Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Toxicol Appl Pharmacol ; 270(1): 23-30, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23566953

RESUMO

Methylmercury (MeHg) is a persistent environmental contaminant with potent neurotoxic action for which the underlying molecular mechanisms remain to be conclusively delineated. Our objectives herein were twofold: first, to corroborate our previous findings of an increased sensitivity of spontaneously-immortalized oxoguanine glycosylase 1-null (Ogg1(-/-)) murine embryonic fibroblasts (MEFs) to MeHg through generation of Simian virus 40 (SV40) large T antigen-immortalized wild-type and Ogg1(-/-) MEFs; and second, to determine whether MeHg toxicity is proliferation-dependent. As with the spontaneously-immortalized cells used previously, the SV40 large T antigen-immortalized cells exhibited similar tendencies to undergo MeHg-initiated cell cycle arrest, with increased sensitivity in the Ogg1(-/-) MEFs as measured by clonogenic survival and DNA damage. Compared to exponentially growing cells, those seeded at a higher density exhibited compromised proliferation, which proved protective against MeHg-mediated cell cycle arrest and induction of DNA double strand breaks (DSBs), measured by phosphorylation of the core histone H2A variant (H2AX) on serine 139 (γH2AX), and by its functional confirmation by micronucleus assessment. This enhanced sensitivity of Ogg1(-/-) MEFs to MeHg toxicity using discrete SV40 immortalization corroborates our previous studies, and suggests a novel role for OGG1 in minimizing MeHg-initiated DNA lesions that trigger replication-associated DSBs. Furthermore, proliferative capacity may determine MeHg toxicity in vivo and in utero. Accordingly, variations in cellular proliferative capacity and interindividual variability in repair activity may modulate the risk of toxicological consequences following MeHg exposure.


Assuntos
Proliferação de Células/efeitos dos fármacos , DNA Glicosilases/deficiência , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Compostos de Metilmercúrio/toxicidade , Animais , Linhagem Celular Transformada , Células Cultivadas , Relação Dose-Resposta a Droga , Camundongos , Camundongos Knockout
2.
Toxicol Appl Pharmacol ; 271(1): 41-8, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23607987

RESUMO

Exposure to methylmercury (MeHg) acutely at high levels, or via chronic low-level dietary exposure from daily fish consumption, can lead to adverse neurological effects in both the adult and developing conceptus. To determine the impact of variable DNA repair capacity, and the role of reactive oxygen species (ROS) and oxidatively damaged DNA in the mechanism of toxicity, transgenic human embryonic kidney (HEK) 293 cells that stably express either human oxoguanine glycosylase 1 (hOgg1) or its bacterial homolog, formamidopyrimidine glycosylase (Fpg), which primarily repair the oxidative lesion 8-oxo-2'-deoxyguanosine (8-oxodG), were used to assess the in vitro effects of MeHg. Western blotting confirmed the expression of hOgg1 or Fpg in both the nuclear and mitochondrial compartments of their respective cell lines. Following acute (1-2h) incubations with 0-10µM MeHg, concentration-dependent decreases in clonogenic survival and cell growth accompanied concentration-dependent increases in lactate dehydrogenase (LDH) release, ROS formation, 8-oxodG levels and apurinic/apyrimidinic (AP) sites, consistent with the onset of cytotoxicity. Paradoxically, hOgg1- and Fpg-expressing HEK 293 cells were more sensitive than wild-type cells stably transfected with the empty vector control to MeHg across all cellular and biochemical parameters, exhibiting reduced clonogenic survival and cell growth, and increased LDH release and DNA damage. Accordingly, upregulation of specific components of the base excision repair (BER) pathway may prove deleterious potentially due to the absence of compensatory enhancement of downstream processes to repair toxic intermediary abasic sites. Thus, interindividual variability in DNA repair activity may constitute an important risk factor for environmentally-initiated, oxidatively damaged DNA and its pathological consequences.


Assuntos
Dano ao DNA/efeitos dos fármacos , DNA Glicosilases/genética , DNA-Formamidopirimidina Glicosilase/genética , Compostos de Metilmercúrio/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Células HEK293 , Humanos , L-Lactato Desidrogenase/metabolismo , Compostos de Metilmercúrio/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Fatores de Risco , Regulação para Cima
3.
Mol Carcinog ; 50(3): 163-72, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21104990

RESUMO

Genotoxicity tests indicate methanol (MeOH) is not mutagenic, but a rodent study has suggested carcinogenic potential, which could result from free radical-initiated oxidative DNA damage. To investigate this possibility we treated male CD-1 mice, New Zealand white rabbits, and cynomolgus monkeys with MeOH (2.0 g/kg ip) and assessed tissue oxidative DNA damage 6 h post-dose, measured as 8-hydroxy-2'-deoxyguanosine (8-oxodG). We found no MeOH-dependent increases in 8-oxodG in bone marrow or spleen of any species. Chronic treatment of CD-1 mice with MeOH (2.0 g/kg ip) daily for 15 d also did not increase 8-oxodG levels in these organs. Further studies in the DNA repair deficient oxoguanine glycosylase 1 (Ogg1) knockout (KO) mice supported these findings. Fibroblasts from Ogg1 KO mice accumulated 8-oxodG following acute exposure to the renal carcinogen potassium bromate (KBrO(3) ; 2.0 mM) but did not accumulate 8-oxodG following exposure to 125 mM MeOH 6 h post-treatment. Ogg1 KO mice accumulated 8-oxodG in bone marrow and spleen with age but not following exposure to MeOH. In addition, free radical-mediated hydroxynonenal-histidine protein adducts were not enhanced by MeOH in primate bone marrow or spleen, or in rabbit bone marrow or mouse spleen, although modest increases were observed in rabbit spleen and mouse bone marrow. Taken together these observations suggest that MeOH exposure does not promote the accumulation of oxidative DNA damage in bone marrow and spleen, and it is unlikely that human environmental exposure to MeOH would lead to lymphomas via this mechanism.


Assuntos
Medula Óssea/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Exposição Ambiental/efeitos adversos , Metanol/toxicidade , Estresse Oxidativo/efeitos dos fármacos , 8-Hidroxi-2'-Desoxiguanosina , Animais , Células Cultivadas , DNA Glicosilases/fisiologia , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Macaca fascicularis , Masculino , Camundongos , Oxirredução , Coelhos , Espécies Reativas de Oxigênio/metabolismo , Especificidade da Espécie , Baço/efeitos dos fármacos , Baço/metabolismo
4.
Birth Defects Res C Embryo Today ; 90(2): 103-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20544694

RESUMO

Several teratogenic agents, including ionizing radiation and xenobiotics such as phenytoin, benzo[a]pyrene, thalidomide, and methamphetamine, can initiate the formation of reactive oxygen species (ROS) that oxidatively damage cellular macromolecules including DNA. Oxidative DNA damage, and particularly the most prevalent 8-oxoguanine lesion, may adversely affect development, likely via alterations in gene transcription rather than via a mutational mechanism. Contributions from oxidative DNA damage do not exclude roles for alternative mechanisms of initiation like receptor-mediated processes or the formation of covalent xenobiotic-macromolecular adducts, damage to other macromolecular targets like proteins and lipids, and other effects of ROS like altered signal transduction. Even in the absence of teratogen exposure, endogenous developmental oxidative stress can have embryopathic consequences in the absence of key pathways for detoxifying ROS or repairing DNA damage. Critical proteins in pathways for DNA damage detection/repair signaling, like p53 and ataxia telangiectasia mutated, and DNA repair itself, like oxoguanine glycosylase 1 and Cockayne syndrome B, can often, but not always, protect the embryo from ROS-initiating teratogens. Protection may be variably dependent upon such factors as the nature of the teratogen and its concentration within the embryo, the stage of development, the species, strain, gender, target tissue and cell type, among other factors.


Assuntos
Dano ao DNA , Doenças Fetais/induzido quimicamente , Animais , Benzo(a)pireno/farmacologia , DNA/metabolismo , DNA/farmacologia , Reparo do DNA , Embrião de Mamíferos , Feminino , Doenças Fetais/genética , Guanina/análogos & derivados , Inativação Metabólica , Masculino , Camundongos , Camundongos Knockout , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Fenitoína/farmacologia , Proteínas/efeitos dos fármacos , Proteínas/metabolismo , Proteínas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Teratogênicos/farmacologia , Talidomida/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/farmacologia , Xenobióticos/farmacologia
5.
Toxicol Sci ; 128(1): 272-83, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22523232

RESUMO

Methylmercury (MeHg) is a potent neurotoxin, teratogen, and probable carcinogen, but the underlying mechanisms of its actions remain unclear. Although MeHg causes several types of DNA damage, the toxicological consequences of this macromolecular damage are unknown. MeHg enhances oxidative stress, which can cause various oxidative DNA lesions that are primarily repaired by oxoguanine glycosylase 1 (OGG1). Herein, we compared the response of wild-type and OGG1 null (Ogg1(-/-)) murine embryonic fibroblasts to environmentally relevant, low micromolar concentrations of MeHg by measuring clonogenic efficiency, cell cycle arrest, DNA double-strand breaks (DSBs), and activation of the DNA damage response pathway.Ogg1(-/-) cells exhibited greater sensitivity to MeHg than wild-type controls, as measured by the clonogenic assay, and showed a greater propensity for MeHg-initiated apoptosis. Both wild-type and Ogg1(-/-) cells underwent cell cycle arrest when exposed to micromolar concentrations of MeHg; however, the extent of DSBs was exacerbated in Ogg1(-/-) cells compared with that in wild-type controls. Pretreatment with the antioxidative enzyme catalase reduced levels of DSBs in both wild-type and Ogg1(-/-) cells but failed to block MeHg-initiated apoptosis at micromolar concentrations. Our findings implicate reactive oxygen species mediated DNA damage in the mechanism of MeHg toxicity; and demonstrate for the first time that impaired DNA repair capacity enhances cellular sensitivity to MeHg. Accordingly, the genotoxic properties of MeHg may contribute to its neurotoxic and teratogenic effects, and an individual's response to oxidative stress and DNA damage may constitute an important determinant of risk.


Assuntos
Dano ao DNA , DNA Glicosilases/metabolismo , Compostos de Metilmercúrio/toxicidade , Animais , Citometria de Fluxo , Humanos , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA