Jeffrey R. Idle
Charles University in Prague
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jeffrey R. Idle.
Drug Metabolism Reviews | 2007
Chi Chen; Frank J. Gonzalez; Jeffrey R. Idle
Xenobiotic metabolism, a ubiquitous natural response to foreign compounds, elicits initiating signals for many pathophysiological events. Currently, most widely used techniques for identifying xenobiotic metabolites and metabolic pathways are empirical and largely based on in vitro incubation assays and in vivo radiotracing experiments. Recent work in our lab has shown that LC-MS-based metabolomic techniques are useful tools for xenobiotic metabolism research since multivariate data analysis in metabolomics can significantly rationalize the processes of xenobiotic metabolite identification and metabolic pathway analysis. In this review, the technological elements of LC-MS-based metabolomics for constructing high-quality datasets and conducting comprehensive data analysis are examined. Four novel approaches of using LC-MS-based metabolomic techniques in xenobiotic metabolism research are proposed and illustrated by case studies and proof-of-concept experiments, and the perspective on their application is further discussed.
Chemical Research in Toxicology | 2009
Chi Chen; Kristopher W. Krausz; Yatrik M. Shah; Jeffrey R. Idle; Frank J. Gonzalez
Metabolic bioactivation, glutathione depletion, and covalent binding are the early hallmark events after acetaminophen (APAP) overdose. However, the subsequent metabolic consequences contributing to APAP-induced hepatic necrosis and apoptosis have not been fully elucidated. In this study, serum metabolomes of control and APAP-treated wild-type and Cyp2e1-null mice were examined by liquid chromatography-mass spectrometry (LC-MS) and multivariate data analysis. A dose-response study showed that the accumulation of long-chain acylcarnitines in serum contributes to the separation of wild-type mice undergoing APAP-induced hepatotoxicity from other mouse groups in a multivariate model. This observation, in conjunction with the increase of triglycerides and free fatty acids in the serum of APAP-treated wild-type mice, suggested that APAP treatment can disrupt fatty acid beta-oxidation. A time-course study further indicated that both wild-type and Cyp2e1-null mice had their serum acylcarnitine levels markedly elevated within the early hours of APAP treatment. While remaining high in wild-type mice, serum acylcarnitine levels gradually returned to normal in Cyp2e1-null mice at the end of the 24 h treatment. Distinct from serum aminotransferase activity and hepatic glutathione levels, the pattern of serum acylcarnitine accumulation suggested that acylcarnitines can function as complementary biomarkers for monitoring the APAP-induced hepatotoxicity. An essential role for peroxisome proliferator-activated receptor alpha (PPARalpha) in the regulation of serum acylcarnitine levels was established by comparing the metabolomic responses of wild-type and Ppara-null mice to a fasting challenge. The upregulation of PPARalpha activity following APAP treatment was transient in wild-type mice but was much more prolonged in Cyp2e1-null mice. Overall, serum metabolomics of APAP-induced hepatotoxicity revealed that the CYP2E1-mediated metabolic activation and oxidative stress following APAP treatment can cause irreversible inhibition of fatty acid oxidation, potentially through suppression of PPARalpha-regulated pathways.
Journal of Biological Chemistry | 2008
Chi Chen; Kristopher W. Krausz; Jeffrey R. Idle; Frank J. Gonzalez
CYP2E1 is recognized as the most important enzyme for initiation of acetaminophen (APAP)-induced toxicity. In this study, the resistance of Cyp2e1-null mice to APAP treatment was confirmed by comparing serum aminotransferase activities and blood urea nitrogen levels in wild-type and Cyp2e1-null mice. However, unexpectedly, profiling of major known APAP metabolites in urine and serum revealed that the contribution of CYP2E1 to APAP metabolism decreased with increasing APAP doses administered. Measurement of hepatic glutathione and hydrogen peroxide levels exposed the importance of oxidative stress in determining the consequence of APAP overdose. Subsequent metabolomic analysis was capable of constructing a principal components analysis (PCA) model that delineated a relationship between urinary metabolomes and the responses to APAP treatment. Urinary ions high in wild-type mice treated with 400 mg/kg APAP were elucidated as 3-methoxy-APAP glucuronide (VII) and three novel APAP metabolites, including S-(5-acetylamino-2-hydroxyphenyl)mercaptopyruvic acid (VI, formed by a Cys-APAP transamination reaction in kidney), 3,3′-biacetaminophen (VIII, an APAP dimer), and a benzothiazine compound (IX, originated from deacetylated APAP), through mass isotopomer analysis, accurate mass measurement, tandem mass spectrometry fragmentation, in vitro reactions, and chemical treatments. Dose-, time-, and genotype-dependent appearance of these minor APAP metabolites implied their association with the APAP-induced toxicity and potential biomarker application. Overall, the oxidative stress elicited by CYP2E1-mediated APAP metabolism might significantly contribute to APAP-induced toxicity. The combination of genetically modified animal models, mass isotopomer analysis, and metabolomics provides a powerful and efficient technical platform to characterize APAP-induced toxicity through identifying novel biomarkers and unraveling novel mechanisms.
Cell Metabolism | 2008
Chi Chen; Yatrik M. Shah; Keiichirou Morimura; Kristopher W. Krausz; Makoto Miyazaki; Terrilyn A. Richardson; Edward T. Morgan; James M. Ntambi; Jeffrey R. Idle; Frank J. Gonzalez
To investigate the pathogenic mechanism of ulcerative colitis, a dextran sulfate sodium (DSS)-induced acute colitis model was examined by serum metabolomic analysis. Higher levels of stearoyl lysophosphatidylcholine and lower levels of oleoyl lysophosphatidylcholine in DSS-treated mice compared to controls led to the identification of DSS-elicited inhibition of stearoyl-CoA desaturase 1 (SCD1) expression in liver. This decrease occurred prior to the symptoms of acute colitis and was well correlated with elevated expression of proinflammatory cytokines. Furthermore, Citrobacter rodentium-induced colitis and lipopolysaccharide treatment also suppressed SCD1 expression in liver. Scd1 null mice were more susceptible to DSS treatment than wild-type mice, while oleic acid feeding and in vivo SCD1 rescue with SCD1 adenovirus alleviated the DSS-induced phenotype. This study reveals that inhibition of SCD1-mediated oleic acid biogenesis exacerbates proinflammatory responses to exogenous challenges, suggesting that SCD1 and its related lipid species may serve as potential targets for intervention or treatment of inflammatory diseases.
Drug Metabolism and Disposition | 2006
Xiaochao Ma; Yatrik M. Shah; Connie Cheung; Grace L. Guo; Lionel Feigenbaum; Kristopher W. Krausz; Jeffrey R. Idle; Frank J. Gonzalez
The most common clinical implication for the activation of the human pregnane X receptor (PXR) is the occurrence of drug-drug interactions mediated by up-regulated cytochromes P450 3A (CYP3A) isozymes. Typical rodent models do not predict drug-drug interactions mediated by human PXR because of species differences in response to PXR ligands. In the current study, a PXR-humanized mouse model was generated by bacterial artificial chromosome (BAC) transgenesis in Pxr-null mice using a BAC clone containing the complete human PXR gene and 5′- and 3′-flanking sequences. In this PXR-humanized mouse model, PXR is selectively expressed in the liver and intestine, the same tissue expression pattern as CYP3A. Treatment of PXR-humanized mice with the PXR ligands mimicked the human response, since both hepatic and intestinal CYP3As were strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16α-carbonitrile, a rodent-specific PXR ligand. In rifampicin-pretreated PXR-humanized mice, an ∼60% decrease was observed for both the maximal midazolam serum concentration (Cmax) and the area under the concentration-time curve, as a result of a 3-fold increase in midazolam 1′-hydroxylation. These results illustrate the potential utility of the PXR-humanized mice in the investigation of drug-drug interactions mediated by CYP3A and suggest that the PXR-humanized mouse model would be an appropriate in vivo tool for evaluation of the overall pharmacokinetic consequences of human PXR activation by drugs.
Expert Opinion on Drug Metabolism & Toxicology | 2008
Xiaochao Ma; Jeffrey R. Idle; Frank J. Gonzalez
Background: The pregnane X receptor (PXR; NR1I2), a member of the nuclear receptor superfamily, regulates the expression of metabolic enzymes and transporters involved in the response of mammals to their chemical environment. Objective: To summarize the functions and clinical implications of PXR. Methods: In the current review, the clinical implications of PXR are discussed, and the use of genetically engineered PXR mouse models is highlighted. Results/conclusion: Recent advances in mouse models, including Pxr-null and PXR-humanized mice, provide in vivo tools for evaluating the physiological functions of PXR and its role in controlling xenobiotic metabolism and transport. By using the PXR knockout and humanized mouse models, PXR was found to influence drug–drug interactions, hepatic steatosis, and the homeostasis of vitamin D, bile acids, and steroid hormones. PXR was also shown to influence inflammatory bowel diseases.
Journal of Pharmacology and Experimental Therapeutics | 2007
Xiaochao Ma; Yatrik M. Shah; Grace L. Guo; Ting Wang; Kristopher W. Krausz; Jeffrey R. Idle; Frank J. Gonzalez
Rifaximin, a rifamycin analog approved for the treatment of travelers diarrhea, is also beneficial in the treatment of multiple chronic gastrointestinal disorders. However, the mechanisms contributing to the effects of rifaximin on chronic gastrointestinal disorders are not fully understood. In the current study, rifaximin was investigated for its role in activation of the pregnane X receptor (PXR), a nuclear receptor that regulates genes involved in xenobiotic and limited endobiotic deposition and detoxication. PXR-humanized (hPXR), Pxr-null, and wild-type mice were treated orally with rifaximin, and rifampicin, a well characterized human PXR ligand. Rifaximin was highly concentrated in the intestinal tract compared with rifampicin. Rifaximin treatment resulted in significant induction of PXR target genes in the intestine of hPXR mice, but not in wild-type and Pxr-null mice. However, rifaximin treatment demonstrated no significant effect on hepatic PXR target genes in wild-type, Pxr-null, and hPXR mice. Consistent with the in vivo data, cell-based reporter gene assay revealed rifaximin-mediated activation of human PXR, but not the other xenobiotic nuclear receptors constitutive androstane receptor, peroxisome proliferator-activated receptor (PPAR)α, PPARγ, and farnesoid X receptor. Pretreatment with rifaximin did not affect the pharmacokinetics of the CYP3A substrate midazolam, but it increased the Cmax and decreased Tmax of 1′-hydroxymidazolam. Collectively, the current study identified rifaximin as a gut-specific human PXR ligand, and it provided further evidence for the utility of hPXR mice as a critical tool for the study of human PXR activators. Further human studies are suggested to assess the potential role of rifaximin-mediated gut PXR activation in therapeutics of chronic gastrointestinal disorders.
Journal of Pharmacology and Experimental Therapeutics | 2006
Chi Chen; LingHua Meng; Xiaochao Ma; Kristopher W. Krausz; Yves Pommier; Jeffrey R. Idle; Frank J. Gonzalez
NSC686288 [aminoflavone (AF)], a candidate chemotherapeutic agent, possesses a unique antiproliferative profile against tumor cells. Metabolic bioactivation of AF by drug-metabolizing enzymes, especially CYP1A monooxygenases, has been implicated as an underlying mechanism for its selective cytotoxicity in several cell culture-based studies. However, in vivo metabolism of AF has not been investigated in detail. In this study, the structural identities of 13 AF metabolites (12 of which are novel) in mouse urine or from microsomal incubations, including three monohydroxy-AFs, two dihydroxy-AFs and their sulfate and glucuronide conjugates, as well as one N-glucuronide, were determined by accurate mass measurements and liquid chromatography-tandem mass spectrometry fragmentation patterns, and a comprehensive map of the AF metabolic pathways was constructed. Significant differences between wild-type and Cyp1a2-null mice, within the relative composition of urinary metabolites of AF, demonstrated that CYP1A2-mediated regioselective oxidation was a major contributor to the metabolism of AF. Comparisons between wild-type and CYP1A2-humanized mice further revealed interspecies differences in CYP1A2-mediated catalytic activity. Incubation of AF with liver microsomes from all three mouse lines and with pooled human liver microsomes confirmed the observations from urinary metabolite profiling. Results from enzyme kinetic analysis further indicated that in addition to CYP1A P450s, CYP2C P450s may also play some role in the metabolism of AF.
Drug Metabolism and Disposition | 2008
Xiaochao Ma; Connie Cheung; Kristopher W. Krausz; Yatrik M. Shah; Ting Wang; Jeffrey R. Idle; Frank J. Gonzalez
Cytochrome P450 3A4 (CYP3A4), the most abundant human cytochrome P450 in liver, participates in the metabolism of ∼50% of clinically used drugs. The pregnane X receptor (PXR), a member of the nuclear receptor superfamily, is the major activator of CYP3A4 transcription. However, because of species differences in response to PXR ligands, it is problematic to use rodents to assess CYP3A4 regulation and function. The generation of double transgenic mice expressing human PXR and CYP3A4 (TgCYP3A4/hPXR) would provide a solution to this problem. In the current study, a TgCYP3A4/hPXR mouse model was generated by bacterial artificial chromosome transgenesis in Pxr-null mice. In TgCYP3A4/hPXR mice, CYP3A4 was strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16α-carbonitrile, a rodent-specific PXR ligand. Consistent with CYP3A expression, hepatic CYP3A activity increased ∼5-fold in TgCYP3A4/hPXR mice pretreated with rifampicin. Most antihuman immunodeficiency virus protease inhibitors are CYP3A substrates and their interactions with rifamycins are a source of major concern in patients coinfected with human immunodeficiency virus and Mycobacterium tuberculosis. By using TgCYP3A4/hPXR mice, human PXR-CYP3A4-mediated rifampicin-protease inhibitor interactions were recapitulated, as the metabolic stability of amprenavir, nelfinavir, and saquinavir decreased 52, 53, and 99%, respectively, in the liver microsomes of TgCYP3A4/hPXR mice pretreated with rifampicin. In vivo, rifampicin pretreatment resulted in an ∼80% decrease in the area under the serum amprenavir concentration-time curve in TgCYP3A4/hPXR mice. These results suggest that the TgCYP3A4/hPXR mouse model could serve as a useful tool for studies on CYP3A4 transcription and function in vivo.
European Journal of Clinical Pharmacology | 2007
Ondřej Slanař; Milan Nobilis; Jaroslav Kvetina; Olga Matoušková; Jeffrey R. Idle; František Perlík
Dear Professor Dahlqvist, We have read with interest the articles by Pedersen et al. and Wang et al. who highlighted the importance of functional polymorphisms of CYP2D6 on the pharmacokinetics of tramadol and its major metabolite O-demethyltramadol in recent issues of the European Journal of Clinical Pharmacology [1, 2]. The pharmacology of tramadol is unusually complex, having at least 11 unconjugated metabolites and 12 conjugated compounds [3]. There are three major metabolic pathways, CYP2D6, CYP3A, and CYP2B6, forming Oand N-demethylated metabolites. Tramadol is believed to undergo first-pass metabolism, reducing its bioavailability to approximately 80% after oral administration. The CYP2D6-dependent pharmacokinetics of tramadol is usually also reflected in increased bioavailability in poor metabolizers (PM) compared with extensive metabolizers (EM). Surprisingly, Pedersen et al. did not observe any significant difference between bioavailability of tramadol in EMs and PMs, while large interindividual variability was noted [1]. It is recognized that the bioavailabity of some drugs can be substantially affected by active transporters expressed in the gut lumen, like P-glycoprotein. We recently conducted a study in order to uncover MDR1 genotype-dependent variations in pharmacokinetic parameters of tramadol and O-demethyltramadol. Twenty-one healthy young volunteers selected from our database participated in the study after providing informed consent. Presence of CYP2D6*3, *4, *5, *6 alleles and gene duplications was analyzed using PCRand RFLPbased methods. MDR1 polymorphisms C3435T and G2677T/A were also detected. Three groups of seven CYP2D6 EM, heterozygous EM, and PM subjects were investigated. Four and nine subjects were homozygous carriers of C3435 and T3435 alleles, respectively. Each volunteer was administered a 100-mg sustainedrelease tramadol tablet (Tramal Retard 100 mg, Zentiva Praha a.s.), and plasma concentrations of (R,S)-(±)-tramadol (TMD) and (±)-O-demethyltramadol (M1) were analyzed by HPLC at baseline and at 2.5, 4, 8, 12, and 24 h post-dose. The average Cmax and AUC0–24 values of TMD increased slightly in groups with increasing numbers of 3435T alleles of MDR1 irrespective of CYP2D6 status. The mean (SD) Cmax values of TMD were 495.4 (91.1), 529.3 (161.7), and 600.2 (179.9) nmol/l in 3435CC, 3435CT, and 3435TT groups, respectively. Corresponding values for AUC0–24 in the respective groups were 7,393.9 (2,299.1), 7,710.1 (3,304.7), and 8,478.8 (3,771.0) nmol·h/l. The differences, however, did not reach the level of statistical significance. Interestingly, a similar trend was not observed for M1, for which production was more dependent on relative numbers of CYP2D6 extensive metabolizers. Detailed analysis focused on comparison of subjects according to mixed CYP2D6 and MDR1 genotypes. Figure 1 shows pharmacokinetics profiles of TMD and Eur J Clin Pharmacol (2007) 63:419–421 DOI 10.1007/s00228-006-0255-3