Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nature ; 475(7354): 106-9, 2011 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-21734707

RESUMO

Reactive oxygen species (ROS) are mutagenic and may thereby promote cancer. Normally, ROS levels are tightly controlled by an inducible antioxidant program that responds to cellular stressors and is predominantly regulated by the transcription factor Nrf2 (also known as Nfe2l2) and its repressor protein Keap1 (refs 2-5). In contrast to the acute physiological regulation of Nrf2, in neoplasia there is evidence for increased basal activation of Nrf2. Indeed, somatic mutations that disrupt the Nrf2-Keap1 interaction to stabilize Nrf2 and increase the constitutive transcription of Nrf2 target genes were recently identified, indicating that enhanced ROS detoxification and additional Nrf2 functions may in fact be pro-tumorigenic. Here, we investigated ROS metabolism in primary murine cells following the expression of endogenous oncogenic alleles of Kras, Braf and Myc, and found that ROS are actively suppressed by these oncogenes. K-Ras(G12D), B-Raf(V619E) and Myc(ERT2) each increased the transcription of Nrf2 to stably elevate the basal Nrf2 antioxidant program and thereby lower intracellular ROS and confer a more reduced intracellular environment. Oncogene-directed increased expression of Nrf2 is a new mechanism for the activation of the Nrf2 antioxidant program, and is evident in primary cells and tissues of mice expressing K-Ras(G12D) and B-Raf(V619E), and in human pancreatic cancer. Furthermore, genetic targeting of the Nrf2 pathway impairs K-Ras(G12D)-induced proliferation and tumorigenesis in vivo. Thus, the Nrf2 antioxidant and cellular detoxification program represents a previously unappreciated mediator of oncogenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Oncogenes/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Alelos , Animais , Antioxidantes/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Células Cultivadas , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/metabolismo , Genes myc/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Sistema de Sinalização das MAP Quinases , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/genética , Células NIH 3T3 , Oxirredução , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-24530239

RESUMO

Interleukin-1ß (IL-1ß) is a pro-inflammatory cytokine. It induces the synthesis of prostaglandin E2 (PGE2) catalyzed by cyclooxygenase (COX) and microsomal prostaglandin E synthase (m-PGES). Besides its pro-inflammatory properties, PGE2 also exhibits anti-inflammatory properties by inhibiting synthesis of 5-lipooxygenase (5-LO) products which are in themselves, pro-inflammatory mediators. Thus, inhibition of 5-LO products is beneficial in regulating immune-responses and pro-inflammatory processes. To investigate the hypothesis that IL-1ß is responsible for the increase in the synthesis of PGE2 and in the reduction of 5-LO products, equine whole blood (EWB) was treated with lipopolysaccharide (LPS). In vitro treatment of EWB with LPS resulted in increased expression of IL-1ß while expression of 5-LO was suppressed. Quantification of eicosanoids using liquid-chromatography-mass spectrometry/multiple reaction monitoring (LC-MS/MRM) showed increased concentrations of prostaglandins and decreased 5-LO products in LPS-treated EWB. Pretreatment of EWB with IL-1ß followed by calcium ionophore A23187 (CI) reduced synthesis of 5-LO products. However, pretreatment of EWB with COX-2 inhibitor (NS-398) or m-PGES-1 inhibitor (CAY 10526) and IL-1ß followed with CI resulted in a significant (p<0.0001) increase in 5-LO products. Pretreatment of EWB with phospholipase C inhibitor (U73122) followed with LPS reduced PGE2 production but increased 5-LO products. The result of this study indicated that increased PGE2 production led to reduction in 5-LO products in LPS-treated EWB via IL-1ß. However, other pathways, cytokines and mediators may be involved in inhibiting 5-LO products but the present study did not include those other potential pathways. Inhibition of 5-LO products by PGE2 in EWB may regulate the initiation and pathogenesis of inflammatory responses in the horse.


Assuntos
Araquidonato 5-Lipoxigenase/biossíntese , Interleucina-1beta/fisiologia , Lipopolissacarídeos/farmacologia , Animais , Araquidonato 5-Lipoxigenase/genética , Ionóforos de Cálcio/farmacologia , Eicosanoides/biossíntese , Eicosanoides/sangue , Repressão Enzimática , Estrenos/farmacologia , Cavalos , Pirrolidinonas/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores
3.
Rapid Commun Mass Spectrom ; 25(5): 585-98, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21290445

RESUMO

Oxidative metabolites of arachidonic acid (AA) are implicated in inflammation. Thus, we evaluated cycloxygenases (COXs) and lipoxygenases (LOs) mediated metabolism of AA to eicosanoids in equine plasma. Eicosanoids were extracted from plasma by two liquid-liquid extraction (LLE) steps; first was by chloroform/isopropanol and second by methyl-tert-butyl ether. For identification and quantification of 25 eicosanoids, a highly specific, selective and sensitive stable isotope dilution liquid chromatography (LC) multiple reaction monitoring (MRM) mass spectrometric (MS) method was developed. To avoid artifact formation of eicosanoids, deferoxamine was added to plasma to chelate residual transition metal ions. The calibration curve showed excellent linearity within 0.1 to 10 ng/mL. Slopes of the calibration curves generated by adding known quantities of eicosanoids in plasma were higher than those prepared in methanol/mobile phase A. Addition of deferoxamine decreased the slope of calibration curves generated using plasma. Limit of detection (LOD) was 1-10 pg on-column for 25 different eicosanoids. Inter-day accuracy was 86-111%, whereas intra-day accuracy was from 88-110%, and precision did not exceed 15% for all quality control (QC) samples. To evaluate the formation of eicosanoids, AA was exogenously added or endogenous AA was released from esterified lipids by calcium ionophore (CI) A23187 treatment of equine whole blood. Pre-treatment of equine whole blood with dexamethasone (DEX) significantly inhibited AA or CI A23187- mediated formation of eicosanoids. The validated method is now employed in studies undertaken to better understand the mechanism of action and pharmacokinetics/pharmacodynamics of eicosanoids after administration of glucocorticoids to horses. This method is reliably reproducible.


Assuntos
Cromatografia de Fase Reversa/métodos , Eicosanoides/sangue , Marcação por Isótopo/métodos , Espectrometria de Massas/métodos , Animais , Calibragem , Fracionamento Químico , Eicosanoides/metabolismo , Cavalos , Modelos Lineares , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
4.
Proc Natl Acad Sci U S A ; 105(19): 6846-51, 2008 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-18474869

RESUMO

Polycyclic aromatic hydrocarbons (PAHs) are tobacco carcinogens implicated in the causation of human lung cancer. Metabolic activation is a key prerequisite for PAHs to cause their deleterious effects. Using human lung adenocarcinoma (A549) cells, we provide evidence for the metabolic activation of (+/-)-trans-7,8dihydroxy-7,8-dihydrobenzo[a]pyrene (B[a]P-7,8-trans-dihydrodiol) by aldo-keto reductases (AKRs) to yield benzo[a]pyrene-7,8-dione (B[a]P-7,8-dione), a redox-active o-quinone. We show that B[a]P-7,8-trans-dihydrodiol (AKR substrate) and B[a]P-7,8-dione (AKR product) lead to the production of intracellular reactive oxygen species (ROS) (measured as an increase in dichlorofluorescin diacetate fluores-cence) and that similar changes were not observed with the regioisomer (+/-)-trans-4,5-dihydroxy-4,5-dihydrobenzo[a]pyrene or the diol-epoxide, (+/-)-anti-7,8-dihydroxy-9alpha,10beta-epoxy-7,8,9,10-tetrahydro-B[a]P. B[a]P-7,8-trans-dihydrodiol and B[a]P-7,8-dione also caused a decrease in glutathione levels and an increase in NADP(+)/NADPH ratios, with a concomitant increase in single-strand breaks (as measured by the comet assay) and 7,8-dihydro-8-oxo-2'-deoxyguanosine (8-oxo-dGuo). The specificity of the comet assay was validated by coupling it to human 8-oxo-guanine glycosylase (hOGG1), which excises 8-oxo-Gua to yield single-strand breaks. The levels of 8-oxo-dGuo observed were confirmed by an immunoaffinity purification stable isotope dilution ([(15)N(5)]-8-oxo-dGuo) liquid chromatography-electrospray ionization/multiple reaction monitoring/mass spectrometry (LC-ESI/MRM/MS) assay. B[a]P-7,8-trans-dihydrodiol produced DNA strand breaks in the hOGG1-coupled comet assay as well as 8-oxo-dGuo (as measured by LC-ESI/MRM/MS) and was enhanced by a catechol O-methyl transferase (COMT) inhibitor, suggesting that COMT protects against o-quinone-mediated redox cycling. We conclude that activation of PAH-trans-dihydrodiols by AKRs in lung cells leads to ROS-mediated genotoxicity and contributes to lung carcinogenesis.


Assuntos
Oxirredutases do Álcool/metabolismo , Di-Hidroxi-Di-Hidrobenzopirenos/metabolismo , Pulmão/enzimologia , 8-Hidroxi-2'-Desoxiguanosina , Aldeído Redutase , Aldo-Ceto Redutases , Benzopirenos/farmacologia , Biotransformação/efeitos dos fármacos , Inibidores de Catecol O-Metiltransferase , Linhagem Celular Tumoral , Ensaio Cometa , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , DNA Glicosilases/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Di-Hidroxi-Di-Hidrobenzopirenos/farmacologia , Inibidores Enzimáticos/farmacologia , Fluoresceínas/metabolismo , Fluorescência , Humanos , Isoenzimas/metabolismo , Pulmão/patologia , Oxirredução/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização por Electrospray
5.
J Biol Chem ; 284(43): 29725-34, 2009 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-19726680

RESUMO

Polycyclic aromatic hydrocarbon (PAH) o-quinones produced by aldo-keto reductases are ligands for the aryl hydrocarbon receptor (AhR) (Burczynski, M. E., and Penning, T. M. (2000) Cancer Res. 60, 908-915). They induce oxidative DNA lesions (reactive oxygen species-mediated DNA strand breaks and 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dGuo) formation) in human lung cells. We tested whether the AhR enhances PAH o-quinone-mediated oxidative DNA damage by translocating these ligands to the nucleus. Using the single cell gel electrophoresis (comet) assay to detect DNA strand breaks in murine hepatoma Hepa1c1c7 cells and its AhR- and aryl hydrocarbon receptor nuclear translocator-deficient variants, benzo[a]pyrene-7,8-dione (B[a]P-7,8-dione) produced fewer DNA strand breaks in AhR-deficient cells compared with aryl hydrocarbon receptor nuclear translocator-deficient and wild type Hepa1c1c7 cells. Decreased DNA strand breaks were also observed in human bronchoalveolar H358 cells in which the AhR was silenced by siRNA. The antioxidant alpha-tocopherol and the iron chelator/antioxidant desferal decreased the formation of B[a]P-7,8-dione-mediated DNA strand breaks indicating that they were reactive oxygen species-dependent. By coupling the comet assay to 8-oxoguanine glycosylase (hOGG1), which excises 8-oxo-Gua, strand breaks dependent upon this lesion were measured. hOGG1 treatment produced more DNA single strand breaks in B[a]P-7,8-dione-treated Hepa cells and H358 cells than in its absence. The levels of hOGG1-dependent DNA strand breaks mediated by B[a]P-7,8-dione were lower in AhR-deficient Hepa and AhR knockdown H358 cells. The AhR antagonist alpha-naphthoflavone also attenuated B[a]P-7,8-dione-mediated DNA strand breaks. The decrease in 8-oxo-dGuo levels in AhR-deficient Hepa cells and AhR knockdown H358 cells was validated by immunoaffinity capture stable isotope dilution ([(15)N(5)]8-oxo-dGuo) liquid chromatography-electrospray ionization/multiple reaction monitoring/mass spectrometry. We conclude that the AhR shuttles PAH o-quinone genotoxins to the nucleus and enhances oxidative DNA damage.


Assuntos
Benzopirenos/farmacologia , Quebras de DNA/efeitos dos fármacos , Desoxiguanosina/análogos & derivados , Espécies Reativas de Oxigênio/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Animais , Antioxidantes/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Linhagem Celular , Linhagem Celular Tumoral , DNA Glicosilases/genética , DNA Glicosilases/metabolismo , Desferroxamina/farmacologia , Desoxiguanosina/genética , Desoxiguanosina/metabolismo , Humanos , Camundongos , Receptores de Hidrocarboneto Arílico/genética , Sideróforos/farmacologia , alfa-Tocoferol/farmacologia
6.
Chem Res Toxicol ; 22(5): 788-97, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19309085

RESUMO

Analysis of cellular 7,8-dihydro-8-oxo-2'-deoxyguanosine (8-oxo-dGuo) as a biomarker of oxidative DNA damage has been fraught with numerous methodological problems. This is primarily due to artifactual oxidation of dGuo that occurs during DNA isolation and hydrolysis. Therefore, it has become necessary to rely on using the comet assay, which is not necessarily specific for 8-oxo-dGuo. A highly specific and sensitive method based on immunoaffinity purification and stable isotope dilution liquid chromatography (LC)-multiple reaction monitoring (MRM)/mass spectrometry (MS) that avoids artifact formation has now been developed. Cellular DNA was isolated using cold DNAzol (a proprietary product that contains guanidine thiocyanate) instead of chaotropic- or phenol-based methodology. Chelex-treated buffers were used to prevent Fenton chemistry-mediated generation of reactive oxygen species (ROS) and artifactual oxidation of DNA bases. Deferoxamine was also added to all buffers in order to complex any residual transition metal ions remaining after Chelex treatment. The LC-MRM/MS method was used to determine that the basal 8-oxo-dGuo level in DNA from human bronchoalveolar H358 cells was 2.2 +/- 0.4 8-oxo-dGuo/10(7) dGuo (mean +/- standard deviation) or 5.5 +/- 1.0 8-oxo-dGuo/10(8) nucleotides. Similar levels were observed in human lung adenocarcinoma A549 cells, mouse hepatoma Hepa-1c1c7 cells, and human HeLa cervical epithelial adenocarcinoma cells. These values are an order of magnitude lower than is typically reported for basal 8-oxo-dGuo levels in DNA as determined by other MS- or chromatography-based assays. H358 cells were treated with increasing concentrations of potassium bromate (KBrO3) as a positive control or with the methylating agent methyl methanesulfonate (MMS) as a negative control. A linear dose-response for 8-oxo-dGuo formation (r(2) = 0.962) was obtained with increasing concentrations of KBrO3 in the range of 0.05 mM to 2.50 mM. In contrast, no 8-oxo-dGuo was observed in H358 cell DNA after treatment with MMS. At low levels of oxidative DNA damage, there was an excellent correlation between a comet assay that measured DNA single strand breaks (SSBs) after treatment with human 8-oxo-guanine glycosylase-1 (hOGG1) when compared with 8-oxo-dGuo in the DNA as measured by the stable isotope dilution LC-MRM/MS method. Availability of the new LC-MRM/MS assay made it possible to show that the benzo[a]pyrene (B[a]P)-derived quinone, B[a]P-7,8-dione, could induce 8-oxo-dGuo formation in H358 cells. This most likely occurred through redox cycling between B[a]P-7,8-dione and B[a]P-7,8-catechol with concomitant generation of DNA damaging ROS. In keeping with this concept, inhibition of catechol-O-methyl transferase (COMT)-mediated detoxification of B[a]P-7,8-catechol with Ro 410961 caused increased 8-oxo-dGuo formation in the H358 cell DNA.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Dano ao DNA , Desoxiguanosina/análogos & derivados , Estresse Oxidativo , Espectrometria de Massas por Ionização por Electrospray/métodos , 8-Hidroxi-2'-Desoxiguanosina , Animais , Linhagem Celular Tumoral , Cromatografia de Afinidade , Ensaio Cometa , Desoxiguanosina/análise , Desoxiguanosina/imunologia , Desoxiguanosina/isolamento & purificação , Humanos , Camundongos
7.
Eur J Pharmacol ; 736: 1-9, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-24751711

RESUMO

Glucocorticoids (corticosteroids) are widely used anti-inflammatory agents in veterinary medical practice. These drugs are considered doping agents because they mask pain and thus, increase injury potential in equine athletes. They exhibit anti-inflammatory property by binding to glucocorticoids receptor (GR) to control the transcription of pro- and anti-inflammatory cytokines and enzymes involved in the synthesis of bioactive eicosanoids. To evaluate the role of triamcinolone acetonide (TA) on concentrations of bioactive eicosanoids in equine plasma, TA (0.04 mg/kg) was intravenously administered to horses. Before (0 h) and after TA administration, equine whole blood (EWB) samples were collected and challenged with either methanol (vehicle), calcium ionophore A-23187 (CI) or lipopolysaccharide (LPS) to stimulate ex-vivo synthesis of eicosanoids. Plasma concentrations of eicosanoids were quantified using LC-MS/MRM. Results showed that thromboxane B2 (TXB2) was not affected by TA administration when EWB was stimulated with CI. However, after LPS treatment, TXB2, PGE2, PGF2α and 15-(s)-HETE decreased during 2-8 h post-TA administration but recovered to concentrations which were not significantly different from those of pre-TA administration (0 h), after 24 h. When EWB was treated with CI, LTB4 was suppressed post-TA administration compared to 0 h. When EWB collected after TA administration was stimulated with LPS, LTB4 was not significantly different from those of 0 h. Administration of a therapeutic dose of TA (0.04 mg/kg, iv) in the horse suppressed biosynthesis of bioactive eicosanoids indicating the anti-inflammatory role of TA in the horse.


Assuntos
Anti-Inflamatórios/farmacologia , Eicosanoides/antagonistas & inibidores , Glucocorticoides/farmacologia , Triancinolona Acetonida/farmacologia , Animais , Anti-Inflamatórios/sangue , Anti-Inflamatórios/farmacocinética , Calcimicina/farmacologia , Eicosanoides/sangue , Eicosanoides/metabolismo , Glucocorticoides/sangue , Glucocorticoides/farmacocinética , Cavalos , Lipopolissacarídeos/farmacologia , Metanol/farmacologia , Triancinolona Acetonida/sangue , Triancinolona Acetonida/farmacocinética
8.
Chem Res Toxicol ; 21(3): 668-77, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18251511

RESUMO

This study demonstrates that benzo[g]chrysene-11,12-dihydrodiol (B[g]C-11,12-dihydrodiol) derived from the fjord-region parent hydrocarbon B[g]C is oxidized by rat AKR1C9 with a k c a t/ K m 100 times greater than that observed with the commonly studied bay-region benzo[ a]pyrene-7,8-dihydrodiol (B[a]P-7,8-dihydrodiol). Conversely, despite its strikingly similar structure to B[ g]C-11,12-dihydrodiol, benzo[ c]phenanthrene-3,4-dihydrodiol (B[ c]Ph-3,4-dihydrodiol) is consumed by AKR1C9 at sluggish rates comparable to those observed with B[ a]P-7,8-dihydrodiol. CD spectroscopy revealed that only the (+)-B[ g]C-11,12-dihydrodiol stereoisomer was oxidized, while AKR1C9 oxidized both stereoisomers of B[a]P-7,8-dihydrodiol and B[ c]Ph-3,4-dihydrodiol. The (+)- S, S- and (-)- R, R-stereoisomers of B[g]C-11,12-dihydrodiol were purified by chiral RP-HPLC. The 11 S,12 S-stereoisomer was oxidized at the same rate as the racemate. The 11 R,12 R-stereoisomer did not act as an inhibitor to AKR1C9, indicating that the (-)- R, R-stereoisomer was excluded from the active site. To understand the basis of stereochemical preference, we screened alanine-scanning mutants of active site residues of AKR1C9. These studies revealed that in comparison to the wild type, F129A, W227A, and Y310A enabled the oxidation of both the B[g]C-11 S,12 S-dihydrodiol and the B[g]C-11 R,12 R-dihydrodiol. Molecular modeling revealed that unlike B[a]P-7,8-dihydrodiol and B[ c]Ph-3,4-dihydrodiol, B[g]C-11,12-dihydrodiol enantiomers are significantly bent out of plane. As a consequence, the (-)- R, R-stereoisomer was prevented from binding to the active site because of unfavorable interactions with F129, W227, or Y310. Additionally, LC/MS validated that the product of the reaction of B[g]C-11,12-dihydrodiol oxidation catalyzed by AKR1C9 was B[g]C-11,12-dione, which was trapped in vitro with the nucleophile 2-mercaptoethanol. The similarity between rates of trans-dihydrodiol oxidation by the rat and human liver specific AKRs (AKR1C9 and AKR1C4) implicate these enzymes in hepatocarcinogenesis in rats observed with the fjord-region PAH.


Assuntos
Crisenos/metabolismo , Fígado/enzimologia , Oxirredutases/metabolismo , Fenantrenos/metabolismo , Alanina/genética , Oxirredutases do Álcool/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Crisenos/química , Dicroísmo Circular , Humanos , Técnicas In Vitro , Indicadores e Reagentes , Cinética , Fígado/metabolismo , Espectrometria de Massas , Mercaptoetanol/metabolismo , Modelos Moleculares , Mutação/genética , Oxirredutases/genética , Fenantrenos/química , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Estereoisomerismo , Relação Estrutura-Atividade , Especificidade por Substrato
9.
Chem Res Toxicol ; 20(9): 1331-41, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17702526

RESUMO

Benzo[ a]pyrene (B[ a]P), a representative polycyclic aromatic hydrocarbon (PAH), is metabolically activated by three enzymatic pathways: by peroxidases (e.g., cytochrome P450 peroxidase) to yield radical cations, by P4501A1/1B1 monooxygenation and epoxide hydrolase to yield diol epoxides, and by P4501A1/1B1 monooxygenation, epoxide hydrolase, and aldo-keto reductases (AKRs) to yield o-quinones. In humans, a major exposure site for environmental and tobacco smoke PAH is the lung; however, the profile of B[ a]P metabolites formed at this site has not been well characterized. In this study, human bronchoalveolar H358 cells were exposed to B[ a]P, and metabolites generated by peroxidase (B[ a]P-1,6- and B[ a]P-3,6-diones), from cytochrome P4501A1/1B1 monooxygenation [3-hydroxy-B[ a]P, B[ a]P-7,8- and 9,10- trans-dihydrodiols, and B[ a]P- r-7, t-8, t-9, c-10-tetrahydrotetrol (B[ a]P-tetraol-1)], and from AKRs (B[ a]P-7,8-dione) were detected and quantified by RP-HPLC, with in-line photo-diode array and radiometric detection, and identified by liquid chromatography-mass spectrometry (LC-MS). Progress curves showed a lag phase in the formation of 3-hydroxy-B[ a]P, B[ a]P-7,8- trans-dihydrodiol, B[ a]P-tetraol-1, and B[ a]P-7,8-dione over 24 h. Northern blot analysis showed that B[ a]P induced P4501B1 and AKR1C isoforms in H358 cells in a time-dependent manner, providing an explanation for the lag phase. Pretreatment of H358 cells with 10 nM 2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD) eliminated this lag phase but did not alter the levels of the individual metabolites observed, suggesting that both B[ a]P and TCDD induction ultimately yield the same B[ a]P metabolic profile. The one exception was B[ a]P-3,6-dione which was formed without a lag phase in the absence and presence of TCDD, suggesting that the peroxidase responsible for its formation was neither P4501A1 nor 1B1. Candidate peroxidases that remain include PGH synthases and uninduced P450 isoforms. This study shows that the P4501A1/1B1 and AKR pathways are inducible in human lung cells and that the peroxidase pathway was not. It also provides evidence that each of the pathways of PAH activation yields their distinctive metabolites in H358 human lung cells and that each pathway may contribute to the carcinogenic process.


Assuntos
Benzo(a)pireno/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , 20-Hidroxiesteroide Desidrogenases/metabolismo , Hidrocarboneto de Aril Hidroxilases/metabolismo , Benzo(a)pireno/química , Benzo(a)pireno/toxicidade , Linhagem Celular Tumoral , Cromatografia Líquida , Citocromo P-450 CYP1B1 , Humanos , Espectrometria de Massas , Estrutura Molecular , Peroxidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA