Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Int J Mol Sci ; 22(14)2021 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-34299079

RESUMEN

In order to tackle the study of DNA repair pathways, the physical and chemical agents creating DNA damage, the genotoxins, are frequently employed. Despite their utility, their effects are rarely restricted to DNA, and therefore simultaneously harm other cell biomolecules. Methyl methanesulfonate (MMS) is an alkylating agent that acts on DNA by preferentially methylating guanine and adenine bases. It is broadly used both in basic genome stability research and as a model for mechanistic studies to understand how alkylating agents work, such as those used in chemotherapy. Nevertheless, MMS exerts additional actions, such as oxidation and acetylation of proteins. In this work, we introduce the important notion that MMS also triggers a lipid stress that stems from and affects the inner nuclear membrane. The inner nuclear membrane plays an essential role in virtually all genome stability maintenance pathways. Thus, we want to raise awareness that the relative contribution of lipid and genotoxic stresses when using MMS may be difficult to dissect and will matter in the conclusions drawn from those studies.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Daño del ADN , Lípidos/análisis , Metilmetanosulfonato/efectos adversos , Mutágenos/efectos adversos , Membrana Nuclear/patología , Epitelio Pigmentado de la Retina/patología , Reparación del ADN , Células Hep G2 , Humanos , Membrana Nuclear/efectos de los fármacos , Epitelio Pigmentado de la Retina/efectos de los fármacos
2.
FEBS J ; 286(6): 1154-1173, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30536627

RESUMEN

Chromatin regulates gene expression and genome maintenance, and consists of histones and other components. The post-translational modification of histones plays a key role in maintaining the structure and function of chromatin under different pathophysiological stress conditions. Here, we investigate the functions of previously unexplored amino acid residues in histones H3 and H4. To do so, we screened a library of yeast histone mutants following DNA damage and identified that substitution mutations of histone H3 (H3Q5A/E and H3Q120A) and H4 (H4Y88A/E and H4R78K) render yeast cells sensitive to DNA-damaging agents. These histone mutants show an activated DNA damage response, Rad53 phosphorylation and Sml1 degradation in the presence of methyl methanesulfonate (MMS). In histone H3Q5A/E mutants, RNR2 and RNR3 genes were induced at low level, as was RNR3 in H4 histone mutants following DNA damage. In H3 mutant cells, the cell cycle was deregulated, leading to inefficient cell cycle arrest in the presence of MMS, and genes involved in aging and DNA damage repair pathways were constitutively upregulated. In H3 mutants (H3Q5A, H3Q5E and H3Q120A), we observed reduced chronological lifespan (CLS), compared with extended CLS in the H4R78K mutant. Histone mutants also showed altered H3K4me and H3K56ac modifications and improper activation of the stress responsive Slt2 and Hog1 kinases. Thus, we have determined the significance of previously uncharacterized residues of H3 and H4 in DNA damage response, cell cycle progression and cellular aging.


Asunto(s)
Aminoácidos/genética , Senescencia Celular , Daño del ADN , Reparación del ADN , Histonas/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Aminoácidos/química , Aminoácidos/metabolismo , Puntos de Control del Ciclo Celular , Cromatina , Histonas/metabolismo , Metilmetanosulfonato/efectos adversos , Mutación , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
3.
Nat Commun ; 5: 4901, 2014 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-25249323

RESUMEN

ATM- and RAD3-related (ATR)/Chk1 and ataxia-telangiectasia mutated (ATM)/Chk2 signalling pathways play critical roles in the DNA damage response. Here we report that the E3 ubiquitin ligase Smurf1 determines cell apoptosis rates downstream of DNA damage-induced ATR/Chk1 signalling by promoting degradation of RhoB, a small GTPase recognized as tumour suppressor by promoting death of transformed cells. We show that Smurf1 targets RhoB for degradation to control its abundance in the basal state. DNA damage caused by ultraviolet light or the alkylating agent methyl methanesulphonate strongly activates Chk1, leading to phosphorylation of Smurf1 that enhances its self-degradation, hence resulting in a RhoB accumulation to promote apoptosis. Suppressing RhoB levels by overexpressing Smurf1 or blocking Chk1-dependent Smurf1 self-degradation significantly inhibits apoptosis. Hence, our study unravels a novel ATR/Chk1/Smurf1/RhoB pathway that determines cell fate after DNA damage, and raises the possibility that aberrant upregulation of Smurf1 promotes tumorigenesis by excessively targeting RhoB for degradation.


Asunto(s)
Daño del ADN/fisiología , Proteínas Quinasas/metabolismo , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Proteína de Unión al GTP rhoB/metabolismo , Análisis de Varianza , Apoptosis/fisiología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Fraccionamiento Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Daño del ADN/efectos de los fármacos , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Silenciador del Gen , Células HEK293 , Humanos , Immunoblotting , Metilmetanosulfonato/efectos adversos , Proteínas Quinasas/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteína de Unión al GTP rhoB/genética
4.
Mutat Res ; 760: 48-56, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24361396

RESUMEN

The genotoxicity and mutagenicity of formaldehyde (FA) has been well-characterized during the last years. Besides its known direct DNA-damaging and mutagenic activity in sufficiently exposed cells, FA at low concentrations might also enhance the mutagenic and carcinogenic effects of other environmental mutagens by interfering with the repair of DNA lesions induced by these mutagens. To further assess potential co-mutagenic effects of FA, we exposed A549 human lung cells to FA in combination with various mutagens and measured the induction and removal of DNA damage by the comet assay and the production of chromosomal mutations by the cytokinesis-block micronucleus assay (CBMN assay). The mutagens tested were ionizing radiation (IR), (±)-anti-B[a]P-7,8-dihydrodiol-9,10-epoxide (BPDE), N-nitroso-N-methylurea (methyl nitrosourea; MNU) and methyl methanesulfonate (MMS). FA (10-75µM) did not enhance the genotoxic and mutagenic activity of these mutagens under the test conditions applied. FA alone and in combination with MNU or MMS did not affect the expression (mRNA level) of the gene of the O(6)-methylguanine-DNA methyltransferase (MGMT) in A549 cells. The results of these experiments do not support the assumption that low FA concentrations might interfere with the repair of DNA damage induced by other mutagens.


Asunto(s)
Aberraciones Cromosómicas/efectos de los fármacos , Desinfectantes/efectos adversos , Sinergismo Farmacológico , Formaldehído/efectos adversos , Neoplasias Pulmonares/patología , Mutágenos/efectos adversos , 7,8-Dihidro-7,8-dihidroxibenzo(a)pireno 9,10-óxido/efectos adversos , Alquilantes/efectos adversos , Ensayo Cometa , Combinación de Medicamentos , Rayos gamma/efectos adversos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Metilmetanosulfonato/efectos adversos , Metilnitrosourea/efectos adversos , Pruebas de Micronúcleos , O(6)-Metilguanina-ADN Metiltransferasa/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
5.
Gene ; 519(1): 182-6, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23403232

RESUMEN

Here we report the physical mapping of the rad56-1 mutation to the NAT3 gene, which encodes the catalytic subunit of the NatB N-terminal acetyltransferase in Saccharomyces cerevisiae. Mutation of RAD56 causes sensitivity to X-rays, methyl methanesulfonate, zeocin, camptothecin and hydroxyurea, but not to UV light, suggesting that N-terminal acetylation of specific DNA repair proteins is important for efficient DNA repair.


Asunto(s)
Mapeo Cromosómico , Clonación Molecular , Mutación , Acetiltransferasa B N-Terminal/genética , Proteínas de Saccharomyces cerevisiae/genética , Acetilación , Bleomicina/efectos adversos , Camptotecina/efectos adversos , Daño del ADN , Reparación del ADN , ADN de Hongos/genética , Hidroxiurea/efectos adversos , Metilmetanosulfonato/efectos adversos , Acetiltransferasa B N-Terminal/metabolismo , Fenotipo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Análisis de Secuencia de ADN , Rayos X/efectos adversos
6.
Cell Biol Int ; 36(10): 873-81, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22657828

RESUMEN

The HRDC (helicase and RNase D C-terminal) domain at the C-terminal of WRNp (Werner protein) (1150-1229 amino acids) and BLMp (Bloom protein) (1212-1292 amino acids) recognize laser microirradiation-induced DNA dsbs (double-strand breaks). However, their role in the recognition of DNA damage other than dsbs has not been reported. In this work, we show that HRDC domain of both the proteins can be recruited to the DNA damage induced by MMS (methyl methanesulfonate) and MMC (methyl mitomycin C). GFP (green fluorescent protein)-tagged HRDC domain produces distinct foci-like respective wild-types after DNA damage induced by the said agents and co-localize with γ-H2AX. However, in time course experiment, we observed that the foci of HRDC domain exist after 24 h of removal of the damaging agents, while the foci of full-length protein disappear completely. This indicates that the repair events are not completed by the presence of protein corresponding to only the HRDC domain. Consequently, cells overexpressing the HRDC domain fail to survive after DNA damage, as determined by MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide] assay. Moreover, 24 h after removal of damaging agents, the extent of DNA damage is greater in cells overexpressing HRDC domain compared with corresponding wild-types, as observed by comet assay. Thus, our observations suggest that HRDC domain of both WRN and BLM can also recognize different types of DNA damages, but for the successful repair they fail to respond to subsequent repair events.


Asunto(s)
Antineoplásicos/efectos adversos , Daño del ADN/efectos de los fármacos , Exodesoxirribonucleasas/metabolismo , Metilmetanosulfonato/efectos adversos , Mitomicina/efectos adversos , RecQ Helicasas/metabolismo , ADN Helicasas/química , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/genética , Células HeLa , Humanos , Estructura Terciaria de Proteína , RecQ Helicasas/química , RecQ Helicasas/genética , Ribonucleasa III/química , Transfección , Helicasa del Síndrome de Werner
7.
Am J Orthod Dentofacial Orthop ; 139(4): 504-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21457861

RESUMEN

INTRODUCTION: The purpose of this study was to evaluate whether corrosion eluates obtained from commercially available orthodontic brackets are able to induce genetic damage in vitro. MATERIAL AND METHODS: Genotoxicity was assessed by the single cell gel (comet) assay using Chinese hamster ovary (CHO) cells. The following orthodontic metallic brackets were used: Morelli (Sorocaba, Brazil); Abzil (São José do Rio Preto, Brazil); Dentaurum (Pforzheim, Germany); and 3M Unitek (Puchheim, Germany). Each dental bracket was submitted to a corrosion process in a solution containing equal amounts of acetic acid and sodium chloride at 0.1 M concentration for 1, 3, 7, 14, 21, 35, and 70 days. CHO cells were exposed to eluates for 30 minutes at 37°C. The negative control was treated with the same solution used for corrosion process for 30 minutes at 37°C. Independent positive control was performed with methyl methanesulfonate (MMS) (Sigma Aldrich, St. Louis, Mo) at 1 ug/mL for 1 hour. RESULTS: None of the eluates was found to exhibit genotoxicity, regardless of the different commercial brands of orthodontic appliance used. CONCLUSIONS: In summary, our results indicate corrosion eluates obtained from orthodontic brackets do not induce genetic damage as assessed by single cell gel (comet) assay.


Asunto(s)
Células CHO/efectos de los fármacos , Aleaciones Dentales/química , Mutágenos/química , Soportes Ortodóncicos , Ácido Acético/química , Animales , Supervivencia Celular/efectos de los fármacos , Ensayo Cometa , Corrosión , Cricetinae , Cricetulus , Daño del ADN , Aleaciones Dentales/farmacología , Relación Dosis-Respuesta a Droga , Ensayo de Materiales , Metilmetanosulfonato/efectos adversos , Mutágenos/efectos adversos , Mutágenos/farmacología , Cloruro de Sodio/química , Temperatura , Factores de Tiempo
8.
J Med Food ; 12(2): 340-4, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19459735

RESUMEN

Selenomethionine (SeMet) has been identified as a chemopreventive antioxidant to activate p53-mediated nucleotide excision repair. In this study, we examined whether p53-mediated base excision repair (BER) might be induced by SeMet. When methyl methanesulfonate, a BER-inducing agent, was treated in the cells, DNA damage was rapidly decreased in the presence of SeMet. In addition, our data showed that the removal of apurinic/apyrimidinic sites was significantly enhanced in the presence of SeMet. Furthermore, we observed that the expression of gadd45a, known to involve BER as one of the p53 downstream genes, was increased by SeMet in p53 wild-type RKO cells. Those results supported the proposal that BER activity might be dependent on wild-type p53 under the modulation of gadd45a expression in response to SeMet. We suggested that p53-dependent BER activity as a distinct mechanism of SeMet might play an important role to prevent cancer caused by various oxidative stresses.


Asunto(s)
Antioxidantes/farmacología , Proteínas de Ciclo Celular/metabolismo , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Proteínas Nucleares/metabolismo , Selenometionina/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Daño del ADN/genética , Reparación del ADN/genética , Expresión Génica , Humanos , Metilmetanosulfonato/efectos adversos , Mutación , Proteínas Nucleares/genética , ARN Interferente Pequeño , Transfección , Proteína p53 Supresora de Tumor/genética
9.
DNA Repair (Amst) ; 4(2): 191-201, 2005 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-15590327

RESUMEN

SGS1 encodes a protein having DNA helicase activity, and a mutant allele of SGS1 was identified as a suppressor of the slow growth phenotype of top3 mutants. In this study, we examined whether Sgs1 prevents formation of DNA double strand breaks (DSBs) or is involved in DSB repair following exposure to methyl methanesulfonate (MMS). An analysis by pulsed-field gel electrophoresis and epistasis analyses indicated that Sgs1 is required for DSB repair that involves Rad52. In addition, analyses on the relationship between Sgs1 and proteins involved in DSB repair suggested that Sgs1 and Mre11 function via independent pathways both of which require Rad52. In sgs1 mutants, interchromosomal heteroallelic recombination and sister chromatid recombination (SCR) were not induced upon exposure to MMS, though both were induced in wild type cells, indicating the involvement of Sgs1 in heteroallelic recombination and SCR. Surprisingly, the ability of Sgs1 to bind to DNA topoisomerase III (Top3) was absolutely required for the induction of heteroallelic recombination and SCR and suppression of MMS sensitivity but its helicase activity was not, suggesting that Top3 plays a more important role in both recombinations than the DNA helicase activity of Sgs1.


Asunto(s)
Daño del ADN , ADN Helicasas/metabolismo , Reparación del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/metabolismo , Recombinación Genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Alelos , Antígenos Nucleares/metabolismo , Reparación del ADN/genética , ADN de Hongos/fisiología , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/metabolismo , Autoantígeno Ku , Metilmetanosulfonato/efectos adversos , Proteína Recombinante y Reparadora de ADN Rad52 , RecQ Helicasas , Saccharomyces cerevisiae/enzimología , Transducción de Señal , Intercambio de Cromátides Hermanas
10.
Indian J Exp Biol ; 42(4): 437-8, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15088697

RESUMEN

Allicin, one of the sulphur compounds of garlic (Allium sativum), possesses antioxidant and thiol disulphide exchange activity and is also shown to cause a variety of activities potentially useful for human health. In this investigation, the effect of 1,5,10 and 20 microM of allicin was determined for inhibiting the rate of SCE induced by 60 microM of MMS. Cultured human lymphocytes from two female donors were used for the experiment. The levels of SCEs were lowered by allicin suggesting its antigenotoxic activity in mammalian cells in vitro.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Antioxidantes/farmacología , Metilmetanosulfonato/efectos adversos , Mutágenos/efectos adversos , Intercambio de Cromátides Hermanas , Ácidos Sulfínicos/farmacología , Células Cultivadas , Disulfuros , Femenino , Humanos , Linfocitos/efectos de los fármacos
12.
Ecotoxicol Environ Saf ; 54(3): 323-9, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12651188

RESUMEN

Gill cells and hemocytes from the blue mussel Mytilus edulis were examined for DNA damage using the comet assay after laboratory exposure in vitro and in vivo to methyl methansulfonate (MMS). Hydrogen peroxide and UV radiation were used as positive control. Comet assay was also carried out on hemocytes from blue mussels sampled at polluted and unpolluted coastal areas. After 60 min in vitro exposure of gill cells to MMS, the highest response, a tail moment of 6.70+/-4.25, was obtained at 1.0mg/L. At higher doses the response decreased. After 2 days in vivo exposure a dose response was seen at concentrations between 1.0 and 33.0mg/L MMS for both gill cells and hemocytes. However, after 4 days in vivo exposure using the same concentrations of MMS, a maximum effect was seen at a 10 times lower concentration of 3.3mg/L. At the higher doses, the effect decreased. Hemocytes from blue mussels sampled at four polluted sites in Køge Bay had a great variation in tail moments with the highest value of 5.38+/-4.39. The average of all samples from Køge Bay had tail moments of 2.75+/-1.00(n=19), which was significantly higher (P<0.05) than the average, 1.72+/-1.16(n=10), of samples from unpolluted coastal waters.


Asunto(s)
Bivalvos/genética , Bivalvos/fisiología , Daño del ADN , Branquias/citología , Branquias/efectos de los fármacos , Hemocitos/efectos de los fármacos , Metilmetanosulfonato/efectos adversos , Mutágenos/efectos adversos , Animales , Técnicas de Cultivo de Célula , Ensayo Cometa , Relación Dosis-Respuesta a Droga
13.
Toxicol Lett ; 135(1-2): 103-110, 2002 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-12243869

RESUMEN

Different preparations of chamomile (Matricaria chamomilla) are used to treat various diseases, including inflammation and cancer; however, no studies on the plant's antigenotoxic capacity have been made. The aim of the present work was to determine the inhibitory effect of the chamomile essential oil (CO), on the sister chromatid exchanges (SCEs) produced by daunorubicin and methyl methanesulfonate (MMS) in mouse bone marrow cells. CO was analyzed and was found to contain 13 compounds, mainly bisabolol and its oxides, chamazulene, farnesene, germacrene and other sesquiterpenes. Initially, a toxic and a genotoxic analysis of CO were made; both showed negative results. To determine whether CO can inhibit the mutagenic effects induced by daunorubicin, one group of mice was administered corn oil, another group was treated with the mutagen (10 mg/kg), a third group was treated with 500 mg/kg of CO; three other groups were treated first with CO (5, 50 and 500 mg/kg) and then with 10 mg/kg of daunorubicin. In the case of MMS, the experimental groups consisted of the following: the negative control group which was administered corn oil, a group treated with 25 mg/kg of MMS, a group treated with 1000 mg/kg of CO, and three groups treated first with CO (250, 500 and 1000 mg/kg) and then with MMS (25 mg/kg). The results indicated a dose-dependent inhibitory effect on the SCEs formed by both mutagens. In the case of daunorubicin, a statistically significant result was observed in the three tested doses: from the lowest to the highest dose, the inhibitory values corresponded to 25.7, 63.1 and 75.5%. No alterations were found with respect to the cellular proliferation kinetics, but a reduction in the mitotic index was detected. As regards MMS, the inhibitory values were 24.8, 45.8 and 60.6%; no alterations were found in either the cellular proliferation kinetics or in the mitotic indices. Our results suggest that CO may be an effective antimutagen that could be considered for further study.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Antineoplásicos Alquilantes/efectos adversos , Células de la Médula Ósea/efectos de los fármacos , Manzanilla , Daunorrubicina/efectos adversos , Metilmetanosulfonato/efectos adversos , Intercambio de Cromátides Hermanas/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/antagonistas & inhibidores , Antineoplásicos Alquilantes/antagonistas & inhibidores , Células de la Médula Ósea/fisiología , División Celular/efectos de los fármacos , Daunorrubicina/antagonistas & inhibidores , Dosificación Letal Mediana , Masculino , Metilmetanosulfonato/antagonistas & inhibidores , Ratones , Índice Mitótico
14.
Mutat Res ; 429(1): 13-26, 1999 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-10434021

RESUMEN

Cell proliferation has been recognized as an important factor in human and experimental carcinogenesis. Point mutations as well as larger chromosomal rearrangements are involved in the initiation of cancer. In this paper we compared the relative potencies of radiation and chemical carcinogens for inducing point mutations vs. deletions in cell cycle arrested with dividing cells of Saccharomyces cerevisiae. Point mutation substrates and deletion (DEL) recombination substrates were constructed with the genes CDC28 and TUB2 that are required for cell cycle progression through G1 and G2, respectively. The carcinogens ionizing radiation, UV, MMS, EMS and 4-NQO induced point mutations in G1 and in G2 arrested as well as in dividing cells. UV, MMS, EMS and 4-NQO caused very weak if any increases in DEL recombination in G1 or G2 arrested cells, but large increases in dividing cells. When cells treated with carcinogen either in G1 or G2 were allowed to progress through the cell cycle, a time-dependent increase in DEL recombination was seen. Ionizing radiation and the site-specific endonuclease I-SceI, which both directly create double-strand breaks, induced DEL recombination in G1 as well as in G2 arrested cells. In conclusion, UV-, MMS-, EMS- and 4-NQO-induced DNA damage was converted during DNA replication to a lesion capable of inducing DEL recombination which is probably a DNA strand break. Thus, cell proliferation is not necessary to turn DNA alkylation or UV damage into a mutagenic lesion but to convert the damage into a lesion that induces DNA deletions. These results are discussed with respect to mechanisms of carcinogenesis.


Asunto(s)
División Celular/genética , Deleción Cromosómica , Recombinación Genética/genética , Saccharomyces cerevisiae/genética , 4-Nitroquinolina-1-Óxido/efectos adversos , 4-Nitroquinolina-1-Óxido/análogos & derivados , Proteína Quinasa CDC28 de Saccharomyces cerevisiae/efectos de los fármacos , Proteína Quinasa CDC28 de Saccharomyces cerevisiae/genética , Proteína Quinasa CDC28 de Saccharomyces cerevisiae/efectos de la radiación , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , ADN de Hongos/efectos de los fármacos , ADN de Hongos/efectos de la radiación , Metanosulfonato de Etilo/efectos adversos , Proteínas Fúngicas/efectos de los fármacos , Proteínas Fúngicas/genética , Proteínas Fúngicas/efectos de la radiación , Fase G1/efectos de los fármacos , Fase G1/genética , Fase G1/efectos de la radiación , Fase G2/efectos de los fármacos , Fase G2/genética , Fase G2/efectos de la radiación , Rayos gamma , Metilmetanosulfonato/efectos adversos , Mutagénesis , Pruebas de Mutagenicidad , Mutágenos/efectos adversos , Mutación Puntual/efectos de los fármacos , Mutación Puntual/efectos de la radiación , Quinolonas/efectos adversos , Recombinación Genética/efectos de los fármacos , Recombinación Genética/efectos de la radiación , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/efectos de la radiación , Rayos Ultravioleta
15.
Curr Genet ; 34(5): 368-74, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9871118

RESUMEN

The Saccharomyces cerevisiae ngs1-1 mutant was previously identified by its enhanced sensitivity to simple DNA-alkylating agents such as methyl methanesulfonate but not to UV. Molecular cloning and sequencing of NGS1 as a putative DNA-alkylation repair gene revealed that it isidentical to MRE11, a gene that is involved in DNA recombinational repair. In order to investigate functional domains of the Mre11 protein, nucleotide-sequence alterations of a number of mre11 mutant alleles, including ngs1-1, mre11-1 (ts), mre11-2, mre11-3 and mre11-58, were determined. Most of these mutations map to the N-terminus ofMre11, emphasizing the importance of this highly conserved domain. The ngs1-1 and mre11-3 mutants carry nonsense mutations resulting in truncated proteins. Missense mutations were found in mre11-1 (ts), mre11-2 and mre11-58, of which mre11-2 and mre11-58 mapped to the conserved phosphoesterase domains, indicating the involvement of these motifs in the formation and/or processing of DNA double-strand breaks. Finally, mitotic-recombination assays show that the mre11 delta mutation enhances inter-chromosomal recombination but decreases the intra-chromosomal deletion frequency. In addition, MRE11 appears to play different roles during spontaneous and alkylation-induced homologous mitotic recombination.


Asunto(s)
Endodesoxirribonucleasas , Exodesoxirribonucleasas , Proteínas Fúngicas/genética , Genes Fúngicos/genética , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Antineoplásicos Alquilantes/efectos adversos , Cromosomas Fúngicos/efectos de los fármacos , Cromosomas Fúngicos/genética , Clonación Molecular , ADN/genética , ADN/metabolismo , Reparación del ADN/genética , Metilmetanosulfonato/efectos adversos , Mutación , Recombinación Genética/efectos de los fármacos , Secuencias Repetitivas de Ácidos Nucleicos
16.
Cancer Lett ; 92(1): 87-90, 1995 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-7757964

RESUMEN

Effects of [4-chloro-6-(2,3-xylidino)-2-pyrimidinylthio] acetic acid (Wy-14643) or di(2-ethylhexyl) phthalate (DEHP) as peroxisome proliferators on the susceptibility of liver DNA to damage by N-nitrosodimethylamine (DMN) or methyl methane sulfonate (MMS) were investigated. Male F344 rats were administered Wy-14643 or DEHP, orally, for 1 or 10 weeks. At 1 week, the susceptibility to hepatic DNA damage by DMN or MMS was significantly increased in the Wy-14643- or DEHP-treated rats. The enhancement of the susceptibility persisted for up to 10 weeks in both peroxisome proliferator-treated groups. These findings suggest that the high susceptibility to DNA damage caused by peroxisome proliferators would amplify the DNA damage action such as spontaneous damage, leading to an increase in the risk of initiation in the hepatocarcinogenesis.


Asunto(s)
Carcinógenos/farmacología , Daño del ADN , Dietilhexil Ftalato/farmacología , Hígado/efectos de los fármacos , Microcuerpos/efectos de los fármacos , Pirimidinas/farmacología , Animales , Dimetilnitrosamina/efectos adversos , Hígado/ultraestructura , Masculino , Metilmetanosulfonato/efectos adversos , Ratas , Ratas Endogámicas F344
17.
Mutat Res ; 281(1): 17-23, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1371586

RESUMEN

The sensitivity and reliability of UDS and FADU in detecting mutagenic effects were compared by measuring DNA damage and repair in PBL treated in vitro with UV light, MMS and BPDE. The results indicate that FADU is more sensitive than UDS, as it is able to detect DNA damage at doses 3-4-fold lower. We also determined the DNA damage and repair induced by the above agents on lymphocyte samples from different donors by FADU and UDS, confirming that the DNA repair process in humans is characterized by interindividually variable efficiency.


Asunto(s)
Daño del ADN , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Replicación del ADN/efectos de los fármacos , Replicación del ADN/efectos de la radiación , ADN/efectos de los fármacos , ADN/efectos de la radiación , Mutágenos/efectos adversos , Benzopirenos/toxicidad , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas In Vitro , Metilmetanosulfonato/efectos adversos , Factores de Tiempo , Rayos Ultravioleta/efectos adversos
19.
Br J Cancer ; 57(5): 512-3, 1988 May.
Artículo en Inglés | MEDLINE | ID: mdl-3395557

RESUMEN

Methylene dimethane sulphonate (MDMS), the first member of the homologous series of dimethane sulphonic acid esters, was administered to 19 patients with advanced epithelial ovarian cancer. All patients had received prior chemotherapy and in addition 3 had received prior radiotherapy. MDMS was given as an i.v. bolus injection at a dose of 125mg m-2 and repeated in a q35 day schedule. Ten patients received only one course, six two courses, two three courses and one four courses. The major toxicity was thrombocytopenia which was cumulative. Serious neutropenia did not occur and no infective episodes requiring i.v. antibiotics were seen. Seven patients experience hair loss and four nausea and vomiting. Sixteen patients were evaluable for response but no objective remissions were seen although three patients had stable disease lasting at least 8 weeks. MDMS is therefore not recommended for further trial in epithelial ovarian carcinoma.


Asunto(s)
Alquilantes/uso terapéutico , Metilmetanosulfonato/análogos & derivados , Neoplasias Ováricas/tratamiento farmacológico , Adulto , Anciano , Alquilantes/efectos adversos , Evaluación de Medicamentos , Femenino , Humanos , Metilmetanosulfonato/efectos adversos , Metilmetanosulfonato/uso terapéutico , Persona de Mediana Edad , Trombocitopenia/inducido químicamente
20.
Cancer Treat Rep ; 71(9): 817-20, 1987 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-3621213

RESUMEN

Methylene dimethane sulfonate is the first member of the homologous series of straight-chain diol sulfonates. It was selected for phase I testing because of high activity in the rat Yoshida sarcoma system and a possible novel site of alkylation due to its small molecular size. Methylene dimethane sulfonate was administered as a rapid iv bolus infusion to 39 patients at doses ranging from 14 to 225 mg/m2. Nausea and vomiting were not severe but total alopecia occurred in the majority of patients at doses greater than 100 mg/m2. The dose-limiting toxic effect was thrombocytopenia, which was cumulative with lower, more prolonged nadirs following successive courses. The median time to platelet count nadir was 21 days, with recovery by 35 days. A minor response was seen in one patient with an adenocarcinoma of the lung. The recommended dose for phase II studies is 125 mg/m2 for patients who have received prior chemotherapy and 150 mg/m2 for those who have not, in an every 35-day schedule.


Asunto(s)
Alquilantes/uso terapéutico , Metilmetanosulfonato/análogos & derivados , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Alquilantes/efectos adversos , Evaluación de Medicamentos , Femenino , Humanos , Masculino , Metilmetanosulfonato/efectos adversos , Metilmetanosulfonato/uso terapéutico , Persona de Mediana Edad , Trombocitopenia/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA