Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Ethnopharmacol ; 267: 113537, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33137430

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Aristolochic acid nephropathy (AAN) is a kidney disease caused by the administration of plants containing aristolochic acids (AAs). Aristolochic acid I (AAI) is the main toxic component in AAs. Organic anion transporters (OATs) 1 and 3 mediate the renal uptake of AAI, which is related to AAN. In our previous study, we found that anthraquinones derived from the herbal medicine Rheum palmatum L. (RP) inhibited both OAT1 and OAT3, with rhein exhibiting the greatest potency among the components. AIM OF THE STUDY: This study aimed to investigate the effects of rhein and RP extract on the pharmacokinetics and tissue distribution of AAI and its demethylated metabolite (8-hydroxy-aristolochic acid I [AAIa]) in rats. MATERIALS AND METHODS: Rhein and RP extract were used as OAT inhibitors, and AAI was used as the toxic substrate. The pharmacokinetics and tissue distribution of AAI and AAIa in rats following the intravenous injection of AAI (10 mg/kg) in the presence and absence of rhein (100 mg/kg) or RP extract (5 g crude drug/kg) were investigated. RESULTS: Co-administration with rhein increased AUC0-∞ of AAI and AAIa by 39 and 44%, respectively. However, the renal level of AAI was decreased to 50, 42, and 58% of those in rats treated with AAI alone at 5, 10, and 20 min after treatment, respectively, and the renal level of AAIa was decreased to 58, 57, and 61% of the level in rats treated with AAI alone, respectively, at these time points. In the RP extract co-administration group, AAI and AAIa plasma exposure was not significantly increased, but renal accumulation of AAI was decreased to 63, 58, and 68% of that in rats treated with AAI alone at 5, 10, and 20 min after treatment, respectively. In addition, renal accumulation of AAIa was decreased to 74, 70, and 70% of that in rats treated with AAI alone at 5, 10, and 20 min after treatment, respectively. CONCLUSIONS: This study indicated that co-administration with rhein significantly increased the plasma exposure of AAI and AAIa while decreased their renal accumulation in rats. RP extract reduced the renal accumulation of AAI and AAIa, but have no significant effect on their plasma exposure levels in rats.


Assuntos
Antraquinonas/farmacologia , Ácidos Aristolóquicos/farmacocinética , Proteína 1 Transportadora de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos Sódio-Independentes/antagonistas & inibidores , Extratos Vegetais/farmacologia , Rheum , Animais , Antraquinonas/isolamento & purificação , Ácidos Aristolóquicos/administração & dosagem , Ácidos Aristolóquicos/sangue , Ácidos Aristolóquicos/toxicidade , Biotransformação , Desmetilação , Injeções Intravenosas , Rim/metabolismo , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Nefropatias/prevenção & controle , Masculino , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Extratos Vegetais/isolamento & purificação , Ratos Sprague-Dawley , Rheum/química , Distribuição Tecidual
2.
Chem Res Toxicol ; 33(6): 1374-1381, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32048847

RESUMO

There is accumulating evidence that Balkan endemic nephropathy (BEN) is an environmental disease caused by aristolochic acids (AAs) released from the decomposition of Aristolochia clematitis L., an AA-containing weed that grows abundantly in the Balkan Peninsula. AA exposure has also been associated with carcinoma development in the upper urinary tract of some patients suffering from BEN. It is believed that an aristolactam-nitrenium ion intermediate with a delocalized positive charge produced in the hepatic metabolism of AAs binds to DNA and the resulting DNA adduct is responsible for initiating the carcinoma development process. In this study, we demonstrated for the first time that the aristolactam-nitrenium ion intermediate will also react with endogenous aminothiols, for example, cysteine, N-acetylcysteine, and glutathione in vitro, and in rats, producing phase II-conjugated metabolites in a dosage-dependent manner. It is highly possible that this conjugation process consumes and ultimately deactivates this carcinogenic intermediate and acts as an important, but previously unreported, detoxification mechanism of AAs. Results also showed AAs, phase I metabolites, and the aminothiol-conjugated metabolites are rapidly eliminated from AA-exposed rats. Furthermore, we found evidence that AA exposure induced oxidative stress in rats, as indicated by the glutathione depletion in rat serum samples.


Assuntos
Ácidos Aristolóquicos/farmacocinética , Ácidos Aristolóquicos/toxicidade , Cisteína/metabolismo , Glutationa/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Ácidos Aristolóquicos/sangue , Glutationa/sangue , Inativação Metabólica , Masculino , Ratos Sprague-Dawley
3.
Anal Chem ; 91(13): 8154-8161, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31140784

RESUMO

Aristolochic acids (AAs) contained in herbal plants are implicated in multiple organ injuries and have a high mutational burden in upper tract urothelial cancers. The currently available techniques for monitoring AAs include LC (liquid chromatography) and LC/MS (mass spectrometry), but the application of these approaches are limited due to the complex sample preparation and derivatization steps. Therefore, there is an urgent need to develop efficient methods for identifying and quantifying AAs. Here, we present a new dual-spectroscopic approach for the direct detection of AAs from blood and tissue samples; the detection of aristolochic acid I (AAI) is performed by surface-enhanced Raman spectroscopy (SERS), and its bioproduct, aristololactam (AAT), is detected by fluorescence spectroscopy based on their distinctive spectral response. Furthermore, a graphene assisted enrichment coupled with a magnetic retrieval strategy was developed to enhance SERS sensitivity toward AAI. Our method was successfully applied to directly determine both AAI and AAT from the blood, liver, and kidney of rats. The potential for real-world application was demonstrated by continuously monitoring AAI and AAT in rat blood and tissues after AAI feeding. The results showed that AAI was gradually metabolized to AAT and transported to different organs. It was found that the metabolism of AAI took place in the kidney, but AAT residue was detected in both liver and kidney, which might be related to long-term toxicity and gene mutation. The proposed dual-spectroscopic strategy is applicable to long-term toxicology research and to the direct diagnosis of AAI-induced organ injury.


Assuntos
Ácidos Aristolóquicos/farmacocinética , Espectrometria de Fluorescência/métodos , Análise Espectral Raman/métodos , Animais , Ácidos Aristolóquicos/sangue , Rim/metabolismo , Fígado/metabolismo , Masculino , Modelos Moleculares , Ratos Sprague-Dawley , Distribuição Tecidual
4.
BMC Complement Altern Med ; 17(1): 107, 2017 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-28187732

RESUMO

BACKGROUND: Aristolochiae Fructus (AF) and honey-fried Aristolochiae Fructus (HAF) have been used in China for a long time as anti-tussive and expectorant drugs. Few clinical cases have been reported to be associated with the toxicity of AF and HAF, although relatively high amounts of aristolochic acids (AAs) have been found in them. Our previous experiments have verified from the chemical changes and from traditional toxicology that honey-processing can significantly reduce the toxicity of AF. To further elucidate the detoxification mechanism of honey-processing, comparative pharmacokinetics of AAs in AF and HAF are performed in this study. METHODS: An HPLC-MS/MS (high-performance liquid chromatography-tandem mass spectrometry) method was developed and validated for the determination of AA I, AA II, AA C, AA D and 7-OH AA I in rat plasma. The multi-component pharmacokinetics of AAs in AF and HAF extracts were investigated after the oral administration of three doses to rats. The relative pharmacokinetic parameters were compared systematically. RESULTS: The five AAs shared a similar nonlinear PK (pharmacokinetic) process. They involve rapid absorption and elimination, and they were fit into a two-compartmental open model. Some significant pharmacokinetic differences were observed between the AF and HAF groups: the C max and AUC values of AA I and AA II in the AF groups were much higher than those of the HAF groups. CONCLUSIONS: Honey-frying technology can reduce the toxicity of AF by significantly decreasing the absorption of AA I and AA II. The PK parameters obtained in this work could provide valuable references for the toxicity research and clinical use of Aristolochiaceae herbs, including AF and HAF. Process diagram of comparative pharmacokinetics study.


Assuntos
Aristolochia/química , Ácidos Aristolóquicos/farmacocinética , Frutas/química , Mel , Extratos Vegetais/farmacocinética , Administração Oral , Animais , Ácidos Aristolóquicos/sangue , Ácidos Aristolóquicos/química , Limite de Detecção , Modelos Lineares , Masculino , Extratos Vegetais/administração & dosagem , Extratos Vegetais/química , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
5.
Mol Med Rep ; 14(4): 3243-50, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27510522

RESUMO

The genotoxicity and cytotoxicity of aristolochic acids is well documented, and the Aristolochiaceae plant family has been widely used in China and India for medical purposes. However, the mechanisms of aristolochic acid I (AAI) in treatment and toxicity remain to be fully elucidated. According to the theory of traditional Chinese medicine (TCM), the spleen is responsible for transportation and transformation, in which a substance is transformed, absorbed and distributed in the body. In the present study, rats were randomized into a blank group without spleen deficiency and a spleen deficiency group to investigate the metabolism of AAI. The results showed that the concentration of AAI was higher in the spleen deficiency group, compared with that of the blank group. To further elucidate this process, the expression of organic anion transporting peptide (oatp)2a1 in the rats of the two groups were examined following oral administration of AAI. It was observed that the mRNA level of oatp2a1 in the small intestine of the blank+AAI 60 min group was downregulated, compared with that in the blank group. Compared with the mRNA level of oatp2a1 in the spleen deficiency group, the expression levels in the lung and liver were downregulated in the spleen deficiency+AAI 5 min group, whereas expression levels in the kidney in the spleen deficiency+AAI 60 min group were upregulated. Based on the above results, it was hypothesized that the expression of oatp2a1 may be one of the mechanisms of AAI metabolism in rats. In TCM, the spleen and certain functions of the small intestine, are important in AAI metabolism, and affect the toxicity of AAI. In addition, the lung, liver and kidney may also be involved in spleen deficiency syndrome in rats.


Assuntos
Ácidos Aristolóquicos/metabolismo , Medicamentos de Ervas Chinesas/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Baço/metabolismo , Animais , Ácidos Aristolóquicos/administração & dosagem , Ácidos Aristolóquicos/química , Ácidos Aristolóquicos/farmacocinética , Asarum/química , Medicamentos de Ervas Chinesas/administração & dosagem , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacocinética , Masculino , Medicina Tradicional Chinesa , Transportadores de Ânions Orgânicos/análise , Transportadores de Ânions Orgânicos/genética , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
6.
J Pharm Pharmacol ; 68(7): 883-9, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27166429

RESUMO

OBJECTIVE: The purpose of this study was to determine whether quercetin decreases the uptake of aristolochic acid I (AAI) from the apical membranes of Caco-2 cells via H(+) -linked MCTs at neutral pH as well as to confirm the secretion of AAI through the Caco-2 cell monolayers via ABC transporters. METHODS: Caco-2 cells cultured on the dishes or permeable membranes were incubated with AAI in the absence or presence of quercetin or transporter inhibitors. KEY FINDINGS: Coincubation with quercetin decreased the uptake of AAI by Caco-2 cells cultured on the dishes at pH 7.4, and a similar decrease in AAI uptake was found when the cells were coincubated with acetic acid or benzoic acid. In contrast, the basolateral-to-apical transport of AAI was higher than the apical-to-basolateral transport of AAI at pH 7.4, and the former transport was decreased by quercetin and the BCRP inhibitors of Ko-143 and mitoxantrone, but not by P-gp or MRP2 inhibitors. CONCLUSIONS: AAI appears to be secreted from the apical membranes of Caco-2 cells via BCRP at neutral pH, although a small amount of AAI is taken up from the apical membranes via H(+) -linked MCTs, and quercetin may decrease both the BCRP-mediated efflux and the MCT-mediated influx of AAI.


Assuntos
Ácidos Aristolóquicos/farmacocinética , Transporte Biológico/efeitos dos fármacos , Absorção Intestinal/efeitos dos fármacos , Quercetina/farmacologia , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Ácido Acético/farmacologia , Ácido Benzoico/farmacologia , Células CACO-2 , Células Cultivadas , Ciclosporina/farmacologia , Dicetopiperazinas/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Indometacina/farmacologia , Mitoxantrona/farmacologia , Pravastatina/farmacologia , Propionatos/farmacologia , Quinidina/farmacologia , Quinolinas/farmacologia
7.
Sci Rep ; 5: 17646, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26620132

RESUMO

Aristolochic acid (AA) is the major active component of medicinal plants from the Aristolochiaceae family of flowering plants widely utilized for medicinal purposes. However, the molecular mechanisms of AA systems effects remain poorly understood. Here, we employed a joint network analysis that combines network pharmacology, a protein-protein interaction (PPI) database, biological processes analysis and functional annotation analysis to explore system effects. Firstly, we selected 15 protein targets (14 genes) in the PubChem database as the potential target genes and used PPI knowledge to incorporate these genes into an AA-specific gene network that contains 129 genes. Secondly, we performed biological processes analysis for these AA-related targets using ClueGO, some of new targeted genes were randomly selected and experimentally verified by employing the Quantitative Real-Time PCR assay for targeting the systems effects of AA in HK-2 cells with observed dependency of concentration. Thirdly, the pathway-based functional enrichment analysis was manipulated using WebGestalt to identify the mostly significant pathways associated with AA. At last, we built an AA target pathway network of significant pathways to predict the system effects. Taken together, this joint network analysis revealed that the systematic regulatory effects of AA on multidimensional pathways involving both therapeutic action and toxicity.


Assuntos
Ácidos Aristolóquicos , Bases de Dados Genéticas , Redes Reguladoras de Genes/efeitos dos fármacos , Ácidos Aristolóquicos/efeitos adversos , Ácidos Aristolóquicos/farmacocinética , Ácidos Aristolóquicos/farmacologia , Linhagem Celular , Humanos
8.
J Ethnopharmacol ; 172: 430-5, 2015 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-26183576

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Aristolochic acid nephropathy is a severe kidney disease caused by the administration of aristolochic acid, which is widely existed in plants of the Aristolochiaceae family. Aristolochic acid I (AAI) is the main toxic component in aristolochic acid. AIM OF THE STUDY: The roles of intestinal efflux drug transporters in the transport of AAI are unclear. This study investigates the interaction between AAI and main intestinal efflux transporters. MATERIALS AND METHODS: Firstly, bidirectional transport of AAI in Caco-2 cell monolayers was investigated. Then, MDCK-MDR1 (gene of P-glycoprotein (P-gp)), MDCK-MRP2 and LLC-PK1-BCRP cell lines were used for further investigation. RESULTS: In this study, we observed that the efflux ratio of AAI in Caco-2 cell monolayers was 5.8, which indicated that efflux transporters might be involved in the transport of AAI. AAI did not inhibit Rho123 efflux by P-gp and calcein efflux by MRP2, and intracellular accumulation of AAI in P-gp or MRP2 overexpressing cells was not different from their parental cells. These results indicated that AAI was not a substrate of P-gp or MRP2. In contrast, intracellular accumulation of AAI in LLC-PK1-BCRP cells was significantly lower than in their parental cells. The presence of GF120918, a BCRP inhibitor, significantly increased AAI accumulation in BCRP overexpressing cells but not in their parental cells. In addition, bidirectional transport assay of AAI in LLC-PK1-BCRP monolayers showed that the net efflux ratios of AAI were 13.8, 8.0 and 7.0 at 20, 40 and 80 µM AAI, respectively, and decreased to 3.0, 1.9 and 2.0 by the addition of 10 µM GF120918. CONCLUSIONS: These results indicated that AAI was a substrate of BCRP but not P-gp or MRP2.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Aristolochiaceae/química , Ácidos Aristolóquicos/farmacocinética , Mucosa Intestinal/metabolismo , Proteínas de Neoplasias/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Ácidos Aristolóquicos/isolamento & purificação , Transporte Biológico , Células CACO-2 , Cães , Humanos , Células LLC-PK1 , Células Madin Darby de Rim Canino , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Suínos
9.
Ren Fail ; 37(2): 209-13, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25540870

RESUMO

BACKGROUND: The purpose of this study was to investigate the cancer incidence in patients with end-stage aristolochic acid nephropathy (AAN). METHODS: A total of 102 patients with end-stage AAN treated in our hospital between 2004 and 2013 were included in this study. The correlation of cancer incidence with age, gender, dosage of aristolochic acid (AA), the type of renal replacement therapies, and the polymorphisms of quinone oxidoreductase 1 (NQO1) C609T and cytochrome P450 1A1 (CYP1A1) A4889G was examined. RESULTS: The cancer incidence rate in our patients was 41.2% (42 in 102) including 39 cases of urinary cancer. The mortality rate in the patients with cancer was significantly higher than that in the patients without cancer (31%, 13/42 vs. 11.7%, 7/60, p<0.05). Thirteen patients developed cancer before entering end-stage renal disease (ESRD). Cancer incidence was significantly associated with the dosage of AA consumption (p=0.091). Hemodialysis, peritoneal dialysis and renal transplant did not affect the cancer incidence in our patients differently, but appeared to be associated with cancer at particular locations of urinary system. The patients undergoing hemodialysis seemed to more likely have bladder cancer (72.72%), while the patients receiving peritoneal dialysis appeared to develop cancer predominantly in the upper urinary tract (66.67%). CONCLUSIONS: The cancer initiation in our patients seems significantly correlate with the dosage of AA consumption. Different renal replacement therapies appear to be associated with cancer at particular locations of urinary system in our patients.


Assuntos
Ácidos Aristolóquicos , Neoplasias Hematológicas , Falência Renal Crônica , Neoplasias Urológicas , Idoso , Ácidos Aristolóquicos/efeitos adversos , Ácidos Aristolóquicos/farmacocinética , Carcinógenos/farmacocinética , China/epidemiologia , Citocromo P-450 CYP1A1/análise , Feminino , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/epidemiologia , Neoplasias Hematológicas/etiologia , Humanos , Incidência , Falência Renal Crônica/induzido quimicamente , Falência Renal Crônica/complicações , Falência Renal Crônica/mortalidade , Masculino , Pessoa de Meia-Idade , NAD(P)H Desidrogenase (Quinona)/análise , Diálise Renal/estatística & dados numéricos , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Análise de Sobrevida , Neoplasias Urológicas/diagnóstico , Neoplasias Urológicas/epidemiologia , Neoplasias Urológicas/etiologia
10.
Am J Chin Med ; 42(6): 1453-69, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25427624

RESUMO

Aristolochic acids (AAs) are found in herbal medicines of Aristolochiaceae plants, including Aristolochia and Asarum species. AAs are associated with a rapidly progressive interstitial nephritis, which is called aristolochic acid nephropathy (AAN). However, the in-situ localization of AAs in the target organ, the kidney, has not been investigated yet. In the present study, the accumulation of aristolochic acid I (AA-I) in mouse kidney was revealed by immunoperoxidase light microscopy as well as colloidal gold immunoelectron microscopy (IEM) based on an anti-AA-I and AA-II monoclonal antibody (mAb). Male BALB/c mice were treated with 1.25 or 2.50 mg kg(-1) of AA-I per day for 5 days. Paraffin sections and ultra-thin sections of kidney tissue were respectively prepared. Under light microscopy, the apical surface of proximal tubules was strongly stained for AA-I, whereas no obvious immunostaining was found in the distal tubules and glomerulus, which remained relatively intact. Under electron microscopy, epithelial cells of the proximal tubules, distal tubules and collecting tubules were broken to various degrees. Gold labeling in the proximal and distal tubules was stronger than that in the collecting tubules. In renal tubules, immunogold signals of AA-I tended to accumulate in the mitochondria and peroxisomes, though the signals could be observed all over the cell. Gold signals were also found in the erythrocytes of glomeruli. The MAb against AA-I and AA-II provides a clue for the identification of proteins or factors which might interact with AA-I and thus induce targeted damage of kidney.


Assuntos
Anticorpos Monoclonais , Ácidos Aristolóquicos/análise , Ácidos Aristolóquicos/toxicidade , Imuno-Histoquímica/métodos , Rim/metabolismo , Animais , Aristolochiaceae/química , Ácidos Aristolóquicos/imunologia , Ácidos Aristolóquicos/farmacocinética , Túbulos Renais Proximais/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Microscopia Imunoeletrônica , Mitocôndrias/metabolismo , Peroxissomos/metabolismo , Distribuição Tecidual
11.
Mutagenesis ; 29(3): 189-200, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24598128

RESUMO

Aristolochic acid I (AAI) is the major toxic component of the plant extract AA, which leads to the development of nephropathy and urothelial cancer in human. Individual susceptibility to AAI-induced disease might reflect variability in enzymes that metabolise AAI. In vitro NAD(P)H: quinone oxidoreductase (NQO1) is the most potent enzyme that activates AAI by catalyzing formation of AAI-DNA adducts, which are found in kidneys of patients exposed to AAI. Inhibition of renal NQO1 activity by dicoumarol has been shown in mice. Here, we studied the influence of dicoumarol on metabolic activation of AAI in Wistar rats in vivo. In contrast to previous in vitro findings, dicoumarol did not inhibit AAI-DNA adduct formation in rats. Compared with rats treated with AAI alone, 11- and 5.4-fold higher AAI-DNA adduct levels were detected in liver and kidney, respectively, of rats pretreated with dicoumarol prior to exposure to AAI. Cytosols and microsomes isolated from liver and kidney of these rats were analysed for activity and protein levels of enzymes known to be involved in AAI metabolism. The combination of dicoumarol with AAI induced NQO1 protein level and activity in both organs. This was paralleled by an increase in AAI-DNA adduct levels found in ex vivo incubations with cytosols from rats pretreated with dicoumarol compared to cytosols from untreated rats. Microsomal ex vivo incubations showed a lower AAI detoxication to its oxidative metabolite, 8-hydroxyaristolochic acid (AAIa), although cytochrome P450 (CYP) 1A was practically unchanged. Because of these unexpected results, we examined CYP2C activity in microsomes and found that treatment of rats with dicoumarol alone and in combination with AAI inhibited CYP2C6/11 in liver. Therefore, these results indicate that CYP2C enzymes might contribute to AAI detoxication.


Assuntos
Ácidos Aristolóquicos/toxicidade , Carcinógenos/toxicidade , Dicumarol/farmacologia , Ativação Metabólica/efeitos dos fármacos , Animais , Ácidos Aristolóquicos/farmacocinética , Carcinógenos/farmacocinética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2 , Citocromos/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Adutos de DNA/efeitos dos fármacos , Adutos de DNA/metabolismo , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Testes de Mutagenicidade , Mutagênicos/farmacocinética , Mutagênicos/toxicidade , NAD(P)H Desidrogenase (Quinona)/metabolismo , Ratos , Ratos Wistar
12.
Xenobiotica ; 44(2): 135-45, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24152141

RESUMO

1. Aristolochic acid I (AAI) is the predominant component in plant extract of Aristolochia genus that is involved in development of aristolochic acid nephropathy, Balkan endemic nephropathy and urothelial cancer. The diseases do not develop in all individuals exposed to AAI and patients exhibit different clinical outcomes. Differences in the activities of enzymes catalyzing the metabolism of AAI might be one of the reasons for this individual susceptibility. 2. Understanding which human enzymes are involved in reductive activation of AAI generating AAI-DNA adducts, and/or its detoxication to the O-demethylated metabolite, aristolochic acid Ia (AAIa), is necessary in the assessment of the susceptibility to this compound. 3. This review summarizes the results of the latest studies utilizing genetically engineered mouse models to identify which human and rodent enzymes catalyze the reductive activation of AAI to AAI-DNA adducts and its oxidative detoxication to AAIa in vivo. 4. The use of hepatic cytochrome P450 (Cyp) reductase null (HRN) mice, in which NADPH:Cyp oxidoreductase (Por) is deleted in hepatocytes, Cyp1a1((-/-)), Cyp1a2((-/-)) single-knockout, Cyp1a1/1a2((-/-)) double-knockout and CYP1A-humanized mice revealed that mouse and human CYP1A1 and 1A2, besides mouse NAD(P)H: quinone oxidoreductase, were involved in the activation of AAI but CYP1A1 and 1A2 also oxidatively detoxified AAI.


Assuntos
Ácidos Aristolóquicos/farmacocinética , Carcinógenos/farmacocinética , Enzimas/metabolismo , Inativação Metabólica , Camundongos Knockout , Animais , Ácidos Aristolóquicos/metabolismo , Ácidos Aristolóquicos/toxicidade , Carcinógenos/metabolismo , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Adutos de DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Enzimas/genética , Humanos , Nefropatias/induzido quimicamente , Camundongos , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , NADPH-Ferri-Hemoproteína Redutase/genética , NADPH-Ferri-Hemoproteína Redutase/metabolismo
13.
Environ Toxicol Pharmacol ; 36(3): 850-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23981375

RESUMO

Hepatic CYP1A especially CYP1A2 plays an important role in the reduction of aristolochic acid I (AAI) nephrotoxicity. In this study, we investigated the effects of tanshinone I, a strong inducer of Cyp1a, on the nephrotoxicity induced by AAI. Histopathology and blood biochemistry assays showed that tanshinone I could reduce AAI-induced acute kidney injury. Pharmacokinetics analysis revealed that tanshinone I markedly decreased AUC of AAI in plasma and the content of AAI in both liver and kidney, indicating the enhancement of AAI metabolism. Real-time PCR and Western blot analysis confirmed that tanshinone I effectively increased the mRNA and protein levels of hepatic CYP1A1 and CYP1A2 in vivo. Luciferase assay showed that tanshinone I strongly increased the transcriptional activity of CYP1A1 and CYP1A2 in the similar extent. In summary, our data suggested that tanshinone I facilitated the metabolism of AAI and prevented AAI-induced kidney injury by induction of hepatic CYP1A 1/2 in vivo.


Assuntos
Abietanos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Ácidos Aristolóquicos/antagonistas & inibidores , Ácidos Aristolóquicos/toxicidade , Carcinógenos/antagonistas & inibidores , Carcinógenos/toxicidade , Citocromo P-450 CYP1A1/biossíntese , Citocromo P-450 CYP1A2/biossíntese , Nefropatias/induzido quimicamente , Nefropatias/enzimologia , Abietanos/farmacocinética , Animais , Antineoplásicos Fitogênicos/farmacocinética , Área Sob a Curva , Ácidos Aristolóquicos/farmacocinética , Análise Química do Sangue , Western Blotting , Carcinógenos/farmacocinética , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Indução Enzimática/efeitos dos fármacos , Rim/química , Rim/metabolismo , Rim/patologia , Nefropatias/patologia , Fígado/química , Fígado/metabolismo , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/genética , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Espectrofotometria Ultravioleta
14.
Curr Drug Metab ; 14(6): 695-705, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23701164

RESUMO

Aristolochic acid (AA), a plant nephrotoxin and carcinogen, causes aristolochic acid nephropathy (AAN) and its associated urothelial malignancy, and is hypothesized to be responsible for Balkan endemic nephropathy (BEN). The major component of AA, aristolochic acid I (AAI), is the predominant compound responsible for these diseases. The reductive activation of AAI leads to the formation of covalent DNA adducts. The most abundant DNA adduct, 7-(deoxyadenosin-N6-yl)aristolactam I, causes characteristic AT→TA transversions found in the TP53 tumor suppressor gene in tumors from AAN and BEN patients. Understanding which human enzymes are involved in AAI activation to species forming DNA adducts and/or detoxication to the AAI O-demethylated metabolite, aristolochic acid Ia (AAIa), is important in the assessment of the susceptibility to this carcinogen. This review summarizes the latest data on identifying human and rodent enzymes participating in AAI metabolism. NAD(P)H:quinone oxidoreductase (NQO1) is the most efficient cytosolic nitroreductase activating AAI in vitro and in vivo. In human hepatic microsomes, AAI is activated by cytochrome P450 1A2 (CYP1A2) and, to a lesser extent, by CYP1A1; NADPH:CYP oxidoreductase also plays a minor role. Human and rodent CYP1A1 and 1A2 are also the principal enzymes involved in oxidative detoxication of AAI to AAIa in vitro and in vivo. The orientation of AAI in the active sites of human CYP1A1/2 and NQO1 was predicted from molecular modeling and is consistent with the efficient reduction of AAI by them observed experimentally. Molecular modeling also shows why CYP1A2 plays an important role in the oxidation of AAI to AAIa.


Assuntos
Ácidos Aristolóquicos/farmacocinética , Nefropatias/metabolismo , Neoplasias Epiteliais e Glandulares/metabolismo , Oxirredutases/metabolismo , Animais , Ácidos Aristolóquicos/toxicidade , Humanos , Nefropatias/induzido quimicamente , Neoplasias Epiteliais e Glandulares/induzido quimicamente , Urotélio/metabolismo
15.
Toxicol Appl Pharmacol ; 265(3): 360-7, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22982977

RESUMO

Aristolochic acid causes a specific nephropathy (AAN), Balkan endemic nephropathy, and urothelial malignancies. Using Western blotting suitable to determine protein expression, we investigated in several transgenic mouse lines expression of NAD(P)H:quinone oxidoreductase (NQO1)-the most efficient cytosolic enzyme that reductively activates aristolochic acid I (AAI). The mouse tissues used were from previous studies [Arlt et al., Chem. Res. Toxicol. 24 (2011) 1710; Stiborova et al., Toxicol. Sci. 125 (2012) 345], in which the role of microsomal cytochrome P450 (CYP) enzymes in AAI metabolism in vivo had been determined. We found that NQO1 levels in liver, kidney and lung of Cyp1a1⁻/⁻, Cyp1a2⁻/⁻ and Cyp1a1/1a2⁻/⁻ knockout mouse lines, as well as in two CYP1A-humanized mouse lines harboring functional human CYP1A1 and CYP1A2 and lacking the mouse Cyp1a1/1a2 orthologs, differed from NQO1 levels in wild-type mice. NQO1 protein and enzymic activity were induced in hepatic and renal cytosolic fractions isolated from AAI-pretreated mice, compared with those in untreated mice. Furthermore, this increase in hepatic NQO1 enzyme activity was associated with bioactivation of AAI and elevated AAI-DNA adduct levels in ex vivo incubations of cytosolic fractions with DNA and AAI. In conclusion, AAI appears to increase its own metabolic activation by inducing NQO1, thereby enhancing its own genotoxic potential.


Assuntos
Ácidos Aristolóquicos/farmacocinética , Nefropatia dos Bálcãs/enzimologia , Nefropatia dos Bálcãs/genética , Citocromo P-450 CYP1A1/deficiência , Citocromo P-450 CYP1A2/deficiência , Fígado/metabolismo , NAD(P)H Desidrogenase (Quinona)/biossíntese , Animais , Ácidos Aristolóquicos/toxicidade , Nefropatia dos Bálcãs/metabolismo , Western Blotting , Linhagem Celular , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Citosol/enzimologia , Citosol/metabolismo , Adutos de DNA/metabolismo , Feminino , Humanos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NAD(P)H Desidrogenase (Quinona)/metabolismo
16.
Mutat Res ; 743(1-2): 52-8, 2012 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-22245565

RESUMO

Aristolochic acid (AA) is known to be a potent mutagen and carcinogen. Aristolochic acid I (AAI) and aristolochic acid II (AAII), the two major components of AA, differ from each other by a single methoxy group. However, their individual mutagenic characteristics in vivo are unclear. In the present study, we compared their DNA adduct formation and mutagenicities in the gpt delta transgenic mouse kidney. The dA-AAI, dG-AAI, dA-AAII and dG-AAII were identified in the kidney two days after intragastric administration of AAI or AAII at 5mg/kg. The concentration of DNA adducts formed by AAII was approximately 2.5-fold higher than that formed by AAI (p<0.05). The mutant frequency induced by AAII was nearly two-fold higher than that induced by AAI (p<0.05) following administration of 5mg/kg AAI or AAII, five times per week for six weeks. Investigation of the mutation spectra showed no statistically significant difference between AAI- and AAII-treated mice (p>0.05). A:T to T:A transversion was the predominant type of mutation in both treated groups, the GC-associated mutation rates, however, differed between the AAI and AAII treatments. The in vivo metabolic pathways of AAI and AAII are different, and this may affect their mutagenicity. In the present study, we measured the levels of AAI and AAII in the kidney and plasma of gpt delta transgenic mice at multiple time points after a single intragastric dose of 1 or 5mg/kg of either component. Our results showed that the levels of AAII in both kidney and plasma were considerably higher than those of AAI (p<0.01). The present study indicated that AAII showed more carcinogenic risk than AAI in vivo, and this may be, at least partly, the result of its increased levels in kidney and plasma.


Assuntos
Ácidos Aristolóquicos/toxicidade , Carcinógenos/toxicidade , Proteínas de Escherichia coli/genética , Rim/efeitos dos fármacos , Mutagênicos/toxicidade , Pentosiltransferases/genética , Animais , Ácidos Aristolóquicos/farmacocinética , Adutos de DNA , Dano ao DNA , Camundongos , Camundongos Transgênicos
17.
Neuro Endocrinol Lett ; 33 Suppl 3: 25-32, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23353840

RESUMO

OBJECTIVES: The herbal drug aristolochic acid (AA) derived from Aristolochia species has been shown to be the cause of aristolochic acid nephropathy (AAN), Balkan endemic nephropathy (BEN) and their urothelial malignancies. One of the common features of AAN and BEN is that not all individuals exposed to AA suffer from nephropathy and tumor development. One cause for these different responses may be individual differences in the activities of the enzymes catalyzing the biotransformation of AA. Thus, the identification of enzymes principally involved in the metabolism of AAI, the major toxic component of AA, and detailed knowledge of their catalytic specificities is of major importance. Human cytochrome P450 (CYP) 1A1 and 1A2 enzymes were found to be responsible for the AAI reductive activation to form AAI-DNA adducts, while its structurally related analogue, CYP1B1 is almost without such activity. However, knowledge of the differences in mechanistic details of CYP1A1-, 1A2-, and 1B1- mediated reduction is still lacking. Therefore, this feature is the aim of the present study. METHODS: Molecular modeling capable of evaluating interactions of AAI with the active site of human CYP1A1, 1A2 and 1B1 under the reductive conditions was used. In silico docking, employing soft-soft (flexible) docking procedure was used to study the interactions of AAI with the active sites of these human enzymes. RESULTS: The predicted binding free energies and distances between an AAI ligand and a heme cofactor are similar for all CYPs evaluated. AAI also binds to the active sites of CYP1A1, 1A2 and 1B1 in similar orientations. The carboxylic group of AAI is in the binding position situated directly above heme iron. This ligand orientation is in CYP1A1/1A2 further stabilized by two hydrogen bonds; one between an oxygen atom of the AAI nitro-group and the hydroxyl group of Ser122/Thr124; and the second bond between an oxygen atom of dioxolane ring of AAI and the hydroxyl group of Thr497/Thr498. For the CYP1B1:AAI complex, however, any hydrogen bonding of the nitro-group of AAI is prevented as Ser122/Thr124 residues are in CYP1B1 protein replaced by hydrophobic residue Ala133. CONCLUSION: The experimental observations indicate that CYP1B1 is more than 10× less efficient in reductive activation of AAI than CYP1A2. The docking simulation however predicts the binding pose and binding energy of AAI in the CYP1B1 pocket to be analogous to that found in CYP1A1/2. We believe that the hydroxyl group of S122/T124 residue, with its polar hydrogen placed close to the nitro group of the substrate (AAI), is mechanistically important, for example it could provide a proton required for the stepwise reduction process. The absence of a suitable proton donor in the AAI-CYP1B1 binary complex could be the key difference, as the nitro group is in this complex surrounded only by the hydrophobic residues with potential hydrogen donors not closer than 5 Å.


Assuntos
Ácidos Aristolóquicos/efeitos adversos , Ácidos Aristolóquicos/farmacocinética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Sequência de Aminoácidos , Aristolochia/química , Ácidos Aristolóquicos/química , Hidrocarboneto de Aril Hidroxilases/química , Hidrocarboneto de Aril Hidroxilases/genética , Domínio Catalítico/efeitos dos fármacos , Simulação por Computador , Citocromo P-450 CYP1A1/química , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/química , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1B1 , Adutos de DNA/química , Adutos de DNA/metabolismo , Medicamentos de Ervas Chinesas/efeitos adversos , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacocinética , Humanos , Ligação de Hidrogênio/efeitos dos fármacos , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Nefropatias/induzido quimicamente , Modelos Químicos , Dados de Sequência Molecular , Nitrorredutases/efeitos adversos , Nitrorredutases/química , Nitrorredutases/farmacocinética , Estrutura Terciária de Proteína/efeitos dos fármacos
18.
Chem Res Toxicol ; 25(1): 130-9, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22118289

RESUMO

Aristolochic acids are natural nitro-compounds found globally in the plant genus Aristolochia that have been implicated in the severe illness in humans termed aristolochic acid nephropathy (AAN). Aristolochic acids undergo nitroreduction, among other metabolic reactions, and active intermediates arise that are carcinogenic. Previous experiments with rats showed that aristolochic acid I (AA-I), after oral administration or injection, is subjected to detoxication reactions to give aristolochic acid Ia, aristolactam Ia, aristolactam I, and their glucuronide and sulfate conjugates that can be found in urine and feces. Results obtained with whole rats do not clearly define the role of liver and kidney in such metabolic transformation. In this study, in order to determine the specific role of the kidney on the renal disposition of AA-I and to study the biotransformations suffered by AA-I in this organ, isolated kidneys of rats were perfused with AA-I. AA-I and metabolite concentrations were determined in perfusates and urine using HPLC procedures. The isolated perfused rat kidney model showed that AA-I distributes rapidly and extensively in kidney tissues by uptake from the peritubular capillaries and the tubules. It was also established that the kidney is able to metabolize AA-I into aristolochic acid Ia, aristolochic acid Ia O-sulfate, aristolactam Ia, aristolactam I, and aristolactam Ia O-glucuronide. Rapid demethylation and sulfation of AA-I in the kidney generate aristolochic acid Ia and its sulfate conjugate that are voided to the urine. Reduction reactions to give the aristolactam metabolites occur to a slower rate. Renal clearances showed that filtered AA-I is reabsorbed at the tubules, whereas the metabolites are secreted. The unconjugated metabolites produced in the renal tissues are transported to both urine and perfusate, whereas the conjugated metabolites are almost exclusively secreted to the urine.


Assuntos
Ácidos Aristolóquicos/metabolismo , Carcinógenos/metabolismo , Rim/metabolismo , Animais , Ácidos Aristolóquicos/farmacocinética , Ácidos Aristolóquicos/urina , Biotransformação , Carcinógenos/farmacocinética , Cromatografia Líquida de Alta Pressão , Medicamentos de Ervas Chinesas/metabolismo , Medicamentos de Ervas Chinesas/farmacocinética , Técnicas In Vitro , Fígado/metabolismo , Masculino , Perfusão , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
19.
Neuro Endocrinol Lett ; 32 Suppl 1: 57-70, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22167209

RESUMO

OBJECTIVES: Ingestion of aristolochic acid (AA) is associated with development of urothelial tumors linked with aristolochic acid nephropathy, and is implicated in the development of Balkan endemic nephropathy-associated urothelial tumors. Aristolochic acid I (AAI), the major toxic component of AA, is more toxic than its demethoxylated derivate AAII. A different enzymatic conversion of both carcinogens might be one of the reasons explaining this feature. Therefore, the present study has been designed to compare efficiency of human NAD(P)H:quinone oxidoreductase (NQO1) and phase II enzymes such as sulfotransferases (SULTs) and N,O-acetyltransferases (NATs) to activate AAI and AAII in vitro. In addition, to investigate the molecular mechanisms of AAI and AAII reduction by human NQO1, molecular modeling was used to compare interactions of AAI and AAII with the active site of this enzyme. METHODS: DNA adduct formation by AAI and AAII was investigated by the nuclease P1 version of the 32P-postlabeling method. In silico docking, employing soft-soft (flexible) docking procedure, was used to study the interactions of AAI and AAII with the active site of human NQO1. RESULTS: Human NQO1 activated AAI and AAII, generating DNA adduct patterns reproducing those found in several species including human exposed to these compounds. These results demonstrate that NQO1 is capable of reducing both AAs to reactive species binding to DNA. However, concentrations required for half-maximum DNA binding mediated by NQO1 were higher for AAII (158 µM) than for AAI (17 µM). One of the reasons causing this phenomenon is a lower efficiency of NQO1 to reduce AAII than AAI we found in this work; although both AAI and AAII are bound with similar binding affinities to the NQO1 active site, the binding orientation of AAII in the active site of NQO1 does not favor the effective reduction of its nitro group. Because reduced nitro-aromatics are often further activated by SULTs or NATs, their roles in AAI and AAII activation were investigated. Our results indicate that phase II reactions do not stimulate the bioactivation of AAs; neither enzymes present in human hepatic cytosols nor human SULT1A1, 1A2, 1A3, 1E, or 2A nor NAT1 or NAT2 further enhanced DNA adduct formation by AAs. In contrast, human SULT1A1, 1A2 and 1A3 as well as NAT1 and NAT2 enzymes even inhibited NQO1-mediated bioactivation of AAII. Therefore, under the in vitro conditions used, DNA adducts arise by enzymatic reduction of AAs through the formation of N-hydroxyaristolactams that are spontaneously decomposed to the reactive species forming DNA adducts. CONCLUSION: The results found in this study emphasize the importance of NQO1 in the metabolic activation of AAI and AAII and provide the evidence that initial nitroreduction is the rate limiting step in their activation. This enzyme is more effective in activation of AAI relative to AAII, which might contribute to its lower binding to DNA found both in vitro and in vivo, Moreover, inhibition effects of conjugation reactions on AAII activation might further contribute to its decreased capability of forming DNA adducts and its lower toxicity comparing with AAI.


Assuntos
Acetiltransferases/metabolismo , Ácidos Aristolóquicos/farmacocinética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Sulfotransferases/metabolismo , Acetiltransferases/química , Acetiltransferases/fisiologia , Animais , Ácidos Aristolóquicos/química , Biotransformação/fisiologia , Domínio Catalítico , Células Cultivadas , Adutos de DNA/metabolismo , Ativação Enzimática , Humanos , Lactamas/metabolismo , Lactamas/farmacocinética , Modelos Moleculares , Conformação Molecular , NAD(P)H Desidrogenase (Quinona)/química , NAD(P)H Desidrogenase (Quinona)/fisiologia , Ligação Proteica , Sulfotransferases/química , Sulfotransferases/fisiologia
20.
Neuro Endocrinol Lett ; 32 Suppl 1: 121-30, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22167220

RESUMO

OBJECTIVE: The herbal drug aristolochic acid (AA) derived from Aristolochia species has been shown to be the cause of aristolochic acid nephropathy (AAN), Balkan endemic nephropathy (BEN) and their urothelial malignancies. One of the common features of AAN and BEN is that not all individuals exposed to AA suffer from nephropathy and tumor development. One cause for these different responses may be individual differences in the activities of the enzymes catalyzing the biotransformation of AA. Thus, the identification of enzymes principally involved in the metabolism of AAI, the major toxic component of AA, and detailed knowledge of their catalytic specificities is of major importance. Therefore, the present study has been designed to evaluate the cytochrome P450 (CYP)-mediated oxidative detoxification and reductive activation of AAI in a rat model. METHODS: DNA adduct formation was investigated by the nuclease P1 version of the 32P-postlabeling method. The CYP-mediated formation of a detoxication metabolite of AAI, 8-hydroxyaristolochic acid I (AAIa), in vitro in rat hepatic microsomes was determined by HPLC. RESULTS: Rat hepatic CYPs both detoxicate AAI by its oxidation to AAIa and reductively activate this carcinogen to a cyclic N-acylnitrenium ion forming AAI-DNA adducts in vitro. To define the role of hepatic CYPs in AAI demethylation and activation, the modulation of AAIa and AAI-DNA adduct formation by CYP inducers and selective CYP inhibitors was investigated. Based on these studies, we attribute the major role of CYP1A1 and 1A2 in AAI detoxication by its demethylation to AAIa, and, under hypoxic conditions also to AAI activation to species forming DNA adducts. Using microsomes of Baculovirus transfected insect cells (Supersomes™) containing recombinantly expressed rat CYPs, NADPH:CYP reductase and/or cytochrome b5, a major role of CYP1A1 and 1A2 in both reactions in vitro was confirmed. CONCLUSION: Based on the results found in this and former studies we propose that AAI activation and detoxication in rats are dictated mainly by AAI binding affinity to CYP1A1/2 or NADPH(P)H:quinone oxidoreductase, by their turnover and by the balance between oxidation and reduction of AAI by CYP1A.


Assuntos
Ácidos Aristolóquicos/farmacocinética , Sistema Enzimático do Citocromo P-450/fisiologia , Fígado/efeitos dos fármacos , Animais , Biotransformação , Carcinógenos/farmacocinética , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2 , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Citocromos/genética , Citocromos/metabolismo , Adutos de DNA/metabolismo , Avaliação Pré-Clínica de Medicamentos , Inativação Metabólica , Fígado/enzimologia , Fígado/metabolismo , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Modelos Biológicos , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...