Acessibilidade / Reportar erro

Study the effect of 3,4-Methylenedioxy methamphetamine on cytochrome P450 2E1 activity

Abstract

Evaluating the effects of ecstasy on CYP2E1 activity is of great concern, mainly due to growing trends in abuse and co-administration of MDMA with ethanol and the dominant role of this isoenzyme on ethanol metabolism. This study aimed to evaluate the effects of MDMA on CYP2E1 activity. A total of 24 male rats were selected and divided into three groups. The first and second groups consisted of 12 rats and were employed to optimize the perfusion method, and the third group was employed for studying the alteration of CYP2E1 activity after liver exposure to MDMA (300 and 600 ng/ml). The amount of chlorzoxazone and 6-hydroxy chlorzoxazone in a sample obtained from liver perfusion before and after exposure to a buffer containing MDMA was determined by HPLC-FL. The enzymatic activity of rat CYP2E1 decreased after liver perfusion with a buffer containing 600 ng/ml of MDMA. However, no significant changes were observed in chlorzoxazone and 6-hydroxy chlorzoxazone concentration in perfusate before and after liver perfusion with a buffer containing 300 ng/ml of MDMA. Our findings suggest that the activity of CYP2E1 in rats might decrease only after administration of MDMA at a lethal dose. However, further animal and human studies are needed to confirm our assumption.

Keywords:
MDMA; Ecstasy; Chlorzoxazone; Isolated perfused rat liver (IPRL); Rat CYP2E1

Graphical Abstract

INTRODUCTION

MDMA (3, 4-methylenedioxymethamphetamine), also known as ecstasy, is recognized as one of the most significant trends in drug abuse among young people (Palamar, Kamboukos, 2014Palamar JJ, Kamboukos D. An examination of sociodemographic correlates of ecstasy use among high school seniors in the United States. Subst Use Misuse. 2014;49(13):1774-83.). According to the World Drug Report published by United Nations Office on Drug and Crime in 2017 (Han, Parker, 2017Han X, Parker TL. Antiinflammatory activity of cinnamon (Cinnamomum zeylanicum) bark essential oil in a human skin disease model. Phytother Res. 2017;31(7):1034-8.), the number of ecstasy users was estimated to be around 21.6 million.

The liver is the prime target for MDMA injury since this organ is the major site for the metabolism and bioactivation of MDMA (Rouini M et al., 2020). Several mechanisms have been proposed to explain the toxicities associated with the MDMA abuse, including increased outflow of neurotransmitters, impairment of mitochondrial function, oxidation of biogenic amines, drug-drug interactions from polydrug abuse, metabolic bioactivation, and hyperthermia (Baumann, Rothman, 2009Baumann MH, Rothman RB. Neural and cardiac toxicities associated with 3,4-methylenedioxymethamphetamine (MDMA). Int Rev Neurobiol. 2009;88:257-96.; Docherty, Green, 2010Docherty JR, Green AR. The role of monoamines in the changes in body temperature induced by 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) and its derivatives. Br J Pharmacol. 2010;160(5):1029-44.). Unfortunately, estimating the MDMA-induced liver damage is complicated since the initiation and extent of the liver damage associated with MDMA abuse are not proportional to the amount and interval of the drug ingestion. Besides, abusers tend to have different polydrug abuse habits, and the MDMA content of illicit tablets varies considerably (Henry, Jeffreys, Dawling, 1992Henry JA, Jeffreys KJ, Dawling S. Toxicity and deaths from 3,4-methylenedioxymethamphetamine (“ecstasy”). Lancet. 1992;340(8816):384-7.; Morefield et al., 2011Morefield KM, Keane M, Felgate P, White JM, Irvine RJ. Pill content, dose and resulting plasma concentrations of 3,4-methylendioxymethamphetamine (MDMA) in recreational ‘ecstasy’ users. Addiction. 2011;106(7):1293- 300.). It has been clearly shown that biotransformation of parent compound has a profound role in MDMA-related hepatotoxicity because of the formation of active redox metabolites, among the other mechanism proposed for MDMA toxicities (Antolino-Lobo et al., 2010Antolino-Lobo I, Meulenbelt J, Nijmeijer SM, Scherpenisse P, van den Berg M, van Duursen MB. Differential roles of phase I and phase II enzymes in 3,4-methylendioxymethamphetamine-induced cytotoxicity. Drug Metab Dispos. 2010;38(7):1105-12.; Carvalho et al., 2010Carvalho M, Pontes H, Remiao F, Bastos ML, Carvalho F. Mechanisms underlying the hepatotoxic effects of ecstasy. Curr Pharm Biotechnol. 2010;11(5):476-95.). Phase I metabolism of MDMA involves two main routes, including N-demethylation to 3,4-methylenedioxy amphetamine (MDA) catalyzed mainly by CYP1A2 and to a lesser extent by CYP2B6 and CYP2C19, and/or O-demethylation of the ring substitute of both MDMA and MDA into the highly reactive catechol metabolites of 3,4-dihydroxymethamphetamine (HHMA) and 3,4-dihydroxyamphetamine (HHA) predominantly by CYP2D6 and to a less extent by CYP3A4 and CYP2C19 isoenzymes (Meyer, Peters, Maurer, 2008Meyer MR, Peters FT, Maurer HH. The role of human hepatic cytochrome P450 isozymes in the metabolism of racemic 3,4-methylenedioxy-methamphetamine and its enantiomers. Drug Metab Dispos. 2008;36(11):2345-54.; Van et al., 2007Van LM, Hargreaves JA, Lennard MS, Tucker GT, Rostami-Hodjegan A. Inactivation of CYP2D6 by methylenedioxymethamphetamine in different recombinant expression systems. Eur J Pharm Sci. 2007;32(1):8-16.). The two latter could be further oxidized to the corresponding ortho-quinones, which can enter in a redox cycling leading to the formation of reactive oxygen and nitrogen species, which has been proposed to be the reason for cytotoxicity in several tissues such as liver, brain, kidney, and heart (Antolino-Lobo et al., 2010Antolino-Lobo I, Meulenbelt J, Nijmeijer SM, Scherpenisse P, van den Berg M, van Duursen MB. Differential roles of phase I and phase II enzymes in 3,4-methylendioxymethamphetamine-induced cytotoxicity. Drug Metab Dispos. 2010;38(7):1105-12.; Carvalho et al., 2002Carvalho M, Hawksworth G, Milhazes N, Borges F, Monks TJ, Fernandes E, et al. Role of metabolites in MDMA (ecstasy)-induced nephrotoxicity: an in vitro study using rat and human renal proximal tubular cells. Arch Toxicol. 2002;76(10):581-8.). While several studies have focused on the interaction of MDMA with CYP2D6 and CYP3A4 enzymes (Antolino-Lobo et al., 2011bAntolino-Lobo I, Meulenbelt J, van den Berg M, van Duursen MB. A mechanistic insight into 3,4-methylenedioxymethamphetamine (“ecstasy”)-mediated hepatotoxicity. Vet Q. 2011b;31(4):193-205.; Downey, Daly, O’Boyle, 2014Downey C, Daly F, O’Boyle KM. An in vitro approach to assessing a potential drug interaction between MDMA (ecstasy) and caffeine. Toxicol In Vitro. 2014;28(2):231-9.; Rietjens et al., 2012 Rietjens SJ, Hondebrink L, Westerink RH, Meulenbelt J. Pharmacokinetics and pharmacodynamics of 3,4-methylenedioxymethamphetamine (MDMA): interindividual differences due to polymorphisms and drug-drug interactions. Crit Rev Toxicol. 2012;42(10):854-76.), we believe that studying the damage to the other CYP450 isoenzymes not involved in MDMA biotransformation would also be important.

MDMA is mainly metabolized in the liver by several cytochrome P450 isoenzymes, including CYP2D6, CYP3A4, CYP1A2, CYP2B6, and CYP2C19. Several studies elucidated the inhibitory effect of MDMA on the enzyme involved in its metabolism, which are CYP2D6, CYP3A4, and CYP1A2 (Antolino-Lobo et al., 2011bAntolino-Lobo I, Meulenbelt J, van den Berg M, van Duursen MB. A mechanistic insight into 3,4-methylenedioxymethamphetamine (“ecstasy”)-mediated hepatotoxicity. Vet Q. 2011b;31(4):193-205.; Meyer et al., 2010Meyer MR, Du P, Schuster F, Maurer HH. Studies on the metabolism of the alpha-pyrrolidinophenone designer drug methylenedioxy-pyrovalerone (MDPV) in rat and human urine and human liver microsomes using GC-MS and LC- high-resolution MS and its detectability in urine by GC-MS. J Mass Spectrom. 2010;45(12):1426-42.).

Simultaneous use of MDMA with other substances should be of great concern since MDMA is generally taken with other agents to enhance psychological effects; there is also likely the deliberate co-administration of MDMA with other drugs among ecstasy abuser in order to attenuate the aversive effects (Gouzoulis-Mayfrank, Daumann, 2006Gouzoulis-Mayfrank E, Daumann J. The confounding problem of polydrug use in recreational ecstasy/MDMA users: a brief overview. J Psychopharmacol. 2006;20(2):188-93.; Yuki et al., 2013Yuki F, Rie I, Miki K, Mitsuhiro W, Naotaka K, Kenichiro N. Warning against co-administration of 3,4-methylenedioxymethamphetamine (MDMA) with methamphetamine from the perspective of pharmacokinetic and pharmacodynamic evaluations in rat brain. Eur J Pharm Sci. 2013;49(1):57-64.). Thus, it is feasible to propose that exposure to MDMA may increase the potential for drug-drug interaction adverse effects through alteration in the activity of the enzymes which are not involved in this drug metabolism. To evaluate the extent of activity alteration for enzymes that are not involved in MDMA metabolism, we focused on investigating the activity of Cytochrome P450 2E1 upon exposure to MDMA in a series of ex-vivo experiments using the isolated perfused rat liver (IPRL) method. Liver perfusion has been extensively used in pharmacological and toxicological studies in recent years (Neyshaburinezhad et al., 2021Neyshaburinezhad N, Rouini M, Lavasani H, H. Ardakani Y. Evaluation of Cinnamon (Cinnamomum Verum) effects on liver CYP450 2D1 activity and hepatic clearance in diabetic rats. Jundishapur J Nat Pharm Prod. 2021;16(1):e101797.; Neyshaburinezhad et al., 2020aNeyshaburinezhad N, Rouini M, Entezari H, Lavasani H, Hosseinzadeh Ardakani Y. Evaluation of changes in cytochrome P450 2C19 activity in type 2 diabetic rats before and after treatment, by using isolated perfused liver model. Iran J Basic Med Sci. 2020;23(5):629-35.; Neyshaburinezhad et al., 2020bNeyshaburinezhad N, Seidabadi M, Rouini M, Lavasani H, Foroumadi A, Ardakani YH. Evaluation of hepatic CYP2D1 activity and hepatic clearance in type I and type II diabetic rat models, before and after treatment with insulin and metformin. Daru. 2020;28(2):479-487.; Rezai et al., 2020Rezai, S., Neyshaburinezhad, N., Rouini, M. et al. Can combination therapy with insulin and metformin improve metabolic function of the liver, in type I diabetic patients? An animal model study on CYP2D1 activity. J Diabetes Metab Disord. 2020;19:2049-2056. https://doi.org/10.1007/ s40200-020-00678-y
https://doi.org/https://doi.org/10.1007/...
).

As the fatal toxicity associated with the simultaneous use of ethanol and ecstasy has been frequently reported, the study of the potential increase in MDMA-induced hepatotoxicity caused by ethanol is of great clinical importance (Pontes et al., 2008Pontes H, Duarte JA, de Pinho PG, Soares ME, Fernandes E, Dinis-Oliveira RJ, et al. Chronic exposure to ethanol exacerbates MDMA-induced hyperthermia and exposes liver to severe MDMA-induced toxicity in CD1 mice. Toxicology. 2008;252(1-3):64-71.; Upreti et al., 2009Upreti VV, Eddington ND, Moon KH, Song BJ, Lee IJ. Drug interaction between ethanol and 3,4-methylenedioxymethamphetamine (“ecstasy”). Toxicol Lett. 2009;188(2):167-72.).

CYP2E1 is involved in the biotransformation of ethanol and a miscellaneous group of drugs, including chlorzoxazone, acetaminophen, and volatile anesthetics. It is also reported that CYP2E1 is important for the inactivation of the group of environmental carcinogens such as nitrosamines, benzene, and styrene. Among these drugs, chlorzoxazone (CZX) has been extensively used as an appropriate probe substrate in most surveys to determine enzyme activity in both in vitro and in vivo studies (Ernstgard, Warholm, Johanson, 2004Ernstgard L, Warholm M, Johanson G. Robustness of chlorzoxazone as an in vivo measure of cytochrome P450 2E1 activity. Br J Clin Pharmacol. 2004;58(2):190-200.; Muzeeb et al., 2005Muzeeb S, Pasha MK, Basha SJ, Mullangi R, Srinivas NR. Effect of 1-aminobenzotriazole on the in vitro metabolism and single-dose pharmacokinetics of chlorzoxazone, a selective CYP2E1 substrate in Wistar rats. Xenobiotica. 2005;35(8):825-38.). Reports indicated that rats are the best animal models for human CYP2E1 (Martignoni, Groothuis, de Kanter, 2006Martignoni M, Groothuis GM, de Kanter R. Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin Drug Metab Toxicol. 2006;2(6):875-94.), and therefore rat liver was employed in this study. It was demonstrated that the formation of 6-OH-chlorzoxazone (6-OH-CZX) is more specific for CYP2E1 activity at high substrate concentrations (500 µM) in contrast to low substrate concentrations (10 µM) (Yuan et al., 2002Yuan R, Madani S, Wei XX, Reynolds K, Huang SM. Evaluation of cytochrome P450 probe substrates commonly used by the pharmaceutical industry to study in vitro drug interactions. Drug Metab Dispos. 2002;30(12):1311-9.).

The present study was carried out to investigate the alteration in enzymatic activity of the hepatic CYP2E1 by using CZX as a probe drug with isolated perfused rat liver (IPRL) to different inlet concentrations of MDMA.

MATERIAL AND METHODS

Material

CZX and 6-OH-CZX were obtained from Sigma- Aldrich (Poole, Dorset, UK). Standards of MDMA, MDA, HMA, and HHMA were synthesized in the Medicinal Chemistry Department, Faculty of Pharmacy, Tehran University of Medical Sciences. The structure of the compound was confirmed by IR, 1H-NMR, and 13C-NMR spectra. All other chemicals were supplied by Merck (Darmstadt, Germany). The water used in all experiments was of Direct-Q® quality (Millipore, France). Xylazine 2% and ketamine 10% (Alfasan, Holand) were acquired from a local supplier.

Animal studies

Animals

Twenty-four male Sprague Dawley rats (weighing 250-300 g) were maintained in a clean room with 12 hours light-dark cycle, controlled temperature (20-23˚c), relative humidity of 50%, and free access to standard laboratory chow and water for this experiment. Rats were divided into three groups.

In the first group, eight rats were divided into four subgroups and given CZX-containing medium (100, 500, 1000, and 2000 µM) in a single-pass experiment (n=2 in each study) to justify the probe substrate concentration and the time of perfusion.

In the second group, other four rats were employed and split into two subgroups to determine the time required for the first washing step (after liver perfusion with 2000 µM CZX) and second washing step (after liver perfusion with 600 ng/ml of MDMA) in order to minimize the total time of perfusion (n=2 in each study).

In the third group, the remaining 12 rats were divided into two subgroups and received a medium containing MDMA (300 and 600 ng/ml) by liver perfusion to estimate the alteration in CYP2E1 activity.

The animals in the first and second groups were employed to optimize the perfusion method, while the animals in the third group were utilized to study the alteration of the CYP2E1 enzymatic activity.

This study was approved by the Institutional Review Board of Pharmaceutical Research Centre of Tehran University of Medical Sciences [Project Code: 93-02- 33-21634].

Liver perfusion

The rats were anesthetized using an intraperitoneal injection of xylazine (15mg/kg) and ketamine (75 mg/ kg) mixture. The portal vein and superior vena cava (as input and output of the perfusion medium) were catheterized using intravenous 16-and 18-gauge catheters, respectively. Five hundred units of heparin were injected into the inferior vena cava. The perfusion medium was freshly prepared Krebs-Henseleit buffer in a total volume of about 200 ml reservoir. The perfusion medium was passed through the portal vein with a constant flow rate of 10 ml/min using a peristaltic pump.

The temperature (37°±0.5˚C), pH (7.4±0.2), and perfusion pressure (14 mmHg) were intermittently monitored and maintained throughout the experiment. Liver viability was proven by the overall macroscopic appearance of the liver (wet liver weights of < 4% of body weight at the end of perfusion) and monitoring the liver enzyme activities (AST and ALT) throughout the perfusion at different time intervals (Jamshidfar et al., 2017).

Experimental protocols

The study was conducted in accordance with the Basic & Clinical Pharmacology & Toxicology Policy for Experimental and Clinical studies (Tveden-Nyborg, Bergmann, Lykkesfeldt, 2018Tveden-Nyborg P, Bergmann TK, Lykkesfeldt J. Basic & Clinical Pharmacology & Toxicology Policy for Experimental and Clinical studies. Basic Clin Pharmacol Toxicol. 2018;123(3):233-5.).

To reduce between-subject variations, this study was designed such that any alterations in CYP2E1 activity before and after MDMA exposure could be measurable in all rat perfused livers. Hence, any changes in probe substrate metabolism before and after liver perfusion with MDMA-containing medium encountering of each perfused liver with MDMA were calculated to demonstrate any induction or inhibition on CYP2E1 activity

Five experimental protocols were planned based on evaluations to address different concerns during the course of experiments for this investigation, and all initial results were used to justify the final MDMA and probe substrate concentrations used in this study along with the minimum time need for each designed step.

Time and Concentration dependent metabolism of probe substrate (CZX) in isolated perfused rat liver (Pre-exposure to MDMA)

In cytochrome P450 probe reaction studies, special care must be taken with the substrate concentrations, and the measured enzyme-substrate affinity data should be provided. For this reason, selecting a saturated substrate concentration with respect to Michaelis-Menten constant (Km) for the test system is recommended in common microsomal or cell culture studies to activate mainly the considered CYP450 pathway and maintain high selectivity.

As there are few enzyme kinetics studies in isolated perfused rat liver conditions, this experiment was designed to calculate the appropriate probe substrate concentration (CZX) that should be used in this study. After pre-stabilization of the perfused liver by the drug-free medium for 10 min, the perfusion was continued with the CZX-containing medium (100, 500, 1000, and 2000 µM) in a single-pass experiment (n=2 in each study). Samples were taken for 30 minutes from the start of the perfusion at 5 minutes intervals.

All samples were centrifuged and refrigerated at −70°C until assayed. Additionally, the inlet concentrations remained relatively constant during the entire perfusion time for all mentioned conditions.

In order to determine the time required for reaching the steady-state, the medium containing CZX (100, 500, 1000, and 2000 µM) was perfused through the liver for 1 hour (n=2 in each study), and samples were collected at a 5-minute interval.

Formation of the 6-OH-CZX by CYP2E1 increased when a higher concentration of probe drug was used in perfusion. However, the rise in the amount of 6-OH-CZX is not proportional to the increase in the concentration of CZX in the medium. Hence, we can assume that maximum enzyme-substrate affinity is reached with 2000 µM concentration of probe drug. The changes in the area under the curve (AUC(0-30)) for CZX and 6-OH-CZX after liver perfusion with the medium containing 100, 500, 1000, and 2000 µM and the corresponding metabolic ratios.

The obtained results indicated that an increase in the AUC of the CZX and 6-OH-CZX is not proportional to the increase of the CZX concentration in the perfusion medium. Besides, the mean metabolic ratio and extraction value declined by increasing the CZX concentration in the perfusion medium. Thus the 2000 µM probe concentration in rat perfusion medium was used as a saturation concentration to study the alteration in CYP2E1 activity and chosen as appropriate probe concentration for rat liver perfusion.

Both CZX and metabolite reached relatively steady concentrations in the outlet soon after the start of perfusion. Therefore, the time for liver perfusion with a medium with CZX as a probe substrate (pre-and post-exposure to MDMA) was reduced to 30 minutes to decrease the total time of perfusion.

First washing step

In order to determine the time required for the first washing step, liver perfused with medium with the highest amount of the CZX employed in this study (2000 µM) for 30 minutes. Consequently, a drug-free medium was passed through the perfused liver for a further 60 minutes, and samples were collected at 5 min time intervals to determine the minimum time required for the first washing step to achieve minimum CZX concentration (below 5% of initial CZX concentration).

Analysis of samples collected in the first washing step indicated that after 20 minutes of liver perfusion with the drug-free medium, the CZX concentration decreased to below 5% of its initial concentration after applying the highest probe substrate concentration (2000 µM).

Hence, after the liver exposure to the medium with CZX in the first phase, the perfusion was continued for a further 20 min with a drug-free medium to ensure removing of CZX effect on MDMA metabolism in all experiments.

Liver exposure to MDMA

Since the maximum plasma concentration after a common recreational dose of MDMA (100 mg) was reported between 200-300 ng/ml and the reported plasma concentrations of this compound in the mortality associated with MDMA was between 600-2800 ng/ mL, we decided to study the effect of MDMA on other metabolism pathways in two definite concentrations of 300 and 600 ng/ml of MDMA.

Our unpublished data on the inhibition of CYP2D6 by MDMA in rat perfused liver indicated that the maximum changes in the outlet concentration of this drug occurred in the first 30 minutes of the perfusion, and after this time, the MDMA and its main metabolites (MDA, HMA, and HHMA) reached relatively steady concentrations in the outlet perfusates. Hence, after the first washing step, the MDMA-containing medium (300 and 600 ng/ml) was passed through the liver for 30 min, and samples were collected at 5 min time intervals. All collected samples were centrifuged and refrigerated at −70 °C until analysis.

Effect of MDMA on the metabolism of CZX (second washing step)

In order to determine the time required for the second washing step, liver perfused with medium containing the highest amount of the MDMA employed in this study (600 ng/ml) for 30 minutes. Consequently, a drug-free medium was passed through the perfused liver for a further 60 min, and samples were collected at 5 min intervals to determine the minimum time required for the washing step to achieve minimum MDMA concentration (below 5% of initial MDMA concentration).

Analysis of samples collected in the second washing step indicated that after 20 minutes of liver perfusion with the drug-free medium, the MDMA concentration declined to below 5% of its initial concentration even after applying the highest concentration of MDMA employed in this study (600 ng/ml).

Thus, after liver perfusion with a medium containing MDMA, the perfusion was continued for a further 20 min with a drug-free medium to ensure removal of MDMA residue on perfusate.

Metabolism of CZX after exposure to MDMA (post-exposure step)

To evaluate any alteration in enzymatic activity after liver perfusion with Krebs-Henseleit buffer containing MDMA, the perfusion was continued with the medium containing CZX (2000 µM) for 30 minutes, and samples were collected at 5 min intervals. All collected samples were centrifuged and refrigerated at −70 °C until analysis.

HPLC methods

MDMA

MDMA concentration in the samples was determined by a previously developed HPLC-Fluorescence method (Jamali et al., 2017Jamali B, Sheikholeslami B, Hosseinzadeh Ardakani Y, Lavasani H, Rouini MR. Evaluation of the Ecstasy influence on tramadol and its main metabolite plasma concentration in rats. Drug Metab Pers Ther. 2017;32(3):137-45.).

CZX and 5-OH- CZX

The HPLC method used to determine the determination of the CZX and 6-OH-CZX in the samples was previously described by Leclercq et al. with some modifications (Leclercq, Horsmans, Desager, 1998Leclercq I, Horsmans Y, Desager JP. Estimation of chlorzoxazone hydroxylase activity in liver microsomes and of the plasma pharmacokinetics of chlorzoxazone by the same high-performance liquid chromatographic method. J Chromatogr A. 1998;828(1-2):291-6.).

Pharmacokinetic analysis

The mean concentration at steady state

The mean concentration of CZX and 6-OH-CZX at the steady-state was the first pharmacokinetic parameter used in this study; hence the mean CZX and 6-OH- CZX concentration in outlet perfusate for the last three sampling time (20, 25, and 30 minutes) before and after liver exposure to 300 and 600 ng/ml calculated.

Metabolic ratio

The metabolic ratio was the second parameter employed in our study using the following formula:

Metabolic ratio = Metabolite concentration at a specific time point Chlorzoxazone concentration at the same time point × 100

The concentration CZX and 6-OH-CZX at the steady-state were used in these formulas.

Statistical Analysis

A paired t-test was employed using SPSS 20 to determine whether the changes in pharmacokinetic parameters before and after liver exposure to MDMA were statistically significant. A P-value of less than 0.05 was considered statistically significant. The data were expressed as mean ± SD.

RESULTS

Effect of rat liver perfusion with 300 ng/ml of MDMA on the CYP2E1 activity

The changes in CZX and 6-OH- CZX metabolic ratios for each sampling time before and after liver exposure to 300 ng/ml of MDMA are given in Figure 1A.

The mean CZX concentration decreased slightly after MDMA exposure from 1580.7 ± 77 to 1545±65.7 µM while the 6-OH-CZX concentration increased from 9.1 ± 1 to 9.73 ± 0.9 µM after MDMA exposure. Therefore, the metabolic ratio increased from 0.58% ±0.07 before MDMA exposure to 0.63% ± 0.05 after liver exposure to MDMA (Figure. 2). None of these changes were statistically significant (P-value > 0.05).

FIGURE 1
Perfusate concentration-time profile of mean metabolic ratios before and after exposure of rats’ liver to 300 ng/ml (A) and 600 ng/ml (B) of MDMA (Ecstasy) following the passage of the perfusion buffer containing 2000 μM chlorzoxazone through the portal vein.

FIGURE 2
Perfusate mean metabolic ratio profile (3 end-points), before and after exposure of rats’ liver to 300 ng/ml and 600 ng/ml of MDMA (Ecstasy), following the passage of the perfusion buffer containing 2000 μM chlorzoxazone through the portal vein (n= 12, mean ± SEM). Each experiment was repeated independently three times in triplicate tests and data are shown as mean ± SEM. *P≤ 0.05.

Effect of rat liver perfusion with 600 ng/ml of MDMA on the CYP2E1 activity

The concentration profiles of CZX, 6-OH-CZX metabolic ratios before and after liver exposure to 600 ng/ml of MDMA are shown in Figure 1B.

The mean CZX concentration in perfusate at steady-state raised significantly from 1384 ± 21.8 to 1657.1 ± 74.2 µM after liver perfusion with a medium containing 600 ng/ ml MDMA. The mean 6-OH-CZX concentration for the last three sampling times in perfusate before and after liver exposure to MDMA (600 ng/ml) was determined to be 8.1± 0.9 and 5.9 ± 1.2 µM, respectively, and the corresponding metabolic ratios decreased from 0.59% ± 0.08 to 0.36% ±0.07 after liver exposure to MDMA (Figure. 2). This change was statistically significant (P-value = 0.03).

DISCUSSION

As mentioned before, studying the effects of MDMA on CYP2E1 isoenzyme is of great importance for several reasons.

Firstly, CYP2E1 is involved in the metabolism of ethanol, and the reports indicated the high frequency of polydrug use among MDMA abusers, notably ethanol users (Barrett, Darredeau, Pihl, 2006Barrett SP, Darredeau C, Pihl RO. Patterns of simultaneous polysubstance use in drug using university students. Hum Psychopharmacol. 2006;21(4):255-63.; Lieber, 1997Lieber CS. Cytochrome P-4502E1: its physiological and pathological role. Physiol Rev. 1997;77(2):517-44.). The extensive co-administration of these two substances increases the risk of hepatotoxicity and neurotoxicity associated with MDMA abuse (Vollenweider et al., 2002 Vollenweider FX, Liechti ME, Gamma A, Greer G, Geyer M. Acute psychological and neurophysiological effects of MDMA in humans. J Psychoactive Drugs. 2002;34(2):171- 84.).

Secondly, several studies have indicated that MDMA administration leads to the induction or inhibition of an enzyme involved in its own mechanism and may affect the enzymatic activity of other isoenzymes in the same manner (Nilchi et al., 2018Nilchi S, Behdarvand D, Lavasani H, Rouini M, Y HA. Assessment of CYP2D6 re-activation after inhibitory effect of MDMA using tramadol as a probe. Drug Metab Pers Ther. 2018;33(3):119-25.; Yang et al., 2006Yang J, Jamei M, Heydari A, Yeo KR, de la Torre R, Farre M, et al. Implications of mechanism-based inhibition of CYP2D6 for the pharmacokinetics and toxicity of MDMA. J Psychopharmacol. 2006;20(6):842-9.). In this research, we conducted an ex-vivo study on the CYP2E1 enzymatic activity alteration of MDMA by using CZX as a well-established probe drug with the aim of isolated perfused rat liver method.

Finally, CYP2E1 is essential for the inactivation of a group of carcinogens, namely nitrosamines, benzene, and styrene; hence studying the alteration in the activity of this isozyme is of great concern (Lieber, 1997Lieber CS. Cytochrome P-4502E1: its physiological and pathological role. Physiol Rev. 1997;77(2):517-44.).

The MDMA plasma level after common recreational use and lethal dose reported to be 300 ng/ml and 600ng/ml, respectively (Milroy, 2011Milroy CM. “Ecstasy” associated deaths: what is a fatal concentration ? Analysis of a case series. Forensic Sci Med Pathol. 2011;7(3):248-52.). Thus, in this study, animals were subjected to Krebs-Henseleit buffer containing 300ng/ml and 600ng/ml MDMA.

The technique used in this study was the isolated perfused rat liver since we wanted to study the enzymatic activity alteration of CYP2E1 in a rat’s liver. In order to minimize the within-subject variation, the CYP2E1 activity was estimated before and after exposure to MDMA in the same rat with two washing steps.

The data obtained from this study demonstrate that CYP2E1 activity after liver exposure to 600ng/ml MDMA decreased; however after liver perfusion with 300 ng/ml MDMA, no significant change in CYP2E1 activity was observed (Figure 1 and 2).

The reduction in CYP2E1 activity after perfusion with 600 ng/ml of MDMA was indicated by a significant increase in CZX concentration, while an insignificant decrease in 6-OH-CZX concentration in the last three sampling times was observed. The rise in CZX concentration simultaneously with the reduction of 6-OH- CZX after liver perfusion with 600 ng/ml of MDMA could also demonstrate the reduction in the CYP2E1 enzymatic activity.

There are several explanations for the variation between CYP2E1 enzymatic activities after liver exposure to 300 ng/ml and 600 ng/ml.

First of all, as we stated earlier, CZX is metabolized primarily to 6-OH-CZX in the presence of the hepatic CYP2E1; it is then rapidly glucuronidated and eliminated by the kidney (Conney, Burns, 1960Conney AH, Burns JJ. Biochemical pharmacological considerations of zoxazolamine and chlorzoxazone metabolism. Ann N Y Acad Sci. 1960;86:167-77.; Mishin et al., 1998Mishin VM, Rosman AS, Basu P, Kessova I, Oneta CM, Lieber CS. Chlorzoxazone pharmacokinetics as a marker of hepatic cytochrome P4502E1 in humans. Am J Gastroenterol. 1998;93(11):2154-61.). In this study, we estimated the amount of 6-OH-CZX, which is known as a free metabolite in perfusate, and did not assess the amount of conjugated metabolite. Since phase II metabolism occurs rapidly, the estimation of the conjugated and unconjugated metabolite together might be more reliable for the assessment of CYP2E1 activity.

Secondly, it looks as if MDMA could change the activity of the enzymes involved in phase II metabolism in addition to alteration the activity of phase I enzymes (Antolino-Lobo et al., 2011aAntolino-Lobo I, Meulenbelt J, Nijmeijer SM, Maas- Bakker RF, Meijerman I, van den Berg M, et al. 3,4-methylenedioxymethamphetamine (MDMA) interacts with therapeutic drugs on CYP3A by inhibition of pregnane X receptor (PXR) activation and catalytic enzyme inhibition. Toxicol Lett. 2011a;203(1):82-91.). According to the result of this study, reduction in the activity of phase II enzymes is more probable even with lower doses of MDMA in comparison to phase I enzymes (CYP2E1) since the apparent increase of 6-OH-CZX with negligible changes in CZX concentration was occurred after liver perfusion with 300 ng/ml of the compound. However, after liver perfusion with a buffer containing 600 ng/ml MDMA, the CZX concentration increased significantly, and the 6-OH-CZX concentration reduced, which could be explained by a reduction in the activity of both phase I and II enzymes at this concentration. Further animal studies are required to confirm this hypothesis.

CONCLUSION

The data obtained from this study indicated that MDMA could not reduce the activity of the CYP2E1 at recreational doses (300 ng/ml), while at the lethal doses (600 ng/ml), it could reduce the enzyme activity. We can extrapolate the data from this study to humans since CZX is a well-established probe drug for CYP2E1 and reports indicated that hepatic rat’s CYP2E1 is a major enzyme involved in the hydroxylation of CZX. Thus, we can presume that at a higher dose, MDMA may reduce CYP2E1 activity and alter the ethanol metabolism in humans since rat CYP2E1 is the best animal model for human CYP2E1. Although to support our hypothesis, further human and animal studies are required.

REFERENCES

  • Antolino-Lobo I, Meulenbelt J, Nijmeijer SM, Scherpenisse P, van den Berg M, van Duursen MB. Differential roles of phase I and phase II enzymes in 3,4-methylendioxymethamphetamine-induced cytotoxicity. Drug Metab Dispos. 2010;38(7):1105-12.
  • Antolino-Lobo I, Meulenbelt J, Nijmeijer SM, Maas- Bakker RF, Meijerman I, van den Berg M, et al. 3,4-methylenedioxymethamphetamine (MDMA) interacts with therapeutic drugs on CYP3A by inhibition of pregnane X receptor (PXR) activation and catalytic enzyme inhibition. Toxicol Lett. 2011a;203(1):82-91.
  • Antolino-Lobo I, Meulenbelt J, van den Berg M, van Duursen MB. A mechanistic insight into 3,4-methylenedioxymethamphetamine (“ecstasy”)-mediated hepatotoxicity. Vet Q. 2011b;31(4):193-205.
  • Barrett SP, Darredeau C, Pihl RO. Patterns of simultaneous polysubstance use in drug using university students. Hum Psychopharmacol. 2006;21(4):255-63.
  • Baumann MH, Rothman RB. Neural and cardiac toxicities associated with 3,4-methylenedioxymethamphetamine (MDMA). Int Rev Neurobiol. 2009;88:257-96.
  • Carvalho M, Hawksworth G, Milhazes N, Borges F, Monks TJ, Fernandes E, et al. Role of metabolites in MDMA (ecstasy)-induced nephrotoxicity: an in vitro study using rat and human renal proximal tubular cells. Arch Toxicol. 2002;76(10):581-8.
  • Carvalho M, Pontes H, Remiao F, Bastos ML, Carvalho F. Mechanisms underlying the hepatotoxic effects of ecstasy. Curr Pharm Biotechnol. 2010;11(5):476-95.
  • Conney AH, Burns JJ. Biochemical pharmacological considerations of zoxazolamine and chlorzoxazone metabolism. Ann N Y Acad Sci. 1960;86:167-77.
  • Docherty JR, Green AR. The role of monoamines in the changes in body temperature induced by 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) and its derivatives. Br J Pharmacol. 2010;160(5):1029-44.
  • Downey C, Daly F, O’Boyle KM. An in vitro approach to assessing a potential drug interaction between MDMA (ecstasy) and caffeine. Toxicol In Vitro. 2014;28(2):231-9.
  • Ernstgard L, Warholm M, Johanson G. Robustness of chlorzoxazone as an in vivo measure of cytochrome P450 2E1 activity. Br J Clin Pharmacol. 2004;58(2):190-200.
  • Gouzoulis-Mayfrank E, Daumann J. The confounding problem of polydrug use in recreational ecstasy/MDMA users: a brief overview. J Psychopharmacol. 2006;20(2):188-93.
  • Han X, Parker TL. Antiinflammatory activity of cinnamon (Cinnamomum zeylanicum) bark essential oil in a human skin disease model. Phytother Res. 2017;31(7):1034-8.
  • Henry JA, Jeffreys KJ, Dawling S. Toxicity and deaths from 3,4-methylenedioxymethamphetamine (“ecstasy”). Lancet. 1992;340(8816):384-7.
  • Jamali B, Sheikholeslami B, Hosseinzadeh Ardakani Y, Lavasani H, Rouini MR. Evaluation of the Ecstasy influence on tramadol and its main metabolite plasma concentration in rats. Drug Metab Pers Ther. 2017;32(3):137-45.
  • Leclercq I, Horsmans Y, Desager JP. Estimation of chlorzoxazone hydroxylase activity in liver microsomes and of the plasma pharmacokinetics of chlorzoxazone by the same high-performance liquid chromatographic method. J Chromatogr A. 1998;828(1-2):291-6.
  • Lieber CS. Cytochrome P-4502E1: its physiological and pathological role. Physiol Rev. 1997;77(2):517-44.
  • Martignoni M, Groothuis GM, de Kanter R. Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin Drug Metab Toxicol. 2006;2(6):875-94.
  • Meyer MR, Du P, Schuster F, Maurer HH. Studies on the metabolism of the alpha-pyrrolidinophenone designer drug methylenedioxy-pyrovalerone (MDPV) in rat and human urine and human liver microsomes using GC-MS and LC- high-resolution MS and its detectability in urine by GC-MS. J Mass Spectrom. 2010;45(12):1426-42.
  • Meyer MR, Peters FT, Maurer HH. The role of human hepatic cytochrome P450 isozymes in the metabolism of racemic 3,4-methylenedioxy-methamphetamine and its enantiomers. Drug Metab Dispos. 2008;36(11):2345-54.
  • Milroy CM. “Ecstasy” associated deaths: what is a fatal concentration ? Analysis of a case series. Forensic Sci Med Pathol. 2011;7(3):248-52.
  • Mishin VM, Rosman AS, Basu P, Kessova I, Oneta CM, Lieber CS. Chlorzoxazone pharmacokinetics as a marker of hepatic cytochrome P4502E1 in humans. Am J Gastroenterol. 1998;93(11):2154-61.
  • Morefield KM, Keane M, Felgate P, White JM, Irvine RJ. Pill content, dose and resulting plasma concentrations of 3,4-methylendioxymethamphetamine (MDMA) in recreational ‘ecstasy’ users. Addiction. 2011;106(7):1293- 300.
  • Muzeeb S, Pasha MK, Basha SJ, Mullangi R, Srinivas NR. Effect of 1-aminobenzotriazole on the in vitro metabolism and single-dose pharmacokinetics of chlorzoxazone, a selective CYP2E1 substrate in Wistar rats. Xenobiotica. 2005;35(8):825-38.
  • Neyshaburinezhad N, Rouini M, Entezari H, Lavasani H, Hosseinzadeh Ardakani Y. Evaluation of changes in cytochrome P450 2C19 activity in type 2 diabetic rats before and after treatment, by using isolated perfused liver model. Iran J Basic Med Sci. 2020;23(5):629-35.
  • Neyshaburinezhad N, Seidabadi M, Rouini M, Lavasani H, Foroumadi A, Ardakani YH. Evaluation of hepatic CYP2D1 activity and hepatic clearance in type I and type II diabetic rat models, before and after treatment with insulin and metformin. Daru. 2020;28(2):479-487.
  • Neyshaburinezhad N, Rouini M, Lavasani H, H. Ardakani Y. Evaluation of Cinnamon (Cinnamomum Verum) effects on liver CYP450 2D1 activity and hepatic clearance in diabetic rats. Jundishapur J Nat Pharm Prod. 2021;16(1):e101797.
  • Nilchi S, Behdarvand D, Lavasani H, Rouini M, Y HA. Assessment of CYP2D6 re-activation after inhibitory effect of MDMA using tramadol as a probe. Drug Metab Pers Ther. 2018;33(3):119-25.
  • Palamar JJ, Kamboukos D. An examination of sociodemographic correlates of ecstasy use among high school seniors in the United States. Subst Use Misuse. 2014;49(13):1774-83.
  • Pontes H, Duarte JA, de Pinho PG, Soares ME, Fernandes E, Dinis-Oliveira RJ, et al. Chronic exposure to ethanol exacerbates MDMA-induced hyperthermia and exposes liver to severe MDMA-induced toxicity in CD1 mice. Toxicology. 2008;252(1-3):64-71.
  • Rezai, S., Neyshaburinezhad, N., Rouini, M. et al. Can combination therapy with insulin and metformin improve metabolic function of the liver, in type I diabetic patients? An animal model study on CYP2D1 activity. J Diabetes Metab Disord. 2020;19:2049-2056. https://doi.org/10.1007/ s40200-020-00678-y
    » https://doi.org/https://doi.org/10.1007/ s40200-020-00678-y
  • Rietjens SJ, Hondebrink L, Westerink RH, Meulenbelt J. Pharmacokinetics and pharmacodynamics of 3,4-methylenedioxymethamphetamine (MDMA): interindividual differences due to polymorphisms and drug-drug interactions. Crit Rev Toxicol. 2012;42(10):854-76.
  • Tveden-Nyborg P, Bergmann TK, Lykkesfeldt J. Basic & Clinical Pharmacology & Toxicology Policy for Experimental and Clinical studies. Basic Clin Pharmacol Toxicol. 2018;123(3):233-5.
  • Upreti VV, Eddington ND, Moon KH, Song BJ, Lee IJ. Drug interaction between ethanol and 3,4-methylenedioxymethamphetamine (“ecstasy”). Toxicol Lett. 2009;188(2):167-72.
  • Van LM, Hargreaves JA, Lennard MS, Tucker GT, Rostami-Hodjegan A. Inactivation of CYP2D6 by methylenedioxymethamphetamine in different recombinant expression systems. Eur J Pharm Sci. 2007;32(1):8-16.
  • Vollenweider FX, Liechti ME, Gamma A, Greer G, Geyer M. Acute psychological and neurophysiological effects of MDMA in humans. J Psychoactive Drugs. 2002;34(2):171- 84.
  • Yang J, Jamei M, Heydari A, Yeo KR, de la Torre R, Farre M, et al. Implications of mechanism-based inhibition of CYP2D6 for the pharmacokinetics and toxicity of MDMA. J Psychopharmacol. 2006;20(6):842-9.
  • Yuan R, Madani S, Wei XX, Reynolds K, Huang SM. Evaluation of cytochrome P450 probe substrates commonly used by the pharmaceutical industry to study in vitro drug interactions. Drug Metab Dispos. 2002;30(12):1311-9.
  • Yuki F, Rie I, Miki K, Mitsuhiro W, Naotaka K, Kenichiro N. Warning against co-administration of 3,4-methylenedioxymethamphetamine (MDMA) with methamphetamine from the perspective of pharmacokinetic and pharmacodynamic evaluations in rat brain. Eur J Pharm Sci. 2013;49(1):57-64.

Publication Dates

  • Publication in this collection
    19 Dec 2022
  • Date of issue
    2022

History

  • Received
    20 May 2019
  • Accepted
    03 Feb 2021
Universidade de São Paulo, Faculdade de Ciências Farmacêuticas Av. Prof. Lineu Prestes, n. 580, 05508-000 S. Paulo/SP Brasil, Tel.: (55 11) 3091-3824 - São Paulo - SP - Brazil
E-mail: bjps@usp.br