Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Chem Biol Interact ; 382: 110644, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37499995

ABSTRACT

Cytochrome P450 4A11 (CYP4A11) has many endogenous and exogenous compounds containing a carboxyl group in their structure as substrates. If drugs with this characteristic potently attenuate the catalytic function of CYP4A11, drug-drug interactions may occur. Acidic non-steroidal anti-inflammatory drugs (NSAIDs) possess a carboxylic acid in their structure. However, it remains unclear whether these drugs inhibit CYP4A11 activity. The present study examined the inhibitory effects of acidic NSAIDs on CYP4A11 activity using human liver microsomes (HLMs) and recombinant CYP4A11. Sulindac sulfide, ibuprofen, and flurbiprofen effectively decreased the luciferin-4A O-demethylase activity of HLMs and recombinant CYP4A11 (inhibition rates of 30-96% at an inhibitor concentration of 100 µM), while salicylic acid, aspirin, diclofenac, mefenamic acid, indomethacin, etodolac, ketoprofen, loxoprofen, S-naproxen, pranoprofen, zaltoprofen, and oxaprozin exhibited weaker inhibitory activity (inhibition rates up to 23%). Among the drugs tested, sulindac sulfide was the most potent inhibitor of CYP4A11 activity. A kinetic analysis of the inhibition of CYP4A11 by sulindac sulfide revealed mixed-type inhibition for HLMs (Ki = 3.38 µM) and recombinant CYP4A11 (Ki = 4.19 µM). Sulindac sulfide is a pharmacologically active metabolite of sulindac (sulfoxide form), which is also oxidized to sulindac sulfone. To elucidate the role of a sulfur atom of sulindac sulfide in the inhibition of CYP4A11, the inhibitory effects of sulindac sulfide and its oxidized forms on CYP4A11 activity were examined. The potency of inhibition against HLMs was greater in the order of sulindac sulfide, sulindac, and sulindac sulfone; IC50 values were 6.16, 52.7, and 71.6 µM, respectively. The present results indicate that sulindac sulfide is a potent inhibitor of CYP4A11. These results and the molecular modeling of CYP4A11 with sulindac sulfide and its oxidized forms suggest that a sulfur atom of sulindac sulfide as well as its carboxylic acid play important roles in the inhibition of CYP4A11.


Subject(s)
Carboxylic Acids , Sulindac , Humans , Sulindac/pharmacology , Sulindac/metabolism , Kinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology
2.
J Clin Pharm Ther ; 47(1): 70-80, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34664726

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Drug-induced long QT syndrome (diLQTS) is a rare but serious adverse drug reaction. Drug-drug interaction (DDI) is one of the risk factors for the development of diLQTS. However, the combinations of drugs that increase the risk of diLQTS have not been extensively investigated. This study was performed to analyse the potential DDIs that elevate the incidence of diLQTS using a spontaneous reporting system. METHODS: The Japanese Adverse Drug Event Report database from April 2004 to January 2020 was used to assess adverse event reports. We calculated the reporting odds ratio and 95% confidence interval for signal detection. RESULTS AND DISCUSSION: Signals for concomitant use risk were detected in 31 drug combinations. Combinations of antipsychotics and antidepressants were the most common (olanzapine & fluvoxamine, olanzapine & trazodone, quetiapine & paroxetine, sulpiride & fluvoxamine, sulpiride & trazodone). Sixteen, 17 and 21 combinations were designated as requiring precaution for concomitant use in at least one of the package inserts in Japan, the United States and the United Kingdom, respectively, although no such precautions were described for the remaining combinations. On the contrary, a combination of bepridil & clarithromycin was categorized as "X (avoid combination)" and two combinations (chlorpromazine & haloperidol, amiodarone & metildigoxin) were classified as "D (modify regimen)" in the Lexicomp® risk rating. WHAT IS NEW AND CONCLUSION: This study identified 31 combinations of drugs that may elevate the risk of diLQTS. The use of these drug combinations should be monitored more carefully in future.


Subject(s)
Adverse Drug Reaction Reporting Systems/statistics & numerical data , Long QT Syndrome/chemically induced , Drug Interactions , Humans , Japan , Pharmacovigilance , Risk Factors
3.
Drug Metab Pharmacokinet ; 36: 100364, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33341662

ABSTRACT

The inhibition of CYP2C9-mediated warfarin metabolism by acid or lactone forms of statin converted in the body and effects of CYP2C9 genetic variants on their inhibition are not fully understood. Here, the effects of acid and lactone forms of statins on S-warfarin 7-hydroxylation were investigated in vitro. S-Warfarin 7-hydroxylase activities of human liver microsomes (HLMs), recombinant CYP2C9.1 (rCYP2C9.1), and rCYP2C9.3 (Ile359Leu variant) in the presence of statins were determined by high-performance liquid chromatography. Lactone forms of atorvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin inhibited the activity of HLMs more potently than the corresponding acid forms, whereas fluvastatin acid showed stronger inhibition than fluvastatin lactone. When the effects of statins on rCYP2C9 variants were examined, inhibition profiles of acid versus lactone forms of statins except for fluvastatin were similar between rCYP2C9.1 and rCYP2C9.3. However, the degrees of inhibition by atorvastatin lactone, fluvastatin acid, fluvastatin lactone, lovastatin lactone, and pitavastatin lactone (Ki values) were significantly different between these variants. These results indicated that lactone forms of statins other than fluvastatin showed more potent inhibition of CYP2C9-catalyzed S-warfarin 7-hydroxylation than the corresponding acid forms. Furthermore, our results indicated that Ile359Leu substitution in CYP2C9 affected the inhibitory potencies of statins.


Subject(s)
Cytochrome P-450 CYP2C9/genetics , Cytochrome P-450 CYP2C9/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Microsomes, Liver/metabolism , Pharmacogenomic Variants/physiology , Warfarin/metabolism , Acids/metabolism , Catalysis , Dose-Response Relationship, Drug , Drug Interactions/physiology , Female , Humans , Hydroxylation/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Lactones/metabolism , Male , Microsomes, Liver/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Warfarin/pharmacology
4.
Life Sci ; 258: 118140, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32730838

ABSTRACT

AIMS: This study was performed to investigate the expression profile of cytochrome P450 (CYP) isoforms and effects of polycyclic aromatic hydrocarbons (PAHs) and antiepileptic drugs on CYP1 expression in human astrocytoma MOG-G-CCM cells. MAIN METHODS: CYP1A1 and CYP1B1 expression were determined by quantitative real-time polymerase chain reaction, Western blotting, and immunocytochemistry. KEY FINDINGS: MOG-G-CCM cells expressed various CYP isoforms. Among the CYP isoforms analyzed, CYP1B1 showed the highest expression level, followed by CYP1A1. Furthermore, CYP1B1 was localized in both the endoplasmic reticulum and mitochondria. 3-Methylcholanthrene (3-MC), benz[a]anthracene (B[a]A), benzo[a]pyrene (B[a]P), and valproic acid (VPA) increased the expression of CYP1B1 and CYP1A1. The potent aryl hydrocarbon receptor antagonist GNF351 significantly suppressed the 3-MC- and VPA-mediated upregulation of CYP1B1 and CYP1A1. In addition, VPA potentiated the induction of CYP1B1 and CYP1A1 by 3-MC, B[a]A, and B[a]P, although the augmentation of CYP1A1 was more remarkable than that of CYP1B1. In contrast, other antiepileptic drugs (carbamazepine, lamotrigine, levetiracetam, phenytoin) did not affect the 3-MC-mediated upregulation of CYP1B1 and CYP1A1. VPA is known to act as a histone deacetylase (HDAC) inhibitor. Therefore, the effects of trichostatin A, a representative HDAC inhibitor, on CYP1 induction by 3-MC were examined. Trichostatin A enhanced the 3-MC-mediated upregulation of CYP1A1 but not CYP1B1. SIGNIFICANCE: These results partially indicated that VPA may augment the PAH-mediated induction of CYP1B1 and CYP1A1 through the activation of transcription by HDAC inhibition.


Subject(s)
Anticonvulsants/pharmacology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1/genetics , Polycyclic Aromatic Hydrocarbons/pharmacology , Up-Regulation/drug effects , Valproic Acid/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Humans , Transcriptome/drug effects
5.
Biol Pharm Bull ; 43(4): 688-692, 2020.
Article in English | MEDLINE | ID: mdl-32238710

ABSTRACT

Sesamin is a major lignan in sesame seeds, and a recent meta-analysis of controlled trials indicated that sesamin intake decreases blood pressure. The antihypertensive effect of sesamin has been suggested to be due to sesamin-mediated suppression of 20-hydroxyeicosatetraenoic acid production catalyzed by CYP4F2. However, the detailed mechanism underlying inhibition of CYP4F2 function by sesamin remains unclear. In this study, the effects of sesamin on catalytic activity of CYP4F2 were investigated in vitro. Sesamin inhibited luciferin-4F2/3 O-dealkylase activity of recombinant human CYP4F2 with an IC50 value of 0.381 µM. When preincubated in the presence of reduced nicotinamide adenine dinucleotide phosphate (NADPH) for 20 min, sesamin potentiated the inhibition of CYP4F2 activity. Moreover, kinetic analysis of the inactivation revealed that sesamin showed a preincubation time- and concentration-dependent inhibition of CYP4F2 activity yielding a maximal inactivation rate constant (kinact) value of 0.354 min-1 and half-maximal inhibitory concentration (KI) value of 1.12 µM. The inactivation of CYP4F2 by sesamin required NADPH. These results indicated that sesamin is a mechanism-based inactivator of human CYP4F2.


Subject(s)
Cytochrome P450 Family 4/antagonists & inhibitors , Dioxoles/pharmacology , Lignans/pharmacology , Animals , Baculoviridae , Cytochrome P450 Family 4/genetics , Cytochrome P450 Family 4/metabolism , Insecta , Microsomes/metabolism , NADP/metabolism , Recombinant Proteins/metabolism
6.
J Clin Pharm Ther ; 44(4): 565-571, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30950099

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Azole antifungal drugs are often co-administered with tacrolimus after allogeneic hematopoietic stem cell transplantation (HSCT). However, the influence of azole antifungal drugs on variation in tacrolimus pharmacokinetics when switching from intravenous tacrolimus (Tac-iv) to once-daily modified release tacrolimus (Tac-MR) remains to be elucidated. This study was performed to evaluate the effects of oral azole antifungal drugs on variation in tacrolimus pharmacokinetics after conversion to Tac-MR in HSCT patients. METHODS: Patients concomitantly receiving fluconazole (FLCZ) or voriconazole (VRCZ) along with tacrolimus were evaluated retrospectively. Blood tacrolimus concentrations before and after changing to oral administration were compared between FLCZ and VRCZ groups. RESULTS AND DISCUSSION: A total of 52 patients (34 FLCZ and 18 VRCZ) were included in the analysis. There were no significant differences in the most recent daily dose (Div ) and blood level (Civ ) of Tac-iv, Civ /Div , and ratio of daily dose of tacrolimus on the first to second day after changing to Tac-MR (Dpo1-2 ) to Div between FLCZ and VRCZ groups (P > 0.2). The trough levels of tacrolimus on the first to second day after switching to Tac-MR (Cpo1-2 ) and on the third to fifth day after the switch (Cpo3-5 ) were significantly higher in the VRCZ group than the FLCZ group (P < 0.05). The values of (Civ /Div )/(Cpo1-2 /Dpo1-2 ) and (Civ /Div )/(Cpo3-5 /Dpo3-5 ) in the VRCZ group were significantly lower compared with those in the FLCZ group (P < 0.05). Furthermore, individual values of (Civ /Div )/(Cpo3-5 /Dpo3-5 ) in the FLCZ group varied widely. WHAT IS NEW AND CONCLUSION: Voriconazole increased blood tacrolimus level more markedly than FLCZ after switching to Tac-MR, whereas FLCZ caused a large variation in tacrolimus blood level. These results suggest that therapeutic monitoring of tacrolimus after the switch may need to be performed carefully considering that orally co-administered VRCZ and FLCZ exhibit different change in blood tacrolimus level just after the switch.


Subject(s)
Antifungal Agents/therapeutic use , Azoles/therapeutic use , Tacrolimus/blood , Administration, Intravenous , Adult , Female , Fluconazole/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood , Immunosuppressive Agents/pharmacokinetics , Male , Middle Aged , Retrospective Studies , Tacrolimus/administration & dosage , Tacrolimus/pharmacokinetics , Voriconazole/therapeutic use , Young Adult
7.
Chem Biol Interact ; 306: 1-9, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30965050

ABSTRACT

The inhibitory effects of antihypertensive drugs (dihydropyridine calcium channel blockers, angiotensin II receptor blockers, and angiotensin-converting enzyme inhibitors) on cytochrome P450 2J2 (CYP2J2) activity were examined. Amlodipine, azelnidipine, barnidipine, benidipine, cilnidipine, efonidipine, felodipine, manidipine, nicardipine, nifedipine, nilvadipine, nisoldipine, nitrendipine, telmisartan, delapril, and quinapril inhibited luciferin-2J2/4F12 O-dealkylase activity of recombinant human CYP2J2 in a concentration-dependent manner (IC50 = 0.116-9.19 µM). Kinetic analyses of the inhibition indicated that azelnidipine, barnidipine, benidipine, cilnidipine, efonidipine, manidipine, nicardipine, telmisartan, delapril, and quinapril competitively inhibited CYP2J2 activity, while amlodipine, felodipine, nifedipine, nilvadipine, nisoldipine, and nitrendipine showed mixed inhibition. Among these drugs, manidipine showed the strongest reversible inhibition with Ki value of 0.0294 µM. The docking simulation data supported the potent inhibition of CYP2J2 by these drugs. Next, the effect of preincubation on CYP2J2 inhibition was investigated to determine whether these antihypertensive drugs inhibited CYP2J2 activity in a metabolism-dependent manner. A 20-min preincubation of azelnidipine and felodipine in the presence of NADPH potentiated the inhibition of CYP2J2. Furthermore, kinetic analysis of the inactivation showed that azelnidipine caused a preincubation time- and concentration-dependent decrease in CYP2J2 activity yielding kinact/KI value of 105 l/mmol/min, although felodipine showed no preincubation time-dependent inhibition. The azelnidipine-mediated inactivation required NADPH. These results indicated that manidipine is a potent competitive reversible inhibitor while azelnidipine is a potent mechanism-based inactivator of human CYP2J2.


Subject(s)
Antihypertensive Agents/pharmacology , Azetidinecarboxylic Acid/analogs & derivatives , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Dihydropyridines/pharmacology , Animals , Antihypertensive Agents/chemistry , Azetidinecarboxylic Acid/chemistry , Azetidinecarboxylic Acid/pharmacology , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme Inhibitors/chemistry , Dihydropyridines/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Nitrobenzenes , Piperazines , Recombinant Proteins/metabolism , Structure-Activity Relationship
8.
J Pharmacol Exp Ther ; 366(3): 446-457, 2018 09.
Article in English | MEDLINE | ID: mdl-29976573

ABSTRACT

The specificity of cytochrome P450 4A11 (CYP4A11) against luciferin-4A O-demethylation in human liver microsomes (HLMs) and human renal microsomes (HRMs) and selectivity of CYP4A11 inhibition by epalrestat were investigated. Kinetic analysis of luciferin-4A O-demethylation yielded Vmax and S50 values of 39.7 pmol/min per milligram protein and 43.2 µM for HLMs (Hill coefficient 1.24) and 39.4 pmol/min per milligram protein and 33.8 µM for HRMs (Hill coefficient 1.34), respectively. Among the selective CYP inhibitors tested, HET0016 (CYP4 inhibitor) exclusively inhibited luciferin-4A O-demethylation by HLMs and HRMs. Furthermore, anti-CYP4A11 antibody nearly abolished the activity of both tissue microsomes. Luciferin-4A O-demethylase activity of HLMs was significantly correlated with lauric acid ω-hydroxylase activity, a marker of CYP4A11 activity (r = 0.904, P < 0.0001). Next, effects of epalrestat on CYP-mediated drug oxidations were examined. Epalrestat showed the most potent inhibition against CYP4A11 (IC50 = 1.82 µM) among the 17 recombinant enzymes tested. The inhibitory effect of epalrestat on CYP4A11 was at least 10-fold stronger than those on CYP4F2, CYP4F3B, and CYP4F12. For known CYP4 inhibitors, in contrast, HET0016 inhibited the activities of CYP4A11 and CYP4F2 (IC50 = 0.0137-0.0182 µM); 17-octadecynoic acid reduced activities of CYP4A11, CYP4F2, CYP4F3B, and CYP4F12 to a similar extent (IC50 = 5.70-17.7 µM). Epalrestat selectively and effectively inhibited the CYP4A11 activity of HLMs (IC50 = 0.913 µM) and HRMs (IC50 = 0.659 µM). These results indicated that luciferin-4A O-demethylase activity is a good CYP4A11 marker of HLMs and HRMs, and that epalrestat is a more selective CYP4A11 inhibitor compared with known CYP4 inhibitors.


Subject(s)
Cytochrome P-450 CYP4A/antagonists & inhibitors , Cytochrome P-450 CYP4A/metabolism , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Microsomes/enzymology , Molecular Probes/metabolism , Quinolines/pharmacology , Rhodanine/analogs & derivatives , Thiazoles/pharmacology , Thiazolidines/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Kidney/cytology , Kinetics , Liver/cytology , Rhodanine/pharmacology
9.
Biochem Biophys Res Commun ; 491(2): 271-276, 2017 09 16.
Article in English | MEDLINE | ID: mdl-28739257

ABSTRACT

Peroxisome proliferator-activated receptor-γ (PPARγ) plays an important role in lipid and glucose metabolism. In this study, the function of PPARγ on lung development was investigated. Lung-specific Pparg conditional knockout mice (PpargΔLuEpC) were developed using Cre-Lox system. PpargΔLuEpC mice showed abnormal lung development with enlarged airspaces and followed by increase of apoptotic cells at E14.5 to E18.5. Gene analysis revealed that expression of Pmaip1, a gene related to apoptosis, was significantly increased while expression of Retnla, a gene related to anti-apoptosis, was dramatically decreased in the fetal lung (E14.5) of PpargΔLuEpC mice. In addition, expression of Pthlh, a gene phenotypically expressed in the congenital cystic adenomatoid malformation (CCAM), was increased at E14.5 to E18.5 in the lung of PpargΔLuEpC mice. Cell culture studies revealed that PPARγ could bind to promoter region of Pthlh gene as a repressor in the immortalized mouse lung epithelial cell line MLE-15. Surprisingly, phenotypic changes in MLE-15-shPparg cells, stably transfected with shPparg plasmid, were similar to the PpargΔLuEpC mice model. In addition, MLE-15-shPparg cells were easily detached from the cultured plate when cold phosphate buffered saline was applied. Furthermore, expression of Cdh1, a gene related to cell adhesion, was significantly reduced in the MLE-15-shPparg cells. Taken together, PPARγ may play an important role in fetal lung development via alveolar cell-to-cell adhesion system.


Subject(s)
Cystic Adenomatoid Malformation of Lung, Congenital/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , PPAR gamma/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Apoptosis , Binding Sites , Cdh1 Proteins/genetics , Cdh1 Proteins/metabolism , Cell Adhesion , Cell Line, Transformed , Cystic Adenomatoid Malformation of Lung, Congenital/metabolism , Cystic Adenomatoid Malformation of Lung, Congenital/pathology , Embryo, Mammalian , Epithelial Cells/pathology , Fetus , Genes, Reporter , Intercellular Signaling Peptides and Proteins/metabolism , Luciferases/genetics , Luciferases/metabolism , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , PPAR gamma/deficiency , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction
10.
Anesthesiology ; 125(1): 204-18, 2016 07.
Article in English | MEDLINE | ID: mdl-27176211

ABSTRACT

BACKGROUND: The aim of this study was to determine whether systemic administration of QX-314 reduces bone cancer pain through selective inhibition of transient receptor potential vanilloid subfamily 1 (TRPV1)-expressing afferents. METHODS: A mouse model of bone cancer pain was used. The authors examined the effects of bolus (0.01 to 3 mg/kg, n = 6 to 10) and continuous (5 mg kg h, n = 5) administration of QX-314 on both bone cancer pain-related behaviors and phosphorylated cyclic adenosine monophosphate response element-binding protein expression in dorsal root ganglion neurons (n = 3 or 6) and the effects of ablation of TRPV1-expressing afferents on bone cancer pain-related behaviors (n = 10). RESULTS: The numbers of flinches indicative of ongoing pain in QX-314-treated mice were smaller than those in vehicle-treated mice at 10 min (3 mg/kg, 4 ± 3; 1 mg/kg, 5 ± 3 vs. 12 ± 3; P < 0.001; n = 8 to 9), 24 h (3 ± 2 vs. 13 ± 3, P < 0.001), and 48 h (4 ± 1 vs. 12 ± 2, P < 0.001; n = 5 in each group) after QX-314 administration, but impaired limb use, weight-bearing including that examined by the CatWalk system, and rotarod performance indicative of movement-evoked pain were comparable. QX-314 selectively inhibited the increase in phosphorylated cyclic adenosine monophosphate response element-binding protein expression in TRPV1-positive, but not in TRPV1-negative, dorsal root ganglion neurons compared to that in the case of vehicle administration (32.2 ± 3.0% vs. 52.6 ± 5.9%, P < 0.001; n = 6 in each group). Ablation of TRPV1-expressing afferents mimicked the effects of QX-314. CONCLUSION: This study showed that systemic administration of QX-314 in mice inhibits some behavioral aspects of bone cancer pain through selective inhibition of TRPV1-expressing afferents without coadministration of TRPV1 agonists.


Subject(s)
Anesthetics, Local/therapeutic use , Bone Neoplasms/complications , Lidocaine/analogs & derivatives , Pain/drug therapy , Pain/etiology , TRPV Cation Channels/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Bone Neoplasms/pathology , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/biosynthesis , Ganglia, Spinal/drug effects , Lidocaine/therapeutic use , Male , Mice , Mice, Inbred C3H , Movement , Pain/psychology , Pain Measurement/drug effects , Postural Balance/drug effects , Psychomotor Performance/drug effects , Weight-Bearing
11.
Life Sci ; 136: 87-93, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26187180

ABSTRACT

AIMS: We herein investigated the inducibility of cytochrome P450 1A1 (CYP1A1) by Δ(9)-tetrahydrocannabinol, cannabidiol (CBD), and cannabinol, three major phytocannabinoids, using human hepatoma HepG2 cells. MAIN METHODS: The expression of CYP1A1 and the aryl hydrocarbon receptor (AhR) was measured by a quantitative real-time polymerase chain reaction and/or Western blotting. KEY FINDINGS: Δ(9)-Tetrahydrocannabinol and CBD concentration-dependently induced the expression of CYP1A1 mRNA, whereas cannabinol showed little or no induction. Among the phytocannabinoids tested, CBD was the most potent inducer of CYP1A1 expression. The induction of CYP1A1 expression by CBD was significantly attenuated by the knockdown of AhR expression with AhR small interfering RNAs. The role of protein tyrosine kinases (PTKs) in the CBD-mediated induction of CYP1A1 was then examined using herbimycin A, a PTK inhibitor. The upregulation of CYP1A1 by CBD was significantly suppressed by herbimycin A as was the induction by omeprazole but not 3-methylcholanthrene. The inducibility of CYP1A1 by CBD-related compounds was examined to clarify the structural requirements for CBD-mediated CYP1A1 induction. Olivetol, which corresponds to the pentylresorcinol moiety of CBD, significantly induced the expression of CYP1A1, whereas d-limonene, CBD-2'-monomethyl ether, and CBD-2',6'-dimethyl ether did not. SIGNIFICANCE: These results showed that CBD may have induced human CYP1A1 expression through the activation of PTK-dependent AhR signaling, in which two phenolic hydroxyl groups in the pentylresorcinol moiety of CBD may play structurally important roles.


Subject(s)
Cannabidiol/pharmacology , Cytochrome P-450 CYP1A1/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Cytochrome P-450 CYP1A1/genetics , Enzyme Induction/drug effects , Hep G2 Cells , Humans , Signal Transduction
12.
Biol Pharm Bull ; 38(3): 441-7, 2015.
Article in English | MEDLINE | ID: mdl-25757926

ABSTRACT

Iguratimod is a novel disease-modifying antirheumatic drug. A blue letter (safety advisory) for drug interaction between iguratimod and warfarin was issued by the Ministry of Health, Labour and Welfare of Japan in May 2013. Iguratimod may affect warfarin metabolism catalyzed by CYP. However, it is not clear whether iguratimod inhibits warfarin oxidation. This study was performed to investigate the effects of iguratimod on warfarin 7-hydroxylation with human liver microsomes (HLMs) and recombinant CYP enzymes. Iguratimod concentration-dependently inhibited R,S-warfarin 7-hydroxylase activity of HLMs with an IC50 value of 15.2 µM. The inhibitory effect was examined with S-warfarin and R-warfarin to determine which enantiomer was more potently inhibited by iguratimod. Iguratimod potently inhibited the S-warfarin 7-hydroxylase activity of HLMs with an IC50 value of 14.1 µM, but showed only slight inhibition of R-warfarin 7-hydroxylation. Furthermore, iguratimod inhibited the S-warfarin 7-hydroxylase activity of recombinant CYP2C9.1 (rCYP2C9.1) and rCYP2C9.3 in a concentration-dependent manner with IC50 values of 10.8 and 20.1 µM, respectively. Kinetic analysis of the inhibition of S-warfarin 7-hydroxylation by iguratimod indicated competitive-type inhibition for HLMs and rCYP2C9.1 but mixed-type inhibition for rCYP2C9.3. The Ki values for HLMs, rCYP2C9.1, and rCYP2C9.3 were 6.74, 4.23, and 14.2 µM, respectively. Iguratimod did not exert metabolism-dependent inhibition of S-warfarin 7-hydroxylation. These results indicated that iguratimod is a potent direct inhibitor of CYP2C9-mediated warfarin 7-hydroxylation and that its inhibitory effect on CYP2C9.1 was more sensitive than that on CYP2C9.3.


Subject(s)
Chromones/pharmacology , Cytochrome P-450 CYP2C9/metabolism , Drug Interactions , Liver/drug effects , Sulfonamides/pharmacology , Warfarin/pharmacokinetics , Aged , Antirheumatic Agents/pharmacology , Humans , Hydroxylation , Immunosuppressive Agents/pharmacology , Inhibitory Concentration 50 , Kinetics , Liver/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Stereoisomerism , Warfarin/metabolism
13.
Chem Biol Interact ; 215: 62-8, 2014 May 25.
Article in English | MEDLINE | ID: mdl-24667653

ABSTRACT

We previously demonstrated that cannabidiol (CBD) was a potent mechanism-based inhibitor of human cytochrome P450 1A1 (CYP1A1). However, the moiety of CBD that contributes to the potent mechanism-based inhibition of human CYP1A1 remains unknown. Thus, the effects of compounds structurally related to CBD on CYP1A1 activity were examined with recombinant human CYP1A1 in order to characterize the structural requirements for potent inactivation by CBD. When preincubated in the presence of NADPH for 20min, olivetol, which corresponds to the pentylresorcinol moiety of CBD, enhanced the inhibition of the 7-ethoxyresorufin O-deethylase activity of CYP1A1. In contrast, d-limonene, which corresponds to the terpene moiety of CBD, failed to inhibit CYP1A1 activity in a metabolism-dependent manner. Pentylbenzene, which lacks two free phenolic hydroxyl groups, also did not enhance CYP1A1 inhibition. On the other hand, preincubation of the CBD-2'-monomethyl ether (CBDM) and CBD-2',6'-dimethyl ether (CBDD) enhanced the inhibition of CYP1A1 activity. Inhibition by cannabidivarin (CBDV), which possessed a propyl side chain, was strongly potentiated by its preincubation. Orcinol, which has a methyl group, augmented CYP1A1 inhibition, whereas its derivative without an alkyl side chain, resorcinol, did not exhibit any metabolism-dependent inhibition. The preincubation of CBD-hydroxyquinone did not markedly enhance CYP1A1 inhibition. We further confirmed that olivetol, CBDM, CBDD, CBDV, and orcinol, as well as CBD (kinact=0.215min(-1)), inactivated CYP1A1 activity; their kinact values were 0.154, 0.0638, 0.0643, 0.226, and 0.0353min(-1), respectively. These results suggest that the methylresorcinol structure in CBD may have structurally important roles in the inactivation of CYP1A1.


Subject(s)
Cannabidiol/chemistry , Cannabidiol/pharmacology , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Animals , Enzyme Activation/drug effects , Humans , Resorcinols/chemistry , Structure-Activity Relationship
14.
Toxicology ; 314(2-3): 254-61, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24184660

ABSTRACT

Tetrahydrocannabinol (THC), a psychoactive component of marijuana, is known to exert cytotoxicity in immune cells. In the present study, we examined the cytotoxicity of Δ8-THC in mouse macrophage J774-1 cells and a possible involvement of cannabinoid receptors and stress-responsive mitogen-activated protein kinases (MAPKs) in the cytotoxic process. J774-1 cells were treated with Δ8-THC (0-20 µM) for up to 6 h. As measured by the MTT and LDH assays, Δ8-THC induced cell death of J774-1 cells in a concentration- and/or exposure time-dependent manner. Δ8-THC-induced cell damage was associated with vacuole formation, cell swelling, chromatin condensation, and nuclear fragmentation. The cytotoxic effect of Δ8-THC was significantly prevented by a caspase-1 inhibitor Ac-YVAD-cmk but not a caspase-3 inhibitor z-DEVD-fmk. The pretreatment with SR144528, a CB2 receptor-selective antagonist, effectively suppressed Δ8-THC-induced cytotoxicity in J774-1 cells, which exclusively expressed CB2 receptors as indicated by real-time polymerase chain reaction analysis. In contrast, AM251, a CB1 receptor-selective antagonist, did not affect the cytotoxicity. Pertussis toxin and α-tocopherol significantly attenuated Δ8-THC-induced cytotoxicity suggesting that G(i/o) protein coupling signal transduction and oxidative stress are responsible for the cytotoxicity. Δ8-THC stimulated the phosphorylation of p38 MAPK and c-Jun N-terminal kinase (JNK) in J774-1 cells, which were effectively antagonized by the pretreatment with SR144528. In addition, SB203580, a p38 MARK inhibitor, significantly attenuated the cytotoxic effect of Δ8-THC, whereas SP600125, a JNK inhibitor, significantly enhanced the cytotoxicity. These results suggest that the cytotoxicity of Δ8-THC to J774-1 cells is exerted mediated through the CB2 receptor followed by the activation of p38 MAPK.


Subject(s)
Dronabinol/toxicity , Macrophages/drug effects , Macrophages/metabolism , Receptor, Cannabinoid, CB2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Mice , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
15.
Biol Pharm Bull ; 36(7): 1197-203, 2013.
Article in English | MEDLINE | ID: mdl-23811569

ABSTRACT

Our recent work has shown that cannabidiol (CBD) exhibits the most potent direct inhibition of human cytochrome P450 1A1 (CYP1A1) among the CYP enzymes examined. However, the mechanism underlying this CBD inhibition remains to be clarified. Thus, to elucidate the structural requirements for the potent inhibition by CBD, the effects of CBD and its structurally related compounds on CYP1A1 activity were investigated with recombinant human CYP1A1. Olivetol, which corresponds to the pentylresorcinol moiety of CBD, inhibited the 7-ethoxyresorufin O-deethylase activity of CYP1A1; its inhibitory effect (IC50=13.8 µM) was less potent than that of CBD (IC50=0.355 µM). In contrast, d-limonene, which corresponds to the terpene moiety of CBD, only slightly inhibited CYP1A1 activity. CBD-2'-monomethyl ether (CBDM) and CBD-2',6'-dimethyl ether inhibited CYP1A1 activity with IC50 values of 4.07 and 23.0 µM, respectively, indicating that their inhibitory effects attenuated depending on the level of methylation on the free phenolic hydroxyl groups in the pentylresorcinol moiety of CBD. Cannabidivarin inhibited CYP1A1 activity, although its inhibitory potency (IC50=1.85 µM) was lower than that of CBD. The inhibitory effects of Δ(9)-tetrahydrocannabinol and cannabielsoin (IC50s ≈10 µM), which contain a free phenolic hydroxyl group and are structurally constrained, were less potent than that of CBDM, which contains a free phenolic hydroxyl group and is rotatable between pentylresorcinol and terpene moieties. These results suggest that the pentylresorcinol structure in CBD may have structurally important roles in direct CYP1A1 inhibition, although the whole structure of CBD is required for overall inhibition.


Subject(s)
Cannabidiol/chemistry , Cannabidiol/pharmacology , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Resorcinols/chemistry , Cytochrome P-450 CYP1A1/chemistry , Humans , Models, Molecular , Molecular Structure , Protein Conformation , Structure-Activity Relationship
16.
Drug Metab Pharmacokinet ; 28(4): 332-8, 2013.
Article in English | MEDLINE | ID: mdl-23318708

ABSTRACT

The present study investigated the inhibitory effect of cannabidiol (CBD), a major constituent of marijuana, on the catalytic activity of cytochrome P450 2C19 (CYP2C19). (S)-Mephenytoin 4'-hydroxylase activities of human liver microsomes (HLMs) and recombinant CYP2C19 were inhibited by CBD in a concentration-dependent manner (IC50 = 8.70 and 2.51 µM, respectively). Omeprazole 5-hydroxylase and 3-O-methylfluorescein O-demethylase activities in recombinant CYP2C19 were also strongly inhibited by CBD (IC50 = 1.55 and 1.79 µM, respectively). Kinetic analysis for inhibition revealed that CBD showed a mixed-type inhibition against (S)-mephenytoin 4'-hydroxylation by recombinant CYP2C19. To clarify the structural requirements for CBD-mediated CYP2C19 inhibition, the effects of CBD-related compounds on CYP2C19 activity were examined. Olivetol inhibited the (S)-mephenytoin 4'-hydroxylase activity of recombinant CYP2C19 with the IC50 value of 15.3 µM, whereas d-limonene slightly inhibited the activity (IC50 > 50 µM). The inhibitory effect of CBD-2'-monomethyl ether (IC50 = 1.88 µM) on CYP2C19 was comparable to that of CBD, although the inhibitory potency of CBD-2',6'-dimethyl ether (IC50 = 14.8 µM) was lower than that of CBD. Cannabidivarin, possessing a propyl side chain, showed slightly less potent inhibition (IC50 = 3.45 µM) as compared with CBD, whereas orcinol and resorcinol did not inhibit CYP2C19 activity at all. These results indicate that CBD caused potent CYP2C19 inhibition, in which one free phenolic hydroxyl group and the pentyl side chain of CBD may play important roles.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Cannabidiol/pharmacology , Cannabidiol/analogs & derivatives , Cytochrome P-450 CYP2C19 , Female , Humans , Inhibitory Concentration 50 , Kinetics , Male , Microsomes, Liver/drug effects , Omeprazole/metabolism , Recombinant Proteins/metabolism , Structure-Activity Relationship
17.
Biochim Biophys Acta ; 1830(1): 2118-28, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23041070

ABSTRACT

BACKGROUND: To investigate the function of the intestinal Vdr gene in inflammatory bowel disease (IBD), in conjunction with the discovery of possible metabolic markers for IBD using intestine-specific Vdr knockout mice. METHODS: Vdr(ΔIEpC) mice were generated, phenotyped and treated with a time-course of 3% dextran sulfate sodium (DSS) to induce colitis. Colitis was diagnosed by evaluating clinical symptoms and intestinal histopathology. Gene expression analysis was carried out. In addition, metabolic markers of IBD were explored by metabolomics. RESULTS: Vdr(ΔIEpC) mice showed abnormal body size, colon structures and feces color. Calcium, collagen, and intestinal proliferation-related gene expression were all decreased, and serum alkaline phosphatase was highly increased. In the acute model which was treated with 3% DSS for six days, Vdr(ΔIEpC) mice showed a high score of IBD symptoms; enlarged mucosal layer and damaged muscularis layer. In the recovery experiment model, where mice were treated with 3% DSS for four days and water for three days, Vdr(ΔIEpC) mice showed a high score of IBD symptoms; severe damage of mucosal layer and increased expression of genes encoding proinflammatory cytokines. Feces metabolomics revealed decreased concentrations of taurine, taurocholic acid, taurodeoxycholic acid and cholic acid in Vdr(ΔIEpC) mice. CONCLUSIONS: Disruption of the intestinal Vdr gene showed phenotypical changes that may exacerbate IBD. These results suggest that VDR may play an important role in IBD. GENERAL SIGNIFICANCE: VDR function has been implicated in IBD. This is of value for understanding the etiology of IBD and for development of diagnostic biomarkers for IBD.


Subject(s)
Gene Expression Regulation , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Receptors, Calcitriol/genetics , Animals , Biomarkers/metabolism , Dextran Sulfate/toxicity , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Mice , Mice, Mutant Strains
18.
Biol Pharm Bull ; 36(2): 292-8, 2013.
Article in English | MEDLINE | ID: mdl-23229390

ABSTRACT

We reported previously that monkey embryonic stem cells (ESCs) were differentiated into hepatocytes by formation of embryoid bodies (EBs). However, this EB formation method is not always efficient for assays using a large number of samples simultaneously. A dispersion culture system, one of the differentiation methods without EB formation, is able to more efficiently provide a large number of feeder-free undifferentiated cells. A previous study demonstrated the effectiveness of the Rho-associated kinase inhibitor Y-27632 for feeder-free dispersion culture and induction of differentiation of monkey ESCs into neural cells. In the present study, the induction of differentiation of cynomolgus monkey ESCs (cmESCs) into hepatocytes was performed by the dispersion culture method, and the expression and drug inducibility of cytochrome P450 (CYP) enzymes in these hepatocytes were examined. The cmESCs were successfully differentiated into hepatocytes under feeder-free dispersion culture conditions supplemented with Y-27632. The hepatocytes differentiated from cmESCs expressed the mRNAs for three hepatocyte marker genes (α-fetoprotein, albumin, CYP7A1) and several CYP enzymes, as measured by real-time polymerase chain reaction. In particular, the basal expression of cmCYP3A4 (3A8) in these hepatocytes was detected at mRNA and enzyme activity (testosterone 6ß-hydroxylation) levels. Furthermore, the expression and activity of cmCYP3A4 (3A8) were significantly upregulated by rifampicin. These results indicated the effectiveness of Y-27632 supplementation for feeder-free dispersed culture and induction of differentiation into hepatocytes, and the expression of functional CYP enzyme(s) in cmESC-derived hepatic cells.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Embryonic Stem Cells/cytology , Hepatocytes/cytology , Albumins/genetics , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Haplorhini , Hepatocytes/metabolism , Pregnane X Receptor , Receptors, Aryl Hydrocarbon/genetics , Receptors, Steroid/genetics , Rifampin/pharmacology , Steroid Hydroxylases/metabolism , alpha-Fetoproteins/genetics
19.
Drug Metab Pharmacokinet ; 27(3): 294-300, 2012.
Article in English | MEDLINE | ID: mdl-22166891

ABSTRACT

Inhibitory effects of Δ9-tetrahydrocannabinol (Δ9-THC), cannabidiol (CBD), and cannabinol (CBN), the three major constituents in marijuana, and polycyclic aromatic hydrocarbons (PAHs) contained in marijuana smoke on catalytic activity of human cytochrome P450 (CYP) 2C9 were investigated. These phytocannabinoids concentration-dependently inhibited S-warfarin 7-hydroxylase and diclofenac 4'-hydroxylase activities of human liver microsomes (HLMs) and recombinant CYP2C9 (rCYP2C9). In contrast, none of the twelve PAHs including benz[a]anthracene and benzo[a]pyrene exerted substantial inhibition (IC50 > 10 µM). The inhibitory potentials of Δ9-THC (Ki = 0.937-1.50 µM) and CBN (Ki = 0.882-1.29 µM) were almost equivalent regardless of the enzyme sources used, whereas the inhibitory potency of CBD (Ki > = 0.954-9.88 µM) varied depending on the enzyme sources and substrates used. Δ9-THC inhibited both S-warfarin 7-hydroxylase and diclofenac 4'-hydroxylase activities of HLMs and rCYP2C9 in a mixed manner. CBD and CBN competitively inhibited the activities of HLMs and rCYP2C9, with the only notable difference being that CBD and CBN exhibited mixed-type inhibitions against diclofenac 4'-hydroxylation and S-warfarin 7-hydroxylation, respectively, by rCYP2C9. None of Δ9-THC, CBD, and CBN exerted metabolism-dependent inhibition. These results indicated that the three major phytocannabinoids but not PAHs contained in marijuana smoke potently inhibited CYP2C9 activity and that these cannabinoids can be characterized as direct inhibitors for CYP2C9.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Cannabinoids/pharmacology , Cannabis/chemistry , Enzyme Inhibitors/pharmacology , Plant Leaves/chemistry , Polycyclic Aromatic Hydrocarbons/pharmacology , Smoke/analysis , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Cannabidiol/pharmacology , Cannabinol/pharmacology , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1A2 Inhibitors , Cytochrome P-450 CYP2C9 , Diclofenac/metabolism , Dronabinol/pharmacology , Female , Humans , Hydroxylation/drug effects , Kinetics , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Middle Aged , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Warfarin/metabolism
20.
Drug Metab Dispos ; 39(11): 2049-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21821735

ABSTRACT

Δ(9)-Tetrahydrocannabinol, cannabidiol (CBD), and cannabinol are the three major cannabinoids contained in marijuana, which are devoid of nitrogen atoms in their structures. In this study, we investigated the inhibitory effects of the major phytocannabinoids on the catalytic activity of human CYP2D6. These major cannabinoids inhibited the 3-[2-(N,N-diethyl-N-methylammonium)ethyl]-7-methoxy-4-methylcoumarin (AMMC) and dextromethorphan O-demethylase activities of recombinant CYP2D6 and pooled human liver microsomes in a concentration-dependent manner (IC(50) = 4.01-24.9 µM), indicating the strongest inhibitory potency of CBD. However, these cannabinoids showed no or weak metabolism-dependent inhibition. CBD competitively inhibited the CYP2D6 activities with the apparent K(i) values of 1.16 to 2.69 µM. To clarify the structural requirement for CBD-mediated CYP2D6 inhibition, effects of CBD-related compounds on the AMMC O-demethylase activity of recombinant CYP2D6 were examined. Olivetol (IC(50) = 7.21 µM) inhibited CYP2D6 activity as potently as CBD did (IC(50) = 6.52 µM), whereas d-limonene did not show any inhibitory effect. Pentylbenzene failed to inhibit CYP2D6 activity. Furthermore, neither monomethyl nor dimethyl ethers of CBD inhibited the activity. Cannabidivarin having a propyl side chain inhibited CYP2D6 activity; its inhibitory effect (IC(50) = 10.2 µM) was less potent than that of CBD. On the other hand, orcinol and resorcinol showed lack of inhibition. The inhibitory effect of CBD on CYP2D6 activity was more potent than those of 16 compounds without nitrogen atoms tested, such as progesterone. These results indicated that CBD caused potent direct CYP2D6 inhibition, in which two phenolic hydroxyl groups and the pentyl side chain of CBD may play important roles.


Subject(s)
Cannabidiol/analogs & derivatives , Cannabidiol/pharmacology , Cannabinoids/pharmacology , Cytochrome P-450 CYP2D6 Inhibitors , Cannabidiol/chemistry , Cannabinoids/chemistry , Coumarins/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Dronabinol/chemistry , Dronabinol/pharmacology , Humans , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Oxidoreductases, O-Demethylating/antagonists & inhibitors , Oxidoreductases, O-Demethylating/metabolism , Pharmacokinetics , Polycyclic Aromatic Hydrocarbons/metabolism , Quaternary Ammonium Compounds/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Steroids/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL