Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 41.461
Filtrer
1.
Chem Res Toxicol ; 37(7): 1104-1112, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38885202

RÉSUMÉ

Chlortoluron (CTU) is an herbicide extensively used in agricultural settings for crop cultivation. Its presence in water has been identified as a pollutant detrimental to aquatic species. The objective of the present study was to explore the metabolic activation and hepatotoxicity of CTU. Through human and rat liver microsomal incubations supplemented with CTU, nicotinamide adenine dinucleotide phosphate (NADPH), and either glutathione or N-acetyl cysteine, a benzylic alcohol metabolite (M1) was discerned, alongside a phenol metabolite (M2), a glutathione conjugate (M3), and an N-acetyl cysteine conjugate (M4). In rats exposed to CTU, biliary M3 and urinary M4 were detected in their bile and urine, respectively. The generation of M1 was detected in the presence of NADPH. The observation of M3 and M4 suggests the formation of an iminoquinone methide intermediate arising from the oxidation of M1. CYP3A4 was found to be the principal enzyme catalyzing the metabolic activation of CTU. Furthermore, CTU exhibited cytotoxic properties in cultured rat primary hepatocytes in a concentration-dependent pattern. Concomitant treatment of hepatocytes with ketoconazole mitigated their susceptibility to the cytotoxic effects of CTU.


Sujet(s)
Cytochrome P-450 CYP3A , Hépatocytes , Microsomes du foie , Animaux , Rats , Cytochrome P-450 CYP3A/métabolisme , Humains , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Mâle , Microsomes du foie/métabolisme , Rat Sprague-Dawley , Activation métabolique , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Structure moléculaire , Herbicides/toxicité , Herbicides/métabolisme , Relation dose-effet des médicaments
2.
PeerJ ; 12: e17446, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827306

RÉSUMÉ

Objectives: To investigate the interaction between tramadol and representative tyrosine kinase inhibitors, and to study the inhibition mode of drug-interaction. Methods: Liver microsomal catalyzing assay was developed. Sprague-Dawley rats were administrated tramadol with or without selected tyrosine kinase inhibitors. Samples were prepared and ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was used for analysis. Besides, liver, kidney, and small intestine were collected and morphology was examined by hematoxyline-eosin (H&E) staining. Meanwhile, liver microsomes were prepared and carbon monoxide differential ultraviolet radiation (UV) spectrophotometric quantification was performed. Results: Among the screened inhibitors, crizotinib takes the highest potency in suppressing the metabolism of tramadol in rat/human liver microsome, following non-competitive inhibitory mechanism. In vivo, when crizotinib was co-administered, the AUC value of tramadol increased compared with the control group. Besides, no obvious pathological changes were observed, including cell morphology, size, arrangement, nuclear morphology with the levels of alanine transaminase (ALT) and aspartate transaminase (AST) increased after multiple administration of crizotinib. Meanwhile, the activities of CYP2D1 and CYP3A2 as well as the total cytochrome P450 abundance were found to be decreased in rat liver of combinational group. Conclusions: Crizotinib can inhibit the metabolism of tramadol. Therefore, this recipe should be vigilant to prevent adverse reactions.


Sujet(s)
Crizotinib , Cytochrome P-450 CYP3A , Microsomes du foie , Rat Sprague-Dawley , Tramadol , Animaux , Tramadol/pharmacologie , Crizotinib/pharmacologie , Rats , Microsomes du foie/effets des médicaments et des substances chimiques , Microsomes du foie/métabolisme , Cytochrome P-450 CYP3A/métabolisme , Mâle , Interactions médicamenteuses , Humains , Spectrométrie de masse en tandem , Famille-2 de cytochromes P450/métabolisme , Famille-2 de cytochromes P450/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/pharmacocinétique , Analgésiques morphiniques/pharmacologie
3.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38928485

RÉSUMÉ

Gyrophoric acid (GA), a lichen secondary metabolite, has attracted more attention during the last years because of its potential biological effects. Until now, its effect in vivo has not yet been demonstrated. The aim of our study was to evaluate the basic physicochemical and pharmacokinetic properties of GA, which are directly associated with its biological activities. The stability of the GA in various pH was assessed by conducting repeated UV-VIS spectral measurements. Microsomal stability in rat liver microsomes was performed using Ultra-Performance LC/MS. Binding to human serum albumin (HSA) was assessed using synchronous fluorescence spectra, and molecular docking analysis was used to reveal the binding site of GA to HSA. In the in vivo experiment, 24 Sprague-Dawley rats (Velaz, Únetice, Czech Republic) were used. The animals were divided as follows. The first group (n = 6) included healthy males as control intact rats (♂INT), and the second group (n = 6) included healthy females as controls (♀INT). Groups three and four (♂GA/n = 6 and ♀GA/n = 6) consisted of animals with daily administered GA (10 mg/kg body weight) in an ethanol-water solution per os for a one-month period. We found that GA remained stable under various pH and temperature conditions. It bonded to human serum albumin with the binding constant 1.788 × 106 dm3mol-1 to reach the target tissue via this mechanism. In vivo, GA did not influence body mass gain, food, or fluid intake during the experiment. No liver toxicity was observed. However, GA increased the rearing frequency in behavioral tests (p < 0.01) and center crossings in the elevated plus-maze (p < 0.01 and p < 0.001, respectively). In addition, the time spent in the open arm was prolonged (p < 0.01 and p < 0.001, respectively). Notably, GA was able to pass through the blood-brain barrier, indicating its ability to permeate into the brain and to stimulate neurogenesis in the hilus and subgranular zone of the hippocampus. These observations highlight the potential role of GA in influencing brain function and neurogenesis.


Sujet(s)
Simulation de docking moléculaire , Rat Sprague-Dawley , Animaux , Rats , Mâle , Femelle , Humains , Microsomes du foie/métabolisme , Concentration en ions d'hydrogène , Sérum-albumine humaine/métabolisme , Sérum-albumine humaine/composition chimique , Liaison aux protéines
4.
Toxins (Basel) ; 16(6)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38922153

RÉSUMÉ

Physiologically based pharmacokinetic (PBPK) models were utilized to investigate potential interactions between aflatoxin B1 (AFB1) and efavirenz (EFV), a non-nucleoside reverse transcriptase inhibitor drug and inducer of several CYP enzymes, including CYP3A4. PBPK simulations were conducted in a North European Caucasian and Black South African population, considering different dosing scenarios. The simulations predicted the impact of EFV on AFB1 metabolism via CYP3A4 and CYP1A2. In vitro experiments using human liver microsomes (HLM) were performed to verify the PBPK predictions for both single- and multiple-dose exposures to EFV. Results showed no significant difference in the formation of AFB1 metabolites when combined with EFV (0.15 µM) compared to AFB1 alone. However, exposure to 5 µM of EFV, mimicking chronic exposure, resulted in increased CYP3A4 activity, affecting metabolite formation. While co-incubation with EFV reduced the formation of certain AFB1 metabolites, other outcomes varied and could not be fully attributed to CYP3A4 induction. Overall, this study provides evidence that EFV, and potentially other CYP1A2/CYP3A4 perpetrators, can impact AFB1 metabolism, leading to altered exposure to toxic metabolites. The results emphasize the importance of considering drug interactions when assessing the risks associated with mycotoxin exposure in individuals undergoing HIV therapy in a European and African context.


Sujet(s)
Aflatoxine B1 , Alcynes , Benzoxazines , Cyclopropanes , Interactions médicamenteuses , Microsomes du foie , Modèles biologiques , Inhibiteurs de la transcriptase inverse , Aflatoxine B1/pharmacocinétique , Aflatoxine B1/toxicité , Humains , Benzoxazines/pharmacocinétique , Benzoxazines/métabolisme , Microsomes du foie/métabolisme , Microsomes du foie/effets des médicaments et des substances chimiques , Inhibiteurs de la transcriptase inverse/pharmacocinétique , Mâle , Cytochrome P-450 CYP3A/métabolisme , Adulte , Femelle , Cytochrome P-450 CYP1A2/métabolisme , Adulte d'âge moyen , Jeune adulte ,
5.
Biopharm Drug Dispos ; 45(3): 149-158, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38886878

RÉSUMÉ

Icaritin is a prenylflavonoid derivative of the genus Epimedium (Berberidaceae) and has a variety of pharmacological actions. Icaritin is approved by the National Medical Products Administration as an anticancer drug that exhibits efficacy and safety advantages in patients with hepatocellular carcinoma cells. This study aimed to evaluate the inhibitory effects of icaritin on UDP-glucuronosyltransferase (UGT) isoforms. 4-Methylumbelliferone (4-MU) was employed as a probe drug for all the tested UGT isoforms using in vitro human liver microsomes (HLM). The inhibition potentials of UGT1A1 and 1A9 in HLM were further tested by employing 17ß-estradiol (E2) and propofol (PRO) as probe substrates, respectively. The results showed that icaritin inhibits UGT1A1, 1A3, 1A4, 1A7, 1A8, 1A10, 2B7, and 2B15. Furthermore, icaritin exhibited a mixed inhibition of UGT1A1, 1A3, and 1A9, and the inhibition kinetic parameters (Ki) were calculated to be 3.538, 2.117, and 0.306 (µM), respectively. The inhibition of human liver microsomal UGT1A1 and 1A9 both followed mixed mechanism, with Ki values of 2.694 and 1.431 (µM). This study provides supporting information for understanding the drug-drug interaction (DDI) potential of the flavonoid icaritin and other UGT-metabolized drugs in clinical settings. In addition, the findings provide safety evidence for DDI when liver cancer patients receive a combination therapy including icaritin.


Sujet(s)
Interactions médicamenteuses , Flavonoïdes , Glucuronosyltransferase , Microsomes du foie , Glucuronosyltransferase/antagonistes et inhibiteurs , Glucuronosyltransferase/métabolisme , Humains , Flavonoïdes/pharmacologie , Microsomes du foie/métabolisme , Oestradiol/pharmacologie , Hymécromone/pharmacologie , Propofol/pharmacologie , Antienzymes/pharmacologie
6.
Toxicol Appl Pharmacol ; 489: 117016, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38925514

RÉSUMÉ

To elucidate the impact of CYP3A4 activity inhibition and genetic polymorphism on the metabolism of crizotinib. Enzymatic incubation systems for crizotinib were established, and Sprague-Dawley rats were utilized for in vivo experiments. Analytes were quantified using LC-MS/MS. Upon screening 122 drugs and natural compounds, proanthocyanidins emerged as inhibitor of crizotinib metabolism, exhibiting a relative inhibition rate of 93.7%. The IC50 values were 24.53 ± 0.32 µM in rat liver microsomes and 18.24 ± 0.12 µM in human liver microsomes. In vivo studies revealed that proanthocyanidins markedly affected the pharmacokinetic parameters of crizotinib. Co-administration led to a significant reduction in the AUC(0-t), Cmax of PF-06260182 (the primary metabolite of crizotinib), and the urinary metabolic ratio. This interaction is attributed to the mixed-type inhibition of liver microsome activity by proanthocyanidins. CYP3A4, being the principal metabolic enzyme for crizotinib, has its genetic polymorphisms significantly influencing crizotinib's pharmacokinetics. Kinetic data showed that the relative metabolic rates of crizotinib across 26 CYP3A4 variants ranged from 13.14% (CYP3A4.12, 13) to 188.57% (CYP3A4.33) when compared to the wild-type CYP3A4.1. Additionally, the inhibitory effects of proanthocyanidins varied between CYP3A4.12 and CYP3A4.33, when compared to the wild type. Our findings indicate that proanthocyanidins coadministration and CYP3A4 genetic polymorphism can significantly influence crizotinib metabolism.


Sujet(s)
Crizotinib , Cytochrome P-450 CYP3A , Interactions médicamenteuses , Microsomes du foie , Polymorphisme génétique , Rat Sprague-Dawley , Crizotinib/pharmacocinétique , Cytochrome P-450 CYP3A/génétique , Cytochrome P-450 CYP3A/métabolisme , Animaux , Humains , Mâle , Microsomes du foie/métabolisme , Microsomes du foie/enzymologie , Microsomes du foie/effets des médicaments et des substances chimiques , Rats , Pyridines/pharmacocinétique , Pyrazoles/pharmacocinétique , Pyrazoles/pharmacologie
7.
Food Chem Toxicol ; 190: 114785, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38849047

RÉSUMÉ

In drug discovery, metabolite profiling unveils biotransformation pathways and potential toxicant formation, guiding selection of candidates with optimal pharmacokinetics and safety profiles. Tazemetostat (TAZ) is employed in treating locally advanced or metastatic epithelioid sarcoma. Identification of drug metabolites are of significant importance in improving safety, efficacy and reduced toxicity of drugs. The current study aimed to investigate the comprehensive metabolic fate of TAZ using different in vivo (rat) and in vitro (RLM, HLM, HS9) models. For in vivo studies, drug was orally administered to Sprague-Dawley rats with subsequent analysis of plasma, feces and urine samples. A total of 21 new metabolites were detected across various matrices and were separated on Phenomenex kinetex C18 (2.5 µm; 150 × 4.6 mm) column using acetonitrile and 0.1% formic acid in water as mobile phase. LC-QTOF-MS/MS and NMR techniques were employed to identify and characterize the metabolites from extracted samples. The major metabolic routes found in biotransformation of TAZ were hydroxylation, N-dealkylation, N-oxidation, hydrogenation, hydrolysis and N-acetylation. In silico toxicity revealed potential immunotoxicity for TAZ and few of its metabolites. This research article is the first time to discuss the complete metabolite profiling including identification and characterization of TAZ metabolites as well as its biotransformation mechanism.


Sujet(s)
Rat Sprague-Dawley , Spectrométrie de masse en tandem , Animaux , Rats , Spectrométrie de masse en tandem/méthodes , Mâle , Chromatographie en phase liquide/méthodes , Humains , Biotransformation , Fèces/composition chimique , Microsomes du foie/métabolisme , Hydroxylation
8.
Biochem Biophys Res Commun ; 725: 150261, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-38897040

RÉSUMÉ

GOAL: The long-term goal of our research is to develop safe and effective soluble epoxide hydrolase (sEH) inhibitors. The objective of this study is to evaluate the potency and selectivity of six natural isothiocyanates (ITCs) as sEH inhibitors. METHODS: Molecular docking was used to model likely interactions between the ligands and receptors. The sEH inhibitory activity was tested using a validated fluorescence-based assay and PHOME as a substrate. To evaluate their selectivity as sEH inhibitors, the inhibitory potential of the ITCs was determined on microsomal epoxide hydrolase (mEH) and cytochrome P450 (CYP) enzymes in human liver microsomes. Probe substrates such as styrene oxide (mEH substrate) and established substrates for CYP2A6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 were used in this study. The metabolites of these substrates were analyzed using validated LC-MS/MS and HPLC-UV assays. RESULTS: Molecular Docking revealed significant differences in binding site preference among the ITCs in silico and pointed to important interactions between the ligands and the catalytic residues of the sEH enzyme. In vitro, the ITCs showed varying degrees of sEH inhibition, but sulforaphane (SFN) and phenyl isothiocyanate (PITC) were the most potent inhibitors with IC50 values of 3.65 and 7.5 µM, respectively. mEH was not significantly inhibited by any of the ITCs. Erucin and iberin were the only ITCs that did not inhibit the activity of any of the tested CYP enzymes. CONCLUSION: Our results demonstrate that natural ITCs have the potential to offer safe, selective, and potent sEH inhibition.


Sujet(s)
Antienzymes , Epoxide hydrolase , Isothiocyanates , Microsomes du foie , Simulation de docking moléculaire , Epoxide hydrolase/antagonistes et inhibiteurs , Epoxide hydrolase/métabolisme , Epoxide hydrolase/composition chimique , Isothiocyanates/pharmacologie , Isothiocyanates/composition chimique , Isothiocyanates/métabolisme , Humains , Microsomes du foie/enzymologie , Microsomes du foie/métabolisme , Microsomes du foie/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Antienzymes/composition chimique , Solubilité
9.
Antimicrob Agents Chemother ; 68(7): e0014324, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38899927

RÉSUMÉ

In response to the spread of artemisinin (ART) resistance, ART-based hybrid drugs were developed, and their activity profile was characterized against drug-sensitive and drug-resistant Plasmodium falciparum parasites. Two hybrids were found to display parasite growth reduction, stage-specificity, speed of activity, additivity of activity in drug combinations, and stability in hepatic microsomes of similar levels to those displayed by dihydroartemisinin (DHA). Conversely, the rate of chemical homolysis of the peroxide bonds is slower in hybrids than in DHA. From a mechanistic perspective, heme plays a central role in the chemical homolysis of peroxide, inhibiting heme detoxification and disrupting parasite heme redox homeostasis. The hybrid exhibiting slow homolysis of peroxide bonds was more potent in reducing the viability of ART-resistant parasites in a ring-stage survival assay than the hybrid exhibiting fast homolysis. However, both hybrids showed limited activity against ART-induced quiescent parasites in the quiescent-stage survival assay. Our findings are consistent with previous results showing that slow homolysis of peroxide-containing drugs may retain activity against proliferating ART-resistant parasites. However, our data suggest that this property does not overcome the limited activity of peroxides in killing non-proliferating parasites in a quiescent state.


Sujet(s)
Antipaludiques , Artémisinines , Plasmodium falciparum , Artémisinines/pharmacologie , Antipaludiques/pharmacologie , Plasmodium falciparum/effets des médicaments et des substances chimiques , Résistance aux substances/effets des médicaments et des substances chimiques , Microsomes du foie/métabolisme , Humains , Tests de sensibilité parasitaire , Animaux , Peroxydes/pharmacologie
10.
Drug Metab Dispos ; 52(7): 634-643, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38830773

RÉSUMÉ

Drug metabolite identification is an integrated part of drug metabolism and pharmacokinetics studies in drug discovery and development. Definitive identification of metabolic modification sides of test compounds such as screening metabolic soft spots and supporting metabolite synthesis are often required. Currently, liquid chromatography-high resolution mass spectrometry is the dominant analytical platform for metabolite identification. However, the interpretation of product ion spectra generated by commonly used collision-induced disassociation (CID) and higher-energy collisional dissociation (HCD) often fails to identify locations of metabolic modifications, especially glucuronidation. Recently, a ZenoTOF 7600 mass spectrometer equipped with electron-activated dissociation (EAD-HRMS) was introduced. The primary objective of this study was to apply EAD-HRMS to identify metabolism sites of vepdegestrant (ARV-471), a model compound that consists of multiple functional groups. ARV-471 was incubated in dog liver microsomes and 12 phase I metabolites and glucuronides were detected. EAD generated unique product ions via orthogonal fragmentation, which allowed for accurately determining the metabolism sites of ARV-471, including phenol glucuronidation, piperazine N-dealkylation, glutarimide hydrolysis, piperidine oxidation, and piperidine lactam formation. In contrast, CID and HCD spectral interpretation failed to identify modification sites of three O-glucuronides and three phase I metabolites. The results demonstrated that EAD has significant advantages over CID and HCD in definitive structural elucidation of glucuronides and phase I metabolites although the utility of EAD-HRMS in identifying various types of drug metabolites remains to be further evaluated. SIGNIFICANCE STATEMENT: Definitive identification of metabolic modification sites by liquid chromatography-high resolution mass spectrometry is highly needed in drug metabolism research, such as screening metabolic soft spots and supporting metabolite synthesis. However, commonly used collision-induced dissociation (CID) and higher-energy collisional dissociation (HCD) fragmentation techniques often fail to provide critical information for definitive structural elucidation. In this study, the electron-activated dissociation (EAD) was applied to identifying glucuronidation and oxidative metabolism sites of vepdegestrant, which generated significantly better results than CID and HCD.


Sujet(s)
Glucuronides , Microsomes du foie , Oxydoréduction , Animaux , Microsomes du foie/métabolisme , Glucuronides/métabolisme , Chiens , Chromatographie en phase liquide/méthodes , Spectrométrie de masse/méthodes , Spectrométrie de masse en tandem/méthodes , Chromatographie en phase liquide à haute performance/méthodes
11.
J Pharm Biomed Anal ; 247: 116241, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38838440

RÉSUMÉ

Senecio scandens Buch.-Ham., a traditional Chinese medicine commonly used clinically, exhibits various pharmacological properties, including anti-inflammatory, anti-tumor, antiviral, and antibacterial activities. However, its water extracts' chemical components and metabolites are inadequately understood, limiting further research. In this study, the chemical components and metabolism processes of Senecio scandens, both in vivo (plasma, feces, urine, and bile) and in vitro (gut microbiota and liver microsomes), were characterized based on ultra-high performance liquid chromatography coupled with hybrid quadrupole-orbitrap high-resolution mass spectrometry. Additionally, metabolites detectable in fecal samples and intestinal microbiota incubated but absent in liver microsomes were identified as characteristic metabolites of intestinal microbiota. The targets of the characteristic metabolites of intestinal microbiota were collected, followed by exploration of potential pathways through KEGG enrichment analysis. As a result, a total of 133 chemical components were preliminarily identified, including 35 organic acids, 21 alkaloids, 19 flavonoids and their glycosides, 17 phenylpropanoids, 10 jacaranda ketones, and 31 other compounds. Notably, 12 of these were potentially novel compounds. In addition, 39 prototype components in rats and 109 metabolites were identified and characterized, including 102 in vivo and 52 metabolites in vitro (51 in rat gut microbiota and 24 in rat liver microsomes). The main metabolic pathways include oxidation, reduction, hydrolysis, methylation, glucuronidation, sulfonation, and acetylation reactions. Furthermore, KEGG enrichment analysis revealed that the characteristic metabolites of intestinal microbiota may be related to the ErbB, FoxO, mTOR, and MAPK signaling pathways, exhibiting anti-inflammatory and anti-tumor effects. In summary, the chemical components and metabolites of Senecio scandens were comprehensively identified using a rapid and accurate method, providing a scientific basis for the in-depth study of the material basis and its clinical application of Senecio scandens.


Sujet(s)
Biotransformation , Biologie informatique , Fèces , Microbiome gastro-intestinal , Microsomes du foie , Senecio , Microbiome gastro-intestinal/physiologie , Animaux , Chromatographie en phase liquide à haute performance/méthodes , Rats , Fèces/microbiologie , Fèces/composition chimique , Microsomes du foie/métabolisme , Senecio/composition chimique , Biologie informatique/méthodes , Mâle , Rat Sprague-Dawley , Médicaments issus de plantes chinoises/métabolisme , Médecine traditionnelle chinoise/méthodes , Spectrométrie de masse/méthodes
12.
Biochem Pharmacol ; 225: 116322, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38815630

RÉSUMÉ

Xenobiotic metabolic reactions in the hepatocyte endoplasmic reticulum (ER) including UDP-glucuronosyltransferase and carboxylesterase play central roles in the detoxification of medical agents with small- and medium-sized molecules. Although the catalytic sites of these enzymes exist inside of ER, the molecular mechanism for membrane permeation in the ER remains enigmatic. Here, we investigated that organic anion transporter 2 (OAT2) regulates the detoxification reactions of xenobiotic agents including anti-cancer capecitabine and antiviral zidovudine, via the permeation process across the ER membrane in the liver. Pharmacokinetic studies in patients with colorectal cancer revealed that the half-lives of capecitabine in rs2270860 (1324C > T) variants was 1.4 times higher than that in the C/C variants. Moreover, the hydrolysis of capecitabine to 5'-deoxy-5-fluorocytidine in primary cultured human hepatocytes was reduced by OAT2 inhibitor ketoprofen, whereas capecitabine hydrolysis directly assessed in human liver microsomes were not affected. The immunostaining of OAT2 was merged with ER marker calnexin in human liver periportal zone. These results suggested that OAT2 is involved in distribution of capecitabine into ER. Furthermore, we clarified that OAT2 plays an essential role in drug-drug interactions between zidovudine and valproic acid, leading to the alteration in zidovudine exposure to the body. Our findings contribute to mechanistically understanding medical agent detoxification, shedding light on the ER membrane permeation process as xenobiotic metabolic machinery to improve chemical changes in hydrophilic compounds.


Sujet(s)
Réticulum endoplasmique , Humains , Réticulum endoplasmique/métabolisme , Interactions médicamenteuses/physiologie , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Mâle , Transporteurs d'anions organiques sodium-indépendants/métabolisme , Transporteurs d'anions organiques sodium-indépendants/génétique , Zidovudine/métabolisme , Zidovudine/pharmacocinétique , Femelle , Microsomes du foie/métabolisme
13.
Drug Metab Dispos ; 52(7): 690-702, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38719744

RÉSUMÉ

Brepocitinib is an oral once-daily Janus kinase 1 and Tyrosine kinase 2 selective inhibitor currently in development for the treatment of several autoimmune disorders. Mass balance and metabolic profiles were determined using accelerator mass spectrometry in six healthy male participants following a single oral 60 mg dose of 14C-brepocitinib (∼300 nCi). The average mass balance recovery was 96.7% ± 6.3%, with the majority of dose (88.0% ± 8.0%) recovered in urine and 8.7% ± 2.1% of the dose recovered in feces. Absorption of brepocitinib was rapid, with maximal plasma concentrations of total radioactivity and brepocitinib achieved within 0.5 hours after dosing. Circulating radioactivity consisted primarily of brepocitinib (47.8%) and metabolite M1 (37.1%) derived from hydroxylation at the C5' position of the pyrazole ring. Fractional contributions to metabolism via cytochrome P450 enzymes were determined to be 0.77 for CYP3A4/5 and 0.14 for CYP1A2 based on phenotyping studies in human liver microsomes. However, additional clinical studies are required to understand the potential contribution of CYP1A1. Approximately 83% of the dose was eliminated as N-methylpyrazolyl oxidative metabolites, with 52.1% of the dose excreted as M1 alone. Notably, M1 was not observed as a circulating metabolite in earlier metabolic profiling of human plasma from a multiple ascending dose study with unlabeled brepocitinib. Mechanistic studies revealed that M1 was highly unstable in human plasma and phosphate buffer, undergoing chemical oxidation leading to loss of the 5-hydroxy-1-methylpyrazole moiety and formation of aminopyrimidine cleavage product M2. Time-dependent inhibition and trapping studies with M1 yielded insights into the mechanism of this unusual and unexpected instability. SIGNIFICANCE STATEMENT: This study provides a detailed understanding of the disposition and metabolism of brepocitinib, a JAK1/TYK2 inhibitor for atopic dermatitis, in humans as well as characterization of clearance pathways and pharmacokinetics of brepocitinib and its metabolites.


Sujet(s)
Inhibiteurs de protéines kinases , Humains , Mâle , Adulte , Inhibiteurs de protéines kinases/pharmacocinétique , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/métabolisme , Jeune adulte , Pyrazoles/pharmacocinétique , Pyrazoles/métabolisme , Pyrazoles/sang , Pyrazoles/administration et posologie , Janus kinase 1/antagonistes et inhibiteurs , Janus kinase 1/métabolisme , Administration par voie orale , Cytochrome P-450 CYP3A/métabolisme , Volontaires sains , Microsomes du foie/métabolisme , Kinase Janus-2/antagonistes et inhibiteurs , Kinase Janus-2/métabolisme , Fèces/composition chimique , Hydroxylation , Cytochrome P-450 CYP1A2/métabolisme , Adulte d'âge moyen
14.
Drug Metab Dispos ; 52(7): 654-661, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38729662

RÉSUMÉ

The delicate balance between ischemic and bleeding risks is a critical factor in antiplatelet therapy administration. Clopidogrel and prasugrel, belonging to the thienopyridine class of antiplatelet drugs, are known for their variability in individual responsiveness and high incidence of bleeding events, respectively. The present study is centered on the development and assessment of a range of deuterated thienopyridine derivatives, leveraging insights from structure-pharmacokinetic relationships of clopidogrel and prasugrel. Our approaches were grounded in the molecular framework of clopidogrel and incorporated the C2-pharmacophore design from prasugrel. The selection of ester or carbamate substituents at the C2-position facilitated the generation of the 2-oxointermediate through hydrolysis, akin to prasugrel, thereby bypassing the issue of CYP2C19 dependency. The bulky C2-pharmacophore in our approach distinguishes itself from prasugrel's acetyloxy substituent by exhibiting a moderated hydrolysis rate, resulting in a more gradual formation of the active metabolite. Excessive and rapid release of the active metabolite, believed to be linked with an elevated risk of bleeding, is thus mitigated. Our proposed structural modification retains the hydrolysis-sensitive methyl ester of clopidogrel but substitutes it with a deuterated methyl group, shown to effectively reduce metabolic deactivation. Three promising compounds demonstrated a pharmacokinetic profile similar to that of clopidogrel at four times the dose, while also augmenting its antiplatelet activity. SIGNIFICANCE STATEMENT: Inspired by the structure-pharmacokinetic relationship of clopidogrel and prasugrel, a range of clopidogrel derivatives were designed, synthesized, and assessed. Among them, three promising compounds have been identified, striking a delicate balance between efficacy and safety for antiplatelet therapy. Additionally, the ozagrel prodrug conjugate was discovered to exert a synergistic therapeutic effect alongside clopidogrel.


Sujet(s)
Clopidogrel , Antiagrégants plaquettaires , Chlorhydrate de prasugrel , Clopidogrel/pharmacocinétique , Clopidogrel/pharmacologie , Antiagrégants plaquettaires/pharmacocinétique , Antiagrégants plaquettaires/pharmacologie , Antiagrégants plaquettaires/composition chimique , Humains , Chlorhydrate de prasugrel/pharmacocinétique , Chlorhydrate de prasugrel/pharmacologie , Cytochrome P-450 CYP2C19/métabolisme , Relation structure-activité , Activation métabolique , Mâle , Hydrolyse , Microsomes du foie/métabolisme , Microsomes du foie/effets des médicaments et des substances chimiques
15.
Environ Pollut ; 355: 124214, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38801883

RÉSUMÉ

Fenpropidin (FPD), a widely employed chiral fungicide, is frequently detected in diverse environments. In an in vitro rat liver microsomes cultivation (RLMs), the metabolism exhibited the order of R-FPD > S-FPD, with respective half-lives of 10.42 ± 0.11 and 12.06 ± 0.15 min, aligning with kinetic analysis results. CYP3A2 has been demonstrated to be the most significant oxidative enzyme through CYP450 enzyme inhibition experiments. Molecular dynamics simulations unveiled the enantioselective metabolic mechanism, demonstrating that R-FPD forms hydrogen bonds with the CYP3A2 protein, resulting in a higher binding affinity (-6.58 kcal mol-1) than S-FPD. Seven new metabolites were identified by Liquid chromatography time-of-flight high-resolution mass spectrometry, which were mainly generated through oxidation, reduction, hydroxylation, and N-dealkylation reactions. The toxicity of the major metabolites predicted by the TEST procedure was found to be stronger than the predicted toxicity of FPD. Moreover, the enantioselective fate of FPD was studied by examining its degradation in three soils with varying physical and chemical properties under aerobic, anaerobic, and sterile conditions. Enantioselective degradation of FPD occurred in soils without enantiomeric transformation, displaying a preference for R-FPD degradation. R-FPD is a low-risk stereoisomer both in the environment and in mammals. The research presented a systematic and comprehensive method for analyzing the metabolic and degradation system of FPD enantiomers. This approach aids in understanding the behavior of FPD in the environment and provides valuable insights into their potential risks to human health.


Sujet(s)
Fongicides industriels , Microsomes du foie , Microsomes du foie/métabolisme , Animaux , Rats , Fongicides industriels/métabolisme , Fongicides industriels/composition chimique , Humains , Polluants du sol/métabolisme , Stéréoisomérie , Appréciation des risques
16.
J Agric Food Chem ; 72(19): 10897-10908, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38691522

RÉSUMÉ

Gramine (GRM), which occurs in Gramineae plants, has been developed to be a biological insecticide. Exposure to GRM was reported to induce elevations of serum ALT and AST in rats, but the mechanisms of the observed hepatotoxicity have not been elucidated. The present study aimed to identify reactive metabolites that potentially participate in the toxicity. In rat liver microsomal incubations fortified with glutathione or N-acetylcysteine, one oxidative metabolite (M1), one glutathione conjugate (M2), and one N-acetylcysteine conjugate (M3) were detected after exposure to GRM. The corresponding conjugates were detected in the bile and urine of rats after GRM administration. CYP3A was the main enzyme mediating the metabolic activation of GRM. The detected GSH and NAC conjugates suggest that GRM was metabolized to a quinone imine intermediate. Both GRM and M1 showed significant toxicity to rat primary hepatocytes.


Sujet(s)
Activation métabolique , Cytochrome P-450 CYP3A , Hépatocytes , Rat Sprague-Dawley , Animaux , Rats , Mâle , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Cytochrome P-450 CYP3A/métabolisme , Cytochrome P-450 CYP3A/génétique , Microsomes du foie/métabolisme , Glutathion/métabolisme , Insecticides/toxicité , Insecticides/métabolisme , Alcaloïdes/métabolisme
17.
Chem Res Toxicol ; 37(6): 935-943, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38761382

RÉSUMÉ

Amitriptyline (ATL), a tricyclic antidepressant, has been reported to cause various adverse effects, particularly hepatotoxicity. The mechanisms of ATL-induced hepatotoxicity remain unknown. The study was performed to identify the olefin epoxidation metabolite of ATL and determine the possible toxicity mechanism. Two glutathione (GSH) conjugates (M1 and M2) and two N-acetylcysteine (NAC) conjugates (M3 and M4) were detected in rat liver microsomal incubations supplemented with GSH and NAC, respectively. Moreover, M1/M2 and M3/M4 were respectively found in ATL-treated rat primary hepatocytes and in bile and urine of rats given ATL. Recombinant P450 enzyme incubations demonstrated that CYP3A4 was the primary enzyme involved in the olefin epoxidation of ATL. Treatment of hepatocytes with ATL resulted in significant cell death. Inhibition of CYP3A attenuated the susceptibility to the observed cytotoxicity of ATL. The metabolic activation of ATL most likely participates in the cytotoxicity of ATL.


Sujet(s)
Amitriptyline , Cytochrome P-450 CYP3A , Composés époxy , Hépatocytes , Microsomes du foie , Rat Sprague-Dawley , Animaux , Amitriptyline/métabolisme , Rats , Cytochrome P-450 CYP3A/métabolisme , Microsomes du foie/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Mâle , Composés époxy/métabolisme , Composés époxy/toxicité , Composés époxy/composition chimique , Glutathion/métabolisme , Cellules cultivées
18.
Toxicology ; 505: 153828, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38740169

RÉSUMÉ

The fungicide fluxapyroxad (BAS 700 F) has been shown to significantly increase the incidence of liver tumours in male Wistar rats at dietary levels of 1500 and 3000 ppm and in female rats at a dietary level of 3000 ppm via a non-genotoxic mechanism. In order to elucidate the mode of action (MOA) for fluxapyroxad-induced rat liver tumour formation a series of in vivo and in vitro investigative studies were undertaken. The treatment of male and female Wistar rats with diets containing 0 (control), 50, 250, 1500 and 3000 ppm fluxapyroxad for 1, 3, 7 and 14 days resulted in a dose-dependent increases in relative weight at 1500 and 3000 ppm from day 3 onwards in both sexes, with an increase in relative liver weight being also observed in male rats given 250 ppm fluxapyroxad for 14 days. Examination of liver sections revealed a centrilobular hepatocyte hypertrophy in some fluxapyroxad treated male and female rats. Hepatocyte replicative DNA synthesis (RDS) was significantly increased in male rats given 1500 and 3000 ppm fluxapyroxad for 3 and 7 days and in female rats given 50-3000 ppm fluxapyroxad for 7 days and 250-3000 ppm fluxapyroxad for 3 and 14 days; the maximal increases in RDS in both sexes being observed after 7 days treatment. The treatment of male and female Wistar rats with 250-3000 ppm fluxapyroxad for 14 days resulted in significant increases in hepatic microsomal total cytochrome P450 (CYP) content and CYP2B subfamily-dependent enzyme activities. Male Wistar rat hepatocytes were treated with control medium and medium containing 1-100 µM fluxapyroxad or 500 µM sodium phenobarbital (NaPB) for 4 days. Treatment with fluxapyroxad and NaPB increased CYP2B and CYP3A enzyme activities and mRNA levels but had little effect on markers of CYP1A and CYP4A subfamily enzymes and of the peroxisomal fatty acid ß-oxidation cycle. Hepatocyte RDS was significantly increased by treatment with fluxapyroxad, NaPB and 25 ng/ml epidermal growth factor (EGF). The treatment of hepatocytes from two male human donors with 1-100 µM fluxapyroxad or 500 µM NaPB for 4 days resulted in some increases in CYP2B and CYP3A enzyme activities and CYP mRNA levels but had no effect on hepatocyte RDS, whereas treatment with EGF resulted in significant increase in RDS in both human hepatocyte preparations. Hepatocytes from male Sprague-Dawley wild type (WT) and constitutive androstane receptor (CAR) knockout (CAR KO) rats were treated with control medium and medium containing 1-16 µM fluxapyroxad or 500 µM NaPB for 4 days. While both fluxapyroxad and NaPB increased CYP2B enzyme activities and mRNA levels in WT hepatocytes, only minor effects were observed in CAR KO rat hepatocytes. Treatment with both fluxapyroxad and NaPB only increased RDS in WT and not in CAR KO rat hepatocytes, whereas treatment with EGF increased RDS in both WT and CAR KO rat hepatocytes. In conclusion, a series of in vivo and in vitro investigative studies have demonstrated that fluxapyroxad is a CAR activator in rat liver, with similar properties to the prototypical CAR activator phenobarbital. A robust MOA for fluxapyroxad-induced rat liver tumour formation has been established. Based on the lack of effect of fluxapyroxad on RDS in human hepatocytes, it is considered that the MOA for fluxapyroxad-induced liver tumour formation is qualitatively not plausible for humans.


Sujet(s)
Récepteur constitutif des androstanes , Fongicides industriels , Hépatocytes , Rat Wistar , Récepteurs cytoplasmiques et nucléaires , Animaux , Mâle , Femelle , Rats , Fongicides industriels/toxicité , Récepteurs cytoplasmiques et nucléaires/métabolisme , Récepteurs cytoplasmiques et nucléaires/génétique , Humains , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Relation dose-effet des médicaments , Taille d'organe/effets des médicaments et des substances chimiques , Tumeurs expérimentales du foie/induit chimiquement , Tumeurs expérimentales du foie/anatomopathologie , Tumeurs expérimentales du foie/métabolisme , Réplication de l'ADN/effets des médicaments et des substances chimiques , Cytochrome P-450 enzyme system/métabolisme , Cytochrome P-450 enzyme system/génétique , Microsomes du foie/effets des médicaments et des substances chimiques , Microsomes du foie/métabolisme , Tumeurs du foie/induit chimiquement , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie
19.
J Pharm Biomed Anal ; 246: 116190, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38735208

RÉSUMÉ

SR9009, a peroxisome proliferator-activated receptor δ (PPARδ) agonist, is known for its potential benefits in energy homeostasis. It failed to receive the United States Food and Drug Administration (USFDA) approval and its illegal distribution has raised concerns. As a result, it has been classified as a prohibited substance by the World Anti-Doping Agency and the International Federation of Horseracing Authorities (IFHA). This study emphasizes the application of the in-silico molecular networking technology to analyze phase I drug metabolites in horses, distinguishing it from conventional methodologies in forensic science. Feature-based molecular networking (FBMN) analysis identified 15 metabolites, with novel major N-dealkylated metabolite (-C8H7NO4S), indicative of diverse metabolic modifications in horse liver microsomes incubation assay. Additionally, a proposed metabolic pathway of SR9009 in the in vitro assay was outlined, including the previously known dehydroxylated metabolite. Finally, the metabolic pathways included in this study were as follows: hydroxylation, dehydrogenation, N-dealkylation dihydroxylation, and combinations. Molecular networking provided insights into MS spectra connectivity, facilitating rapid interpretation and accurate detection of previously undiscovered metabolites. In conclusion, this study contributes to the understanding of SR9009 metabolism in horses and underscores the importance of advanced analytical techniques, such as molecular networking, in enhancing the accuracy and efficiency of metabolite analysis for forensic and doping control purposes.


Sujet(s)
Dopage sportif , Microsomes du foie , Equus caballus , Dopage sportif/prévention et contrôle , Dopage sportif/méthodes , Microsomes du foie/métabolisme , Animaux , Voies et réseaux métaboliques , Détection d'abus de substances/méthodes , Spectrométrie de masse en tandem/méthodes
20.
J Pharm Biomed Anal ; 246: 116221, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38759324

RÉSUMÉ

Lumateperone is a novel agent approved by FDA for treatment of schizophrenia in adults. To elucidate the species differences in the of biotransformation of lumateperone and its pharmacokinetic (PK) characteristics in rats, the metabolite identification of lumateperone was carried out in rat, dog and human liver microsomes, and rat plasma after oral administration using UPLC-Q Exactive Orbitrap high-resolution mass spectrometry HRMS. Furtherly, the PK characteristics of lumateperone and its N-demethylated metabolite (M3) in rat plasma were investigated using a validated LC-MS/MS method following intravenous and oral administration. Fourteen phase I metabolites were found in liver microsomes and ten of them were observed in rat plasma. N-demethylation, carbonylation, dehydrogenation, and piperazine ring cleavage were main metabolic pathway of lumateperone. No unique metabolites were formed in human liver microsomes. After rapid absorption in rats, lumateperone was quickly metabolized and eliminated with bioavailability of less than 5%. The exposure level of M3 was about 1.5-fold higher than that of lumateperone in rat plasma. Lumatperone underwent extensive metabolism and was absorbed rapidly in rats. Metabolite M3 had equivalent or slightly higher exposure levels than lumateperone. This study provides essential PK information to facilitate further pharmacodynamic researches of lumateperone.


Sujet(s)
Microsomes du foie , Rat Sprague-Dawley , Spectrométrie de masse en tandem , Animaux , Microsomes du foie/métabolisme , Spectrométrie de masse en tandem/méthodes , Chiens , Rats , Humains , Mâle , Chromatographie en phase liquide à haute performance/méthodes , Administration par voie orale , Biodisponibilité , Chromatographie en phase liquide/méthodes , Neuroleptiques/pharmacocinétique , Neuroleptiques/sang , Neuroleptiques/administration et posologie , Biotransformation , Pipérazines/pharmacocinétique , Pipérazines/sang ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...