Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
PeerJ ; 12: e17446, 2024.
Article in English | MEDLINE | ID: mdl-38827306

ABSTRACT

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.


Subject(s)
Crizotinib , Cytochrome P-450 CYP3A , Tramadol , Animals , Humans , Male , Rats , Analgesics, Opioid/pharmacology , Crizotinib/pharmacology , Cytochrome P-450 CYP3A/metabolism , Cytochrome P450 Family 2/metabolism , Cytochrome P450 Family 2/antagonists & inhibitors , Drug Interactions , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Tramadol/pharmacology
2.
Future Med Chem ; 14(1): 17-34, 2022 01.
Article in English | MEDLINE | ID: mdl-34818903

ABSTRACT

Aim: Our previous results suggest that phenyl/naphthylacetyl pentanoic acid derivatives may exhibit dual MMP-2 and HDAC8 inhibitory activities and show effective cytotoxic properties. Methodology: Here, 13 new compounds (C1-C13) were synthesized and characterized. Along with these new compounds, 16 previously reported phenyl/napthylacetyl pentanoic acid derivatives (C14-C29) were biologically evaluated. Results: Compounds C6 and C27 showed good cytotoxicity against leukemia cell line Jurkat E6.1. The mechanisms of cytotoxicity of these compounds were confirmed by DNA deformation assay and reactive oxygen species assay. MMP-2 and HDAC8 expression assays suggested the dual inhibiting property of these two compounds. These findings were supported by results of molecular docking studies. In silico pharmacokinetic properties showed compounds C6 and C27 have high gastrointestinal absorption. Conclusion: This study highlights the action of phenyl/naphthylacetyl pentanoic acid derivatives as anticancer agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Molecular Docking Simulation , Pentanoic Acids/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites , Catalytic Domain , Cell Line, Tumor , Cell Survival/drug effects , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 2/metabolism , DNA Damage/drug effects , Drug Screening Assays, Antitumor , Gene Expression/drug effects , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Structure-Activity Relationship
3.
Sci Rep ; 11(1): 18930, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556703

ABSTRACT

Epidemiological studies have shown that head and neck cancer (HNC) is a complex multistage process that in part involves exposure to a combination of carcinogens and the capacity of certain drug-metabolising enzymes including cytochrome P450 (CYP) to detoxify or activate such carcinogens. In this study, CYP1A1, CYP1B1 and CYP2W1 expression in HNC was correlated with potential as target for duocarmycin prodrug activation and selective therapy. In the HNC cell lines, elevated expression was shown at the gene level for CYP1A1 and CYP1B1 whereas CYP2W1 was hardly detected. However, CYP2W1 was expressed in FaDu and Detroit-562 xenografts and in a cohort of human HNC samples. Functional activity was measured in Fadu and Detroit-562 cells using P450-Glo™ assay. Antiproliferative results of duocarmycin prodrugs ICT2700 and ICT2706 revealed FaDu and Detroit-562 as the most sensitive HNC cell lines. Administration of ICT2700 in vivo using a single dose of ICT2700 (150 mg/kg) showed preferential inhibition of small tumour growth (mean size of 60 mm3) in mice bearing FaDu xenografts. Significantly, our findings suggest a potential targeted therapeutic approach to manage HNCs by exploiting intratumoural CYP expression for metabolic activation of duocarmycin-based prodrugs such as ICT2700.


Subject(s)
Antineoplastic Agents/pharmacology , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Cytochrome P450 Family 2/antagonists & inhibitors , Head and Neck Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cohort Studies , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1/metabolism , Cytochrome P450 Family 2/metabolism , Female , Head and Neck Neoplasms/pathology , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Indoles/pharmacology , Indoles/therapeutic use , Mice , Prodrugs/pharmacology , Prodrugs/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Xenograft Model Antitumor Assays
4.
J Pharmacol Exp Ther ; 376(1): 64-73, 2021 01.
Article in English | MEDLINE | ID: mdl-33093187

ABSTRACT

Preparations from the leaves of the kratom plant (Mitragyna speciosa) are consumed for their opioid-like effects. Several deaths have been associated with kratom used concomitantly with some drugs. Pharmacokinetic interactions are potential underlying mechanisms of these fatalities. Accumulating in vitro evidence has demonstrated select kratom alkaloids, including the abundant indole alkaloid mitragynine, as reversible inhibitors of several cytochromes P450 (CYPs). The objective of this work was to refine the mechanistic understanding of potential kratom-drug interactions by considering both reversible and time-dependent inhibition (TDI) of CYPs in the liver and intestine. Mitragynine was tested against CYP2C9 (diclofenac 4'-hydroxylation), CYP2D6 (dextromethorphan O-demethylation), and CYP3A (midazolam 1'-hydroxylation) activities in human liver microsomes (HLMs) and CYP3A activity in human intestinal microsomes (HIMs). Comparing the absence to presence of NADPH during preincubation of mitragynine with HLMs or HIMs, an ∼7-fold leftward shift in IC50 (∼20 to 3 µM) toward CYP3A resulted, prompting determination of TDI parameters (HLMs: K I , 4.1 ± 0.9 µM; k inact , 0.068 ± 0.01 min-1; HIMs: K I , 4.2 ± 2.5 µM; k inact , 0.079 ± 0.02 min-1). Mitragynine caused no leftward shift in IC50 toward CYP2C9 (∼40 µM) and CYP2D6 (∼1 µM) but was a strong competitive inhibitor of CYP2D6 (K i , 1.17 ± 0.07 µM). Using a recommended mechanistic static model, mitragynine (2-g kratom dose) was predicted to increase dextromethorphan and midazolam area under the plasma concentration-time curve by 1.06- and 5.69-fold, respectively. The predicted midazolam area under the plasma concentration-time curve ratio exceeded the recommended cutoff (1.25), which would have been missed if TDI was not considered. SIGNIFICANCE STATEMENT: Kratom, a botanical natural product increasingly consumed for its opioid-like effects, may precipitate potentially serious pharmacokinetic interactions with drugs. The abundant kratom indole alkaloid mitragynine was shown to be a time-dependent inhibitor of hepatic and intestinal cytochrome P450 3A activity. A mechanistic static model predicted mitragynine to increase systemic exposure to the probe drug substrate midazolam by 5.7-fold, necessitating further evaluation via dynamic models and clinical assessment to advance the understanding of consumer safety associated with kratom use.


Subject(s)
Dextromethorphan/pharmacokinetics , Midazolam/pharmacokinetics , Secologanin Tryptamine Alkaloids/pharmacokinetics , Cytochrome P450 Family 2/antagonists & inhibitors , Drug Interactions , Humans , Intestinal Mucosa/metabolism , Microsomes, Liver/metabolism
5.
Brain Res Bull ; 164: 400-406, 2020 11.
Article in English | MEDLINE | ID: mdl-32926950

ABSTRACT

Cytochrome P450 2D (CYP2D) mediates the activation and inactivation of several classes of psychoactive drugs, including opioids, which can alter drug response. Tramadol is a synthetic opioid with analgesic activity of its own as well as being metabolically activated by CYP2D to O-desmethyltramadol (ODMST) an opioid receptor agonist. We investigated the impact of brain CYP2D metabolism on central tramadol and ODSMT levels, and resulting analgesic response after oral tramadol administration in rats. CYP2D inhibitors propranolol and propafenone were administered intracerebroventricularly prior to oral tramadol administration and analgesia was measured by tail-flick latency. Drug levels of tramadol and its metabolites, ODSMT and N-desmethyltramadol, were assessed in plasma and in brain by microdialysis using LC-ESI-MS/MS. Inhibiting brain CYP2D with propafenone pretreatment increased analgesia after oral tramadol administration (ANOVA p = 0.02), resulting in a 1.5-fold increase in area under the analgesia-time curve (AUC0-60, p < 0.01). This effect was associated with changes in the brain levels of tramadol and its metabolites consistent with brain CYP2D inhibition. In conclusion, under oral tramadol dosing pretreatment with a central administration of the CYP2D inhibitor propafenone increased analgesia (without altering plasma drug or metabolite levels), indicating that tramadol itself (and activity of CYP2D within the brain) contributed to analgesia.


Subject(s)
Analgesia/methods , Cytochrome P-450 Enzyme Inhibitors/therapeutic use , Cytochrome P450 Family 2/antagonists & inhibitors , Narcotics/therapeutic use , Pain Threshold/drug effects , Pain/drug therapy , Tramadol/therapeutic use , Animals , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Drug Therapy, Combination , Male , Narcotics/pharmacology , Pain Management , Pain Measurement , Rats , Rats, Wistar , Tramadol/pharmacology
6.
Brain Res Bull ; 163: 57-64, 2020 10.
Article in English | MEDLINE | ID: mdl-32707261

ABSTRACT

Cytochrome P450 (CYP) epoxygenases have been considered the main producers of epoxyeicosatrienoic acids (EETs) through the oxidation of arachidonic acid (AA). EETs display various biological properties, notably their powerful anti-inflammatory activities. In the brain, EETs have proven to be neuroprotective and to improve neuroinflammation. However, it is known that inflammation could modify CYP expression. We have previously reported that an inflammatory process in astrocytes is able to down-regulate CYP2J3 and CYP2C11 mRNA, protein levels, and activity (Navarro-Mabarak et al., 2019). In this work, we evaluated the effect of neuroinflammation in protein expression of CYP epoxygenases in the brain. Neuroinflammation was induced by the intraperitoneal administration of LPS (1 mg/kg) to male Wistar rats and was corroborated by IL-6, GFAP, and Iba-1 protein levels in the cortex over time. CYP2J3 and CYP2C11 protein levels were also evaluated in the cortex after 6, 12, 24, 48, and 72 h of LPS treatment. Our results show for the first time that neuroinflammation is able to downregulate CYP2J3 and CYP2C11 protein expression in the brain cortex.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Brain/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 2/metabolism , Down-Regulation/physiology , Inflammation Mediators/metabolism , Steroid 16-alpha-Hydroxylase/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Brain/drug effects , Cytochrome P450 Family 2/antagonists & inhibitors , Down-Regulation/drug effects , Lipopolysaccharides/toxicity , Male , Rats , Rats, Wistar , Steroid 16-alpha-Hydroxylase/antagonists & inhibitors
7.
Am J Physiol Renal Physiol ; 318(6): F1369-F1376, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32308018

ABSTRACT

Cytochrome P-450 (Cyp) epoxygenase-dependent metabolites of arachidonic acid (AA) have been shown to inhibit renal Na+ transport, and inhibition of Cyp-epoxygenase is associated with salt-sensitive hypertension. We used the patch-clamp technique to examine whether Cyp-epoxygenase-dependent AA metabolites inhibited the basolateral 40-pS K+ channel (Kir4.1/Kir5.1) in the distal convoluted tubule (DCT). Application of AA inhibited the basolateral 40-pS K+ channel in the DCT. The inhibitory effect of AA on the 40-pS K+ channel was specific because neither linoleic nor oleic acid was able to mimic the effect of AA on the K+ channel. Inhibition of Cyp-monooxygenase with N-methylsulfonyl-12,12-dibromododec-11-enamide or inhibition of cyclooxygenase with indomethacin failed to abolish the inhibitory effect of AA on the 40-pS K+ channel. However, the inhibition of Cyp-epoxygenase with N-methylsulfonyl-6-(propargyloxyphenyl)hexanamide abolished the effect of AA on the 40-pS K+ channel in the DCT. Moreover, addition of either 11,12-epoxyeicosatrienoic acid (EET) or 14,15-EET also inhibited the 40-pS K+ channel in the DCT. Whole cell recording demonstrated that application of AA decreased, whereas N-methylsulfonyl-6-(propargyloxyphenyl)hexanamide treatment increased, Ba2+-sensitive K+ currents in the DCT. Finally, application of 14,15-EET but not AA was able to inhibit the basolateral 40-pS K+ channel in the DCT of Cyp2c44-/- mice. We conclude that Cyp-epoxygenase-dependent AA metabolites inhibit the basolateral Kir4.1/Kir5.1 in the DCT and that Cyp2c44-epoxygenase plays a role in the regulation of the basolateral K+ channel in the mouse DCT.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Arachidonic Acid/pharmacology , Cytochrome P450 Family 2/metabolism , Kidney Tubules, Distal/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , 8,11,14-Eicosatrienoic Acid/metabolism , 8,11,14-Eicosatrienoic Acid/pharmacology , Amides/pharmacology , Animals , Arachidonic Acid/metabolism , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 2/genetics , Enzyme Inhibitors/pharmacology , Kidney Tubules, Distal/metabolism , Male , Membrane Potentials , Mice, 129 Strain , Mice, Knockout , Potassium Channel Blockers/metabolism , Potassium Channels, Inwardly Rectifying/metabolism
8.
BMC Cancer ; 19(1): 1166, 2019 Dec 02.
Article in English | MEDLINE | ID: mdl-31791289

ABSTRACT

BACKGROUND: Lung cancer (LC) is one of the leading causes of death worldwide, which highlights the urgent need for better therapies. Peroxisome proliferator-activated nuclear receptor alpha (PPARα), known as a key nuclear transcription factor involved in glucose and lipid metabolism, has been also implicated in endothelial proliferation and angiogenesis. However, the effects and potential mechanisms of the novel PPARα ligand, AVE8134, on LC growth and progression remain unclear. METHODS: A subcutaneous tumour was established in mice by injecting TC-1 lung tumour cells (~ 1 × 106 cells) into their shaved left flank. These mice were treated with three different PPARα ligands: AVE8134 (0.025% in drinking water), Wyeth-14,643 (0.025%), or Bezafibrate (0.3%). Tumour sizes and metastasis between treated and untreated mice were then compared by morphology and histology, and the metabolites of arachidonic acid (AA) were detected by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Inhibition of either Cyp2c44 expression by genetic disruption or cyclooxygenase (COX) activity by indomethacin was used to test the mechanisms by which AVE8134 affects tumour growth. RESULTS: The pharmacodynamics effects of AVE8134, Wyeth-14,643, and Bezafibrate on lipids control were similar. However, their effects on tumour suppression were different. Eicosanoid profile analysis showed that all PPARα ligands reduced the production of AA-derived epoxyeicosatrienoic acids (EETs) and increased the hydroxyl product, 11-hydroxyeicosatetraenoic acids (11-HETE). Moreover, increased 11-HETE promoted endothelial proliferation, angiogenesis, and subsequent tumour deterioration in a dose-dependent manner possibly via activating the AKT/extracellular signal-regulated kinase (ERK) pathway. The increased 11-HETE partly neutralized the benefits provided by the Cyp2c44-EETs system inhibited by PPARα ligands in tumour-bearing mice. AVE8134 treatment worsened the tumour phenotype in Cyp2c44 knockout mice, indicating that AVE8134 has contradictory effects on tumour growth. The COX inhibitor indomethacin strengthened the inhibitory actions of AVE8134 on tumour growth and metastasis by inhibiting the 11-HETE production in vivo and in vitro. CONCLUSION: In this study, we found that the degrees of inhibition on LC growth and metastasis by PPARα ligands depended on their bidirectional regulation on EETs and 11-HETE. Considering their safety and efficacy, the novel PPARα ligand, AVE8134, is a potentially ideal anti-angiogenesis drug for cancer treatment when jointly applied with the COX inhibitor indomethacin.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzoates/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Oxazoles/therapeutic use , PPAR alpha/agonists , Animals , Bezafibrate/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 2/genetics , Cytochrome P450 Family 2/metabolism , Drug Evaluation, Preclinical , Drug Therapy, Combination , Eicosanoids/analysis , Eicosanoids/metabolism , Indomethacin/therapeutic use , Lung Neoplasms/blood supply , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Neovascularization, Pathologic , Pyrimidines/therapeutic use , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Molecules ; 24(23)2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31775347

ABSTRACT

The inhibitory effect of new chemical entities on rat liver P450 marker activities was investigated in a functional approach towards drug development. Treatment of colorectal cancer (CRC) and chemoprevention using salicylic acid has gained a lot of attention, mainly in the prevention of the onset of colon cancer. Thus, an in vitro inhibitory effect of salicylic acid on rat CYP2C11 activity was examined by using high performance liquid chromatography (HPLC). High performance liquid chromatography analysis of a CYP2C11 assay was developed on a reversed phase C18 column (SUPELCO 25 cm × 4.6 mm × 5 µm) at 243 nm using 32% phosphate buffer (pH 3.36) and 68% methanol as a mobile phase. The CYP2C11 assay showed good linearity for all components (R2 > 0.999). Substrates and metabolites were found to be stable for up to 72 hours. Additionally, the method demonstrated good reproducibility, intra- and inter-day precision (<15%), acceptable recovery and accuracy (80%-120%), and low detection (1.3501 µM and 3.2757 µM) and quantitation limit values (4.914 µM and 9.927 µM) for 16α-hydroxytestosterone and testosterone, respectively. Salicylic acid acts reversibly as a noncompetitive (weak) inhibitor with Ki = 84.582 ± 2.67 µM (concentration of inhibitor to cause 50% inhibition of original enzyme activity (IC50) = 82.70 ± 2.67 µM) for CYP2C11 enzyme activity. This indicates a low potential to cause toxicity and drug-drug interactions.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P450 Family 2/antagonists & inhibitors , Liver/drug effects , Salicylic Acid/pharmacology , Steroid 16-alpha-Hydroxylase/antagonists & inhibitors , Animals , Aryl Hydrocarbon Hydroxylases/chemistry , Catalysis , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme Inhibitors/chemistry , Cytochrome P450 Family 2/chemistry , Drug Development , Humans , Liver/enzymology , Rats , Salicylic Acid/chemistry , Steroid 16-alpha-Hydroxylase/chemistry
10.
Eur J Drug Metab Pharmacokinet ; 44(6): 787-796, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31175627

ABSTRACT

BACKGROUND AND OBJECTIVES: Honokiol, a major constituent isolated from Magnolia officinalis, is regarded as a phytochemical marker and bioactive substance present in many traditional Chinese medicines. However, the effect of honokiol on cytochrome P450 (CYP) has not been thoroughly investigated. The aim of this study was to investigate the effect of honokiol on CYP1A2 and CYP2C11 in vitro and in vivo. METHODS: The effect of honokiol on CYP1A2 and CYP2C11 was investigated with rat liver microsomes (RLMs) by measuring phenacetin and tolbutamide metabolism (probe drugs for CYP1A2 and CYP2C11, respectively), and then explored in vivo by measuring the effect of honokiol (2.5 and 5 mg/kg, intravenous injection) on the pharmacokinetics of theophylline and tolbutamide (probe drugs for CYP1A2 and CYP2C11, respectively) in rats in vivo. RESULTS: Honokiol inhibited the formation of acetaminophen from phenacetin and 4-hydroxytolbutamide from tolbutamide in RLMs, with inhibition constant (Ki) values of 1.6 µM and 16.5 µM, respectively. In vivo, honokiol (2.5 or 5.0 mg/kg) increased the half-life (t1/2) of theophylline by 40.9% and 119.9%, decreased the clearance (CL) by 23.8% and 42.9%, and increased the area under the curve (AUC) by 41.3% and 83.4%, respectively. Similarly, the t1/2 of tolbutamide increased by 25.5% and 33.8%, the CL decreased by 14.3% and 19.1%, and the AUC increased by 19.2% and 25.7%, respectively. CONCLUSION: The inhibition of CYP1A2 by honokiol is greater than the inhibition of CYP2C11. The changes in the pharmacokinetics of theophylline and tolbutamide in rats treated with honokiol are due to the inhibition of CYP1A2 and CYP2C11 activity in a dose-dependent manner.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Biphenyl Compounds/pharmacology , Cytochrome P450 Family 2/antagonists & inhibitors , Lignans/pharmacology , Steroid 16-alpha-Hydroxylase/antagonists & inhibitors , Animals , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacokinetics , Cytochrome P-450 CYP1A2 , Cytochromes/antagonists & inhibitors , Lignans/chemistry , Lignans/pharmacokinetics , Male , Rats , Rats, Sprague-Dawley , Theophylline/pharmacokinetics , Tolbutamide/pharmacokinetics
11.
Xenobiotica ; 49(12): 1396-1402, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30747554

ABSTRACT

1. A protocol has been developed and validated for the high-throughput screening of eight major human cytochrome P450 (CYP) isozymes inhibition (CYP 1A2, 2C9, 2C19, 2D6, 3A4, 2B6, 2C8 and 2E1) using an in vitro probe cocktail containing eight substrates by overcoming the unfavorable effect of assay conditions on CYP2E1 inhibition data. 2. The cocktail consisting of selective probe substrates like tacrine (CYP1A2), diclofenac (CYP2C9), S-mephenytoin (CYP2C19), dextromethorphan (CYP2D6), midazolam (CYP3A4), bupropion (CYP2B6), paclitaxel (CYP2C8) and chlorzoxazone (CYP2E1) was incubated with human liver microsomes. 3. The method was investigated by incubating well-known CYP inhibitors {alphanaphthoflavone (CYP1A2), sulfaphenazole (CYP2C9), N-3-benzylnirvanol (CYP2C19), quinidine (CYP2D6), ketoconazole (CYP3A4), ticlopidine (CYP2B6), quercetin (CYP2C8) and 4-methylpyrazole (CYP2E1)} with the substrate cocktail. A fast gradient liquid chromatography tandem mass spectrometry (LC-MS/MS) was used for this study. 4. The IC50 values determined for typical CYP inhibitors were reproducible and consistent with those in the literature. DMSO has significant effect and itself inhibits CYP2E1. DMSO should not exceed 0.1% for the determination of reliable CYP2E1 inhibition profile. This cocktail assay offers an efficient and robust method to determine the CYP450 isoforms inhibition profiles of large numbers of compounds in a quick turnaround time.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P450 Family 2/metabolism , Drug Evaluation, Preclinical/methods , Chromatography, Liquid , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 3/antagonists & inhibitors , Cytochrome P450 Family 3/metabolism , Dimethyl Sulfoxide/pharmacology , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Reproducibility of Results , Substrate Specificity , Tandem Mass Spectrometry
12.
J Vet Pharmacol Ther ; 41(6): 815-824, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30113702

ABSTRACT

We previously showed that (+)-tramadol is metabolized in dog liver to (+)-M1 exclusively by CYP2D15 and to (+)-M2 by multiple CYPs, but primarily CYP2B11. However, (+)-M1 and (+)-M2 are further metabolized in dogs to (+)-M5, which is the major metabolite found in dog plasma and urine. In this study, we identified canine CYPs involved in metabolizing (+)-M1 and (+)-M2 using recombinant enzymes, untreated dog liver microsomes (DLMs), inhibitor-treated DLMs, and DLMs from CYP inducer-treated dogs. A canine P-glycoprotein expressing cell line was also used to evaluate whether (+)-tramadol, (+)-M1, (+)-M2, or (+)-M5 are substrates of canine P-glycoprotein, thereby limiting their distribution into the central nervous system. (+)-M5 was largely formed from (+)-M1 by recombinant CYP2C21 with minor contributions from CYP2C41 and CYP2B11. (+)-M5 formation in DLMs from (+)-M1 was potently inhibited by sulfaphenazole (CYP2C inhibitor) and chloramphenicol (CYP2B11 inhibitor) and was greatly increased in DLMs from phenobarbital-treated dogs. (+)-M5 was formed from (+)-M2 predominantly by CYP2D15. (+)-M5 formation from (+)-M1 in DLMs was potently inhibited by quinidine (CYP2D inhibitor) but had only a minor impact from all CYP inducers tested. Intrinsic clearance estimates showed over 50 times higher values for (+)-M5 formation from (+)-M2 compared with (+)-M1 in DLMs. This was largely attributed to the higher enzyme affinity (lower Km) for (+)-M2 compared with (+)-M1 as substrate. (+)-tramadol, (+)-M1, (+)-M2, or (+)-M5 were not p-glycoprotein substrates. This study provides a clearer picture of the role of individual CYPs in the complex metabolism of tramadol in dogs.


Subject(s)
Analgesics, Opioid/metabolism , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P450 Family 2/metabolism , Dogs/metabolism , Microsomes, Liver/metabolism , Steroid Hydroxylases/metabolism , Tramadol/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Cats/metabolism , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 2/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Male , Species Specificity , Steroid Hydroxylases/antagonists & inhibitors , Steroid Hydroxylases/genetics
13.
Biopharm Drug Dispos ; 39(3): 143-151, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29327455

ABSTRACT

Terminalia arjuna (T. arjuna) is an Indian medicinal plant belonging to the family Combretaceae and possesses numerous therapeutic activities including its immense cardioprotective activity. In the present work, a methanolic bark extract of T. arjuna was evaluated for CYP3A and CYP2D inhibition potential in rat liver microsomes (RLM). Further, the methanolic bark extract was fractionated successively using increasing polarity solvents starting with petroleum ether, chloroform, ethyl acetate and n-butanol. The fractions so obtained were also evaluated for their CYP3A and CYP2D inhibition potential. Probe substrates testosterone and dextromethorphan were used for CYP3A and CYP2D respectively. The IC50 values for the methanolic extract and the fractions were found to be less than 50 µg/ml in RLM for both CYP3A and CYP2D isoenzymes. The most potent n-butanol fraction was further fractionated with column chromatography to isolate the highest active constituent responsible for the activity. Fraction 4 of the n-butanol extract was the most potent fraction with IC50 values of 5.64 ± 0.735 µg/ml and 16.63 ± 0.879 µg/ml for CYP3A and CYP2D in RLM, respectively. Therefore, in vitro data indicated that the Terminalia arjuna extract contains constituents that can potentially inhibit the CYP3A and CYP2D isoenzymes which may in turn lead to pharmacokinetic drug-herb interaction.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P450 Family 2/antagonists & inhibitors , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Plant Extracts/pharmacology , Terminalia/chemistry , Animals , Cytochrome P450 Family 2/metabolism , Isoenzymes/antagonists & inhibitors , Methanol/chemistry , Plant Bark/chemistry , Plant Extracts/chemistry , Rats
14.
Xenobiotica ; 48(1): 1-10, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28010169

ABSTRACT

1. Present investigation was carried out in rats to study influence of corticosteroids after repeated dosing with/without pre-treatment with CYP2D inhibitor quinidine on the CYP2D1 mRNA levels and CYP2D enzyme activity using dextromethorphan as probe substrate. 2. CYP2D1 mRNA was measured in liver homogenate using quantitative real-time polymerase chain reaction [qRT-PCR] and enzymatic reaction was studied ex vivo in liver S-9 fractions of rats treated with oral 10 mg/kg dexamethasone or prednisolone for five days or pre-treated with quinidine and followed by treatment with oral 10 mg/kg corticosteroids for five days. 3. Five days repeat dosing of dexamethasone or prednisolone decreased the activity of the rat liver CYP2D by 37% and 34%, at 30 min incubation and decreased CYP2D1 mRNA levels by 62% and 61%, respectively. 4. Pre-treatment of quinidine decreased the enzymatic activity of rat CYP2D by 58% and did not potentiate CYP2D inhibition by corticosteroids. This observation was further complemented by qRT-PCR data. 5. Corticosteroids caused CYP2D inhibition in rats vs. literature evidence of CYP2D induction in human hepatocytes/pregnant humans demonstrating lack of concordance. In vivo inhibition should be factored for interpretation of pharmacokinetic data of CYP2D substrates when treated with corticosteroids in rats.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Cytochrome P450 Family 2/genetics , Dextromethorphan/pharmacology , Enzyme Inhibitors/pharmacology , Quinidine/pharmacology , Animals , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 2/metabolism , Demethylation , Dextromethorphan/metabolism , Rats
15.
Biochemistry ; 57(5): 817-826, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29215266

ABSTRACT

Human hepatic cytochromes P450 (CYP) are integral to xenobiotic metabolism. CYP2B6 is a major catalyst of biotransformation of environmental toxicants, including polybrominated diphenyl ethers (PBDEs). CYP2B substrates tend to contain halogen atoms, but the biochemical basis for this selectivity and for species specific determinants of metabolism has not been identified. Spectral binding titrations and inhibition studies were performed to investigate interactions of rat CYP2B1, rabbit CYP2B4, and CYP2B6 with a series of phenoxyaniline (POA) congeners that are analogues of PBDEs. For most congeners, there was a <3-fold difference between the spectral binding constants (KS) and IC50 values. In contrast, large discrepancies between these values were observed for POA and 3-chloro-4-phenoxyaniline. CYP2B1 was the enzyme most sensitive to POA congeners, so the Val-363 residue from that enzyme was introduced into CYP2B4 or CYP2B6. This substitution partially altered the protein-ligand interaction profiles to make them more similar to that of CYP2B1. Addition of cytochrome P450 oxidoreductase (POR) to titrations of CYP2B6 with POA or 2'4'5'TCPOA decreased the affinity of both ligands for the enzyme. Addition of cytochrome b5 to a recombinant enzyme system containing POR and CYP2B6 increased the POA IC50 value and decreased the 2'4'5'TCPOA IC50 value. Overall, the inconsistency between KS and IC50 values for POA versus 2'4'5'TCPOA is largely due to the effects of redox partner binding. These results provide insight into the biochemical basis of binding of diphenyl ethers to human CYP2B6 and changes in CYP2B6-mediated metabolism that are dependent on POA congener and redox partner identity.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Cytochrome P-450 CYP2B1/antagonists & inhibitors , Cytochrome P-450 CYP2B6/drug effects , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Halogenated Diphenyl Ethers/pharmacology , Alkylation/drug effects , Amino Acid Substitution , Aniline Compounds , Animals , Aryl Hydrocarbon Hydroxylases/chemistry , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Benzene Derivatives/pharmacology , Cytochrome P-450 CYP2B1/chemistry , Cytochrome P-450 CYP2B1/genetics , Cytochrome P-450 CYP2B1/metabolism , Cytochrome P-450 CYP2B6/chemistry , Cytochrome P-450 CYP2B6/genetics , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP2B6 Inhibitors/metabolism , Cytochrome P-450 CYP2B6 Inhibitors/pharmacology , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 2/chemistry , Cytochrome P450 Family 2/genetics , Cytochrome P450 Family 2/metabolism , Cytochromes b5/metabolism , Environmental Pollutants/metabolism , Halogenated Diphenyl Ethers/metabolism , Humans , Hydrocarbons, Halogenated/metabolism , Inhibitory Concentration 50 , Molecular Structure , Mutagenesis, Site-Directed , NADPH Oxidases/metabolism , Oxidation-Reduction , Rabbits , Rats , Recombinant Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity
16.
Basic Clin Pharmacol Toxicol ; 120(1): 59-63, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27404500

ABSTRACT

Vitamin D metabolism was studied in primary human dermal fibroblasts with focus on drug-mediated gene regulation related to adverse side effects of antiretroviral drugs used in HIV therapy. The fibroblasts expressed mRNA for cytochrome P450 (CYP) enzymes catalysing bioactivating (CYP2R1, CYP27A1 and CYP27B1) and catabolic reactions (CYP24A1). The cells produced both 25-hydroxyvitamin D3 and 1α,25-dihydroxyvitamin D3 . The results demonstrate that primary dermal fibroblasts have an active vitamin D3 -metabolizing system. High incidence of low bone mineral density is a concern for HIV-infected patients treated with antiretroviral drugs. Osteomalacia and severe vitamin D deficiency have been reported. We investigated whether drug-mediated gene regulation could be a possible mechanism behind these adverse drug effects. Fibroblasts were treated with different drugs used in HIV therapy, and the 1α,25-dihydroxyvitamin D3 levels and relative mRNA levels for crucial enzymes were determined. Efavirenz, stavudine and ritonavir significantly down-regulated the bioactivating CYP2R1 and up-regulated the catabolic CYP24A1. The drugs reduced bioactivating enzyme activities and cellular levels of 1α,25-dihydroxyvitamin D3 . The current results indicate that effects on gene expression may lead to disturbed vitamin D metabolism and decreased cellular levels of active vitamin D3 . The data are consistent with the impaired bone health in patients treated with certain antiretroviral drugs.


Subject(s)
Anti-HIV Agents/pharmacology , Cholecalciferol/metabolism , Cholestanetriol 26-Monooxygenase/metabolism , Cytochrome P450 Family 2/metabolism , Dermis/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Vitamin D3 24-Hydroxylase/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Adolescent , Adult , Alkynes , Benzoxazines/pharmacology , Calcifediol/metabolism , Calcitriol/antagonists & inhibitors , Calcitriol/metabolism , Cells, Cultured , Cholestanetriol 26-Monooxygenase/antagonists & inhibitors , Cholestanetriol 26-Monooxygenase/genetics , Cyclopropanes , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 2/genetics , Dermis/cytology , Dermis/metabolism , Female , Humans , Male , RNA, Messenger/metabolism , Reproducibility of Results , Ritonavir/pharmacology , Stavudine/pharmacology , Vitamin D3 24-Hydroxylase/chemistry , Vitamin D3 24-Hydroxylase/genetics , Young Adult
17.
Basic Clin Pharmacol Toxicol ; 119(2): 173-83, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26797788

ABSTRACT

Sunitinib (SUN) is a multi-targeted tyrosine kinase inhibitor that was recently approved for the treatment of gastrointestinal tract and renal cancers. To date, very little is known about the effects of SUN on the expression of hepatic and renal xenobiotic-metabolizing enzymes (XMEs) and transporters. The present study was designed to investigate the capacity of chronic SUN treatment to modulate the mRNA and protein expression levels of phase I cytochrome P450 (CYP), phase II conjugating enzymes, and phase III transporters in rat liver and kidneys. For this purpose, SUN (25, 50 and 100 mg/kg) was injected IP into Wistar albino rats for 4 weeks; thereafter, the mRNA and protein expression levels of several XMEs and transporters were determined by RT-PCR and Western blot analysis, respectively. Real-time PCR analysis showed that SUN significantly induced the hepatic and renal CYP1A1, 1A2, 1B1, 2E1 and 4F4, whereas it inhibited CYP2C11 and 4A2. Furthermore, SUN specifically induced renal, but not hepatic, CYP2J3 and 3A2, while it induced only hepatic CYP4A1. With regard to phase II, SUN induced hepatic GSTA1 and UGT1A and renal NQO1 and UGT1A mRNA levels, whereas it inhibited renal GST1A expression. On the other hand, both renal and hepatic P-gp, MRP2 and BCRP transporters were significantly induced by SUN at the mRNA and protein expression levels. Importantly, these differential effects were associated with changes in oxidative stress genes and lipid peroxidation levels. In conclusion, SUN can serve as XME and transporters modulator, which potentially may counteract the efficacy of the treatment, adverse reactions and drug interactions in SUN treatment.


Subject(s)
Indoles/pharmacology , Kidney/drug effects , Liver/drug effects , Pyrroles/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 2/genetics , Cytochrome P450 Family 2/metabolism , Cytochrome P450 Family 4/genetics , Cytochrome P450 Family 4/metabolism , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Kidney/enzymology , Lipid Peroxidation/drug effects , Liver/enzymology , Male , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Steroid 16-alpha-Hydroxylase/antagonists & inhibitors , Steroid 16-alpha-Hydroxylase/genetics , Steroid 16-alpha-Hydroxylase/metabolism , Sunitinib , Transcriptome , Xenobiotics/metabolism
18.
Interdiscip Sci ; 7(2): 129-35, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26239541

ABSTRACT

Among CYPs, CYP2A sub-family is well known for its function to metabolise xenobiotics. CYP2A includes three members: CYP2A6, CYP2A7 and CYP2A13. Of these three proteins, structure and function of CYP2A6 and CYP2A13 are widely studied, whereas very little study has been carried out on CYP2A7. In the initial in vitro studies on CYP2A7, full protein in its active form could not be expressed. The exact structure and function of CYP2A7 is still not revealed. However, up-regulation of CYP2A7 has been reported in malignant oesophageal cells and colon cancer cells. In the present study, we generated the structure of CYP2A7 protein. The modelled proteins were validated and subjected to molecular docking analyses. The energy and RMSD calculations demonstrated that the protein is highly conserved in nature, i.e., the protein is not much flexible. Here the ligand molecules of NCI Diversity Set II from the ZINC database against the active site of the CYP2A7 protein were screened. Five compounds that possess good inhibitory activity against CYP2A7 active site were identified. The top ranking molecule (ZINC01572309) has a minimum energy score of -12.0 kcal/Mol. This compound is thus a good starting point for further development of strong inhibitors. Our in silico approach could help in better structural and functional analysis of CYP2A7. Apart from structural description of CYP2A7, elaboration of binding sites for inhibitors provides us with an opportunity to utilise binding pockets in targeted inactivation of this protein for further research.


Subject(s)
Antineoplastic Agents/pharmacology , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Computer-Aided Design , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P450 Family 2/antagonists & inhibitors , Drug Design , Molecular Docking Simulation , Molecular Dynamics Simulation , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Aryl Hydrocarbon Hydroxylases/chemistry , Aryl Hydrocarbon Hydroxylases/metabolism , Binding Sites , Cytochrome P-450 Enzyme Inhibitors/chemistry , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P450 Family 2/chemistry , Cytochrome P450 Family 2/metabolism , Humans , Molecular Structure , Protein Binding , Reproducibility of Results , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL