Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Chem Res Toxicol ; 30(2): 552-563, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-27977153

RESUMEN

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.


Asunto(s)
Butanonas/química , Aductos de ADN/química , ADN de Cadena Simple/química , ADN/química , Purinas/análisis , Pirimidinas/análisis , Cromatografía Líquida de Alta Presión , Humanos , Purinas/química , Pirimidinas/química , Espectrometría de Fluorescencia
2.
Chem Res Toxicol ; 29(6): 1041-50, 2016 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-27161607

RESUMEN

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.


Asunto(s)
Butadienos/metabolismo , Butanonas/química , Butanonas/metabolismo , Aductos de ADN/análisis , Aductos de ADN/química , ADN/química , Desoxiadenosinas/análisis , Animales , Butadienos/química , Bovinos , ADN/metabolismo , Aductos de ADN/metabolismo , Desoxiadenosinas/química , Desoxiadenosinas/metabolismo , Células Hep G2 , Humanos , Estructura Molecular
3.
Toxicol Appl Pharmacol ; 267(1): 1-10, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23253325

RESUMEN

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.


Asunto(s)
Acetilcisteína/análogos & derivados , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Tricloroetileno/química , Tricloroetileno/metabolismo , Acetilcisteína/metabolismo , Acetilcisteína/toxicidad , Animales , Enfermedades Renales/patología , Masculino , Ratas , Ratas Sprague-Dawley , Tricloroetileno/toxicidad
4.
Toxicol Appl Pharmacol ; 271(1): 13-9, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23643860

RESUMEN

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.


Asunto(s)
Butanoles/toxicidad , Butanonas/toxicidad , Carcinógenos/toxicidad , Hepatocitos/efectos de los fármacos , Mutágenos/toxicidad , Butadienos/metabolismo , Butadienos/toxicidad , Línea Celular , Ensayo Cometa , Roturas del ADN/efectos de los fármacos , Hepatocitos/patología , Humanos , Pruebas de Mutagenicidad , Mutación Puntual/efectos de los fármacos , Salmonella/genética
5.
Chem Res Toxicol ; 26(10): 1545-53, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-24020501

RESUMEN

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.


Asunto(s)
Butadienos/metabolismo , Butanonas/química , Aductos de ADN/química , Desoxicitidina/química , Biomarcadores/metabolismo , Butadienos/química , Butanonas/metabolismo , Butanonas/toxicidad , Línea Celular , Cromatografía Líquida de Alta Presión , Daño del ADN/efectos de los fármacos , Desoxicitidina/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Hidrólisis , Espectrometría de Masa por Ionización de Electrospray , Temperatura , Factores de Tiempo
6.
Chem Res Toxicol ; 25(11): 2600-7, 2012 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-23110628

RESUMEN

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.


Asunto(s)
Alcohol Deshidrogenasa/metabolismo , Butanoles/metabolismo , Butanonas/metabolismo , Citosol/metabolismo , Hígado/metabolismo , Alcohol Deshidrogenasa/química , Alquilación , Animales , Butanoles/química , Butanonas/química , Citosol/química , Citosol/enzimología , Eritrocitos/química , Eritrocitos/metabolismo , Glutatión/química , Glutatión/metabolismo , Caballos , Hígado/química , Hígado/enzimología , Estructura Molecular , Ratas
7.
Chem Res Toxicol ; 24(11): 1915-23, 2011 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-21988407

RESUMEN

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.


Asunto(s)
Biotina/metabolismo , Cisteína/análogos & derivados , Glutatión Reductasa/sangre , Indicadores y Reactivos/análisis , Neoplasias Renales/sangre , Riñón/metabolismo , Malato Deshidrogenasa/sangre , Animales , Sitios de Unión , Unión Competitiva , Biotina/química , Biotinilación , Western Blotting , Cisteína/efectos adversos , Cisteína/química , Cisteína/metabolismo , Cisteína/toxicidad , Relación Dosis-Respuesta a Droga , Estabilidad de Medicamentos , Electroforesis en Gel de Poliacrilamida , Glutatión Reductasa/química , Semivida , Humanos , Indicadores y Reactivos/química , Riñón/efectos de los fármacos , Riñón/patología , Neoplasias Renales/etiología , Neoplasias Renales/patología , Malato Deshidrogenasa/química , Unión Proteica , Ratas Sprague-Dawley
8.
J Pharmacol Exp Ther ; 329(3): 1148-55, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19307449

RESUMEN

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.


Asunto(s)
Riñón/metabolismo , Hígado/metabolismo , Oxigenasas/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Reacciones Cruzadas/inmunología , Femenino , Aparato de Golgi/metabolismo , Humanos , Corteza Renal/metabolismo , Glomérulos Renales/metabolismo , Médula Renal/metabolismo , Túbulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos , Microsomas/metabolismo , Oxigenasas/inmunología , Isoformas de Proteínas/inmunología , Ratas , Ratas Sprague-Dawley
9.
Toxicol Appl Pharmacol ; 236(3): 358-65, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19236888

RESUMEN

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.


Asunto(s)
Hepatocitos/metabolismo , Metionina/metabolismo , Factores Sexuales , Aminas/metabolismo , Animales , Femenino , Masculino , Ratones
10.
Chem Res Toxicol ; 22(5): 918-25, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19317513

RESUMEN

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.


Asunto(s)
Aminoácidos/química , Butadienos/metabolismo , Butanonas/química , Butadienos/química , Cromatografía Líquida de Alta Presión , Lisina/análogos & derivados
11.
Chem Res Toxicol ; 22(9): 1629-38, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19694420

RESUMEN

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.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Cisteína/análogos & derivados , Globinas/química , Tionas/sangre , Secuencia de Aminoácidos , Animales , Cromatografía Líquida de Alta Presión , Cisteína/administración & dosificación , Cisteína/sangre , Cisteína/química , Cisteína/toxicidad , Eritrocitos/metabolismo , Masculino , Datos de Secuencia Molecular , Péptidos/química , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Ionización de Electrospray , Tionas/química , Tricloroetileno/química , Tricloroetileno/metabolismo , Tripsina/metabolismo
12.
Chem Res Toxicol ; 22(7): 1351-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19572755

RESUMEN

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.


Asunto(s)
Liasas de Carbono-Azufre/metabolismo , Reactivos de Enlaces Cruzados/química , Cisteína/análogos & derivados , Eritrocitos/enzimología , Hemoglobinas/química , Secuencia de Aminoácidos , Animales , Benzotiazoles/química , Benzotiazoles/farmacología , Benzotiazoles/toxicidad , Cromatografía Líquida de Alta Presión , Cisteína/química , Cisteína/farmacología , Cisteína/toxicidad , Masculino , Datos de Secuencia Molecular , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Ionización de Electrospray , Tripsina/metabolismo
13.
Chem Res Toxicol ; 22(6): 1163-71, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19419228

RESUMEN

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.


Asunto(s)
Butanonas/química , Compuestos Epoxi/farmacología , Glicoles/farmacología , Hemoglobinas/administración & dosificación , Hemoglobinas/química , Compuestos Heterocíclicos con 1 Anillo/administración & dosificación , Cetonas/administración & dosificación , Secuencia de Aminoácidos , Animales , Cromatografía Líquida de Alta Presión , Compuestos Epoxi/química , Glicoles/química , Hemoglobinas/metabolismo , Compuestos Heterocíclicos con 1 Anillo/química , Cetonas/química , Masculino , Datos de Secuencia Molecular , Fragmentos de Péptidos/análisis , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Ionización de Electrospray , Tripsina/metabolismo
14.
J Pharmacol Exp Ther ; 326(3): 809-17, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18552130

RESUMEN

L-methionine (Met) has been implicated in parenteral nutrition-associated cholestasis in infants and, at high levels, it causes liver toxicity by mechanisms that are not clear. In this study, Met toxicity was characterized in freshly isolated male and female mouse hepatocytes incubated with 5 to 30 mM Met for 0 to 5 h. In male hepatocytes, 20 mM Met was cytotoxic at 4 h as indicated by trypan blue exclusion and lactate dehydrogenase leakage assays. Cytotoxicity was preceded by reduced glutathione (GSH) depletion at 3 h without glutathione disulfide formation. Exposure to 30 mM Met resulted in increased cytotoxicity and GSH depletion. It is interesting to note that female hepatocytes were resistant to Met-induced cytotoxicity at these concentrations and showed increased cellular GSH levels compared with hepatocytes exposed to medium alone. The effects of amino-oxyacetic acid (AOAA), an inhibitor of Met transamination, and 3-deazaadenosine (3-DA), an inhibitor of the Met transmethylation pathway enzyme S-adenosylhomocysteine hydrolase, on Met toxicity in male hepatocytes were then examined. Addition of 0.2 mM AOAA partially blocked Met-induced GSH depletion and cytotoxicity, whereas 0.1 mM 3-DA potentiated Met-induced toxicity. Exposure of male hepatocytes to 0.3 mM 3-methylthiopropionic acid (3-MTP), a known Met transamination metabolite, resulted in cytotoxicity and cellular GSH depletion similar to that observed with 30 mM Met, whereas incubations with D-methionine resulted in no toxicity. Female hepatocytes were less sensitive to 3-MTP toxicity than males, which may partially explain their resistance to Met toxicity. Taken together, these results suggest that Met transamination and not transmethylation plays a major role in Met toxicity in male mouse hepatocytes.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Cetoácidos/metabolismo , Metionina/metabolismo , Metionina/toxicidad , Caracteres Sexuales , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Femenino , Glutatión/metabolismo , Masculino , Metionina/antagonistas & inhibidores , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
15.
Drug Metab Dispos ; 36(12): 2468-74, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18775983

RESUMEN

Rats are a common animal model for metabolism and toxicity studies. Previously, the enzymatic properties of rat flavin-containing monooxygenase (FMO) 1 purified from hepatic and renal microsomes and that of FMO3 purified from hepatic microsomes were characterized. This study investigated the physical, immunological, and enzymatic properties of FMO3 purified from male rat kidney microsomes and compared the results with those obtained with isolated rat liver FMO3. Renal FMO3 was purified via affinity columns based on the elution of L-methionine (Met) S-oxidase activity and reactivity of the eluted proteins with human FMO3 antibody. In general, Met S-oxidase-specific activity was increased 100-fold through the purification steps. The resulting protein had similar mobility (approximately 56 kDa) as isolated rat liver FMO3 and cDNA-expressed human FMO3 by SDS-polyacrylamide gel electrophoresis. When the isolated kidney protein band was subjected to trypsin digestion and matrix-assisted laser desorption ionization/time of flight mass spectral analysis, 34% of the sequence of rat FMO3 was detected. The apparent K(m) and V(max) values for rat kidney FMO3 were determined using the known FMO substrates Met, seleno-L-methionine, S-allyl-L-cysteine (SAC), and methimazole (N-methyl-2-mercaptoimidazole). The stereoselectivity of the reactions with Met and SAC were also examined using high-performance liquid chromatography. The obtained kinetic and stereoselectivity results were similar to those we obtained in the present study, or those previously reported, for rat liver FMO3. Taken together, the results demonstrate many similar properties between rat hepatic and renal FMO3 forms and suggest that renal FMO3 may play an important role in kidney metabolism of xenobiotics containing sulfur and selenium atoms.


Asunto(s)
Riñón/química , Microsomas/química , Oxigenasas/química , Oxigenasas/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Catálisis , Cisteína/análogos & derivados , Cisteína/química , Cinética , Masculino , Metimazol/química , Metionina/química , Metionina/metabolismo , Datos de Secuencia Molecular , Peso Molecular , NADP/química , Ratas , Ratas Sprague-Dawley , Selenometionina/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Estereoisomerismo
16.
Drug Metab Dispos ; 36(11): 2252-60, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18687801

RESUMEN

L-methionine-dl-sulfoxide (MetO) is an L-methionine (Met) metabolite, but its role in Met metabolism and toxicity is not clear. In this study, MetO uptake, metabolism to Met, cytotoxicity, and glutathione (GSH) and glutathione disulfide (GSSG) status were characterized in freshly isolated mouse hepatocytes incubated at 37 degrees C with 0 to 30 mM MetO for 0 to 5 h. In male hepatocytes, dose-dependent cytotoxicity concomitant with GSH depletion without GSSG formation occurred after exposure to 20 or 30 mM MetO but not after exposure to 10 mM MetO. Interestingly, female hepatocytes exposed to 30 mM MetO showed no cytotoxicity and exhibited increased intracellular GSH levels compared with control hepatocytes. Male hepatocytes had approximately 2-fold higher levels of intracellular Met-d-O or Met-l-O after MetO (30 mM) exposure for 0 to 1.5 h compared with female hepatocytes. In hepatocytes of both genders, Met-l-O was detected at nearly 5-fold higher levels than Met-d-O, and no significant increase in cellular Met levels was detected. Addition of aminooxyacetic acid (AOAA), an inhibitor of transamination reactions, to MetO-exposed male hepatocytes resulted in higher cellular Met-d-O and Met-l-O levels and decreased the cytotoxicity of MetO. Interestingly, exposure of control male hepatocytes to AOAA selectively increased cellular Met-d-O levels to levels similar to those observed after exposure to MetO (30 mM). Analysis of MetO transamination activity by glutamine transaminase K in mouse liver cytosol revealed similar rates of MetO transamination in cytosol of both genders. Taken together, these results provide evidence for stereoselective oxidation of Met to Met-d-O under physiological conditions and suggest a major role for MetO transamination in MetO metabolism and toxicity.


Asunto(s)
Ácido Aminooxiacético/farmacología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Metionina/análogos & derivados , Caracteres Sexuales , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Femenino , Masculino , Metionina/antagonistas & inhibidores , Metionina/metabolismo , Metionina/toxicidad , Ratones
17.
Chem Res Toxicol ; 21(9): 1716-25, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18681461

RESUMEN

S-(1,2-dichlorovinyl)- L-cysteine sulfoxide (DCVCS), a Michael acceptor produced by an FMO3-mediated oxidation of the trichloroethylene metabolite S-(1,2-dichlorovinyl)- L-cysteine (DCVC), is a more potent nephrotoxicant than DCVC. Because DCVCS incubations with N-acetyl- L-cysteine at pH 7.4, 37 degrees C resulted in the formation of three diastereomeric monoadducts and one diadduct, globin monoadducts and cross-links formed after in vitro incubations of rat erythrocytes with DCVCS (0.9-450 microM) for 2 h and those present at 30 min after in vivo treatment of rats with DCVCS (23 and 230 micromol/kg) were characterized. ESI/MS of intact globin chains revealed adduction of 1 DCVCS moiety on the beta2 chain at the three lowest DCVCS concentrations and on the beta1 chain after the in vivo treatment with 230 micromol/kg DCVCS. Interestingly, intact globin dimers and trimers were detectable by ESI/MS with all DCVCS concentrations in vitro (also by SDS-PAGE) and in vivo. LC/MS and MALDI/FTICR of trypsin digested peptides from globin samples obtained after in vitro (450 microM DCVCS) or in vivo exposure to DCVCS (230 micromol/kg) suggested the formation of DCVCS monoadducts not only with Cys93 and Cys125 of the beta chains but also with Cys13 of the alpha chains, whereas no monoadducted peptides were detected at lower DCVCS concentrations in vitro or in vivo. However, LC/MS and MALDI-TOF/TOF suggested the presence of several DCVCS-derived peptide cross-links both in vivo and in vitro at all DCVCS exposure levels. Collectively, the results indicate at least 4 out of the 5 cysteine moieties of the rat hemoglobin heterodimer may be alkylated by DCVCS, in reactions that could also lead to the formation of multiple cross-links. DCVCS- and N-acetyl-DCVCS (NA-DCVCS)-derived globin cross-links containing GSH and Cys were also detected by mass spectrometry, providing strong evidence for the reactivity and/or cross-linking ability of DCVCS, NA-DCVCS, and their GSH or Cys conjugates in both the in vitro and the in vivo. Thus, hemoglobin adducts and cross-links may be useful biomarkers to investigate the possible presence of DCVCS in circulation after DCVC or trichloroethylene exposure.


Asunto(s)
Cisteína/análogos & derivados , Eritrocitos/efectos de los fármacos , Globinas/análisis , Globinas/química , Safrol/análogos & derivados , Animales , Cromatografía Líquida de Alta Presión , Cisteína/administración & dosificación , Cisteína/química , Cisteína/farmacología , Dimerización , Electroforesis en Gel de Poliacrilamida , Eritrocitos/metabolismo , Hemoglobinas/química , Inyecciones Intraperitoneales , Masculino , Espectrometría de Masas , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Safrol/administración & dosificación , Safrol/química , Safrol/farmacología , Sensibilidad y Especificidad , Tripsina/química
18.
Expert Opin Drug Discov ; 13(9): 815-824, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30101640

RESUMEN

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.


Asunto(s)
Antineoplásicos/administración & dosificación , Mercaptopurina/administración & dosificación , Tioguanina/administración & dosificación , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/metabolismo , Diseño de Fármacos , Glutatión/metabolismo , Glutatión Transferasa/metabolismo , Humanos , Riñón/metabolismo , Mercaptopurina/efectos adversos , Mercaptopurina/metabolismo , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Profármacos , Tioguanina/efectos adversos , Tioguanina/metabolismo
19.
Biochem Pharmacol ; 73(11): 1829-41, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17433263

RESUMEN

cis-6-(2-Acetylvinylthio)purine (cAVTP) and trans-6-(2-acetylvinylthio)guanine (tAVTG) are thiopurine prodrugs provisionally inactivated by an alpha,beta-unsaturated substituent on the sulfur of the parental thiopurines 6-mercaptopurine (6-MP) and 6-thioguanine (6-TG). The active thiopurines are liberated intracellularly by glutathione (GSH) in reactions catalyzed by glutathione transferases (GSTs) (EC 2.5.1.18). Catalytic activities of 13 human GSTs representing seven distinct classes of soluble GSTs have been determined. The bioactivation of cAVTP and tAVTG occurs via a transient addition of GSH to the activated double bond of the S-substituent of the prodrug, followed by elimination of the thiopurine. The first of these consecutive reactions is rate-limiting for thiopurine release, but GST-activation of this first addition is shifting the rate limitation to the subsequent elimination. Highly active GSTs reveal the transient intermediate, which is detectable by UV spectroscopy and HPLC analysis. LC/MS analysis of the reaction products demonstrates that the primary GSH conjugate, 4-glutathionylbuten-2-one, can react with a second GSH molecule to form the 4-(bis-glutathionyl)butan-2-one. GST M1-1 and GST A4-4 were the most efficient enzymes with tAVTG, and GST M1-1 and GST M2-2 had highest activity with cAVTP. The highly efficient GST M1-1 is polymorphic and is absent in approximately half of the human population. GST P1-1, which is overexpressed in many cancer cells, had no detectable activity with cAVTP and only minor activity with tAVTG. Other GST-activated prodrugs have targeted GST P1-1-expressing cancer cells. Tumors expressing high levels of GST M1-1 or GST A4-4 can be predicted to be particularly vulnerable to chemotherapy with cAVTP or tAVTG.


Asunto(s)
Antineoplásicos/metabolismo , Glutatión Transferasa/metabolismo , Profármacos/metabolismo , Azatioprina/metabolismo , Catálisis , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Glutatión/metabolismo , Humanos , Cinética , Espectrometría de Masas , Mercaptopurina/metabolismo , Espectrofotometría Ultravioleta , Tioguanina/metabolismo
20.
Chem Biol Interact ; 166(1-3): 176-81, 2007 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-16735035

RESUMEN

Hydroxymethylvinyl ketone (HMVK) is a reactive oxidation product of 3-butene-1,2-diol, a metabolite of 1,3-butadiene. The potential for HMVK (0.1 and 1mM) to form hemoglobin (Hb) adducts in erythrocytes from Sprague-Dawley rats was investigated at physiological conditions (pH 7.4, 37 degrees C) using electrospray ionization mass spectrometry (ESI/MS). With the 0.1mM HMVK globin samples, the results indicate HMVK adduction on the alpha2, beta2 and beta3 chains. With the 1.0mM HMVK globin samples, adducts were detected on the beta2 and beta3 chains. However, no correlation was observed between incubation time and the extent of adduct formation, and additional adducts were detected when globin samples were fractionated by HPLC before the ESI/MS analyses. For specific localizations of adducts on the globin chains, trypsin digested peptides from the 1mM HMVK globin samples were subjected to liquid chromatography/mass spectrometry analyses. The results, which are consistent with formation of HMVK adducts on several specific peptides within the alpha- and beta-chains, suggest selectivity in the interaction of HMVK with the different cysteine residues in Hb. Because adducts were also detected in peptides containing no cysteine residues and multiple HMVK moieties were detected on some of the cysteine-containing peptides, the results suggest other amino acids may be also reactive with HMVK. Adduct profiles and their relative intensities were consistent between the 1 and 2h samples providing evidence for the HMVK reactions being fast and selective. The finding that fewer peptides were adducted in the 0.1mM HMVK globin samples provides further evidence for selectivity of the HMVK reaction. Collectively, the results show HMVK readily and selectively forms adducts on Hb. Characterization of these adducts will facilitate development of useful biomarkers of exposure to HMVK and its precursor 1,3-butadiene.


Asunto(s)
Butanonas/toxicidad , Eritrocitos/efectos de los fármacos , Hemoglobinas/análisis , Espectrometría de Masa por Ionización de Electrospray , Secuencia de Aminoácidos , Animales , Butanonas/análisis , Butanonas/química , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Eritrocitos/química , Hemoglobinas/química , Masculino , Datos de Secuencia Molecular , Péptidos/química , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda