , Células de la Médula Ósea , Análisis Mutacional de ADN , Cartilla de ADN , Desoxiuridina/química , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
17.
Toxicol Appl Pharmacol ; 192(1): 69-77, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14554104

RESUMEN

Nonciliated bronchiolar (Clara cells) are progenitor cells during development. During differentiation, they are more susceptible to injury by environmental toxicants metabolized by the cytochrome P450 monooxygenase system, and injury results in altered bronchiolar repair and development. Squamous cells and abnormal cuboidal epithelium persist into early adulthood. The hypothesis tested in this study was that the failure of bronchiolar epithelium to repair normally in neonates following injury is due to an inhibition of proliferation. A model of differential repair in rabbit kits was used. Proliferation was followed for 1 week post injury in rabbit kits treated with a single dose of the P450-mediated cytotoxicant 4-ipomeanol (IPO) at 7 days old (repair abnormal) and compared to rabbits treated with a single dose of IPO at 21 days old (repair normal). Proliferation was measured by the nuclear incorporation of 5-chloro-2'-deoxyuridine (CldU) within epithelium at the target site (terminal bronchiole). The repair pattern between the two age groups was histologically defined. There was no difference in the CldU labeling index during the week of repair between the two age groups, even though the bronchiolar epithelium did not return to normal in the animals treated at 7 days old. In summary, proliferation (through S-phase) is not inhibited during repair in neonatal rabbits treated with IPO at 7 days old compared to animals treated at 21 days old, and we conclude that other factors may be responsible for the altered repair in the young neonates injured by a P450-mediated cytotoxicant.


Asunto(s)
Animales Recién Nacidos/fisiología , Antineoplásicos/toxicidad , Bronquios/citología , Desoxiuridina/análogos & derivados , Terpenos/toxicidad , Animales , Bronquios/efectos de los fármacos , Recuento de Células , División Celular/efectos de los fármacos , Desoxiuridina/toxicidad , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Inmunohistoquímica , Conejos , Regeneración/fisiología
18.
Mutat Res ; 476(1-2): 99-107, 2001 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-11336987

RESUMEN

5-Formyluracil (5-foU) is a major oxidation product of thymine formed in yields comparable to that of 8-oxoguanine in DNA by ionizing radiation. Whereas the mutagenic effects of 8-oxoguanine are well understood, the investigation of the biological implications of 5-foU has so far been limited. Here we demonstrate that 5-formyl-2'-deoxyuridine (5-fodUrd) supplied to the growth medium of Escherichia coli induces several base substitutions at different frequencies at position 461 in the lacZ gene in the following order: A.T-->G.C>G.C-->A.T>G.C-->T.A>>A.T-->T.A>A.T-->C.G. No induction of G.C-->C.G transversions was observed. It is inferred that 5-fodUrd will be incorporated into the DNA during cell growth, forming mispairs with guanine, cytosine and thymine during replication. It, thus, appears that cell growth in the presence of 5-fodUrd may represent a good model for elucidating the cellular effects of 5-foU residues in DNA.


Asunto(s)
Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidad , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Mutación , Disparidad de Par Base , Secuencia de Bases , Daño del ADN , Análisis Mutacional de ADN , ADN Bacteriano/química , ADN Bacteriano/efectos de los fármacos , ADN Bacteriano/genética , Genes Bacterianos/efectos de los fármacos , Operón Lac/efectos de los fármacos , Modelos Químicos , Mutagénesis
19.
Toxicol Lett ; 119(1): 71-8, 2001 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-11275423

RESUMEN

Oxidation of the methyl group of thymine yields 5-(hydroxymethyl)uracil (5-hmU) and 5-formyluracil (5-foU) as major products. Whereas 5-hmU appears to have normal base pairing properties, the biological effects of 5-foU are rather poorly characterised. Here, we show that the colony forming ability of Chinese hamster fibroblast (CHF) cells is greatly reduced by addition of 5-foU, 5-formyluridine (5-foUrd) and 5-formyl-2'-deoxyuridine (5-fodUrd) to the growth medium. There are no toxic effects of 5-fodUrd on cells defective in thymidine kinase or thymidylate synthetase, suggesting that the toxicity may be caused by 5-fodUrd phosphorylation and subsequent inhibition of thymidylate synthetase. Whereas 5-fodUrd was the most effective 5-foU derivative causing cell growth inhibition, the corresponding ribonucleoside 5-foUrd was more effective in inhibiting [3H]uridine incorporation in non-dividing rat nerve cells in culture, suggesting that 5-foUrd exerts its toxicity through interference with RNA rather than DNA synthesis. Addition of 5-foU and 5-fodUrd was also found to promote mutagenicity at the hypoxanthine-guanine phosphoribosyltransferase (HPRT) locus of CHF cells; 5-fodUrd being three orders of magnitude more potent than 5-foU. In contrast, neither 5-hmU nor 5-(hydroxymethyl)-2'-deoxyuridine induced HPRT mutations. The mutation induction indicates that 5-foU will be incorporated into DNA and has base pairing properties different from that of thymine. These results suggest that 5-foU residues, originating from incorporation of oxidised bases, nucleosides or nucleotides or by oxidation of DNA, may contribute significantly to the damaging effects of oxygen radical species in mammalian cells.


Asunto(s)
ADN/metabolismo , Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidad , Mutágenos/toxicidad , ARN/metabolismo , Uracilo/análogos & derivados , Uracilo/toxicidad , Uridina/análogos & derivados , Uridina/toxicidad , Animales , División Celular/efectos de los fármacos , Cricetinae , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Células Tumorales Cultivadas
20.
Clin Cancer Res ; 6(2): 725-30, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10690559

RESUMEN

beta-5-o-Carboranyl-2'-deoxyuridine (D-CDU) is a nontoxic pyrimidine nucleoside analogue designed for boron neutron capture therapy of brain tumors. In vitro studies indicated that D-CDU accumulates to levels 92- and 117-fold higher than the extracellular concentration in rat 9L and human U-251 glioma cells, respectively, and persists for several hours at levels 5-fold higher than the extracellular concentration. Furthermore, D-CDU was not toxic to rats injected i.p. with up to 150 mg/kg. On the basis of these studies, D-CDU was evaluated as a neutron capture therapy agent using rats bearing stereotactically implanted intracranial 9L tumors at single i.p. doses of 30 mg/kg and 150 mg/kg of D-CDU (20% 10B enriched), given 2 h before irradiation with thermal neutrons. Boron concentrations in tumors 2 h after dosing were 2.3 +/- 1.6 and 7.4 +/- 1.3 micrograms boron/g tissue (mean +/- SD), corresponding to tumor/brain ratios of 11.5 +/- 3.6 and 6.8 +/- 2.0 micrograms boron/g tissue for the low and high doses, respectively. All untreated animals died within 28 days, whereas half survived at days 32, 55, and 38 for groups receiving neutrons only, 30 mg/kg D-CDU, and 150 mg/kg D-CDU, respectively. Odds ratios of all treatment groups differed significantly from the untreated group (P < 0.002; logrank test). The median survival time for the 30 mg/kg-treated group but not for the 150 mg/kg-treated group was significantly longer than for rats treated with neutrons only (P = 0.036), which may correlate with the decreased tumor selectivity for D-CDU observed at the higher dose. Additional pharmacodynamic studies are warranted to determine optimal dosing strategies for D-CDU.


Asunto(s)
Compuestos de Boro/farmacocinética , Compuestos de Boro/uso terapéutico , Terapia por Captura de Neutrón de Boro , Neoplasias Encefálicas/radioterapia , Desoxiuridina/análogos & derivados , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Animales , Compuestos de Boro/toxicidad , Desoxiuridina/farmacocinética , Desoxiuridina/uso terapéutico , Desoxiuridina/toxicidad , Humanos , Masculino , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Fármacos Sensibilizantes a Radiaciones/toxicidad , Ratas , Ratas Endogámicas F344 , Distribución Tisular , Trasplante Isogénico , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...