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












Base de dados
Intervalo de ano de publicação
1.
Chem Res Toxicol ; 37(5): 814-823, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38652696

RESUMO

The major product of DNA-methylating agents, N7-methyl-2'-deoxyguanosine (MdG), is a persistent lesion in vivo, but it is not believed to have a large direct physiological impact. However, MdG reacts with histone proteins to form reversible DNA-protein cross-links (DPCMdG), a family of DNA lesions that can significantly threaten cell survival. In this paper, we developed a tandem mass spectrometry method for quantifying the amounts of MdG and DPCMdG in nuclear DNA by taking advantage of their chemical lability and the concurrent release of N7-methylguanine. Using this method, we determined that DPCMdG is formed in less than 1% yield based upon the levels of MdG in methyl methanesulfonate (MMS)-treated HeLa cells. Despite its low chemical yield, DPCMdG contributes to MMS cytotoxicity. Consequently, cells that lack efficient DPC repair by the DPC protease SPRTN are hypersensitive to MMS. This investigation shows that the downstream chemical and biochemical effects of initially formed DNA damage can have significant biological consequences. With respect to MdG formation, the initial DNA lesion is only the beginning.


Assuntos
DNA , Desoxiguanosina , Metanossulfonato de Metila , Humanos , Células HeLa , DNA/metabolismo , DNA/química , DNA/efeitos dos fármacos , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Desoxiguanosina/química , Metanossulfonato de Metila/química , Metanossulfonato de Metila/farmacologia , Espectrometria de Massas em Tandem , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Reagentes de Ligações Cruzadas/química , Proteínas de Ligação a DNA
2.
Chem Res Toxicol ; 33(12): 3023-3030, 2020 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-33190492

RESUMO

Allergic contact dermatitis (ACD) is a reaction of the immune system resulting from skin sensitization to an exogenous hazardous chemical and leading to the activation of antigen-specific T-lymphocytes. The adverse outcome pathway (AOP) for skin sensitization identified four key events (KEs) associated with the mechanisms of this pathology, the first one being the ability of skin chemical sensitizers to modify epidermal proteins to form antigenic structures that will further trigger the immune system. So far, these interactions have been studied in solution using model nucleophiles such as amino acids or peptides. As a part of our efforts to better understand chemistry taking place during the sensitization process, we have developed a method based on the use of high-resolution magic angle spinning (HRMAS) NMR to monitor in situ the reactions of 13C substituted chemical sensitizers with nucleophilic amino acids of epidermal proteins in reconstructed human epidermis. A quantitative approach, developed so far for liquid NMR applications, has not been developed to our knowledge in a context of a semisolid nonanisotropic environment like the epidermis. We now report a quantitative chemical reactivity mapping of methyl methanesulfonate (MMS), a sensitizing methylating agent, in reconstructed human epidermis by quantitative HRMAS (qHRMAS) NMR. First, the haptenation process appeared to be much faster in RHE than in solution with a maximum concentration of adducts reached between 4 and 8 h. Second, it was observed that the concentration of cysteine adducts did not significantly increase with the dose (2.07 nmol/mg at 0.4 M and 2.14 nmol/mg at 1 M) nor with the incubation time (maximum of 2.27 nmol/mg at 4 h) compared to other nucleophiles, indicating a fast reaction and a potential saturation of targets. Third, when increasing the exposure dose, we observed an increase of adducts up to 12.5 nmol/mg of RHE, excluding cysteine adducts, for 3112 µg/cm2 (1 M solution) of (13C)MMS. This methodology applied to other skin sensitizers could allow for better understanding of the potential links between the amount of chemical modifications formed in the epidermis in relation to exposure and the sensitization potency.


Assuntos
Epiderme/efeitos dos fármacos , Metanossulfonato de Metila/farmacologia , Alquilação , Células Cultivadas , Dermatite Alérgica de Contato/metabolismo , Epiderme/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Metanossulfonato de Metila/síntese química , Metanossulfonato de Metila/química , Estrutura Molecular
3.
Chem Res Toxicol ; 32(10): 2144-2151, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31532638

RESUMO

N7-Methyl-2'-deoxyguanosine (MdG) is the major damage product in DNA produced by methylating agents, but it often thought to be nontoxic and nonmutagenic. MdG is chemically unstable. An abasic site (AP) is the major product produced from MdG under physiologically relevant conditions. AP formation is frequently considered to be responsible for the cytotoxic effects of MdG, but the reaction is suppressed in nucleosome core particles (NCPs). Recently, it was discovered that histone proteins form reversible DNA-protein cross-links (DPCs) with MdG in reconstituted NCPs, as well as in methylmethanesulfonate (MMS) treated cells. In this study, the formation and reactivity of MdG in MMS treated NCPs was examined at single nucleotide resolution. Sequences consisting of three or more consecutive dGs are more reactive with MMS. The efficiency and selectivity of MdG formation by MMS is largely unaffected within a NCP, although reactivity at several dGs is ∼1.5-2.5-fold higher in NCPs. DPC formation from MdG (DPCMdG) predominates over AP at all positions within the NCP. With few exceptions, DPCMdG yield is strongly dependent upon the accessibility of the major groove containing MdG to lysine-rich histone N-terminal tails. These data indicate that histone-MdG DPC formation will depend upon DNA sequence and translational position within an NCP.


Assuntos
Proteínas de Ligação a DNA/química , DNA/química , Metanossulfonato de Metila/química , Nucleossomos/química , DNA/efeitos dos fármacos , Desoxiguanosina/química , Metanossulfonato de Metila/farmacologia , Metanossulfonato de Metila/toxicidade , Modelos Moleculares , Estrutura Molecular , Tamanho da Partícula , Propriedades de Superfície
4.
Biochim Biophys Acta Proteins Proteom ; 1867(11): 140259, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31376523

RESUMO

S-Methyl methanethiosulfonate (MMTS) is used in experimental biochemistry for alkylating thiol groups of protein cysteines. Its applications include mainly trapping of natural thiol-disulfide states of redox-sensitive proteins and proteins which have undergone S-nitrosylation. The reagent can also be employed as an inhibitor of enzymatic activity, since nucleophilic cysteine thiolates are commonly present at active sites of various enzymes. The advantage of using MMTS for this purpose is the reversibility of the formation of methylthio mixed disulfides, compared to irreversible alkylation using conventional agents. Additional benefits include good accessibility of MMTS to buried protein cysteines due to its small size and the simplicity of the protection and deprotection procedures. In this study we report examples of MMTS application in experiments involving oxidoreductase (glyceraldehyde-3-phosphate dehydrogenase, GAPDH), redox-regulated protein (recoverin) and cysteine protease (triticain-α). We demonstrate that on the one hand MMTS can modify functional cysteines in the thiol enzyme GAPDH, thereby preventing thiol oxidation and reversibly inhibiting the enzyme, while on the other hand it can protect the redox-sensitive thiol group of recoverin from oxidation and such modification produces no impact on the activity of the protein. Furthermore, using the example of the papain-like enzyme triticain-α, we report a novel application of MMTS as a protector of the primary structure of active cysteine protease during long-term purification and refolding procedures. Based on the data, we propose new lines of MMTS employment in research, pharmaceuticals and biotechnology for reversible switching off of undesirable activity and antioxidant protection of proteins with functional thiol groups.


Assuntos
Cisteína Proteases/química , Gliceraldeído-3-Fosfato Desidrogenases/química , Metanossulfonato de Metila/análogos & derivados , Proteínas de Plantas/química , Recoverina/química , Triticum/enzimologia , Animais , Humanos , Metanossulfonato de Metila/química , Oxirredução , Coelhos , Compostos de Sulfidrila/química
5.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 6): 419-427, 2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-31204688

RESUMO

Chagas disease, which is caused by Trypanosoma cruzi, affects more than six million people worldwide. Cruzain is the major cysteine protease involved in the survival of this parasite. Here, the expression, purification and crystallization of this enzyme are reported. The cruzain crystals diffracted to 1.2 Šresolution, yielding two novel cruzain structures: apocruzain and cruzain bound to the reversible covalent inhibitor S-methyl thiomethanesulfonate. Mass-spectrometric experiments confirmed the presence of a methylthiol group attached to the catalytic cysteine. Comparison of these structures with previously published structures indicates the rigidity of the cruzain structure. These results provide further structural information about the enzyme and may help in new in silico studies to identify or optimize novel prototypes of cruzain inhibitors.


Assuntos
Apoproteínas/química , Apoproteínas/metabolismo , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Desenho de Fármacos , Metanossulfonato de Metila/análogos & derivados , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Cristalografia por Raios X , Inibidores de Cisteína Proteinase/química , Metanossulfonato de Metila/química , Metanossulfonato de Metila/metabolismo , Modelos Moleculares , Conformação Proteica
6.
Anal Chem ; 91(8): 4987-4994, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30896925

RESUMO

Emerging evidence suggests that cross-links formed by reacting DNA lesions with proteins may play a significant role in the pathophysiology of human cancer and degenerative diseases. The goal of this study was to develop a method involving liquid chromatography-tandem mass spectrometry (LC-MS/MS) coupled with the stable isotope-dilution method to quantify DNA-protein cross-link (DPC). A novel type of cross-link involving a S-glycosidic linkage formed by reacting an abasic site in DNA with the cysteine residues in protein was targeted in this study. The method entails hydrolysis of the cross-link to a 2'-deoxyribose-cysteine adduct, addition of isotopically labeled internal standard, and quantitation by LC-MS/MS analysis. The accuracy and precision of the method were evaluated with a synthetic peptide containing the cross-link. The validated method was then applied to quantitate the levels of the DNA-protein cross-link in vitro and in HeLa cells exposed to alkylating agent methylmethanesulfonate (MMS). The analysis detected dosage-dependent formation of the cross-link in both purified DNA (6.0 ± 0.6 DPC per 106 nt µM-1 MMS) and in human cells (7.8 ± 1.2 DPC per 106 nt mM-1 MMS). With the abasic site being one of the most common DNA lesions produced continuously by multiple pathways, the results provide significant new knowledge for better understanding the potential biological implications of its associated DNA-protein cross-link.


Assuntos
Cromatografia Líquida , Cisteína/química , DNA/química , Espectrometria de Massas em Tandem , Sequência de Aminoácidos , DNA/metabolismo , Células HeLa , Humanos , Isótopos/química , Cinética , Metanossulfonato de Metila/química , Modelos Moleculares , Conformação de Ácido Nucleico , Ligação Proteica , Conformação Proteica
7.
Br J Pharmacol ; 176(4): 646-670, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29909607

RESUMO

BACKGROUND AND PURPOSE: Posttranslational modifications of cysteine residues represent a major aspect of redox biology, and their reliable detection is key in providing mechanistic insights. The metastable character of these modifications and cell lysis-induced artifactual oxidation render current state-of-the-art protocols to rely on alkylation-based stabilization of labile cysteine derivatives before cell/tissue rupture. An untested assumption in these procedures is that for all cysteine derivatives, alkylation rates are faster than their dynamic interchange. However, when the interconversion of cysteine derivatives is not rate limiting, electrophilic labelling is under Curtin-Hammett control; hence, the final alkylated mixture may not represent the speciation that prevailed before alkylation. EXPERIMENTAL APPROACH: Buffered aqueous solutions of inorganic, organic, cysteine, GSH and GAPDH polysulfide species were used. Additional experiments in human plasma and serum revealed that monobromobimane can extract sulfide from the endogenous sulfur pool by shifting speciation equilibria, suggesting caution should be exercised when interpreting experimental results using this tool. KEY RESULTS: In the majority of cases, the speciation of alkylated polysulfide/thiol derivatives depended on the experimental conditions. Alkylation perturbed sulfur speciation in both a concentration- and time-dependent manner and strong alkylating agents cleaved polysulfur chains. Moreover, the labelling of sulfenic acids with dimedone also affected cysteine speciation, suggesting that part of the endogenous pool of products previously believed to represent sulfenic acid species may represent polysulfides. CONCLUSIONS AND IMPLICATIONS: We highlight methodological caveats potentially arising from these pitfalls and conclude that current derivatization strategies often fail to adequately capture physiological speciation of sulfur species. LINKED ARTICLES: This article is part of a themed section on Chemical Biology of Reactive Sulfur Species. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.4/issuetoc.


Assuntos
Alquilantes/química , Compostos de Enxofre/química , Adulto , Humanos , Iodoacetamida/química , Maleimidas/química , Metanossulfonato de Metila/análogos & derivados , Metanossulfonato de Metila/química , Compostos de Enxofre/análise , Compostos de Enxofre/sangue
8.
Methods ; 156: 91-101, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30395967

RESUMO

Ribonucleic acids (RNA) are extensively modified. These modifications are quantified by mass spectrometry (LC-MS/MS) to determine the abundance of a modification under certain conditions or in various genetic backgrounds. With LC-MS/MS the steady state of modifications is determined, and thus we only have a static view of the dynamics of RNA modifications. With nucleic acid isotope labeling coupled mass spectrometry (NAIL-MS) we overcome this limitation and get access to the dynamics of RNA modifications. We describe labeling techniques for E. coli, S. cerevisiae and human cell culture and the current instrumental limitations. We present the power of NAIL-MS but we also outline validation experiments, which are necessary for correct data interpretation. As an example, we apply NAIL-MS to study the demethylation of adenine and cytidine, which are methylated by the damaging agent methyl-methanesulfonate in E. coli. With NAIL-MS we exclude the concurrent processes for removal of RNA methylation, namely RNA degradation, turnover and dilution. We use our tool to study the speed and efficiency of 1-methyladenosine and 3-methylcytidine demethylation. We further outline current limitations of NAIL-MS but also potential future uses for e.g. relative quantification of tRNA isoacceptor abundances.


Assuntos
Adenosina/análogos & derivados , Citidina/análogos & derivados , Marcação por Isótopo/métodos , Espectrometria de Massas/métodos , Processamento Pós-Transcricional do RNA , RNA Mensageiro/química , RNA de Transferência/química , Adenosina/química , Adenosina/metabolismo , Isótopos de Carbono , Citidina/química , Citidina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Células HEK293 , Humanos , Hidrólise , Metanossulfonato de Metila/química , Isótopos de Nitrogênio , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transcriptoma
9.
Oxid Med Cell Longev ; 2018: 7820890, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29849914

RESUMO

Statins are 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, and this class of drugs has been studied as protective agents against DNA damages. Alkylating agents (AAs) are able to induce alkylation in macromolecules, causing DNA damage, as DNA methylation. Our objective was to evaluate atorvastatin (AVA) antimutagenic, cytoprotective, and antigenotoxic potentials against DNA lesions caused by AA. AVA chemopreventive ability was evaluated using antimutagenicity assays (Salmonella/microsome assay), cytotoxicity, cell cycle, and genotoxicity assays in HepG2 cells. The cells were cotreated with AVA and the AA methyl methanesulfonate (MMS) or cyclophosphamide (CPA). Our datum showed that AVA reduces the alkylation-mediated DNA damage in different in vitro experimental models. Cytoprotection of AVA at low doses (0.1-1.0 µM) was observed after 24 h of cotreatment with MMS or CPA at their LC50, causing an increase in HepG2 survival rates. After all, AVA at 10 µM and 25 µM had decreased effect in micronucleus formation in HepG2 cells and restored cell cycle alterations induced by MMS and CPA. This study supports the hypothesis that statins can be chemopreventive agents, acting as antimutagenic, antigenotoxic, and cytoprotective components, specifically against alkylating agents of DNA.


Assuntos
Atorvastatina/farmacologia , Ciclofosfamida/farmacologia , Dano ao DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Metanossulfonato de Metila/farmacologia , Alquilantes/química , Alquilantes/farmacologia , Alquilação , Atorvastatina/química , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Núcleo Celular/química , Núcleo Celular/efeitos dos fármacos , Ciclofosfamida/química , Células Hep G2 , Humanos , Metanossulfonato de Metila/química , Salmonella enterica/efeitos dos fármacos , Salmonella enterica/genética
10.
J Toxicol Environ Health A ; 80(23-24): 1290-1300, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28956726

RESUMO

The popular use of Annona muricata L. is based upon a range of medicinal purposes, and the plant exhibits biological activities including antihyperglycemic, antiparasitic, and antitumor activities. The objectives of this study were to examine the antioxidant, cytotoxic, and genotoxic potential of the hydroalcoholic extract of A. muricata leaves (AMEs), as well as its effects on genotoxicity induced by methyl methanesulfonate (MMS) and hydrogen peroxide (H2O2). The results using 2,2-diphenyl-1-picrylhydrazyl assay showed that AME was able to scavenge 44.71% of free radicals. The extract significantly reduced the viability of V79 cells in the clonogenic assay at concentrations ≥8 µg/ml. No significant differences in micronucleus (MN) frequency were observed between V79 cell cultures treated with different concentrations of the extract (0.125, 0.25, 0.5, and 1 µg/ml) and negative control. When AME concentrations were combined with MMS, data revealed no marked differences from mutagen alone. In contrast, significant reductions in the frequencies of MN were noted in cultures treated with AME combined with H2O2 compared to H2O2 alone. In vivo studies found no significant differences in the frequencies of micronucleated polychromatic erythrocytes (MNPCEs) between animals treated with different AME doses compared to control. Animals treated with AME doses of 125 and 250 mg/kg and MMS exhibited significantly higher frequencies of MNPCE compared to mutagen alone. In conclusion, under current experimental conditions, AME was not genotoxic and exerted a modulatory effect on DNA damage depending upon the experimental conditions. The extract did not influence markedly MMS-induced genotoxicity in in vitro test system. However, the extract increased DNA damage induced by mutagen in mice. In V79 cells, AME reduced the genotoxicity produced by H2O2, and this protective effect was attributed in part to the antioxidant activity of AME.


Assuntos
Annona/química , Antioxidantes/química , Dano ao DNA , Genoma de Planta/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Folhas de Planta/química , Compostos de Bifenilo/química , Peróxido de Hidrogênio/química , Metanossulfonato de Metila/química , Picratos/química , Extratos Vegetais/toxicidade
11.
Angew Chem Int Ed Engl ; 56(39): 11749-11753, 2017 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-28700817

RESUMO

A strategy to deliver a well-defined persulfide species in a biological medium is described. Under near physiological conditions, the persulfide prodrug can be activated by an esterase to generate a "hydroxymethyl persulfide" intermediate, which rapidly collapses to form a defined persulfide. Such persulfide prodrugs can be used either as chemical tools to study persulfide chemistry and biology or for future development as H2 S-based therapeutic reagents. Using the persulfide prodrugs developed in this study, the reactivity between S-methyl methanethiosulfonate (MMTS) with persulfide was unambiguously demonstrated. Furthermore, a representative prodrug exhibited potent cardioprotective effects in a murine model of myocardial ischemia-reperfusion (MI/R) injury with a bell shape therapeutic profile.


Assuntos
Esterases/metabolismo , Pró-Fármacos/farmacocinética , Sulfetos/administração & dosagem , Ativação Metabólica , Animais , Cardiotônicos/administração & dosagem , Cardiotônicos/química , Linhagem Celular , Relação Dose-Resposta a Droga , Desenvolvimento de Medicamentos , Metanossulfonato de Metila/análogos & derivados , Metanossulfonato de Metila/química , Camundongos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Pró-Fármacos/administração & dosagem , Sulfetos/química
12.
Biochim Biophys Acta ; 1858(1): 38-46, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26476106

RESUMO

We present molecular dynamics (MD) simulation studies of the interaction of a chemo preventive and protective agent, S-methyl methanethiosulfonate (MMTS), with a model bilayer of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC). We analyzed and compared its diffusion mechanisms with the related molecule dimethyl sulfoxide (DMSO). We obtained spatially resolved free energy profiles of MMTS partition into a DPPC bilayer in the liquid-crystalline phase through potential of mean force (PMF) calculations using an umbrella sampling technique. These profiles showed a minimum for MMTS close to the carbonyl region of DPPC. The location of MMTS molecules in the DPPC bilayer observed in the MD was confirmed by previous SERS studies. We decomposed PMF profiles into entropic and enthalpic contributions. These results showed that the driving force for the partitioning of MMTS into the upper region of DPPC is driven by a favorable entropy change while partitioning into the acyl chains is driven by enthalpy. On the other hand, the partition of DMSO into the membrane is not favored, and is driven by entropy instead of enthalpy. Free diffusion MD simulations using all atom and coarse grained (CG) models of DPPC in presence of MMTS were used to analyze the effect of DPPC-MMTS interaction. Density profiles showed that MMTS locates preferentially in the carbonyl region, as expected according to the PMF profile and the experimental evidence. MMTS presented two differential effects over the packing of DPPC hydrocarbonate chains at low or at high molar ratios. An ordering effect was observed when a CG MMTS model was used. Finally, free diffusion MD and PMF decomposition for DMSO were used for comparison.


Assuntos
1,2-Dipalmitoilfosfatidilcolina/análogos & derivados , Bicamadas Lipídicas/química , Metanossulfonato de Metila/análogos & derivados , 1,2-Dipalmitoilfosfatidilcolina/química , Difusão , Dimetil Sulfóxido/química , Metanossulfonato de Metila/química , Simulação de Dinâmica Molecular , Termodinâmica
13.
Proc Natl Acad Sci U S A ; 112(18): 5779-84, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25902524

RESUMO

DNA replication initiates at defined replication origins along eukaryotic chromosomes, ensuring complete genome duplication within a single S-phase. A key feature of replication origins is their ability to control the onset of DNA synthesis mediated by DNA polymerase-α and its intrinsic RNA primase activity. Here, we describe a novel origin-independent replication process that is mediated by transcription. RNA polymerase I transcription constraints lead to persistent RNA:DNA hybrids (R-loops) that prime replication in the ribosomal DNA locus. Our results suggest that eukaryotic genomes have developed tools to prevent R-loop-mediated replication events that potentially contribute to copy number variation, particularly relevant to carcinogenesis.


Assuntos
Replicação do DNA , DNA Ribossômico/química , DNA/química , Instabilidade Genômica , RNA/química , Ribonuclease H/química , Proteínas de Bactérias , Camptotecina/química , Carcinogênese/metabolismo , Ciclo Celular , Separação Celular , Cromossomos/química , Variações do Número de Cópias de DNA , Eletroforese em Gel Bidimensional , Citometria de Fluxo , Dosagem de Genes , Humanos , Hidroxiureia/química , Proteínas Luminescentes , Metanossulfonato de Metila/química , Mutação , Origem de Replicação , Saccharomyces cerevisiae/metabolismo
14.
Environ Mol Mutagen ; 56(2): 97-110, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25196723

RESUMO

Increased levels of oxidatively damaged DNA have been documented in studies of metal, metal oxide, carbon-based and ceramic engineered nanomaterials (ENMs). In particular, 8-oxo-7,8-dihydroguanine-2'-deoxyguanosine (8-oxodG) is widely assessed as a DNA nucleobase oxidation product, measured by chromatographic assays, antibody-based methods or the comet assay with DNA repair enzymes. However, spurious oxidation of DNA has been a problem in certain studies applying chromatographic assays, yielding high baseline levels of 8-oxodG. Antibody-based assays detect high 8-oxodG baseline levels, related to cross-reactivity with other molecules in cells. This review provides an overview of efforts to reliably detect oxidatively damaged DNA and a critical assessment of the published studies on DNA damage levels. Animal studies with high baseline levels of oxidatively damaged DNA are more likely to show positive associations between exposure to ENMs and oxidized DNA in tissue than studies showing acceptable baseline levels (odds ratio = 12.1, 95% confidence interval: 1.2-124). Nevertheless, reliable studies indicate that intratracheal instillation of nanosized carbon black is associated with increased levels of oxidatively damaged DNA in lung tissue. Oral exposure to nanosized carbon black, TiO2 , carbon nanotubes and ZnO is associated with elevated levels of oxidatively damaged DNA in tissues. These observations are supported by cell culture studies showing concentration-dependent associations between ENM exposure and oxidatively damaged DNA measured by the comet assay. Cell culture studies show relatively high variation in the ability of ENMs to oxidatively damage DNA; hence, it is currently impossible to group ENMs according to their DNA damaging potential.


Assuntos
Células Cultivadas/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Nanoestruturas/toxicidade , Estresse Oxidativo/efeitos dos fármacos , 8-Hidroxi-2'-Desoxiguanosina , Animais , DNA Glicosilases/biossíntese , Desoxiguanosina/análogos & derivados , Desoxiguanosina/química , Metanossulfonato de Metila/química , Nanoestruturas/química , Emissões de Veículos/toxicidade
15.
Artigo em Inglês | MEDLINE | ID: mdl-24534338

RESUMO

Organophosphate compounds, which induce organophosphate poisoning, were originally used as pesticides. But this type of product has also been used as warfare nerve agent like sarin, soman, Russian VX, or tabun. HI-6-dimethanesulfonate is a salt of the oxime HI-6 used in the treatment of nerve-agent poisoning. It is known to be the best re-activator component of inactivated acetyl cholinesterase. HI-6-dimethanesulfonate has shown a higher level of solubility with similar potency to reactivate acetyl cholinesterase and a similar pharmacokinetics profile compared with HI-6 dichloride. HI-6 dimethanesulfonate was tested for its mutagenic and genotoxic potential by use of the standard ICH S2R (1) battery for the evaluation of pharmaceuticals. HI-6-dimethanesulfonate was mutagenic in the Ames test only in the presence of metabolic activation. In the mutation assay at the Tk locus in L5178Y mouse-lymphoma cells, HI-6-dimethanesulfonate showed mutagenic activity both with and without metabolic activation, with a significant increase in small colonies. The effects were in favour of a clastogenic activity. It was concluded that the compound was mutagenic and possibly clastogenic in vitro. In contrast, the in vivo micronucleus test in rat bone-marrow did not demonstrate any genotoxic activity and the Comet assay performed in rat liver did not show any statistically or biologically significant increases in DNA strand-breaks. The results of both in vivo studies performed on two different organs with two endpoints are sufficient to conclude the absence of a genotoxic hazard in vivo and to consider that there is no genotoxic concern in humans for HI-6-dimethanesulfonate.


Assuntos
Dano ao DNA , Metanossulfonato de Metila/toxicidade , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Mutagênicos/toxicidade , Oximas/toxicidade , Compostos de Piridínio/toxicidade , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Linhagem Celular Tumoral , Ensaio Cometa , Relação Dose-Resposta a Droga , Feminino , Injeções Intraperitoneais , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Metanossulfonato de Metila/química , Camundongos , Estrutura Molecular , Mutagênicos/química , Oximas/química , Compostos de Piridínio/química , Ratos , Ratos Sprague-Dawley , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética
16.
PLoS One ; 9(2): e88633, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24533124

RESUMO

The function of the replication clamp loaders in the semi-conservative telomere replication and their relationship to telomerase- and recombination mechanisms of telomere addition remains ambiguous. We have investigated the variant clamp loader Ctf18 RFC (Replication Factor C). To understand the role of Ctf18 at the telomere, we first investigated genetic interactions after loss of Ctf18 and TLC1 (the yeast telomerase RNA). We find that the tlc1Δ ctf18Δ double mutant confers a rapid >1000-fold decrease in viability. The rate of loss was similar to the kinetics of cell death in rad52Δ tlc1Δ cells. However, the Ctf18 pathway is distinct from Rad52, required for the repair of DSBs, as demonstrated by the synthetic lethality of rad52▵ tlc1Δ ctf18Δ triple mutants. These data suggest that each mutant elicits non-redundant defects acting on the same substrate. Second, interactions of the yeast hyper-recombinational mutant, mre11A470T, with ctf18▵ confer a synergistic cold sensitivity. The phenotype of these double mutants ultimately results in telomere loss and the generation of recombinational survivors. We observed a similar synergism between single mutants that led to hypersensitivity to the DNA alkylating agent, methane methyl sulphonate (MMS), the replication fork inhibitor hydroxyurea (HU), and to a failure to separate telomeres of sister chromatids. Hence, ctf18Δ and mre11A470T act in different pathways on telomere substrates for multiple phenotypes. The mre11A470T cells also displayed a DNA damage response (DDR) at 15°C but not at 30°C while ctf18Δ mutants conferred a constitutive DDR activity. Both the 15°C DDR pattern and growth rate were reversible at 30°C and displayed telomerase activity in vivo. We hypothesize that Ctf18 confers protection against stalling and/or breaks at the replication fork in cells that either lack, or are compromised for, telomerase activity. This Ctf18-based function is likely to contribute another level to telomere size homeostasis.


Assuntos
Endodesoxirribonucleases/genética , Exodesoxirribonucleases/genética , Mutação , Proteína de Replicação C/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Telomerase/metabolismo , Telômero/ultraestrutura , Alelos , Proteínas de Ciclo Celular/metabolismo , Quinase do Ponto de Checagem 2/metabolismo , Cromátides/química , Reparo do DNA , Endodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/metabolismo , Hidroxiureia/química , Cinética , Metanossulfonato de Metila/química , Fenótipo , Saccharomyces cerevisiae/metabolismo , Temperatura
17.
FEBS J ; 280(23): 6150-61, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24103186

RESUMO

Cellular exposure to reactive oxygen species induces rapid oxidation of DNA, proteins, lipids and other biomolecules. At the proteome level, cysteine thiol oxidation is a prominent post-translational process that is implicated in normal physiology and numerous pathologies. Methods for investigating protein oxidation include direct labeling with selective chemical probes and indirect tag-switch techniques. Common to both approaches is chemical blocking of free thiols using reactive electrophiles to prevent post-lysis oxidation or other thiol-mediated cross-reactions. These reagents are used in large excess, and their reactivity with cysteine sulfenic acid, a critical oxoform in numerous proteins, has not been investigated. Here we report the reactivity of three thiol-blocking electrophiles, iodoacetamide, N-ethylmaleimide and methyl methanethiosulfonate, with protein sulfenic acid and dimedone, the structural core of many sulfenic acid probes. We demonstrate that covalent cysteine -SOR (product) species are partially or fully susceptible to reduction by dithiothreitol, tris(2-carboxyethyl)phosphine and ascorbate, regenerating protein thiols, or, in the case of ascorbate, more highly oxidized species. The implications of this reactivity on detection methods for protein sulfenic acids and S-nitrosothiols are discussed.


Assuntos
Cisteína/química , Ditiotreitol/química , Proteínas/química , Ácidos Sulfênicos/química , Compostos de Sulfidrila/química , Ácido Ascórbico/química , Ácido Ascórbico/metabolismo , Cicloexanonas/química , Cicloexanonas/metabolismo , Cisteína/metabolismo , Ditiotreitol/metabolismo , Etilmaleimida/química , Etilmaleimida/metabolismo , Iodoacetamida/química , Iodoacetamida/metabolismo , Metanossulfonato de Metila/análogos & derivados , Metanossulfonato de Metila/química , Metanossulfonato de Metila/metabolismo , Oxirredução , Proteínas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Espectrometria de Massas por Ionização por Electrospray , Ácidos Sulfênicos/metabolismo , Compostos de Sulfidrila/metabolismo
18.
BMC Med Genet ; 14: 93, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-24053728

RESUMO

BACKGROUND: We investigated a potential link between genetic polymorphisms in genes XRCC1 (Arg399Gln), OGG1 (Ser326Cys), XRCC3 (Thr241Met), and XRCC4 (Ile401Thr) with the level of DNA damage and repair, accessed by comet and micronucleus test, in 51 COPD patients and 51 controls. METHODS: Peripheral blood was used to perform the alkaline and neutral comet assay; and genetic polymorphisms by PCR/RFLP. To assess the susceptibility to exogenous DNA damage, the cells were treated with methyl methanesulphonate for 1-h or 3-h. After 3-h treatment the % residual damage was calculated assuming the value of 1-h treatment as 100%. The cytogenetic damage was evaluated by buccal micronucleus cytome assay (BMCyt). RESULTS: COPD patients with the risk allele XRCC1 (Arg399Gln) and XRCC3 (Thr241Met) showed higher DNA damage by comet assay. The residual damage was higher for COPD with risk allele in the four genes. In COPD patients was showed negative correlation between BMCyt (binucleated, nuclear bud, condensed chromatin and karyorrhexic cells) with pulmonary function and some variant genotypes. CONCLUSION: Our results suggest a possible association between variant genotypes in XRCC1 (Arg399Gln), OGG1 (Ser326Cys), XRCC3 (Thr241Met), and XRCC4 (Ile401Thr), DNA damage and progression of COPD.


Assuntos
Dano ao DNA , DNA Glicosilases/genética , Proteínas de Ligação a DNA/genética , Doença Pulmonar Obstrutiva Crônica/genética , Idoso , Alelos , Ensaio Cometa , Reparo do DNA , Genótipo , Humanos , Masculino , Metanossulfonato de Metila/química , Pessoa de Meia-Idade , Polimorfismo Genético , Doença Pulmonar Obstrutiva Crônica/patologia , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
19.
J Biol Chem ; 288(24): 17420-31, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23640880

RESUMO

General anesthetics exert many of their CNS actions by binding to and modulating membrane-embedded pentameric ligand-gated ion channels (pLGICs). The structural mechanisms underlying how anesthetics modulate pLGIC function remain largely unknown. GLIC, a prokaryotic pLGIC homologue, is inhibited by general anesthetics, suggesting anesthetics stabilize a closed channel state, but in anesthetic-bound GLIC crystal structures the channel appears open. Here, using functional GLIC channels expressed in oocytes, we examined whether propofol induces structural rearrangements in the GLIC transmembrane domain (TMD). Residues in the GLIC TMD that frame intrasubunit and intersubunit water-accessible cavities were individually mutated to cysteine. We measured and compared the rates of modification of the introduced cysteines by sulfhydryl-reactive reagents in the absence and presence of propofol. Propofol slowed the rate of modification of L240C (intersubunit) and increased the rate of modification of T254C (intrasubunit), indicating that propofol binding induces structural rearrangements in these cavities that alter the local environment near these residues. Propofol acceleration of T254C modification suggests that in the resting state propofol does not bind in the TMD intrasubunit cavity as observed in the crystal structure of GLIC with bound propofol (Nury, H., Van Renterghem, C., Weng, Y., Tran, A., Baaden, M., Dufresne, V., Changeux, J. P., Sonner, J. M., Delarue, M., and Corringer, P. J. (2011) Nature 469, 428-431). In silico docking using a GLIC closed channel homology model suggests propofol binds to intersubunit sites in the TMD in the resting state. Propofol-induced motions in the intersubunit cavity were distinct from motions associated with channel activation, indicating propofol stabilizes a novel closed state.


Assuntos
Anestésicos Intravenosos/metabolismo , Proteínas de Bactérias/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos de Abertura Ativada por Ligante/metabolismo , Propofol/metabolismo , Regulação Alostérica , Substituição de Aminoácidos , Anestésicos Intravenosos/farmacologia , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Cianobactérias , Cisteína/química , Cisteína/genética , Concentração de Íons de Hidrogênio , Cinética , Canais Iônicos de Abertura Ativada por Ligante/química , Canais Iônicos de Abertura Ativada por Ligante/genética , Potenciais da Membrana/efeitos dos fármacos , Metanossulfonato de Metila/análogos & derivados , Metanossulfonato de Metila/química , Simulação de Acoplamento Molecular , Mutagênese Sítio-Dirigida , Propofol/farmacologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Estrutura Secundária de Proteína , Subunidades Proteicas , Homologia Estrutural de Proteína , Xenopus laevis
20.
ACS Chem Biol ; 8(6): 1110-6, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23557648

RESUMO

Hydrogen sulfide (H2S) has emerged as a new member of the gaseous transmitter family of signaling molecules and appears to play a regulatory role in the cardiovascular and nervous systems. Recent studies suggest that protein cysteine S-sulfhydration may function as a mechanism for transforming the H2S signal into a biological response. However, selective detection of S-sulfhydryl modifications is challenging since the persulfide group (RSSH) exhibits reactivity akin to other sulfur species, especially thiols. A modification of the biotin switch technique, using S-methyl methanethiosulfonate (MMTS) as an alkylating reagent, was recently used to identify a large number of proteins that may undergo S-sulfhydration, but the underlying mechanism of chemical detection was not fully explored. To address this key issue, we have developed a protein persulfide model and analogue of MMTS, S-4-bromobenzyl methanethiosulfonate (BBMTS). Using these new reagents, we investigated the chemistry in the modified biotin switch method and examined the reactivity of protein persulfides toward different electrophile/nucleophile species. Together, our data affirm the nucleophilic properties of the persulfide sulfane sulfur and afford new insights into protein S-sulfhydryl chemistry, which may be exploited in future detection strategies.


Assuntos
Proteína S/química , Sulfetos/análise , Alquilantes/química , Bioquímica/métodos , Biotina/química , Sulfeto de Hidrogênio/química , Mesilatos/química , Metanossulfonato de Metila/análogos & derivados , Metanossulfonato de Metila/química , Compostos de Sulfidrila/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...