Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 40(13): 6016-25, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22447450

RESUMEN

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.


Asunto(s)
Reparación del ADN , Pentoxil (Uracilo)/análogos & derivados , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Animales , Línea Celular , Fibroblastos/efectos de los fármacos , Fluorouracilo/metabolismo , Marcación de Gen , Predisposición Genética a la Enfermedad , Longevidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína 2 Homóloga a MutS/genética , Neoplasias Experimentales/genética , Pentoxil (Uracilo)/metabolismo , Timidina/análogos & derivados , Timidina/toxicidad , beta-Galactosidasa/genética
2.
Chem Res Toxicol ; 26(11): 1730-8, 2013 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-24134187

RESUMEN

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.


Asunto(s)
Replicación del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/metabolismo , Timidina/toxicidad , Secuencia de Bases , Cromatografía Líquida de Alta Presión , ADN/metabolismo , ADN Polimerasa I/metabolismo , Cartilla de ADN/química , Cartilla de ADN/metabolismo , ADN Bacteriano/biosíntesis , Escherichia coli/enzimología , Humanos , Saccharomyces cerevisiae/enzimología , Estereoisomerismo , Espectrometría de Masas en Tándem , Timidina/análogos & derivados , Timidina/análisis , ADN Polimerasa iota
3.
Nucleic Acids Res ; 39(19): 8430-44, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21745813

RESUMEN

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.


Asunto(s)
Reparación del ADN , Fluorouracilo/metabolismo , Uracil-ADN Glicosidasa/metabolismo , Animales , Ciclo Celular , Línea Celular Tumoral , ADN/química , ADN/metabolismo , Daño del ADN , Endodesoxirribonucleasas/genética , Floxuridina/metabolismo , Floxuridina/toxicidad , Fluorouracilo/toxicidad , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteína 2 Homóloga a MutS/genética , ARN/metabolismo , Timidina/análogos & derivados , Timidina/metabolismo , Timidina/toxicidad , Timina ADN Glicosilasa/genética , Timina ADN Glicosilasa/metabolismo , Uracil-ADN Glicosidasa/genética , Uridina/análogos & derivados , Uridina/metabolismo , Uridina/toxicidad
4.
Br J Cancer ; 104(12): 1869-76, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21610703

RESUMEN

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.


Asunto(s)
Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Timidina/análogos & derivados , Terapia Ultravioleta , Neoplasias de la Vejiga Urinaria/terapia , Animales , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Daño del ADN , Femenino , Humanos , Quinazolinas/farmacología , Ratas , Ratas Endogámicas F344 , Tiofenos/farmacología , Timidina/metabolismo , Timidina/uso terapéutico , Timidina/toxicidad , Neoplasias de la Vejiga Urinaria/patología
5.
Exp Cell Res ; 315(15): 2539-43, 2009 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-19481540

RESUMEN

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.


Asunto(s)
Antineoplásicos/toxicidad , Arabinonucleósidos/toxicidad , ADN Mitocondrial/metabolismo , Timidina/análogos & derivados , Animales , Línea Celular Tumoral/efectos de los fármacos , ADN Mitocondrial/genética , Desoxicitidina Quinasa/genética , Desoxicitidina Quinasa/metabolismo , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Timidina/toxicidad , Timidina Quinasa/genética , Timidina Quinasa/metabolismo
6.
Biochemistry ; 48(33): 7833-41, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19618962

RESUMEN

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.


Asunto(s)
Daño del ADN , ADN Bacteriano/efectos de los fármacos , Repeticiones de Dinucleótido/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Mutagénesis , Timidina/análogos & derivados , Regiones no Traducidas 5'/efectos de los fármacos , Regiones no Traducidas 5'/genética , Bacteriófago M13/genética , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , ADN Bacteriano/síntesis química , Repeticiones de Dinucleótido/genética , Genoma Bacteriano/efectos de los fármacos , Radical Hidroxilo/toxicidad , Respuesta SOS en Genética/efectos de los fármacos , Eliminación de Secuencia/efectos de los fármacos , Azúcares Ácidos/toxicidad , Timidina/toxicidad
7.
Science ; 223(4636): 594-7, 1984 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-6695170

RESUMEN

When human lymphocytes were cultured with [3H]thymidine, which acts as a source of low-level chronic radiation, and then exposed to 150 rad of x-rays at 5, 7, 9, or 11 hours before fixation, the yield of chromatid aberrations was less than the sum of the yields of aberrations induced by [3H]thymidine and x-rays separately. Often fewer aberrations were found after exposure to radiation from both sources than were found after exposure to x-rays alone. At the same fixation times, nonradioactive thymidine did not affect the yield of x-ray-induced aberrations. The same phenomenon occurred at earlier fixation times, after exposure to 30 or 40 rad of x-rays and [3H]thymidine. This response is analogous to the adaptive response to alkylating agents whereby prior treatment with small doses for a long period reduces the damage occurring from large doses of similar agents given for a short time.


Asunto(s)
Aberraciones Cromosómicas , Linfocitos/efectos de la radiación , Timidina/toxicidad , Células Cultivadas , Deleción Cromosómica , Humanos , Linfocitos/efectos de los fármacos , Linfocitos/fisiología , Metafase/efectos de los fármacos , Metafase/efectos de la radiación , Tritio
8.
Nucleic Acids Res ; 35(12): 4103-13, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17567611

RESUMEN

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.


Asunto(s)
ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Línea Celular Transformada , ADN Helicasas/fisiología , Enzimas Reparadoras del ADN/fisiología , Genoma Humano , Humanos , Metilmetanosulfonato/toxicidad , Proteínas de Unión a Poli-ADP-Ribosa , Timidina/análogos & derivados , Timidina/toxicidad
9.
J Clin Invest ; 68(2): 544-52, 1981 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6114965

RESUMEN

Cultured leukemic T and null lymphocytes are highly sensitive to growth inhibition by thymidine, as well as the other deoxynucleosides, deoxyguanosine and deoxyadenosine. By contrast, Epstein-Barr virus-transformed B lymphocytes are relatively resistant to deoxynucleosides. Growth inhibition is associated with the development of high deoxyribotriphosphate pools after exposure to the respective deoxynucleotides. We show that malignant T and null lymphocytes are deficient in ecto-ATPase activity. We show this cell surface enzyme to be of broad specificity, capable of degrading both ribotriphosphates and deoxyribotriphosphates. High levels of this ecto-enzyme are found in deoxynucleoside-resistant, Epstein-Barr virus-transformed B lymphocytes. Ecto-ATPase deficiency may represent a mechanism for increased sensitivity to deoxynucleoside growth inhibition.


Asunto(s)
Adenosina Trifosfatasas/deficiencia , Leucemia/enzimología , Linfocitos/enzimología , Linfocitos T/enzimología , Timidina/toxicidad , Transformación Celular Viral , Células Cultivadas , Desoxiadenosinas/toxicidad , Desoxiguanosina/toxicidad , Espacio Extracelular/enzimología , Herpesvirus Humano 4 , Humanos , Timidina/metabolismo , Timidina Quinasa/metabolismo , Nucleótidos de Timina/metabolismo
10.
J Clin Invest ; 81(1): 87-91, 1988 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-3335645

RESUMEN

Marrow cells from seven untreated patients with polycythemia vera (PV) were used to initiate standard single inoculum long-term marrow cultures. The numbers, erythropoietin independence, and cycling behavior of all detectable classes of erythroid, granulopoietic, and multilineage progenitors were then evaluated and the results obtained compared with preculture values. Time course studies showed that the long-term marrow culture system supports the continuous proliferation of primitive neoplastic progenitor cells from PV patients for many weeks. However, these progenitors fail to respond to signals from the adherent layer that return their counterparts in normal long-term marrow cultures to a quiescent state 5-7 d after each medium change. This abnormal cycling behavior of PV cells in the long-term culture system appears to mimic that operative in vivo, where primitive hemopoietic progenitors are also in a continuous state of turnover, in contrast to similar primitive progenitor compartments in normal individuals, which are quiescent. The long-term marrow culture system thus offers new possibilities for the further analysis of abnormal cellular and molecular mechanisms underlying clonal expansion at the stem cell level in PV.


Asunto(s)
Médula Ósea/patología , Transformación Celular Neoplásica/patología , Células Madre Neoplásicas/patología , Policitemia Vera/patología , Células Tumorales Cultivadas/patología , Anciano , Transformación Celular Neoplásica/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/efectos de los fármacos , Policitemia Vera/metabolismo , Timidina/toxicidad , Células Tumorales Cultivadas/efectos de los fármacos
11.
Antiviral Res ; 76(2): 124-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17658623

RESUMEN

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.


Asunto(s)
Antivirales/uso terapéutico , Timidina/análogos & derivados , Virus Vaccinia/efectos de los fármacos , Vaccinia/tratamiento farmacológico , Administración Oral , Animales , Cidofovir , Citosina/administración & dosificación , Citosina/análogos & derivados , Citosina/uso terapéutico , Inyecciones Intraperitoneales , Ratones , Organofosfonatos/administración & dosificación , Organofosfonatos/uso terapéutico , Análisis de Supervivencia , Timidina/administración & dosificación , Timidina/uso terapéutico , Timidina/toxicidad , Ensayo de Placa Viral
12.
Radiats Biol Radioecol ; 47(1): 108-16, 2007.
Artículo en Ruso | MEDLINE | ID: mdl-17388002

RESUMEN

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.


Asunto(s)
ADN/efectos de la radiación , Células Endoteliales/efectos de la radiación , Fase G2/efectos de la radiación , Rayos gamma , Partículas beta , Proliferación Celular/efectos de la radiación , Radioisótopos de Cesio/toxicidad , Inestabilidad Cromosómica , Aberraciones Cromosómicas , ADN/química , ADN/metabolismo , Daño del ADN , Humanos , Timidina/análisis , Timidina/metabolismo , Timidina/toxicidad , Tritio
13.
Cancer Res ; 48(19): 5466-70, 1988 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-3416303

RESUMEN

5-Hydroxymethyluracil (HmUra) is formed from thymine in DNA through the action of ionizing radiation or reactive oxygen species generated by activated leukocytes. HmUra is removed from DNA by a specific DNA glycosylase, suggesting that it is also formed from endogenously generated reactive oxygen species and that its formation in DNA is potentially deleterious. To determine whether HmUra residues in DNA are mutagenic, hamster V79 cells were grown in the presence of 5-hydroxymethyl-2'-deoxyuridine (HmdUrd) which is incorporated into DNA, and mutagenicity at the ouabain- and thioguanine-resistant loci was determined. Levels of substitution ranged from 1/500 to 1/5,000 HmUra residues/thymine residues. There was slight mutagenicity at the thioguanine-resistant locus but none at the ouabain-resistant locus. The mutagenicity of HmdUrd, expressed as a function of HmUra substitution in DNA, was 1/30,000 in the hypoxanthine-guanine-phosphoribosyltransferase target gene. This low frequency indicates that the oxidation of thymine to HmUra in a preexisting AT base pair does not contribute significantly to the mutagenicity of ionizing radiation, because the yield of HmUra formed in DNA at mutagenic doses of radiation is too low. To determine whether repair of HmUra might be inhibited by ionizing radiation, cells were grown in medium containing HmdUrd and exposed to as much as 5 Gy of gamma-irradiation, and the removal of HmUra from DNA was measured. No inhibition of repair was noted. Preirradiation of cells neither accelerated the rate of repair nor raised the level of HmUra-DNA glycosylase activity, indicating that repair of HmUra was not induced by this type of oxidative stress. Although the mutagenicity of HmUra residues in DNA is low, even a rare mutation might be sufficiently deleterious to higher organisms to promote the development of HmUra-DNA glycosylase activity.


Asunto(s)
Timidina/análogos & derivados , Animales , Línea Celular , Supervivencia Celular , Cricetinae , Cricetulus , ADN Glicosilasas , Reparación del ADN , Pruebas de Mutagenicidad , N-Glicosil Hidrolasas/metabolismo , Pentoxil (Uracilo)/análogos & derivados , Pentoxil (Uracilo)/metabolismo , Timidina/toxicidad
14.
Cancer Res ; 47(12): 3190-4, 1987 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-3034403

RESUMEN

Due to similarities between human immunodeficiency virus and feline leukemia virus, the etiological agents of acquired immunodeficiency syndromes in humans and cats, the feline system was used as a model to conduct preliminary investigations as to the efficacy of the thymidine analogue 3'-azido-3'-deoxythymidine (AZT) as a therapeutic and preventive agent against retroviruses. In vitro evaluations of AZT cytotoxicity and its antiviral effects were conducted. Subsequently, 50 6-week-old specific pathogen free kittens were inoculated with a highly immunosuppressive strain of Richard-Feline Leukemia Virus. These cats were randomly subdivided into smaller groups with initiation of AZT treatment at variable times postinfection. All animals were periodically monitored for circulating infectious virus particles and virus-neutralizing antibodies. Their clinical condition was closely followed throughout the 6-week AZT treatment phase and for several months thereafter. The results indicate that AZT prevents retrovirus infection if administered immediately following virus exposure, and may also reduce retrovirus replication if administered to previously infected animals. Some of the treated cats developed neutralizing antibodies against the virus and became resistant to subsequent viral challenge. Future trials with this drug, both for the prevention and treatment of retroviral diseases in humans and animals, are warranted.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/prevención & control , Leucemia Experimental/tratamiento farmacológico , Timidina/análogos & derivados , Animales , Anticuerpos Antivirales/análisis , Antineoplásicos/uso terapéutico , Antivirales/uso terapéutico , Gatos , Virus de la Leucemia Felina , Timidina/uso terapéutico , Timidina/toxicidad , Zidovudina
15.
Cancer Res ; 40(3): 818-22, 1980 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7471098

RESUMEN

High-dose thymidine (dThd) was given to 12 patients with advanced hematological and solid tumors. The dose schedule used was 75 g/sq m/day, given i.v. continuously for 5 days or more. Myelosuppression, especially leukopenia, was the dose-limiting toxicity. Nonhematological toxicities affected the gastrointestinal tract (nausea, vomiting, anorexia, diarrhea, and indigestion) and the central nervous system (somnolence, headache, visual illusions, and memory impairment). Patients who had received cumulative doses of dThd developed alopecia. Thymine crystals were noted in the urine after refrigeration. Tumor regression (less than partial remission) occurred in one patient with melanoma. Three of four patients with acute leukemia had a fall in peripheral white blood cell counts and blasts but no marrow improvement. Four patients with adenocarcinoma (three colon, one unknown primary) had stable disease. Pharmacokinetic studies revealed that, at a dThd dose of 75 g/sq m/day, millimolar concentrations of dThd and thymine can be achieved in the plasma. The half-life of dThd was approximately 100 min. One-third of the plasma concentrations was measurable in the cerebrospinal fluid. dThd was mainly excreted by the kidneys.


Asunto(s)
Neoplasias/tratamiento farmacológico , Timidina/metabolismo , Adolescente , Adulto , Anciano , Niño , Evaluación de Medicamentos , Femenino , Humanos , Infusiones Parenterales , Cinética , Masculino , Persona de Mediana Edad , Timidina/administración & dosificación , Timidina/toxicidad
16.
Cancer Res ; 44(6): 2534-9, 1984 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-6722792

RESUMEN

Cytotoxicity and perturbations of the deoxyribonucleoside triphosphate pools caused by thymidine were studied in thymidine-sensitive and -resistant human tumor cells. Incubation with 1 mM thymidine reduced cell viability by more than 90% in the three sensitive cell lines (two melanomas and one adrenal carcinoma) and reduced the growth rate without decreasing the viability of resistant LO melanoma cells. Thymidine (1 mM) greatly increased the ratio of the deoxythymidine 5'-triphosphate to deoxycytidine 5'-triphosphate pools in the sensitive cells compared to LO cells and also caused larger relative increases in the pool sizes of deoxyguanosine 5'-triphosphate and deoxyadenosine 5'-triphosphate in the sensitive compared to the resistant cells. 3-Deazauridine, known to inhibit synthesis of deoxycytidine 5'-triphosphate and cytidine 5'-triphosphate in other cell lines, potentiated the cytotoxicity of thymidine for thymidine-sensitive BE melanoma and LO cells. In LO cells, 3-deazauridine (50 microM) decreased the intracellular pool of deoxycytidine 5'-triphosphate to the level obtained with 1 mM thymidine. Lower concentrations of deoxycytidine as compared to cytidine were required to protect BE and LO cells against the cytotoxicity of thymidine plus 3-deazauridine. Deoxycytidine also was more effective than was cytidine in preventing loss of cell viability after exposure to thymidine or to 3-deazauridine individually. In these human melanoma cells, ribonucleotide reductase may be a major site of action of thymidine, of 3-deazauridine, and of both drugs in combination. These results indicate that in human tumor cells the cytotoxic effect of thymidine correlates with greater perturbations of the pyrimidine deoxyribonucleoside 5'-triphosphate pools and that thymidine and 3-deazauridine, which independently reduce the intracellular levels of deoxycytidine 5'-triphosphate, act synergistically against human tumor cells.


Asunto(s)
3-Desazauridina/toxicidad , Neoplasias de las Glándulas Suprarrenales/fisiopatología , Melanoma/fisiopatología , Timidina/toxicidad , Uridina/análogos & derivados , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Desoxirribonucleótidos/metabolismo , Humanos , Cinética
17.
Cancer Res ; 40(8 Pt 1): 2730-5, 1980 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7388822

RESUMEN

The present paper describes the specific amelioration of 5-flourouracil (FUra)-induced host toxicity (manifest in body weight loss, leukopenia, and mortality) by testosterone in the spontaneous, autochthonous CD8F1 (BALB/c x DBA/8F1) murine breast tumor model. Administration of testosterone did not affect the growth rate of these hormone-independent tumors, and, most importantly the antitumor activity of FUra was not reduced in testosterone-treated mice. Therefore, the net result of treatment with the combination of FUra and testosterone was an increase in the selective antitumor specificity of FUra.


Asunto(s)
Fluorouracilo/toxicidad , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Testosterona/farmacología , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Quimioterapia Combinada , Femenino , Recuento de Leucocitos , Masculino , Ratones , Testosterona/administración & dosificación , Timidina/toxicidad
18.
Cancer Res ; 45(4): 1797-802, 1985 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-3978641

RESUMEN

To avoid infusing large volumes of fluid while treating patients with the standard thymidine solution (30 g/liter), it may be possible to administer this drug in more concentrated form. At 25 degrees C, thymidine is saturating at a concentration of 52 g/liter of 0.6% NaCl solution, and the thymidine concentration at saturation increases with temperature. Nude mice were infused at 29 degrees C with thymidine (60 or 72 g/liter) in cycles consisting of 4 to 5 days infusion followed by 9 days rest. Therapeutically effective doses of concentrated thymidine did not cause significant mortality in mice, and weight loss attributable to treatment was small and reversible. Significant growth inhibition of CA 1 human melanoma heterotransplants was observed after 3 treatment cycles. After 4 or 5 cycles, tumor responses were obtained in 7 mice (6 complete responses) of 12 inoculated with this tumor. These results show that concentrated thymidine solutions are highly effective against human tumor heterotransplants in nude mice and suggest that clinical use of concentrated thymidine may allow practical administration of maximum tolerated doses of this drug.


Asunto(s)
Antineoplásicos/uso terapéutico , Timidina/uso terapéutico , Animales , Antineoplásicos/toxicidad , Masculino , Melanoma/tratamiento farmacológico , Ratones , Ratones Desnudos , Solubilidad , Timidina/sangre , Timidina/toxicidad , Timina/sangre
19.
Cancer Res ; 40(5): 1718-21, 1980 May.
Artículo en Inglés | MEDLINE | ID: mdl-6989492

RESUMEN

Cultured malignant human lymphocytes are highly sensitive to growth inhibition by thymidine (50% inhibitory dose congruent to 10(-5) M). Growth inhibition reflects sustained elevation of the deoxythymidine 5'-triphosphate pool associated with secondary elevation of the deoxyguanosine 5'-triphosphate pool and reduction in the deoxycytidine 5'-triphosphate (dCTP) pool. Deoxycytidine was capable of partially reversing thymidine growth inhibition at a concentration of 0.5 microM, and growth recovery was virtually complete at 8 microM. The dCTP pool remained depressed until growth inhibition reversal by deoxycytidine was complete, and at a higher concentration of deoxycytidine the dCTP rose above control levels, but the deoxythymidine 5'-triphosphate and deoxyguanosine 5'-triphosphate pools remained elevated. These results support the view that thymidine growth inhibition induces a critical deficiency of dCTP via allosteric inhibition of ribonucleotide reductase rather than inhibiting DNA replication directly by elevated deoxythymidine 5'-triphosphate or deoxyguanosine 5'-triphosphate pools.


Asunto(s)
Desoxicitidina/farmacología , Leucemia/patología , Linfocitos/efectos de los fármacos , Timidina/antagonistas & inhibidores , División Celular/efectos de los fármacos , Línea Celular , Replicación del ADN/efectos de los fármacos , Desoxirribonucleótidos/metabolismo , Humanos , Ribonucleótido Reductasas/metabolismo , Timidina/toxicidad
20.
Cancer Res ; 40(1): 107-13, 1980 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7349890

RESUMEN

The serum half-life of 5-fluorouracil (5-FUra) in humans is best described as a biexponential decay function, with t1/2 alpha = 7.8 +/- 2.6 (S.E.) min and t1/2 beta = 36.8 +/- 13.5 min during initial courses of this drug alone. Pharmacokinetics of 5-FUra during courses of daily therapy (for 5 days) revealed prolongation of t1/2 in both components of the decay curve, which has not been previously reported. Despite the efficacy of thymidine (dThd) given as a continous i.v. infusion of 8 g/sq m/day in prevention of high-dose methotrexate toxicity, continuous infusion of dThd at this dose does not prevent the toxicity of 5-FUra orreverse inhibition of DNA and RNA synthesis by 5-fura. On the contrary, continuous infusion of dThd appears to increase the toxicity of 5-FUra during continuous dThd infusion revealed prolongation of the 5-FUra t1/2 which remained stable through the course of 5 days of 5-FUra with dThd. This protracted t1/2 is believed to account at least in part for the increased toxicity of 5-FUra with dThd. Dose-limiting mucositis, myelosuppression, and gastrointestinal toxicity were observed at 5-FUra doses ranging from one-half to two-thirds the customarily tolerated dose of 5-FUra alone in similar courses of daily bolus therapy (for 5 days).


Asunto(s)
Fluorouracilo/sangre , Neoplasias/sangre , Timidina/administración & dosificación , Adulto , Médula Ósea/efectos de los fármacos , Evaluación de Medicamentos , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/toxicidad , Semivida , Humanos , Infusiones Parenterales , Masculino , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Timidina/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA