Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Expert Opin Drug Discov ; 13(9): 815-824, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30101640

RESUMO

INTRODUCTION: 6-Mercaptopurine (6-MP) and 6-thioguanine (6-TG), two anticancer drugs, have high systemic toxicity due to a lack of target specificity. Therefore, increasing target selectivity should improve drug safety. Areas covered: The authors examined the hypothesis that new prodrug designs based upon mechanisms of kidney-selective toxicity of trichloroethylene would reduce systemic toxicity and improve selectivity to kidney and tumor cells. Two approaches specifically were investigated. The first approach was based upon bioactivation of trichloroethylene-cysteine S-conjugate by renal cysteine S-conjugate ß-lyases. The prodrugs obtained were kidney-selective but exhibited low turnover rates. The second approach was based on the toxic mechanism of trichloroethylene-cysteine S-conjugate sulfoxide, a Michael acceptor that undergoes rapid addition-elimination reactions with biological thiols. Expert opinion: Glutathione-dependent Michael addition-elimination reactions appear to be an excellent strategy to design highly efficient anticancer drugs. Targeting glutathione could be a promising approach for the development of anticancer prodrugs because cancer cells usually upregulate glutathione biosynthesis and/or glutathione S-transferases expression.


Assuntos
Antineoplásicos/administração & dosagem , Mercaptopurina/administração & dosagem , Tioguanina/administração & dosagem , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/metabolismo , Desenho de Fármacos , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Humanos , Rim/metabolismo , Mercaptopurina/efeitos adversos , Mercaptopurina/metabolismo , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Pró-Fármacos , Tioguanina/efeitos adversos , Tioguanina/metabolismo
2.
Chem Res Toxicol ; 30(2): 552-563, 2017 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-27977153

RESUMO

We have previously shown that 1-chloro-3-buten-2-one (CBO), a potential reactive metabolite of 1,3-butadiene (BD), exhibits potent cytotoxicity and genotoxicity that have been attributed in part to its reactivity toward DNA. In an effort to identify the DNA adducts of CBO, we characterized the CBO reactions with 2'-deoxyguanosine (dG), 2'-deoxycytidine (dC), and 2'-deoxyadenosine (dA) under in vitro physiological conditions (pH 7.4, 37 °C). In the present study, we investigated the CBO reaction with 2'-deoxythymidine (dT) and compared the rate constants of the reactions of CBO with dA, dC, dG, and dT at both individual- and mixed-nucleosides levels. We also investigated the reactions of CBO with single- and double-stranded DNA using HPLC with UV detection after adducts were released by either acid or enzymatic hydrolysis of DNA. Consistent with the results from the nucleoside reactions and the rate constant experiments, 1,N6-(1-hydroxy-1-chloromethylpropan-1,3-diyl)adenine (A-2D) was identified as the major DNA adduct detected after acid hydrolysis, followed by N7-(4-chloro-3-oxobutyl)guanine (CG-2H) and a small amount of 1,N6-(1-hydroxy-1-hydroxymethylpropan-1,3-diyl)adenine (A-1D). After enzymatic hydrolysis, 1,N6-(1-hydroxy-1-hydroxymethylpropan-1,3-diyl)-2'-dexoyadenosine (dA-1), 3,N4-(1-hydroxy-1-hydroxymethylpropan-1,3-diyl)-2'-deoxycytidine (dC-1/2), and 1,N2-(3-hydroxy-3-hydroxymethylpropan-1,3-diyl)-2'-dexoyguanosine (CG-1) were detected, with dA-1 being the major product, followed by dC-1/2. When a nontoxic concentration of CBO (1 µM) was incubated with HepG2 cells, no adducts could be detected by LC-MS. However, pretreatment of cells with l-buthionine sulfoximine to deplete GSH levels allowed A-2D to be consistently detected in cellular DNA. These results may contribute to a better understanding of the role of the DNA adducts in CBO genotoxicity and mutagenicity. It also suggests that A-2D could be developed as a biomarker of CBO formation after BD exposure in vivo.


Assuntos
Butanonas/química , Adutos de DNA/química , DNA de Cadeia Simples/química , DNA/química , Purinas/análise , Pirimidinas/análise , Cromatografia Líquida de Alta Pressão , Humanos , Purinas/química , Pirimidinas/química , Espectrometria de Fluorescência
3.
Expert Opin Drug Metab Toxicol ; 13(5): 513-524, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27936967

RESUMO

INTRODUCTION: Adverse drug reactions (ADRs) pose a significant health problem and are generally attributed to reactive metabolites. Olefinic moieties in drugs can undergo cytochrome P450-mediated bioactivation to produce reactive metabolites but myeloperoxidase (MPO)-mediated bioactivation of these moieties has not been reported. Thus, small molecules of alkene hydrocarbons are used as model compounds to characterize the MPO-mediated metabolism. Areas covered: The authors focus on MPO-mediated metabolism of alkene hydrocarbons to form chlorohydrins and the potential role of chlorohydrins in alkene toxicity and carcinogenicity. A case study is presented, in which a carcinogenic alkene, 1,3-butadiene, is demonstrated to form 1-chloro-2-hydroxy-3-butene (CHB) through the MPO-mediated pathway. Further bioactivation of CHB yields a cross-linking metabolite, 1-chloro-3-buten-2-one (CBO), which is highly reactive toward glutathione, proteins, nucleosides, and DNA. Toxicity and mutagenicity of CHB and CBO are also presented. Expert opinion: Alkene hydrocarbons readily undergo MPO-mediated bioactivation to form chlorohydrins, which can further be biotransformed into proteins/DNA-modifying reactive metabolites. Therefore, chlorohydrin formation may play an important role in alkene toxicity and carcinogenicity. Olefinic moieties in drugs are expected to undergo similar bioactivation, which may contribute to ADRs. Studies to investigate the roles of MPO and chlorohydrin formation in ADRs are thus warranted.


Assuntos
Alcenos/metabolismo , Ácido Hipocloroso/metabolismo , Peroxidase/metabolismo , Alcenos/efeitos adversos , Alcenos/química , Animais , Butadienos/metabolismo , Butadienos/toxicidade , Carcinógenos/metabolismo , Carcinógenos/toxicidade , Cloridrinas/efeitos adversos , Cloridrinas/química , Cloridrinas/metabolismo , Humanos , Hidrocarbonetos/efeitos adversos , Hidrocarbonetos/química , Hidrocarbonetos/metabolismo
4.
Chem Res Toxicol ; 29(6): 1041-50, 2016 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-27161607

RESUMO

1-Chloro-3-buten-2-one (CBO) is an in vitro metabolite of 1,3-butadiene (BD), a carcinogenic air pollutant. CBO exhibited potent cytotoxicity and genotoxicity that have been attributed in part to its reactivity toward DNA. Previously, we have characterized the CBO adducts with 2'-deoxycytidine and 2'-deoxyguanosine. In the present study, we report on the reaction of CBO with 2'-deoxyadenosine (dA) under in vitro physiological conditions (pH 7.4, 37 °C). We used the synthesized standards and their decomposition and acid-hydrolysis products to characterize the CBO-DNA adducts formed in human cells. The fused-ring dA adducts (dA-1 and dA-2) were readily synthesized and were structurally characterized as 1,N(6)-(1-hydroxy-1-hydroxymethylpropan-1,3-diyl)-2'-deoxyadenosine and 1,N(6)-(1-hydroxy-1-chloromethylpropan-1,3-diyl)-2'-deoxyadenosine, respectively. dA-1 exhibited a half-life of 16.0 ± 0.7 h and decomposed to dA at pH 7.4 and 37 °C. At similar conditions, dA-2 decomposed to dA-1 and dA, and had a half-life of 0.9 ± 0.1 h. These results provide strong evidence for dA-1 being a degradation product of dA-2. dA-1 is formed by replacement of the chlorine atom of dA-2 by a hydroxyl group. The slow decomposition of dA-1 to dA, along with the detection of hydroxymethyl vinyl ketone (HMVK) as another degradation product, suggested equilibrium between dA-1 and a ring-opened carbonyl-containing intermediate that undergoes a retro-Michael reaction to yield dA and HMVK. Acid hydrolysis of dA-1 and dA-2 yielded the corresponding deribosylated products A-1D and A-2D, respectively. In the acid-hydrolyzed reaction mixture of CBO with calf thymus DNA, both A-1D and A-2D could be detected; however, the amount of A-2D was significantly larger than that of A-1D. Interestingly, only A-2D could be detected by LC-MS analysis of acid-hydrolyzed DNA from cells incubated with CBO, suggesting that dA-2 was stable in DNA and thus may play an important role in the genotoxicity and carcinogenicity of BD. In addition, A-2D could be developed as a biomarker of CBO formation in human cells.


Assuntos
Butadienos/metabolismo , Butanonas/química , Butanonas/metabolismo , Adutos de DNA/análise , Adutos de DNA/química , DNA/química , Desoxiadenosinas/análise , Animais , Butadienos/química , Bovinos , DNA/metabolismo , Adutos de DNA/metabolismo , Desoxiadenosinas/química , Desoxiadenosinas/metabolismo , Células Hep G2 , Humanos , Estrutura Molecular
5.
Chem Biol Interact ; 226: 40-8, 2015 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-25500269

RESUMO

1-Chloro-3-buten-2-one (CBO) is a potential reactive metabolite of 1,3-butadiene (BD), a carcinogenic air pollutant. To develop tools that may help investigate the role of CBO in BD carcinogenicity and to develop biomarkers that can be used to assess BD exposure, the reaction of CBO with 2'-deoxyguanosine (dG) under in vitro physiological conditions (pH 7.4, 37°C) was investigated and the products (designated as CG-1, CG-2, CG-3, CG-4, CG-5, and CG-6 based on their retention times on HPLC) were characterized by MS and NMR spectroscopy. The structures of CG-1, CG-2, CG-3, and CG-4 were 1,N2-(3-hydroxy-3-hydroxymethylpropan-1,3-diyl)-2'-deoxyguanosine, N7-(4-chloro-3-oxobutyl)-2'-deoxyguanosine, N7,8-(3-hydroxy-3-chloromethylpropan-1,3-diyl)guanine and N2-(4-chloro-3-oxobutyl)-2'-deoxyguanosine, respectively. CG-5 and CG-6, a pair of diastereomers, were characterized as 1,N2-(3-hydroxy-3-chloromethylpropan-1,3-diyl)-2'-deoxyguanosine. CG-1 was stable under in vitro physiological conditions, whereas CG-2, CG-3, CG-4, and CG-5/6 were unstable and exhibited the half-lives at <1.0, 4.8±0.1, 6.7±0.3, and 2.7±0.1 h, respectively. CG-2 decomposed primarily via a retro-Michael reaction to produce dG and CBO, with only a small fraction of CG-2 degrading to CG-3. Decomposition of CG-4 proceeded via a cyclization reaction and/or replacement of the chlorine atom by a hydroxyl group to form 1,N2-(1-hydroxy-1-hydroxymethylpropan-1,3-diyl)-2'-deoxyguanosine (CG-4D1) and N(2)-(4-hydroxy-3-oxobutyl)-2'-deoxyguanosine (CG-4D2), whereas decomposition of CG-5/6 yielded CG-1. Collectively, the newly characterized CBO adducts could be used to investigate the role of CBO in the mechanism of BD carcinogenicity and could also be used to develop biomarkers for BD exposure.


Assuntos
Butanonas/química , Butanonas/toxicidade , Carcinógenos/química , Carcinógenos/toxicidade , Adutos de DNA/química , Desoxiguanosina/química , Concentração de Íons de Hidrogênio , Cinética
6.
Chem Res Toxicol ; 26(10): 1545-53, 2013 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-24020501

RESUMO

1-Chloro-3-buten-2-one (CBO) is a potential metabolite of 1,3-butadiene (BD), a carcinogenic air pollutant. CBO is a bifunctional alkylating agent that readily reacts with glutathione (GSH) to form mono-GSH and di-GSH adducts. Recently, CBO and its precursor 1-chloro-2-hydroxy-3-butene (CHB) were found to be cytotoxic and genotoxic in human liver cells in culture with CBO being approximately 100-fold more potent than CHB. In the present study, CBO was shown to react readily with 2'-deoxycytidine (dC) under in vitro physiological conditions (pH 7.4, 37 °C) to form four dC adducts with the CBO moieties forming fused rings with the N3 and N(4) atoms of dC. The four products were structurally characterized as 2-hydroxy-2-hydroxymethyl-7-(2-deoxy-ß-d-erythro-pentofuranosyl)-1,2,3,4-tetrahydro-6-oxo-6H,7H-pyrimido[1,6-a]pyrimidin-5-ium (dC-1 and dC-2, a pair of diastereomers), 4-chloromethyl-4-hydroxy-7-(2-deoxy-ß-d-erythro-pentofuranosyl)-1,2,3,4-tetrahydro-6-oxo-6H,7H-pyrimido[1,6-a]pyrimidin-5-ium (dC-3), and 2-chloromethyl-2-hydroxy-7-(2-deoxy-ß-d-erythro-pentofuranosyl)-1,2,3,4-tetrahydro-6-oxo-6H,7H-pyrimido[1,6-a]pyrimidin-5-ium (dC-4). Interestingly, dC-1 and dC-2 were stable under our experimental conditions (pH 7.4, 37 °C, and 6 h) and existed in equilibrium as indicated by HPLC analysis, whereas dC-3 and dC-4 were labile with the half-lives being 3.0 ± 0.36 and 1.7 ± 0.06 h, respectively. Decomposition of dC-4 produced both dC-1 and dC-2, whereas acid hydrolysis of dC-1/dC-2 and dC-4 in 1 M HCl at 100 °C for 30 min yielded the deribosylated adducts dC-1H/dC-2H and dC-4H, respectively. Because fused-ring dC adducts of other chemicals are mutagenic, the characterized CBO-dC adducts could be mutagenic and play a role in the cytotoxicity and genotoxicity of CBO and its precursors, CHB and BD. The CBO-dC adducts may also be used as standards to characterize CBO-DNA adducts and to develop potential biomarkers for CBO formation in vivo.


Assuntos
Butadienos/metabolismo , Butanonas/química , Adutos de DNA/química , Desoxicitidina/química , Biomarcadores/metabolismo , Butadienos/química , Butanonas/metabolismo , Butanonas/toxicidade , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Dano ao DNA/efeitos dos fármacos , Desoxicitidina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Espectrometria de Massas por Ionização por Electrospray , Temperatura , Fatores de Tempo
7.
Toxicol Appl Pharmacol ; 271(1): 13-9, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23643860

RESUMO

The cytotoxicity, genotoxicity, and mutagenicity of 1-chloro-2-hydroxy-3-butene (CHB), a known in vitro metabolite of the human carcinogen 1,3-butadiene, have not previously been investigated. Because CHB can be bioactivated by alcohol dehydrogenases to yield 1-chloro-3-buten-2-one (CBO), a bifunctional alkylating agent that caused globin-chain cross-links in erythrocytes, in the present study we investigated the cytotoxic and genotoxic potential of CHB and CBO in human normal hepatocyte L02 cells using the MTT assay, the relative cloning efficiency assay and the comet assay. We also investigated the mutagenic potential of these compounds with the Ames test using Salmonella strains TA1535 and TA1537. The results provide clear evidence for CHB and CBO being both cytotoxic and genotoxic with CBO being approximately 100-fold more potent than CHB. Interestingly, CHB generated both single-strand breaks and alkali-labile sites on DNA, whereas CBO produced only alkali-labile sites. CHB did not directly result in DNA breaks, whereas CBO was capable of directly generating breaks on DNA. Interestingly, both compounds did not induce DNA cross-links as examined by the comet assay. The Ames test results showed that CHB induced point mutation but not frameshift mutation, whereas the toxic effects of CBO made it difficult to reliably assess the mutagenic potential of CBO in the two strains. Collectively, the results suggest that CHB and CBO may play a role in the mutagenicity and carcinogenicity of 1,3-butadiene.


Assuntos
Butanóis/toxicidade , Butanonas/toxicidade , Carcinógenos/toxicidade , Hepatócitos/efeitos dos fármacos , Mutagênicos/toxicidade , Butadienos/metabolismo , Butadienos/toxicidade , Linhagem Celular , Ensaio Cometa , Quebras de DNA/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Testes de Mutagenicidade , Mutação Puntual/efeitos dos fármacos , Salmonella/genética
8.
Toxicology ; 306: 157-61, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23416178

RESUMO

The nephrotoxicity and nephrocarcinogenicity of trichloroethylene (TCE) and tetrachloroethylene (PCE) are believed to be mediated primarily through the cysteine S-conjugate ß-lyase-dependent bioactivation of the corresponding cysteine S-conjugate metabolites S-(1,2-dichlorovinyl)-l-cysteine (DCVC) and S-(1,2,2-trichlorovinyl)-l-cysteine (TCVC), respectively. DCVC and TCVC have previously been demonstrated to be mutagenic by the Ames Salmonella mutagenicity assay, and reduction in mutagenicity was observed upon treatment with the ß-lyase inhibitor aminooxyacetic acid (AOAA). Because DCVC and TCVC can also be bioactivated through sulfoxidation to yield the potent nephrotoxicants S-(1,2-dichlorovinyl)-l-cysteine sulfoxide (DCVCS) and S-(1,2,2-trichlorovinyl)-l-cysteine sulfoxide (TCVCS), respectively, the mutagenic potential of these two sulfoxides was investigated using the Ames Salmonella typhimurium TA100 mutagenicity assay. The results show both DCVCS and TCVCS were mutagenic, and TCVCS exhibited 3-fold higher mutagenicity than DCVCS. However, DCVCS and TCVCS mutagenic activity was approximately 700-fold and 30-fold lower than DCVC and TCVC, respectively. DCVC and DCVCS appeared to induce toxicity in TA100, as evidenced by increased microcolony formation and decreased mutant frequency above threshold concentrations. TCVC and TCVCS were not toxic in TA100. The toxic effects of DCVC limited the sensitivity of TA100 to DCVC mutagenic effects and rendered it difficult to investigate the effects of AOAA on DCVC mutagenic activity. Collectively, these results suggest that DCVCS and TCVCS exerted a definite but weak mutagenicity in the TA100 strain. Therefore, despite their potent nephrotoxicity, DCVCS and TCVCS are not likely to play a major role in DCVC or TCVC mutagenicity in this strain.


Assuntos
Liases de Carbono-Enxofre/metabolismo , Cisteína/análogos & derivados , Tetracloroetileno/toxicidade , Tricloroetileno/toxicidade , Liases de Carbono-Enxofre/antagonistas & inibidores , Cisteína/metabolismo , Testes de Mutagenicidade , Ácido Oxâmico/farmacologia
9.
Toxicol Appl Pharmacol ; 267(1): 1-10, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23253325

RESUMO

N-Acetyl-S-(1,2-dichlorovinyl)-L-cysteine (NA-DCVC) has been detected in the urine of humans exposed to trichloroethylene and its related sulfoxide, N-acetyl-S-(1,2-dichlorovinyl)-L-cysteine sulfoxide (NA-DCVCS), has been detected as hemoglobin adducts in blood of rats dosed with S-(1,2-dichlorovinyl)-L-cysteine (DCVC) or S-(1,2-dichlorovinyl)-L-cysteine sulfoxide (DCVCS). Because the in vivo nephrotoxicity of NA-DCVCS was unknown, in this study, male Sprague-Dawley rats were dosed (i.p.) with 230 µmol/kg b.w. NA-DCVCS or its potential precursors, DCVCS or NA-DCVC. At 24 h post treatment, rats given NA-DCVC or NA-DCVCS exhibited kidney lesions and effects on renal function distinct from those caused by DCVCS. NA-DCVC and NA-DCVCS primarily affected the cortico-medullary proximal tubules (S(2)-S(3) segments) while DCVCS primarily affected the outer cortical proximal tubules (S(1)-S(2) segments). When NA-DCVCS or DCVCS was incubated with GSH in phosphate buffer pH 7.4 at 37°C, the corresponding glutathione conjugates were detected, but NA-DCVC was not reactive with GSH. Because NA-DCVCS exhibited a longer half-life than DCVCS and addition of rat liver cytosol enhanced GSH conjugate formation, catalysis of GSH conjugate formation by the liver could explain the lower toxicity of NA-DCVCS in comparison with DCVCS. Collectively, these results provide clear evidence that NA-DCVCS formation could play a significant role in DCVC, NA-DCVC, and trichloroethylene nephrotoxicity. They also suggest a role for hepatic metabolism in the mechanism of NA-DCVC nephrotoxicity.


Assuntos
Acetilcisteína/análogos & derivados , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Tricloroetileno/química , Tricloroetileno/metabolismo , Acetilcisteína/metabolismo , Acetilcisteína/toxicidade , Animais , Nefropatias/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Tricloroetileno/toxicidade
10.
Chem Res Toxicol ; 25(11): 2600-7, 2012 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-23110628

RESUMO

1,3-Butadiene (BD) is an air pollutant whose toxicity and carcinogenicity have been considered primarily mediated by its reactive metabolites, 3,4-epoxy-1-butene and 1,2,3,4-diepoxybutane, formed in liver and extrahepatic tissues by cytochromes P450s. A possible alternative metabolic pathway in bone marrow and immune cells is the conversion of BD to the chlorinated allylic alcohol 1-chloro-2-hydroxy-3-butene (CHB) by myeloperoxidase in the presence of hydrogen peroxide and chloride ion. In the present study, we investigated the in vitro bioactivation of CHB by alcohol dehydrogenases (ADH) under in vitro physiological conditions (pH 7.4, 37 °C). The results provide clear evidence for CHB being converted to 1-chloro-3-buten-2-one (CBO) by purified horse liver ADH and rat liver cytosol. CBO readily reacted with glutathione (GSH) under assay conditions to form three products: two CBO-mono-GSH conjugates [1-chloro-4-(S-glutathionyl)butan-2-one (3) and 1-(S-glutathionyl)-3-buten-2-one (4)] and one CBO-di-GSH conjugate [1,4-bis(S-glutathionyl)butan-2-one (5)]. CHB bioactivation and the ratios of the three GSH conjugates formed were dependent upon incubation time, GSH and CHB concentrations, and the presence of ADH or rat liver cytosol. The ADH enzymatic reaction followed Michaelis-Menten kinetics with a K(m) at 3.5 mM and a k(cat) at 0.033 s(-1). After CBO was incubated with freshly isolated mouse erythrocytes, globin dimers were detected using SDS-PAGE and silver staining, providing evidence that CBO can act as a protein cross-linking agent. Collectively, the results provide clear evidence for CHB bioactivation by ADH and rat liver cytosol to yield CBO. The bifunctional alkylating ability of CBO suggests that it may play a role in BD toxicity and/or carcinogenicity.


Assuntos
Álcool Desidrogenase/metabolismo , Butanóis/metabolismo , Butanonas/metabolismo , Citosol/metabolismo , Fígado/metabolismo , Álcool Desidrogenase/química , Alquilação , Animais , Butanóis/química , Butanonas/química , Citosol/química , Citosol/enzimologia , Eritrócitos/química , Eritrócitos/metabolismo , Glutationa/química , Glutationa/metabolismo , Cavalos , Fígado/química , Fígado/enzimologia , Estrutura Molecular , Ratos
11.
Expert Opin Drug Metab Toxicol ; 8(9): 1157-72, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22681489

RESUMO

INTRODUCTION: Reactive metabolite-mediated toxicity is frequently limited to the organ where the electrophilic metabolites are generated. Some reactive metabolites, however, might have the ability to translocate from their site of formation. This suggests that for these reactive metabolites, investigations into the role of organs other than the one directly affected could be relevant to understanding the mechanism of toxicity. AREAS COVERED: The authors discuss the physiological and biochemical factors that can enable reactive metabolites to cause toxicity in an organ distal from the site of generation. Furthermore, the authors present a case study which describes studies that demonstrate that S-(1,2-dichlorovinyl)-L-cysteine sulfoxide (DCVCS) and N-acetyl-S-(1,2-dichlorovinyl-L-cysteine sulfoxide (N-AcDCVCS), reactive metabolites of the known trichloroethylene metabolites S-(1,2-dichlorovinyl)-L-cysteine (DCVC), and N-acetyl-S-(1,2-dichlorovinyl)-L-cysteine (N-AcDCVC), are generated in the liver and translocate through the circulation to the kidney to cause nephrotoxicity. EXPERT OPINION: The ability of reactive metabolites to translocate could be important to consider when investigating mechanisms of toxicity. A mechanistic approach, similar to the one described for DCVCS and N-AcDCVCS, could be useful in determining the role of circulating reactive metabolites in extrahepatic toxicity of drugs and other chemicals. If this is the case, intervention strategies that would not otherwise be feasible might be effective for reducing extrahepatic toxicity.


Assuntos
Cisteína/análogos & derivados , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Acetilcisteína/análogos & derivados , Acetilcisteína/sangue , Acetilcisteína/metabolismo , Acetilcisteína/toxicidade , Cisteína/sangue , Cisteína/metabolismo , Cisteína/toxicidade , Humanos , Rim/metabolismo , Rim/patologia , Fígado/metabolismo , Fígado/patologia , Tricloroetileno/sangue , Tricloroetileno/metabolismo , Tricloroetileno/toxicidade
12.
Chem Res Toxicol ; 24(11): 1915-23, 2011 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-21988407

RESUMO

S-(1,2-Dichlorovinyl)-L-cysteine sulfoxide (DCVCS) is a reactive and potent nephrotoxic metabolite of the human trichloroethylene metabolite S-(1,2-dichlorovinyl)-L-cysteine (DCVC). Because DCVCS covalent binding to kidney proteins likely plays a role in its nephrotoxicity, in this study biotin-tagged DCVCS, N-biotinyl-DCVCS (NB-DCVCS), was synthesized, and its stability in buffer alone and in the presence of rat blood or plasma was characterized in vitro. In addition, reactivity toward GSH and covalent binding to selected model enzymes and isolated kidney proteins were characterized. The half-lives of NB-DCVCS (39.6 min) and the DCVCS (diastereomer 1, 14.4 min; diastereomer 2, 6 min) in the presence of GSH were comparable. Incubating the model enzymes glutathione reductase and malate dehydrogenase with 10 µM NB-DCVCS for 3 h at 37 °C followed by immunoblotting using antibiotin antibodies demonstrated that glutathione reductase and malate dehydrogenase were extensively modified by NB-DCVCS. When rat kidney cytosol (6 µg/µL) was incubated with NB-DCVCS (312.5 nM to 5 µM) for 3 h at 37 °C followed by immunoblotting, a concentration-dependent increase in signal with multiple proteins with different molecular weights was observed, suggesting that NB-DCVCS binds to multiple kidney proteins with different selectivity. Incubating rat kidney cytosol with DCVCS (10-100 µM) prior to the addition of NB-DCVCS (2.5 µM) reduced the immunoblotting signal, suggesting that NB-DCVCS and DCVCS compete for the same binding sites. A comparison of the stability of NB-DCVCS and DCVCS in rat blood and plasma was determined in vitro, and NB-DCVCS exhibited higher stability than DCVCS in both media. Collectively, these results suggest that NB-DCVCS shows sufficient stability, reactivity, and selectivity to warrant further investigations into its possible use as a tool for future characterization of the role of covalent modification of renal proteins by DCVCS in nephrotoxicity.


Assuntos
Biotina/metabolismo , Cisteína/análogos & derivados , Glutationa Redutase/sangue , Indicadores e Reagentes/análise , Neoplasias Renais/sangue , Rim/metabolismo , Malato Desidrogenase/sangue , Animais , Sítios de Ligação , Ligação Competitiva , Biotina/química , Biotinilação , Western Blotting , Cisteína/efeitos adversos , Cisteína/química , Cisteína/metabolismo , Cisteína/toxicidade , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Eletroforese em Gel de Poliacrilamida , Glutationa Redutase/química , Meia-Vida , Humanos , Indicadores e Reagentes/química , Rim/efeitos dos fármacos , Rim/patologia , Neoplasias Renais/etiologia , Neoplasias Renais/patologia , Malato Desidrogenase/química , Ligação Proteica , Ratos Sprague-Dawley
13.
Expert Opin Drug Metab Toxicol ; 6(11): 1333-46, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20874374

RESUMO

IMPORTANCE OF THE FIELD: Disrupted l-methionine (Met) metabolism can lead to hepatic, neurological and cardiovascular dysfunction in humans. Aberrant methyl group flux likely contributes to the development of these pathologies, but when patients also become hypermethionemic, additional toxicological mechanisms may be relevant. AREAS COVERED IN THIS REVIEW: Following a discussion of the causes of hypermethionemia in humans, evidence for the toxicological roles and clinical significance of the Met transmethylation (TM), transamination (TA) and sulfoxidation (SO) pathways will be presented. WHAT THE READER WILL GAIN: Recent data from freshly isolated mouse hepatocytes (FIMHs) confirmed previous in vivo results in rodents that Met TM is a detoxification pathway while Met TA leads to toxicity. Gender-related differences in Met accumulation and metabolism in FIMHs correlated with gender differences in toxicity. Data obtained from FIMHs also implicated Met SO in Met metabolism and toxicity. Currently, little is known about the mechanisms and biological significance of Met sulfoxidation in humans. TAKE HOME MESSAGE: In hypermethionemic patients, clinical and dietary interventions should focus on increasing Met TM and decreasing Met TA and SO. Novel biomarkers of hypermethionemia in humans that correlate with pathological end points are needed to better understand the impact of the condition.


Assuntos
Hepatopatias/etiologia , Fígado/metabolismo , Metionina/sangue , Animais , Biomarcadores/sangue , Feminino , Hepatócitos/metabolismo , Humanos , Fígado/patologia , Hepatopatias/patologia , Masculino , Metionina/metabolismo , Metilação , Camundongos , Fatores Sexuais , Sulfóxidos/metabolismo , Transaminases/metabolismo
14.
Biochem Pharmacol ; 79(9): 1345-51, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20036217

RESUMO

Flavin-containing monooxygenase (FMO) expression in male mouse liver is altered after 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure or castration. Because TCDD is slowly eliminated from the body, we examined hepatic Fmo mRNA alterations for up to 32 days following 10 or 64 microg/kg TCDD exposure by oral gavage in male C57BL/6J mice. Fmo2 mRNA was significantly induced at 1, 4, and 8 days whereas Fmo3 mRNA was also induced at 32 days relative to controls. Fmo3 mRNA levels exhibited a dose-dependent increase at 4, 8, and 32 days after exposure; Fmo1, Fmo4, and Fmo5 mRNA did not exhibit clear trends. Because castration alone also increased Fmo2, Fmo3, and Fmo4 mRNA we examined the combined effects of castration and TCDD treatment on FMO expression. A greater than additive effect was observed with Fmo2 and Fmo3 mRNA expression. Fmo2 mRNA exhibited a 3-5-fold increase after castration or 10 microg/kg TCDD exposure by oral gavage, whereas an approximately 20-fold increase was observed between the sham-castrated control and castrated TCDD-treated mice. Similarly, treatment with 10 microg/kg TCDD alone increased Fmo3 mRNA 130- and 180-fold in the sham-castrated and castrated mice compared to their controls respectively, whereas, Fmo3 mRNA increased approximately 1900-fold between the sham control and castrated TCDD-treated mice. An increase in hepatic Fmo3 protein in TCDD-treated mice was observed by immunoblotting and assaying methionine S-oxidase activity. Collectively, these results provide evidence for isoform distinct time-, dose-, and castration-dependent effects of TCDD on FMO expression and suggest cross-talk between TCDD and testosterone signal transduction pathways.


Assuntos
Fígado/efeitos dos fármacos , Fígado/enzimologia , Orquiectomia , Oxigenases/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Animais , Regulação Enzimológica da Expressão Gênica/fisiologia , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Oxigenases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
Chem Res Toxicol ; 22(9): 1629-38, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19694420

RESUMO

S-(1,2-Dichlorovinyl)-L-cysteine (DCVC), a mutagenic and nephrotoxic metabolite of trichloroethylene, is bioactivated to S-(1,2-dichlorovinyl)-L-cysteine sulfoxide (DCVCS) and chlorothioketene and/or 2-chlorothionoacetyl chloride by cysteine conjugate S-oxidase (S-oxidase) and cysteine conjugate beta-lyase (beta-lyase), respectively. Previously, we identified DCVCS-globin monoadducts and cross-links upon treating rats with DCVCS or incubating erythrocytes with DCVCS. In this study, the formation of DCVC-derived reactive intermediates was investigated after rats were given a single (230 or 460 micromol/kg, i.p.) or multiple (3 or 30 micromol/kg daily for 5 days) DCVC doses. LC/ESI/MS of trypsin-digested globin peptides revealed both S-oxidase and beta-lyase-derived globin monoadducts and cross-links consistent with in vivo DCVC bioactivation by both pathways. MS/MS analyses of trypsin-digested fractions of globin from one of the rats treated with multiple 30 micromol/kg DCVC doses led to identification of beta-lyase-derived monoadducts on both Cys93 and Cys125 of the beta-chains. While rats dosed with the 230 micromol/kg DCVC dose exhibited beta-lyase-dependent monoadducts and cross-links only (four out of four rats), rats given the 460 micromol/kg DCVC dose (two out of four) and rats administered the multiple DCVC doses (two out of four) exhibited both beta-lyase- and S-oxidase-derived monoadducts and cross-links. Because previous incubations of erythrocytes with DCVC did not result in detection of DCVCS-derived monoadducts or cross-links and had only resulted in detection of beta-lyase-derived monoadducts and cross-links, the DCVCS-globin monoadducts and cross-links detected in this study are likely the result of DCVC bioactivation outside the circulation and subsequent translocation of DCVCS and N-acetylated DCVCS into the erythrocytes.


Assuntos
Reagentes de Ligações Cruzadas/química , Cisteína/análogos & derivados , Globinas/química , Tionas/sangue , Sequência de Aminoácidos , Animais , Cromatografia Líquida de Alta Pressão , Cisteína/administração & dosagem , Cisteína/sangue , Cisteína/química , Cisteína/toxicidade , Eritrócitos/metabolismo , Masculino , Dados de Sequência Molecular , Peptídeos/química , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização por Electrospray , Tionas/química , Tricloroetileno/química , Tricloroetileno/metabolismo , Tripsina/metabolismo
16.
Chem Res Toxicol ; 22(7): 1351-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19572755

RESUMO

S-(1,2-Dichlorovinyl)-L-cysteine (DCVC), a mutagenic and nephrotoxic metabolite of trichloroethylene, can be bioactivated to reactive metabolites, S-(1,2-dichlorovinyl)-L-cysteine sulfoxide (DCVCS) or chlorothioketene and/or 2-chlorothionoacetyl chloride, by cysteine conjugate S-oxidase (S-oxidase) and cysteine conjugate beta-lyase (beta-lyase), respectively. Previously, we characterized the reactivity of DCVCS with Hb upon incubation of erythrocytes with DCVCS and provided evidence for the formation of distinct DCVCS-Hb monoadducts and cross-links in both isolated erythrocytes and rats given DCVCS. In the present study, we investigated DCVC bioactivation and Hb adduct formation in isolated rat erythrocytes incubated with DCVC (9 and 450 microM) at 37 degrees C and pH 7.4. The results suggested that no DCVCS monoadducts or cross-links were formed; however, LC/electrospray ionization/MS and matrix-assisted laser desorption/ionization/MS of trypsin-digested globin peptides revealed the presence of beta-lyase-derived globin monoadducts and cross-links. Adducts and cross-links in which the sulfur atom of the reactive sulfur intermediates were replaced by oxygen have also been detected. Use of SDS-PAGE provided additional evidence for globin cross-link formation in the presence of DCVC. Interestingly, the MS results suggest that the observed peptide selectivity of the beta-lyase-derived reactive sulfur/oxygen-containing species was different than that previously observed with DCVCS. While these results suggested that erythrocytes have beta-lyase but not S-oxidase activity, further support for this hypothesis was obtained using S-(2-benzothiazolyl)-L-cysteine, an alternative substrate for beta-lyases. Collectively, the results demonstrate the utility of Hb adducts and cross-links to characterize the metabolic pathway responsible for DCVC bioactivation in erythrocytes and to provide distinct biomarkers for each reactive metabolite.


Assuntos
Liases de Carbono-Enxofre/metabolismo , Reagentes de Ligações Cruzadas/química , Cisteína/análogos & derivados , Eritrócitos/enzimologia , Hemoglobinas/química , Sequência de Aminoácidos , Animais , Benzotiazóis/química , Benzotiazóis/farmacologia , Benzotiazóis/toxicidade , Cromatografia Líquida de Alta Pressão , Cisteína/química , Cisteína/farmacologia , Cisteína/toxicidade , Masculino , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização por Electrospray , Tripsina/metabolismo
17.
Chem Res Toxicol ; 22(6): 1163-71, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19419228

RESUMO

3-Butene-1,2-diol (BDD), a known in vivo metabolite of 1,3-butadiene, is oxidized to a reactive Michael acceptor, hydroxymethylvinyl ketone (HMVK). Previously, we characterized the formation of three HMVK-amino acid monoadducts when HMVK was incubated in vitro with N-acetyl-l-cysteine, l-valinamide, and N-acetyl-l-lysine (NAL) at physiological conditions. One HMVK-NAL cyclic diadduct (cyclic diadduct 1) also formed by sequential Michael addition reactions of two HMVK molecules with the epsilon-amino group of NAL followed by enolization and cyclization. Loss of a water molecule and autoxidation convert cyclic diadduct 1 to a more stable cyclic diadduct 2. In the present study, we used multiple mass spectrometry techniques to investigate the formation of HMVK adducts with nucleophilic residues of Hb in vivo after dosing Sprague-Dawley rats with 25 and 200 mg/kg BDD. Trypsin-digested globin peptides with mass shifts consistent with the presence of HMVK monoadducts and cyclic diadducts were detected by LC/electrospray-quadrupole time-of-flight/MS with all rats given BDD. Use of matrix-assisted laser desorption ionization/Fourier transform ion cyclotron resonance provided further evidence for the formation of HMVK monoadducts and cyclic diadducts, and use of LC/MS/MS provided unequivocal evidence for adduction of HMVK with Cys125 of globin beta chains. Because BDD can also be oxidized to 1,2-dihydroxy-3,4-epoxybutane (EBD), the formation of N(2)-(2,3,4-trihydroxybutyl) (THB)-Hb adducts was also investigated in rats given BDD, and several peptides modified by THB were detected. However, because HMVK incubations with red blood cells in vitro also led to the detection of THB-Hb adducts, the THB adducts formed in vivo could be attributed to formation of HMVK, EBD, or both. Collectively, the results provide new insights into the reaction of HMVK with proteins.


Assuntos
Butanonas/química , Compostos de Epóxi/farmacologia , Glicóis/farmacologia , Hemoglobinas/administração & dosagem , Hemoglobinas/química , Compostos Heterocíclicos com 1 Anel/administração & dosagem , Cetonas/administração & dosagem , Sequência de Aminoácidos , Animais , Cromatografia Líquida de Alta Pressão , Compostos de Epóxi/química , Glicóis/química , Hemoglobinas/metabolismo , Compostos Heterocíclicos com 1 Anel/química , Cetonas/química , Masculino , Dados de Sequência Molecular , Fragmentos de Peptídeos/análise , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização por Electrospray , Tripsina/metabolismo
18.
J Pharmacol Exp Ther ; 329(3): 1148-55, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19307449

RESUMO

Flavin-containing monooxygenases (FMOs) play significant roles in the metabolism of drugs and endogenous or foreign compounds. In this study, the regional distribution of FMO isoforms 1, 3, and 4 was investigated in male Sprague-Dawley rat liver and kidney using immunohistochemistry (IHC). Rabbit polyclonal antibodies to rat FMO1 and FMO4, developed using anti-peptide technology, and commercial anti-human FMO3 antibody were used; specificities of the antibodies were verified using Western blotting, immunoprecipitation, and IHC. In liver, the highest immunoreactivity for FMO1 and FMO3 was detected in the perivenous region, and immunoreactivity decreased in intensity toward the periportal region. In contrast, FMO4 immunoreactivity was detected with the opposite lobular distribution. In the kidney, the highest immunoreactivity for FMO1, -3, and -4 was detected in the distal tubules. FMO1 and FMO4 immunoreactivity was also detected in the proximal tubules with strong staining in the brush borders, whereas less FMO3 immunoreactivity was detected in the proximal tubules. Immunoreactivity for FMO3 and FMO4 was detected in the collecting tubules in the renal medulla and the glomerulus, whereas little FMO1 immunoreactivity was detected in these regions. The FMO1 antibody did not react with human liver or kidney microsomes. However, the FMO4 antibody reacted with male and female mouse and human tissues. These data provided a compelling visual demonstration of the isoform-specific localization patterns of FMO1, -3, and -4 in the rat liver and kidney and the first evidence for expression of FMO4 at the protein level in mouse and human liver and kidney microsomes.


Assuntos
Rim/metabolismo , Fígado/metabolismo , Oxigenases/metabolismo , Isoformas de Proteínas/metabolismo , Animais , Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Reações Cruzadas/imunologia , Feminino , Complexo de Golgi/metabolismo , Humanos , Córtex Renal/metabolismo , Glomérulos Renais/metabolismo , Medula Renal/metabolismo , Túbulos Renais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Microssomos/metabolismo , Oxigenases/imunologia , Isoformas de Proteínas/imunologia , Ratos , Ratos Sprague-Dawley
19.
Chem Res Toxicol ; 22(5): 918-25, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19317513

RESUMO

Previously, our laboratory has shown that hydroxymethylvinyl ketone (HMVK), a Michael acceptor oxidation product of the 1,3-butadiene metabolite, 3-butene-1,2-diol, readily reacts with hemoglobin at physiological conditions and that mass spectrometry of trypsin-digested peptides suggested adduct formation with various nucleophilic amino acids. In the present study, we characterized reactions ofHMVK (3 mM) with three model nucleophilic amino acids (6 and/or 15 mM): N-acetyl-L-cysteine (NAC),L-valinamide, and N-acetyl-L-lysine (NAL). NAC was the most reactive toward HMVK followed by L-valinamide and NAL. HMVK incubations with each amino acid at pH 7.4 and 37 degrees C resulted in the formation of a mono-Michael adduct. In addition, HMVK incubated with NAL gave rise to two additional bis-Michael adducts characterized by LC/MS, LC/MS/MS, 1H NMR, and 1H-detected heteronuclear single quantum correlation. The relative ratios of areas of NAL monoadduct (adduct 1) and diadducts (adducts 2 and 3) at 6 h were 49, 21, and 30% of total product area, respectively. The formation of adduct 2 was dependent upon the presence of both adduct 1 and HMVK, whereas the formation of adduct 3 was dependent upon the presence of adduct 2 only. Monoadducts were formed by a Michael addition reaction of one HMVK moiety with nucleophilic amino acid, whereas NAL diadducts were products of two Michael addition reactions of two HMVK moieties followed by enolization and formation of an octameric cyclic product. NAL diadduct (adduct 3) was formed by loss of a water molecule from adduct 2 followed by autoxidation of one of the hydroxy groups, yielding a diketone conjugated system. Collectively, our results provide strong evidence that HMVK can react with various nucleophilic residues and form different types of adducts, suggesting that a variety of proteins may be subjected to these modifications, which could result in loss of protein function.


Assuntos
Aminoácidos/química , Butadienos/metabolismo , Butanonas/química , Butadienos/química , Cromatografia Líquida de Alta Pressão , Lisina/análogos & derivados
20.
Toxicol Appl Pharmacol ; 236(3): 358-65, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19236888

RESUMO

L-methionine (Met) is hepatotoxic at high concentrations. Because Met toxicity in freshly isolated mouse hepatocytes is gender-dependent, the goal of this study was to assess the roles of Met accumulation and metabolism in the increased sensitivity of male hepatocytes to Met toxicity compared with female hepatocytes. Male hepatocytes incubated with Met (30 mM) at 37 degrees C exhibited higher levels of intracellular Met at 0.5, 1.0, and 1.5 h, respectively, compared to female hepatocytes. Conversely, female hepatocytes had higher levels of S-adenosyl-L-methionine compared to male hepatocytes. Female hepatocytes also exhibited higher L-methionine-L-sulfoxide levels relative to control hepatocytes, whereas the increases in L-methionine-D-sulfoxide (Met-D-O) levels were similar in hepatocytes of both genders. Addition of aminooxyacetic acid (AOAA), an inhibitor of Met transamination, significantly increased Met levels at 1.5 h and increased Met-d-O levels at 1.0 and 1.5 h only in Met-exposed male hepatocytes. No gender differences in cytosolic Met transamination activity by glutamine transaminase K were detected. However, female mouse liver cytosol exhibited higher methionine-dl-sulfoxide (MetO) reductase activity than male mouse liver cytosol at low (0.25 and 0.5 mM) MetO concentrations. Collectively, these results suggest that increased cellular Met accumulation, decreased Met transmethylation, and increased Met and MetO transamination in male mouse hepatocytes may be contributing to the higher sensitivity of the male mouse hepatocytes to Met toxicity in comparison with female mouse hepatocytes.


Assuntos
Hepatócitos/metabolismo , Metionina/metabolismo , Fatores Sexuais , Aminas/metabolismo , Animais , Feminino , Masculino , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...