1
|
Miksys S, McDonald C, Baghai Wadji F, Gonzalez FJ, Tyndale RF. Human CYP2D6 varies across the estrous cycle in brains of transgenic mice altering drug response. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111108. [PMID: 39069248 PMCID: PMC11402587 DOI: 10.1016/j.pnpbp.2024.111108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Cytochrome P450 (CYP) 2Ds are drug metabolizing enzymes found in brain and liver which metabolize numerous centrally acting drugs. Inhibition and induction of CYP2D-mediated metabolism in rodent brain alters brain drug and metabolite concentrations and resulting drug response. In female rats, brain CYP2D metabolism varies across the estrous cycle and with exogenous estrogen, changing brain drug concentrations and response. In this study harmine-induced hypothermia was lower in humanized CYP2D6 transgenic female mice during estrus compared to diestrus. Pretreatment into the cerebral ventricles with propranolol, a selective irreversible inhibitor of human CYP2D6 in brain, increased hypothermia in estrus but not in diestrus. In vivo enzyme activity was higher in brains of transgenic mice in estrus compared to diestrus and was lower after pretreatment with inhibitor in estrus, but not in diestrus. Hepatic activity and plasma harmine concentrations were unaffected by either estrous phase or inhibition of brain CYP2D6. In wild-type female mice, harmine-induced hypothermia was unaffected by either estrous phase or inhibitor pretreatment. Male mice were used as positive controls, where pretreatment with inhibitor increased harmine-induced hypothermia in transgenic but not wild-type, mice. This study provides evidence for female hormone cycle-based regulation of drug metabolism by human CYP2D6 in brain and resulting drug response. This suggests that brain CYP2D6 metabolism may vary, for example, during the menstrual cycle, pregnancy, or menopause, or while taking oral contraceptives or hormone therapy. This variation could contribute to individual differences in response to centrally acting CYP2D6-substrate drugs by altering local brain drug and/or metabolite concentrations.
Collapse
Affiliation(s)
- Sharon Miksys
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| | - Claire McDonald
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| | - Fariba Baghai Wadji
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| | - Frank J Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Rachel F Tyndale
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Zhang M, Vuist IM, Rottschäfer V, de Lange EC. Exploring K p,uu,BBB values smaller than unity in remoxipride: A physiologically-based CNS model approach highlighting brain metabolism in drugs with passive blood-brain barrier transport. Eur J Pharm Sci 2024; 203:106883. [PMID: 39181172 DOI: 10.1016/j.ejps.2024.106883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
(AIM) Kp,uu,BBB values are crucial indicators of drug distribution into the brain, representing the steady-state relationship between unbound concentrations in plasma and in brain extracellular fluid (brainECF). Kp,uu,BBB values < 1 are often interpreted as indicators of dominant active efflux transport processes at the blood-brain barrier (BBB). However, the potential impact of brain metabolism on this value is typically not addressed. In this study, we investigated the brain distribution of remoxipride, as a paradigm compound for passive BBB transport with yet unexplained brain elimination that was hypothesized to represent brain metabolism. (METHODS) The physiologically-based LeiCNS pharmacokinetic predictor (LeiCNS-PK model) was used to compare brain distribution of remoxipride with and without Michaelis-Menten kinetics at the BBB and/or brain cell organelle levels. To that end, multiple in-house (IV 0.7, 3.5, 4, 5.2, 7, 8, 14 and 16 mg kg-1) and external (IV 4 and 8 mg kg-1) rat microdialysis studies plasma and brainECF data were analysed. (RESULTS) The incorporation of active elimination through presumed brain metabolism of remoxipride in the LeiCNS-PK model significantly improved the prediction accuracy of experimentally observed brainECF profiles of this drug. The model integrated with brain metabolism in both barriers and organelles levels is named LeiCNS-PK3.5. (CONCLUSION) For drugs with Kp,uu,BBB values < 1, not only the current interpretation of dominant BBB efflux transport, but also potential brain metabolism needs to be considered, especially because these may be concentration dependent. This will improve the mechanistic understanding of the processes that determine brain PK profiles.
Collapse
Affiliation(s)
- Mengxu Zhang
- Division of Systems Pharmacology and Pharmacy, Predictive Pharmacology Group, Leiden Academic Centre of Drug Research, Leiden University, Gorlaeus Laboratories, Leiden, the Netherlands
| | - Ilona M Vuist
- Charles River Laboratories, Groningen, the Netherlands
| | - Vivi Rottschäfer
- Mathematical Institute, Leiden University, Leiden, the Netherlands; Korteweg-de Vries Institute for Mathematics, University of Amsterdam, P.O. Box 94248, 1090 GE Amsterdam, the Netherlands
| | - Elizabeth Cm de Lange
- Division of Systems Pharmacology and Pharmacy, Predictive Pharmacology Group, Leiden Academic Centre of Drug Research, Leiden University, Gorlaeus Laboratories, Leiden, the Netherlands.
| |
Collapse
|
3
|
Carr LM, Mustafa S, Care A, Collins-Praino LE. More than a number: Incorporating the aged phenotype to improve in vitro and in vivo modeling of neurodegenerative disease. Brain Behav Immun 2024; 119:554-571. [PMID: 38663775 DOI: 10.1016/j.bbi.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 03/04/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
Age is the number one risk factor for developing a neurodegenerative disease (ND), such as Alzheimer's disease (AD) or Parkinson's disease (PD). With our rapidly ageing world population, there will be an increased burden of ND and need for disease-modifying treatments. Currently, however, translation of research from bench to bedside in NDs is poor. This may be due, at least in part, to the failure to account for the potential effect of ageing in preclinical modelling of NDs. While ageing can impact upon physiological response in multiple ways, only a limited number of preclinical studies of ND have incorporated ageing as a factor of interest. Here, we evaluate the aged phenotype and highlight the critical, but unmet, need to incorporate aspects of this phenotype into both the in vitro and in vivo models used in ND research. Given technological advances in the field over the past several years, we discuss how these could be harnessed to create novel models of ND that more readily incorporate aspects of the aged phenotype. This includes a recently described in vitro panel of ageing markers, which could help lead to more standardised models and improve reproducibility across studies. Importantly, we cannot assume that young cells or animals yield the same responses as seen in the context of ageing; thus, an improved understanding of the biology of ageing, and how to appropriately incorporate this into the modelling of ND, will ensure the best chance for successful translation of new therapies to the aged patient.
Collapse
Affiliation(s)
- Laura M Carr
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Sanam Mustafa
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, Adelaide, SA, Australia; Davies Livestock Research Centre, The University of Adelaide, Roseworthy, SA, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Lyndsey E Collins-Praino
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
4
|
Zhang M, Rottschäfer V, C M de Lange E. The potential impact of CYP and UGT drug-metabolizing enzymes on brain target site drug exposure. Drug Metab Rev 2024; 56:1-30. [PMID: 38126313 DOI: 10.1080/03602532.2023.2297154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Drug metabolism is one of the critical determinants of drug disposition throughout the body. While traditionally associated with the liver, recent research has unveiled the presence and functional significance of drug-metabolizing enzymes (DMEs) within the brain. Specifically, cytochrome P-450 enzymes (CYPs) and UDP-glucuronosyltransferases (UGTs) enzymes have emerged as key players in drug biotransformation within the central nervous system (CNS). This comprehensive review explores the cellular and subcellular distribution of CYPs and UGTs within the CNS, emphasizing regional expression and contrasting profiles between the liver and brain, humans and rats. Moreover, we discuss the impact of species and sex differences on CYPs and UGTs within the CNS. This review also provides an overview of methodologies for identifying and quantifying enzyme activities in the brain. Additionally, we present factors influencing CYPs and UGTs activities in the brain, including genetic polymorphisms, physiological variables, pathophysiological conditions, and environmental factors. Examples of CYP- and UGT-mediated drug metabolism within the brain are presented at the end, illustrating the pivotal role of these enzymes in drug therapy and potential toxicity. In conclusion, this review enhances our understanding of drug metabolism's significance in the brain, with a specific focus on CYPs and UGTs. Insights into the expression, activity, and influential factors of these enzymes within the CNS have crucial implications for drug development, the design of safe drug treatment strategies, and the comprehension of drug actions within the CNS. To that end, CNS pharmacokinetic (PK) models can be improved to further advance drug development and personalized therapy.
Collapse
Affiliation(s)
- Mengxu Zhang
- Division of Systems Pharmacology and Pharmacy, Predictive Pharmacology Group, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Vivi Rottschäfer
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Korteweg-de Vries Institute for Mathematics, University of Amsterdam, Amsterdam, The Netherlands
| | - Elizabeth C M de Lange
- Division of Systems Pharmacology and Pharmacy, Predictive Pharmacology Group, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
5
|
Bautista M, Mogul AS, Fowler CD. Beyond the label: current evidence and future directions for the interrelationship between electronic cigarettes and mental health. Front Psychiatry 2023; 14:1134079. [PMID: 37645635 PMCID: PMC10460914 DOI: 10.3389/fpsyt.2023.1134079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 07/31/2023] [Indexed: 08/31/2023] Open
Abstract
Electronic cigarette use has dramatically increased over the last decade. With this recent technological development and wide range of constituents in various products, putative adverse effects on the brain and body have been largely unexplored. Here, we review current evidence linking electronic nicotine cigarette use with potential health consequences and provide evidence supporting an association between drug use and depression in humans. We also examine the biological effects of individual constituents in electronic cigarette aerosols, which include labeled ingredients, such as propylene glycol, vegetable glycerin, nicotine, and flavorants, as well as unlabeled ingredients found in the aerosols, such as carbonyls and heavy metals. Lastly, we examine the effects of electronic cigarette use on endogenous metabolism via changes in cytochrome P450 enzymes, which can thereby impact therapeutic outcomes. While the current evidence offers insight into the potential effects of electronic cigarette use on biological processes, further studies are necessary to determine the long-term clinical relevance of aerosol inhalation.
Collapse
Affiliation(s)
| | | | - Christie D. Fowler
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
6
|
Characterization of the Stereoselective Disposition of Bupropion and Its Metabolites in Rat Plasma and Brain. Eur J Drug Metab Pharmacokinet 2023; 48:171-187. [PMID: 36823342 DOI: 10.1007/s13318-023-00817-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2023] [Indexed: 02/25/2023]
Abstract
BACKGROUND AND OBJECTIVES Bupropion is an atypical antidepressant and smoking cessation aid; its use is associated with wide intersubject variability in efficacy and safety. Knowledge of the brain pharmacokinetics of bupropion and its pharmacologically active metabolites is considered important for understanding the cause-effect relationships driving this variability. METHODS Brain concentrations from rats administered a 10 mg/kg subcutaneous dose of racemic bupropion were analyzed using a stereoselective LC/MS-MS method. A 2 mg/kg dose of (S,S)-hydroxybupropion, which has comparable pharmacologic potency to bupropion, was administered to a separate group of rats. Plasma exposure and unbound concentrations in both matrices from companion equilibrium dialysis experiments were determined to assess potential carrier-mediated transport at the blood-brain barrier. RESULTS Exposures to unbound forms of bupropion enantiomers were similar in plasma; this was also true in brain. This trend held for reductive diastereomer metabolite pairs in the two matrices. Unbound (R,R)-hydroxybupropion exposure was 1.5-fold higher than (S,S)-hydroxybupropion exposure in plasma and brain following bupropion administration. Unbound concentration ratios (Kp,uu) of a given molecular form decreased over time: between 4 and 6 h, these were < 1 for the two bupropion enantiomers, and they were ~ 1 for metabolites that formed. Administration of preformed (S,S)-hydroxybupropion also demonstrated a declining Kp,uu. CONCLUSIONS The temporal shift in Kp,uu among the different molecular forms provides evidence regarding the operation of carrier-mediated transport and/or within-brain metabolism of bupropion, and, thereby, fresh insight regarding the causes of intersubject variability in the safety and efficacy of bupropion therapy.
Collapse
|
7
|
Stocco MR, Tyndale RF. Cytochrome P450 enzymes and metabolism of drugs and neurotoxins within the mammalian brain. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:73-106. [PMID: 35953164 DOI: 10.1016/bs.apha.2022.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cytochrome P450 enzymes (CYPs) that metabolize xenobiotics are expressed and active in the brain. These CYPs contribute to the metabolism of many centrally acting compounds, including clinically used drugs, drugs of abuse, and neurotoxins. Although CYP levels are lower in the brain than in the liver, they may influence central substrate and metabolite concentrations, which could alter resulting centrally-mediated responses to these compounds. Additionally, xenobiotic metabolizing CYPs are highly variable due to genetic polymorphisms and regulation by endogenous and xenobiotic molecules. In the brain, these CYPs are sensitive to xenobiotic induction. As a result, CYPs in the brain vary widely, including among humans, and this CYP variation may influence central metabolism and resulting response to centrally acting compounds. It has been demonstrated, using experimental manipulation of CYP activity in vivo selectively within the brain, that CYP metabolism in the brain alters central substrate and metabolite concentrations, as well as drug response and neurotoxic effects. This suggests that variability in xenobiotic metabolizing CYPs in the human brain may meaningfully contribute to individual differences in response to, and effects of, centrally acting drugs and neurotoxins. This chapter will provide an overview of CYP expression in the brain, endogenous- and xenobiotic-mediated CYP regulation, and the functional impact of CYP-mediated metabolism of drugs and neurotoxins in the brain, with a focus on experimental approaches in mice, rats, and non-human primates, and a discussion regarding the potential role of xenobiotic metabolizing CYPs in the human brain.
Collapse
Affiliation(s)
- Marlaina R Stocco
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Rachel F Tyndale
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Daniel WA, Bromek E, Danek PJ, Haduch A. The mechanisms of interactions of psychotropic drugs with liver and brain cytochrome P450 and their significance for drug effect and drug-drug interactions. Biochem Pharmacol 2022; 199:115006. [PMID: 35314167 DOI: 10.1016/j.bcp.2022.115006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 02/08/2023]
Abstract
Cytochrome P450 (CYP) plays an important role in psychopharmacology. While liver CYP enzymes are responsible for the biotransformation of psychotropic drugs, brain CYP enzymes are involved in the local metabolism of these drugs and endogenous neuroactive substances, such as neurosteroids, and in alternative pathways of neurotransmitter biosynthesis including dopamine and serotonin. Recent studies have revealed a relation between the brain nervous system and cytochrome P450, indicating that CYP enzymes metabolize endogenous neuroactive substances in the brain, while the brain nervous system is engaged in the central neuroendocrine and neuroimmune regulation of cytochrome P450 in the liver. Therefore, the effect of neuroactive drugs on cytochrome P450 should be investigated not only in vitro, but also at in vivo conditions, since only in vivo all mechanisms of drug-enzyme interaction can be observed, including neuroendocrine and neuroimmune modulation. Psychotropic drugs can potentially affect cytochrome P450 via a number of mechanisms operating at the level of the nervous, hormonal and immune systems, and the liver. Their effect on cytochrome P450 in the brain is often different than in the liver and region-dependent. Since psychotropic drugs can affect cytochrome P450 both in the liver and brain, they can modify their own pharmacological effect at both pharmacokinetic and pharmacodynamic level. The article describes the mechanisms by which psychotropic drugs can change the expression/activity of cytochrome P450 in the liver and brain, and discusses the significance of those mechanisms for drug action and drug-drug interactions. Moreover, the brain CYP2D6 is considered as a potential target for psychotropics.
Collapse
Affiliation(s)
- Władysława A Daniel
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland.
| | - Ewa Bromek
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Przemysław J Danek
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Anna Haduch
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| |
Collapse
|
9
|
CYP 450 enzymes influence (R,S)-ketamine brain delivery and its antidepressant activity. Neuropharmacology 2021; 206:108936. [PMID: 34965407 DOI: 10.1016/j.neuropharm.2021.108936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/07/2021] [Accepted: 12/21/2021] [Indexed: 11/23/2022]
Abstract
Esketamine, the S-stereoisomer of (R,S)-ketamine was recently approved by drug agencies (FDA, EMA), as an antidepressant drug with a new mechanism of action. (R,S)-ketamine is a N-methyl-d-aspartate receptor (NMDA-R) antagonist putatively acting on GABAergic inhibitory synapses to increase excitatory synaptic glutamatergic neurotransmission. Unlike monoamine-based antidepressants, (R,S)-ketamine exhibits rapid and persistent antidepressant activity at subanesthetic doses in preclinical rodent models and in treatment-resistant depressed patients. Its major brain metabolite, (2R,6R)-hydroxynorketamine (HNK) is formed following (R,S)-ketamine metabolism by various cytochrome P450 enzymes (CYP) mainly activated in the liver depending on routes of administration [e.g., intravenous (largely used for a better bioavailability), intranasal spray, intracerebral, subcutaneous, intramuscular or oral]. Experimental or clinical studies suggest that (2R,6R)-HNK could be an antidepressant drug candidate. However, questions still remain regarding its molecular and cellular targets in the brain and its role in (R,S)-ketamine's fast-acting antidepressant effects. The purpose of the present review is: 1) to review (R,S)-ketamine pharmacokinetic properties in humans and rodents and its metabolism by CYP enzymes to form norketamine and HNK metabolites; 2) to provide a summary of preclinical strategies challenging the role of these metabolites by modifying (R,S)-ketamine metabolism, e.g., by administering a pre-treatment CYP inducers or inhibitors; 3) to analyze the influence of sex and age on CYP expression and (R,S)-ketamine metabolism. Importantly, this review describes (R,S)-ketamine pharmacodynamics and pharmacokinetics to alert clinicians about possible drug-drug interactions during a concomitant administration of (R,S)-ketamine and CYP inducers/inhibitors that could enhance or blunt, respectively, (R,S)-ketamine's therapeutic antidepressant efficacy in patients.
Collapse
|
10
|
Song Y, Li C, Liu G, Liu R, Chen Y, Li W, Cao Z, Zhao B, Lu C, Liu Y. Drug-Metabolizing Cytochrome P450 Enzymes Have Multifarious Influences on Treatment Outcomes. Clin Pharmacokinet 2021; 60:585-601. [PMID: 33723723 DOI: 10.1007/s40262-021-01001-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Drug metabolism is a critical process for the removal of unwanted substances from the body. In humans, approximately 80% of oxidative metabolism and almost 50% of the overall elimination of commonly used drugs can be attributed to one or more of various cytochrome P450 (CYP) enzymes from CYP families 1-3. In addition to the basic metabolic effects for elimination, CYP enzymes in vivo are capable of affecting the treatment outcomes in many cases. Drug-metabolizing CYP enzymes are mainly expressed in the liver and intestine, the two principal drug oxidation and elimination organs, where they can significantly influence the drug action, safety, and bioavailability by mediating phase I metabolism and first-pass metabolism. Furthermore, CYP-mediated local drug metabolism in the sites of action may also have the potential to impact drug response, according to the literature in recent years. This article underlines the ability of CYP enzymes to influence treatment outcomes by discussing CYP-mediated diversified drug metabolism in primary metabolic sites (liver and intestine) and typical action sites (brain and tumors) according to their expression levels and metabolic activity. Moreover, intrinsic and extrinsic factors of personal differential CYP phenotypes that contribute to interindividual variation of treatment outcomes are also reviewed to introduce the multifarious pivotal role of CYP-mediated metabolism and clearance in drug therapy.
Collapse
Affiliation(s)
- Yurong Song
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chenxi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Guangzhi Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Rui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Youwen Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wen Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhiwen Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Baosheng Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
11
|
Müller-Wirtz LM, Maurer F, Brausch T, Kiefer D, Floss M, Doneit J, Volk T, Sessler DI, Fink T, Lehr T, Kreuer S. Exhaled Propofol Concentrations Correlate With Plasma and Brain Tissue Concentrations in Rats. Anesth Analg 2021; 132:110-118. [PMID: 32118620 DOI: 10.1213/ane.0000000000004701] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Propofol can be measured in exhaled gas. Exhaled and plasma propofol concentrations correlate well, but the relationship with tissue concentrations remains unknown. We thus evaluated the relationship between exhaled, plasma, and various tissue propofol concentrations. Because the drug acts in the brain, we focused on the relationship between exhaled and brain tissue propofol concentrations. METHODS Thirty-six male Sprague-Dawley rats were anesthetized with propofol, ketamine, and rocuronium for 6 hours. Animals were randomly assigned to propofol infusions at 20, 40, or 60 mg·kg·h (n = 12 per group). Exhaled propofol concentrations were measured at 15-minute intervals by multicapillary column-ion mobility spectrometry. Arterial blood samples, 110 µL each, were collected 15, 30, and 45 minutes, and 1, 2, 4, and 6 hours after the propofol infusion started. Propofol concentrations were measured in brain, lung, liver, kidney, muscle, and fat tissue after 6 hours. The last exhaled and plasma concentrations were used for linear regression analyses with tissue concentrations. RESULTS The correlation of exhaled versus plasma concentrations (R = 0.71) was comparable to the correlation of exhaled versus brain tissue concentrations (R = 0.75) at the end of the study. In contrast, correlations between plasma and lung and between lung and exhaled propofol concentrations were poor. Less than a part-per-thousand of propofol was exhaled over 6 hours. CONCLUSIONS Exhaled propofol concentrations correlate reasonably well with brain tissue and plasma concentrations in rats, and may thus be useful to estimate anesthetic drug effect. The equilibration between plasma propofol and exhaled gas is apparently independent of lung tissue concentration. Only a tiny fraction of administered propofol is eliminated via the lungs, and exhaled quantities thus have negligible influence on plasma concentrations.
Collapse
Affiliation(s)
- Lukas M Müller-Wirtz
- From the Department of Anesthesiology, Intensive Care and Pain Therapy, Center of Breath Research, Saarland University Medical Center, Homburg (Saar), Germany
| | - Felix Maurer
- From the Department of Anesthesiology, Intensive Care and Pain Therapy, Center of Breath Research, Saarland University Medical Center, Homburg (Saar), Germany
| | - Timo Brausch
- From the Department of Anesthesiology, Intensive Care and Pain Therapy, Center of Breath Research, Saarland University Medical Center, Homburg (Saar), Germany
| | - Daniel Kiefer
- From the Department of Anesthesiology, Intensive Care and Pain Therapy, Center of Breath Research, Saarland University Medical Center, Homburg (Saar), Germany
| | - Maximilian Floss
- From the Department of Anesthesiology, Intensive Care and Pain Therapy, Center of Breath Research, Saarland University Medical Center, Homburg (Saar), Germany
| | - Jonas Doneit
- From the Department of Anesthesiology, Intensive Care and Pain Therapy, Center of Breath Research, Saarland University Medical Center, Homburg (Saar), Germany
| | - Thomas Volk
- From the Department of Anesthesiology, Intensive Care and Pain Therapy, Center of Breath Research, Saarland University Medical Center, Homburg (Saar), Germany
| | - Daniel I Sessler
- Department of Outcomes Research, Anesthesiology Institute, Cleveland Clinic, Cleveland, Ohio
| | - Tobias Fink
- From the Department of Anesthesiology, Intensive Care and Pain Therapy, Center of Breath Research, Saarland University Medical Center, Homburg (Saar), Germany
| | - Thorsten Lehr
- Clinical Pharmacy, Department of Pharmacy, Saarland University, Saarbrücken, Germany. The Center of Breath Research is part of the Outcomes Research Consortium, Cleveland, Ohio, USA
| | - Sascha Kreuer
- From the Department of Anesthesiology, Intensive Care and Pain Therapy, Center of Breath Research, Saarland University Medical Center, Homburg (Saar), Germany
| |
Collapse
|
12
|
Abstract
The regulation of brain cytochrome P450 enzymes (CYPs) is different compared with respective hepatic enzymes. This may result from anatomical bases and physiological functions of the two organs. The brain is composed of a variety of functional structures built of different interconnected cell types endowed with specific receptors that receive various neuronal signals from other brain regions. Those signals activate transcription factors or alter functioning of enzyme proteins. Moreover, the blood-brain barrier (BBB) does not allow free penetration of all substances from the periphery into the brain. Differences in neurotransmitter signaling, availability to endogenous and exogenous active substances, and levels of transcription factors between neuronal and hepatic cells lead to differentiated expression and susceptibility to the regulation of CYP genes in the brain and liver. Herein, we briefly describe the CYP enzymes of CYP1-3 families, their distribution in the brain, and discuss brain-specific regulation of CYP genes. In parallel, a comparison to liver CYP regulation is presented. CYP enzymes play an essential role in maintaining the levels of bioactive molecules within normal ranges. These enzymes modulate the metabolism of endogenous neurochemicals, such as neurosteroids, dopamine, serotonin, melatonin, anandamide, and exogenous substances, including psychotropics, drugs of abuse, neurotoxins, and carcinogens. The role of these enzymes is not restricted to xenobiotic-induced neurotoxicity, but they are also involved in brain physiology. Therefore, it is crucial to recognize the function and regulation of CYP enzymes in the brain to build a foundation for future medicine and neuroprotection and for personalized treatment of brain diseases.
Collapse
Affiliation(s)
- Wojciech Kuban
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Władysława Anna Daniel
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
13
|
Stocco MR, Tolledo C, Wadji FB, Gonzalez FJ, Miksys S, Tyndale RF. Human CYP2D6 in the Brain Is Protective Against Harmine-Induced Neurotoxicity: Evidence from Humanized CYP2D6 Transgenic Mice. Mol Neurobiol 2020; 57:4608-4621. [PMID: 32761352 PMCID: PMC8865091 DOI: 10.1007/s12035-020-02050-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/28/2020] [Indexed: 10/25/2022]
Abstract
CYP2D6 metabolically inactivates several neurotoxins, including beta-carbolines, which are implicated in neurodegenerative diseases. Variation in CYP2D6 within the brain may alter local inactivation of neurotoxic beta-carbolines, thereby influencing neurotoxicity. The beta-carboline harmine, which induces hypothermia and tremor, is metabolized by CYP2D6 to the non-hypothermic/non-tremorgenic harmol. Transgenic mice (TG), expressing human CYP2D6 in addition to their endogenous mouse CYP2D, experience less harmine-induced hypothermia and tremor compared with wild-type mice (WT). We first sought to elucidate the role of CYP2D in general within the brain in harmine-induced hypothermia and tremor severity. A 4-h intracerebroventricular (ICV) pretreatment with the CYP2D inhibitor propranolol increased harmine-induced hypothermia and tremor in TG and increased harmine-induced hypothermia in WT. We next sought to specifically demonstrate that human CYP2D6 expressed in TG brain altered harmine response severity. A 24-h ICV propranolol pretreatment, which selectively and irreversibly inhibits human CYP2D6 in TG brain, increased harmine-induced hypothermia. This 24-h pretreatment had no impact on harmine response in WT, as propranolol is not an irreversible inhibitor of mouse CYP2D in the brain, thus confirming no off-target effects of ICV propranolol pretreatment. Human CYP2D6 activity in TG brain was sufficient in vivo to mitigate harmine-induced neurotoxicity. These findings suggest that human CYP2D6 in the brain is protective against beta-carboline-induced neurotoxicity and that the extensive interindividual variability in CYP2D6 expression in human brain may contribute to variation in susceptibility to certain neurotoxin-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Marlaina R Stocco
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Cole Tolledo
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Fariba Baghai Wadji
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sharon Miksys
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Rachel F Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
- Department of Psychiatry, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
14
|
Bhattacharya C, Kirby D, Van Stipdonk M, Stratford RE. Comparison of In Vitro Stereoselective Metabolism of Bupropion in Human, Monkey, Rat, and Mouse Liver Microsomes. Eur J Drug Metab Pharmacokinet 2019; 44:261-274. [PMID: 30298475 DOI: 10.1007/s13318-018-0516-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES Bupropion is an atypical antidepressant and smoking cessation aid associated with wide intersubject variability. This study compared the formation kinetics of three phase I metabolites (hydroxybupropion, threohydrobupropion, and erythrohydrobupropion) in human, marmoset, rat, and mouse liver microsomes. The objective was to establish suitability and limitations for subsequent use of nonclinical species to model bupropion central nervous system (CNS) disposition in humans. METHODS Hepatic microsomal incubations were conducted separately for the R- and S-bupropion enantiomers, and the formation of enantiomer-specific metabolites was determined using LC-MS/MS. Intrinsic formation clearance (CLint) of metabolites across the four species was determined from the formation rate versus substrate concentration relationship. RESULTS The total clearance of S-bupropion was higher than that of R-bupropion in monkey and human liver microsomes. The contribution of hydroxybupropion to the total racemic bupropion clearance was 38%, 62%, 17%, and 96% in human, monkey, rat, and mouse, respectively. In the same species order, threohydrobupropion contributed 53%, 23%, 17%, and 3%, and erythrohydrobupropion contributed 9%, 14%, 66%, and 1.3%, respectively, to racemic bupropion clearance. CONCLUSION The results demonstrate that phase I metabolism in monkeys best approximates that observed in humans, and support the preferred use of this species to investigate possible pharmacokinetic factors that influence the CNS disposition of bupropion and contribute to its high intersubject variability.
Collapse
Affiliation(s)
- Chandrali Bhattacharya
- Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA, 15282, USA.,Department of Pharmacy Practice, Purdue University, Indianapolis, IN, 46202, USA
| | - Danielle Kirby
- Department of Chemistry and Biochemistry, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA, 15282, USA
| | - Michael Van Stipdonk
- Department of Chemistry and Biochemistry, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA, 15282, USA
| | - Robert E Stratford
- Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA, 15282, USA. .,Indiana University School of Medicine, Research II, Suite 480, 950 W. Walnut St, Indianapolis, IN, 46202-5188, USA. .,Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
15
|
Vendel E, Rottschäfer V, de Lange ECM. The need for mathematical modelling of spatial drug distribution within the brain. Fluids Barriers CNS 2019; 16:12. [PMID: 31092261 PMCID: PMC6521438 DOI: 10.1186/s12987-019-0133-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/19/2019] [Indexed: 12/17/2022] Open
Abstract
The blood brain barrier (BBB) is the main barrier that separates the blood from the brain. Because of the BBB, the drug concentration-time profile in the brain may be substantially different from that in the blood. Within the brain, the drug is subject to distributional and elimination processes: diffusion, bulk flow of the brain extracellular fluid (ECF), extra-intracellular exchange, bulk flow of the cerebrospinal fluid (CSF), binding and metabolism. Drug effects are driven by the concentration of a drug at the site of its target and by drug-target interactions. Therefore, a quantitative understanding is needed of the distribution of a drug within the brain in order to predict its effect. Mathematical models can help in the understanding of drug distribution within the brain. The aim of this review is to provide a comprehensive overview of system-specific and drug-specific properties that affect the local distribution of drugs in the brain and of currently existing mathematical models that describe local drug distribution within the brain. Furthermore, we provide an overview on which processes have been addressed in these models and which have not. Altogether, we conclude that there is a need for a more comprehensive and integrated model that fills the current gaps in predicting the local drug distribution within the brain.
Collapse
Affiliation(s)
- Esmée Vendel
- Mathematical Institute, Leiden University, Niels Bohrweg 1, 2333CA, Leiden, The Netherlands
| | - Vivi Rottschäfer
- Mathematical Institute, Leiden University, Niels Bohrweg 1, 2333CA, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC, Leiden, The Netherlands.
| |
Collapse
|
16
|
Rath SN, Jena L, Patri M. Understanding ligands driven mechanism of wild and mutant aryl hydrocarbon receptor in presence of phytochemicals combating Parkinson’s disease: an in silico and in vivo study. J Biomol Struct Dyn 2019; 38:807-826. [DOI: 10.1080/07391102.2019.1590240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Surya Narayan Rath
- Department of Bioinformatics, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
- Neurobiology Laboratory, Department of Zoology, School of Life Sciences, Ravenshaw University, Cuttack, Odisha, India
| | - Lingaraja Jena
- Bioinformatics Centre, Mahatma Gandhi Institute of Medical Sciences, Wardha, Maharashtra, India
| | - Manorama Patri
- Neurobiology Laboratory, Department of Zoology, School of Life Sciences, Ravenshaw University, Cuttack, Odisha, India
| |
Collapse
|
17
|
Wu GJ, Lin YW, Tsai HC, Lee YW, Chen JT, Chen RM. Sepsis-induced liver dysfunction was ameliorated by propofol via suppressing hepatic lipid peroxidation, inflammation, and drug interactions. Life Sci 2018; 213:279-286. [PMID: 30352244 DOI: 10.1016/j.lfs.2018.10.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022]
Abstract
AIMS Our previous study showed that propofol can protect against sepsis-induced insults through suppressing liver nitrosation and inflammation. This study further evaluated the mechanisms of propofol-caused protection from sepsis-induced liver dysfunction. MAIN METHODS Male Wistar rats were subjected to cecal ligation and puncture (CLP) and then exposed to propofol. Levels of hepatic oxidative stress and lipid peroxidation were consecutively measured. Expressions of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-4 messenger (m)RNA or proteins were quantified. Effects of propofol on microsomal pentoxyresorufin O-dealkelase (PROD) and ethoxycoumarin O-deethylase (ECOD) activities were determined. KEY FINDINGS Administration of propofol to CLP-treated rats significantly attenuated sepsis-induced insults. CLP caused augmented serum aspartate aminotransferase and alanine aminotransferase activities and concurrently triggered liver damage. In contrast, treatment with propofol protected against CLP-induced liver dysfunction. As to the mechanisms, the CLP-induced increases in oxidative stress and lipid peroxidation levels and TNF-α and IL-1β mRNA and protein expressions were subsequently attenuated by propofol. Furthermore, administration of CLP-treated rats with propofol augmented levels of IL-4 in the liver. Phenobarbital treatment of liver microsomes in CLP-treated rats produced less amplification of PROD and ECOD activities, and a smaller amount of 4-hydroxypropofol was metabolized from propofol by liver microsomes. In contrast, more drug interactions occurred with propofol, which decreased PROD and ECOD activities in liver microsomes of CLP-treated rats. SIGNIFICANCE Taken together, the present study showed that propofol can protect against sepsis-induced liver dysfunction through suppressing hepatic oxidative stress, lipid peroxidation, inflammation, and drug biotransformation and interactions in the liver.
Collapse
Affiliation(s)
- Gong-Jhe Wu
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Wei Lin
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chien Tsai
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Wen Lee
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jui-Tai Chen
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ruei-Ming Chen
- Anesthesiology and Health Policy Research Center, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
18
|
Metabolic Profiles of Propofol and Fospropofol: Clinical and Forensic Interpretative Aspects. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6852857. [PMID: 29992157 PMCID: PMC5994321 DOI: 10.1155/2018/6852857] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/27/2018] [Accepted: 04/15/2018] [Indexed: 02/08/2023]
Abstract
Propofol is an intravenous short-acting anesthetic widely used to induce and maintain general anesthesia and to provide procedural sedation. The potential for propofol dependency and abuse has been recognized, and several cases of accidental overdose and suicide have emerged, mostly among the health professionals. Different studies have demonstrated an unpredictable interindividual variability of propofol pharmacokinetics and pharmacodynamics with forensic and clinical adverse relevant outcomes (e.g., pronounced respiratory and cardiac depression), namely, due to polymorphisms in the UDP-glucuronosyltransferase and cytochrome P450 isoforms and drugs administered concurrently. In this work the pharmacokinetics of propofol and fospropofol with particular focus on metabolic pathways is fully reviewed. It is concluded that knowing the metabolism of propofol may lead to the development of new clues to help further toxicological and clinical interpretations and to reduce serious adverse reactions such as respiratory failure, metabolic acidosis, rhabdomyolysis, cardiac bradyarrhythmias, hypotension and myocardial failure, anaphylaxis, hypertriglyceridemia, renal failure, hepatomegaly, hepatic steatosis, acute pancreatitis, abuse, and death. Particularly, further studies aiming to characterize polymorphic enzymes involved in the metabolic pathway, the development of additional routine forensic toxicological analysis, and the relatively new field of ‘‘omics” technology, namely, metabolomics, can offer more in explaining the unpredictable interindividual variability.
Collapse
|
19
|
Development and validation of brain target controlled infusion of propofol in mice. PLoS One 2018; 13:e0194949. [PMID: 29684039 PMCID: PMC5912730 DOI: 10.1371/journal.pone.0194949] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 03/13/2018] [Indexed: 12/25/2022] Open
Abstract
Mechanisms through which anesthetics disrupt neuronal activity are incompletely understood. In order to study anesthetic mechanisms in the intact brain, tight control over anesthetic pharmacology in a genetically and neurophysiologically accessible animal model is essential. Here, we developed a pharmacokinetic model that quantitatively describes propofol distribution into and elimination out of the brain. To develop the model, we used jugular venous catheters to infuse propofol in mice and measured propofol concentration in serial timed brain and blood samples using high performance liquid chromatography (HPLC). We then used adaptive fitting procedures to find parameters of a three compartment pharmacokinetic model such that all measurements collected in the blood and in the brain across different infusion schemes are fit by a single model. The purpose of the model was to develop target controlled infusion (TCI) capable of maintaining constant brain propofol concentration at the desired level. We validated the model for two different targeted concentrations in independent cohorts of experiments not used for model fitting. The predictions made by the model were unbiased, and the measured brain concentration was indistinguishable from the targeted concentration. We also verified that at the targeted concentration, state of anesthesia evidenced by slowing of the electroencephalogram and behavioral unresponsiveness was attained. Thus, we developed a useful tool for performing experiments necessitating use of anesthetics and for the investigation of mechanisms of action of propofol in mice.
Collapse
|
20
|
McMillan DM, Tyndale RF. CYP-mediated drug metabolism in the brain impacts drug response. Pharmacol Ther 2018; 184:189-200. [DOI: 10.1016/j.pharmthera.2017.10.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
21
|
Garcia KLP, Lê AD, Tyndale RF. Brain CYP2B induction can decrease nicotine levels in the brain. Addict Biol 2017; 22:1257-1266. [PMID: 27230546 DOI: 10.1111/adb.12411] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 03/20/2016] [Accepted: 04/21/2016] [Indexed: 01/07/2023]
Abstract
Nicotine can be metabolized by the enzyme CYP2B; brain CYP2B is higher in rats and monkeys treated with nicotine, and in human smokers. A 7-day nicotine treatment increased CYP2B expression in rat brain but not liver, and decreased the behavioral response and brain levels (ex vivo) to the CYP2B substrate propofol. However, the effect of CYP2B induction on the time course and levels of circulating brain nicotine in vivo has not been demonstrated. Using brain microdialysis, nicotine levels following a subcutaneous nicotine injection were measured on day one and after a 7-day nicotine treatment. There was a significant time x treatment interaction (p = 0.01); peak nicotine levels (15-45 minutes post-injection) were lower after treatment (p = 0.04) consistent with CYP2B induction. Following a two-week washout period, brain nicotine levels increased to day one levels (p = 0.02), consistent with brain CYP2B levels returning to baseline. Brain pretreatment of the CYP2B inhibitor, C8-xanthate, increased brain nicotine levels acutely and after 7-day nicotine treatment, indicating the alterations in brain nicotine levels were due to changes in brain CYP2B activity. Plasma nicotine levels were not altered for any time or treatment sampled, confirming no effect on peripheral nicotine metabolism. These results demonstrate that chronic nicotine, by increasing brain CYP2B activity, reduces brain nicotine levels, which could alter nicotine's reinforcing effects. Higher brain CYP2B levels in smokers could lower brain nicotine levels; as this induction would occur following continued nicotine exposure it could increase withdrawal symptoms and contribute to sustaining smoking behavior.
Collapse
Affiliation(s)
- Kristine L. P. Garcia
- Departments of Pharmacology and Toxicology and Psychiatry; Toronto Ontario Canada
- Campbell Family Mental Health Research Institute; Centre for Addiction and Mental Health; Toronto Ontario Canada
| | - Anh Dzung Lê
- Departments of Pharmacology and Toxicology and Psychiatry; Toronto Ontario Canada
- Campbell Family Mental Health Research Institute; Centre for Addiction and Mental Health; Toronto Ontario Canada
| | - Rachel F. Tyndale
- Departments of Pharmacology and Toxicology and Psychiatry; Toronto Ontario Canada
- Campbell Family Mental Health Research Institute; Centre for Addiction and Mental Health; Toronto Ontario Canada
| |
Collapse
|
22
|
Toselli F, Dodd PR, Gillam EMJ. Emerging roles for brain drug-metabolizing cytochrome P450 enzymes in neuropsychiatric conditions and responses to drugs. Drug Metab Rev 2016; 48:379-404. [DOI: 10.1080/03602532.2016.1221960] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
23
|
Ghosh C, Hossain M, Solanki J, Dadas A, Marchi N, Janigro D. Pathophysiological implications of neurovascular P450 in brain disorders. Drug Discov Today 2016; 21:1609-1619. [PMID: 27312874 DOI: 10.1016/j.drudis.2016.06.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/14/2016] [Accepted: 06/06/2016] [Indexed: 01/12/2023]
Abstract
Over the past decades, the significance of cytochrome P450 (CYP) enzymes has expanded beyond their role as peripheral drug metabolizers in the liver and gut. CYP enzymes are also functionally active at the neurovascular interface. CYP expression is modulated by disease states, impacting cellular functions, detoxification, and reactivity to toxic stimuli and brain drug biotransformation. Unveiling the physiological and molecular complexity of brain P450 enzymes will improve our understanding of the mechanisms underlying brain drug availability, pharmacological efficacy, and neurotoxic adverse effects from pharmacotherapy targeting brain disorders.
Collapse
Affiliation(s)
- Chaitali Ghosh
- Cerebrovascular Research, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA; Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
| | - Mohammed Hossain
- Cerebrovascular Research, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA; Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | | | - Aaron Dadas
- The Ohio State University, Columbus, OH, USA
| | - Nicola Marchi
- Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (CNRS/INSERM), Montpellier, France
| | - Damir Janigro
- Flocel Inc. and Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
24
|
Vanoye-Carlo A, Gutiérrez-Ospina G, Marcial-Quino J, Gómez-Manzo S, Garcia-Torres I, De la Mora-De la Mora I, Enríquez-Flores S, Méndez ST, Phillips-Farfán BV, Gutiérrez-Castrellón P. Analysis of Cyp2b1 gene expression in the rat liver and brain by multiplex PCR. Mol Cell Toxicol 2015. [DOI: 10.1007/s13273-015-0043-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
25
|
Subacute nicotine co-exposure has no effect on 2,2',3,5',6- pentachlorobiphenyl disposition but alters hepatic cytochrome P450 expression in the male rat. Toxicology 2015; 338:59-68. [PMID: 26463278 DOI: 10.1016/j.tox.2015.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/30/2015] [Accepted: 10/06/2015] [Indexed: 01/09/2023]
Abstract
Polychlorinated biphenyls (PCBs) are metabolized by cytochrome P450 2B enzymes (CYP2B) and nicotine is reported to alter CYP2B activity in the brain and liver. To test the hypothesis that nicotine influences PCB disposition, 2,2',3,5',6-pentachlorobiphenyl (PCB 95) and its metabolites were quantified in tissues of adult male Wistar rats exposed to PCB 95 (6mg/kg/d, p.o.) in the absence or presence of nicotine (1.0mg/kg/d of the tartrate salt, s.c.) for 7 consecutive days. PCB 95 was enantioselectively metabolized to hydroxylated (OH-) PCB metabolites, resulting in a pronounced enrichment of E1-PCB 95 in all tissues investigated. OH-PCBs were detected in blood and liver tissue, but were below the detection limit in adipose, brain and muscle tissues. Co-exposure to nicotine did not change PCB 95 disposition. CYP2B1 mRNA and CYP2B protein were not detected in brain tissues but were detected in liver. Co-exposure to nicotine and PCB 95 increased hepatic CYP2B1 mRNA but did not change CYP2B protein levels relative to vehicle control animals. However, hepatic CYP2B protein in animals co-exposed to PCB 95 and nicotine were reduced compared to animals that received only nicotine. Quantification of CYP2B3, CYP3A2 and CYP1A2 mRNA identified significant effects of nicotine and PCB 95 co-exposure on hepatic CYP3A2 and hippocampal CYP1A2 transcripts. Our findings suggest that nicotine co-exposure does not significantly influence PCB 95 disposition in the rat. However, these studies suggest a novel influence of PCB 95 and nicotine co-exposure on hepatic cytochrome P450 (P450) expression that may warrant further attention due to the increasing use of e-cigarettes and related products.
Collapse
|
26
|
Foti RS, Tyndale RF, Garcia KLP, Sweet DH, Nagar S, Sharan S, Rock DA. "Target-Site" Drug Metabolism and Transport. Drug Metab Dispos 2015; 43:1156-68. [PMID: 25986849 PMCID: PMC11024933 DOI: 10.1124/dmd.115.064576] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 05/18/2015] [Indexed: 04/20/2024] Open
Abstract
The recent symposium on "Target-Site" Drug Metabolism and Transport that was sponsored by the American Society for Pharmacology and Experimental Therapeutics at the 2014 Experimental Biology meeting in San Diego is summarized in this report. Emerging evidence has demonstrated that drug-metabolizing enzyme and transporter activity at the site of therapeutic action can affect the efficacy, safety, and metabolic properties of a given drug, with potential outcomes including altered dosing regimens, stricter exclusion criteria, or even the failure of a new chemical entity in clinical trials. Drug metabolism within the brain, for example, can contribute to metabolic activation of therapeutic drugs such as codeine as well as the elimination of potential neurotoxins in the brain. Similarly, the activity of oxidative and conjugative drug-metabolizing enzymes in the lung can have an effect on the efficacy of compounds such as resveratrol. In addition to metabolism, the active transport of compounds into or away from the site of action can also influence the outcome of a given therapeutic regimen or disease progression. For example, organic anion transporter 3 is involved in the initiation of pancreatic β-cell dysfunction and may have a role in how uremic toxins enter pancreatic β-cells and ultimately contribute to the pathogenesis of gestational diabetes. Finally, it is likely that a combination of target-specific metabolism and cellular internalization may have a significant role in determining the pharmacokinetics and efficacy of antibody-drug conjugates, a finding which has resulted in the development of a host of new analytical methods that are now used for characterizing the metabolism and disposition of antibody-drug conjugates. Taken together, the research summarized herein can provide for an increased understanding of potential barriers to drug efficacy and allow for a more rational approach for developing safe and effective therapeutics.
Collapse
Affiliation(s)
- Robert S Foti
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Rachel F Tyndale
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Kristine L P Garcia
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Douglas H Sweet
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Swati Nagar
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Satish Sharan
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Dan A Rock
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| |
Collapse
|
27
|
Garcia KLP, Coen K, Miksys S, Lê AD, Tyndale RF. Effect of Brain CYP2B Inhibition on Brain Nicotine Levels and Nicotine Self-Administration. Neuropsychopharmacology 2015; 40:1910-8. [PMID: 25652250 PMCID: PMC4839514 DOI: 10.1038/npp.2015.40] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/13/2015] [Accepted: 02/02/2015] [Indexed: 11/09/2022]
Abstract
The CYP2B enzyme is expressed in human and rat brain, and metabolizes many CNS-acting drugs. The gene that encodes human CYP2B6 is highly polymorphic, where the variation in brain enzyme levels could result in altered brain drug levels. CYP2B can metabolize nicotine, the main psychoactive ingredient in cigarettes; if altered brain CYP2B activity can influence nicotine brain levels, it could influence nicotine-mediated behaviors. To investigate this, a mechanism-based inhibitor selective for CYP2B, C8-xanthate (20 μg), was administered intracerebroventricularly (ICV) into the brain of rats, and 22 h later, nicotine levels were measured by in vivo microdialysis following nicotine (150 μg/kg intravenous). Brain nicotine levels from 15 to 30 min and the AUC0-45 min were both twofold higher (p<0.05) with C8-xanthate vs vehicle pretreatment; there was no difference in peripheral nicotine levels. Rats were then given ICV pretreatment with C8-xanthate/ASCF and underwent intravenous nicotine self-administration with 3.75-30 μg/kg per infusion dose. C8-xanthate pretreatment increased responding in progressive ratio (15 μg/kg per infusion dose, p<0.05). In a separate cohort, C8-xanthate increased the percentage of rats that acquired self-administration (7.5 μg/kg per infusion dose, p<0.05) from 40% after vehicle pretreatment to 100%, with no difference in peripheral nicotine levels measured at the end of behavior. In a third cohort, C8-xanthate increased the number of sessions required to meet extinction criteria (p<0.05). Together these data demonstrate that the brain CYP2B activity can influence nicotine brain levels and subsequent behaviors independent of hepatic metabolism. This suggests that human smokers with variable CYP2B brain levels could have different nicotine levels and reinforcement, which might have a role in smoking behaviors and dependence.
Collapse
Affiliation(s)
- Kristine L P Garcia
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Kathy Coen
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Sharon Miksys
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Anh Dzung Lê
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Rachel F Tyndale
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Pharmacology and Toxicology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Toronto, Ontario M5S 1A8, Canada, Tel: +1 416 978 6374, Fax: +1 416 978-6395, E-mail:
| |
Collapse
|
28
|
McMillan DM, Tyndale RF. Nicotine Increases Codeine Analgesia Through the Induction of Brain CYP2D and Central Activation of Codeine to Morphine. Neuropsychopharmacology 2015; 40:1804-12. [PMID: 25630571 PMCID: PMC4916647 DOI: 10.1038/npp.2015.32] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 01/22/2023]
Abstract
CYP2D metabolically activates codeine to morphine, which is required for codeine analgesia. Permeability across the blood-brain barrier, and active efflux, suggests that initial morphine in the brain after codeine is due to brain CYP2D metabolism. Human CYP2D is higher in the brains, but not in the livers, of smokers and 7-day nicotine treatment induces rat brain, but not hepatic, CYP2D. The role of nicotine-induced rat brain CYP2D in the central metabolic activation of peripherally administered codeine and resulting analgesia was investigated. Rats received 7-day nicotine (1 mg/kg subcutaneously) and/or a single propranolol (CYP2D mechanism-based inhibitor; 20 μg intracerebroventricularly) pretreatment, and then were tested for analgesia and drug levels following codeine (20 mg/kg intraperitoneally) or morphine (3.5 mg/kg intraperitoneally), matched for peak analgesia. Nicotine increased codeine analgesia (1.59X AUC(0-30 min) vs vehicle; p<0.001), while propranolol decreased analgesia (0.56X; p<0.05); co-pretreatment was similar to vehicle controls (1.23X; p>0.1). Nicotine increased, while propranolol decreased, brain, but not plasma, morphine levels, and analgesia correlated with brain (p<0.02), but not plasma (p>0.4), morphine levels after codeine. Pretreatments did not alter baseline or morphine analgesia. Here we show that brain CYP2D alters drug response despite the presence of substantial first-pass metabolism of codeine and further that nicotine induction of brain CYP2D increases codeine response in vivo. Thus variation in brain CYP2D activity, due to genetics or environment, may contribute to individual differences in response to centrally acting substrates. Exposure to nicotine may increase central drug metabolism, not detected peripherally, contributing to altered drug efficacy, onset time, and/or abuse liability.
Collapse
Affiliation(s)
- Douglas M McMillan
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH) and Departments of Psychiatry, Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Rachel F Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH) and Departments of Psychiatry, Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada,Departments of Psychiatry, Pharmacology and Toxicology, University of Toronto, Medical Sciences Building Room 4326, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada, Tel: 416 978 6374, Fax: 416 978 6395, E-mail:
| |
Collapse
|
29
|
Nicotine regulates the expression of UDP-glucuronosyltransferase (UGT) in humanized UGT1 mouse brain. Drug Metab Pharmacokinet 2015. [PMID: 26210671 DOI: 10.1016/j.dmpk.2015.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
UDP-glucuronosyltransferase (UGT) is a family of enzymes that catalyze the glucuronidation of various compounds, and thereby has an important role in metabolism and detoxification of a large number of xenobiotic and endogenous compounds. UGTs are present highly in the liver and small intestine, while several investigations on quantification of UGT mRNA reported that UGTs were also expressed in the brain. However, reported expression patterns of UGT isoforms in human brain were often incongruous with each other. In the present study, therefore, we investigated UGT mRNA expressions in brains of humanized UGT1 (hUGT1) mice. We found that among the human UGT1 members, UGT1A1, 1A3, and 1A6 were expressed in the brain. We further observed that nicotine (3 mg/kg) induced the expression of UGT1A3 mRNA in the brain, but not liver. While it was not statistically significant, the nicotine treatment resulted in an increase in the chenodeoxycholic acid glucuronide-formation activity in the brain microsomes. UGT1A3 is involved in metabolism of various antidepressants and non-steroidal antiinflammatory drugs, which exhibit their pharmacological effects in the brain. Therefore, nicotine-treated hUGT1 mice might be useful to investigate the role of brain UGT1A3 in the regulation of local levels of these drugs and their response.
Collapse
|
30
|
Agúndez JAG, Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E. Drug and xenobiotic biotransformation in the blood-brain barrier: a neglected issue. Front Cell Neurosci 2014; 8:335. [PMID: 25368552 PMCID: PMC4201098 DOI: 10.3389/fncel.2014.00335] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 09/30/2014] [Indexed: 11/13/2022] Open
Abstract
Drug biotransformation is a crucial mechanism for facilitating the elimination of chemicals from the organism and for decreasing their pharmacological activity. Published evidence suggests that brain drug metabolism may play a role in the development of adverse drug reactions and in the clinical response to drugs and xenobiotics. The blood-brain barrier (BBB) has been regarded mainly as a physical barrier for drugs and xenobiotics, and little attention has been paid to the BBB as a drug-metabolizing barrier. The presence of drug-metabolizing enzymes in the BBB is likely to have functional implications because local metabolism may inactivate drugs or may modify the drug's ability to cross the BBB, thus modifying drug response and the risk of developing adverse drug reactions. In this perspective paper, we discuss the expression of relevant xenobiotic metabolizing enzymes in the brain and in the BBB, and we cover current advances and future directions on the potential role of these BBB drug-metabolizing enzymes as modifiers of drug response.
Collapse
Affiliation(s)
- José A G Agúndez
- Department of Pharmacology, University of Extremadura Cáceres, Spain ; ISCIII Research Network of Adverse Reactions to Allergens and Drugs Madrid, Spain
| | | | | | - Elena García-Martín
- ISCIII Research Network of Adverse Reactions to Allergens and Drugs Madrid, Spain ; Department of Biochemistry, Molecular Biology and Genetics, University of Extremadura Cáceres, Spain
| |
Collapse
|
31
|
Valencia-Olvera AC, Morán J, Camacho-Carranza R, Prospéro-García O, Espinosa-Aguirre JJ. CYP2E1 induction leads to oxidative stress and cytotoxicity in glutathione-depleted cerebellar granule neurons. Toxicol In Vitro 2014; 28:1206-14. [PMID: 24929095 DOI: 10.1016/j.tiv.2014.05.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/25/2014] [Accepted: 05/30/2014] [Indexed: 11/27/2022]
Abstract
Increasing evidence suggests that brain cytochrome P450 (CYP) can contribute to the in situ metabolism of xenobiotics. In the liver, some xenobiotics can be metabolized by CYPs into more reactive products that can damage hepatocytes and induce cell death. In addition, normal CYP activity may produce reactive oxygen species (ROS) that contribute to cell damage through oxidative mechanisms. CYP2E1 is a CYP isoform that can generate ROS leading to cytotoxicity in multiple tissue types. The aim of this study was to determine whether CYP2E1 induction may lead to significant brain cell impairment. Immunological analysis revealed that exposure of primary cerebellar granule neuronal cultures to the CYP inducer isoniazid, increased CYP2E1 expression. In the presence of buthionine sulfoximine, an agent that reduces glutathione levels, isoniazid treatment also resulted in reactive oxygen species (ROS) production, DNA oxidation and cell death. These effects were attenuated by simultaneous exposure to diallyl sulfide, a CYP2E1 inhibitor, or to a mimetic of superoxide dismutase/catalase, (Euka). These results suggest that in cases of reduced antioxidant levels, the induction of brain CYP2E1 could represent a risk of in situ neuronal damage.
Collapse
Affiliation(s)
- Ana Carolina Valencia-Olvera
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, D.F., Mexico
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, D.F., Mexico
| | - Rafael Camacho-Carranza
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, D.F., Mexico
| | - Oscar Prospéro-García
- Grupo de Neurociencias, Laboratorio de Canabinoides, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, DF, Mexico
| | - Jesús Javier Espinosa-Aguirre
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, D.F., Mexico.
| |
Collapse
|
32
|
Miller RT, Miksys S, Hoffmann E, Tyndale RF. Ethanol self-administration and nicotine treatment increase brain levels of CYP2D in African green monkeys. Br J Pharmacol 2014; 171:3077-88. [PMID: 24611668 PMCID: PMC4055207 DOI: 10.1111/bph.12652] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/31/2014] [Accepted: 02/09/2014] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND AND PURPOSE CYP2D6 metabolizes many centrally acting drugs, neurotoxins and endogenous neurochemicals, and differences in brain levels of CYP2D have been associated with brain function and drug response. Alcohol consumers and smokers have higher levels of CYP2D6 in brain, but not liver, suggesting ethanol and/or nicotine may induce human brain CYP2D6. We investigated the independent and combined effects of chronic ethanol self-administration and nicotine treatment on CYP2D expression in African green monkeys. EXPERIMENTAL APPROACH Forty monkeys were randomized into control, ethanol-only, nicotine-only and ethanol + nicotine groups. Two groups voluntarily self-administered 10% ethanol in sucrose solution for 4 h·day(-1) , whereas two groups consumed sucrose solution on the same schedule. Two groups received daily s.c. injections of 0.5 mg·kg(-1) nicotine in saline bid, whereas two groups were injected with saline on the same schedule. KEY RESULTS Both nicotine and ethanol dose-dependently increased CYP2D in brain; brain mRNA was unaffected, and neither drug altered hepatic CYP2D protein or mRNA. The combination of ethanol and nicotine increased brain CYP2D protein levels to a greater extent than either drug alone (1.2-2.2-fold, P < 0.05 among the eight brain regions assessed). Immunohistochemistry revealed the induction of brain CYP2D protein within specific cell types and regions in the treatment groups. CONCLUSIONS AND IMPLICATIONS Ethanol and nicotine increase brain CYP2D protein levels in monkeys, in a region and treatment-specific manner, suggesting that CNS drug responses, neurodegeneration and personality may be affected among people who consume alcohol and/or nicotine.
Collapse
Affiliation(s)
- R T Miller
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | | | | | | |
Collapse
|
33
|
Khokhar JY, Tyndale RF. Intracerebroventricularly and Systemically Delivered Inhibitor of Brain CYP2B (C8-Xanthate), Even Following Chlorpyrifos Exposure, Reduces Chlorpyrifos Activation and Toxicity in Male Rats. Toxicol Sci 2014; 140:49-60. [DOI: 10.1093/toxsci/kfu075] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
34
|
Tripathi VK, Kumar V, Singh AK, Kashyap MP, Jahan S, Pandey A, Alam S, Khan F, Khanna VK, Yadav S, Lohani M, Pant AB. Monocrotophos induces the expression and activity of xenobiotic metabolizing enzymes in pre-sensitized cultured human brain cells. PLoS One 2014; 9:e91946. [PMID: 24663500 PMCID: PMC3963866 DOI: 10.1371/journal.pone.0091946] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 02/16/2014] [Indexed: 01/04/2023] Open
Abstract
The expression and metabolic profile of cytochrome P450s (CYPs) is largely missing in human brain due to non-availability of brain tissue. We attempted to address the issue by using human brain neuronal (SH-SY5Y) and glial (U373-MG) cells. The expression and activity of CYP1A1, 2B6 and 2E1 were carried out in the cells exposed to CYP inducers viz., 3-methylcholanthrene (3-MC), cyclophosphamide (CPA), ethanol and known neurotoxicant- monocrotophos (MCP), a widely used organophosphorous pesticide. Both the cells show significant induction in the expression and CYP-specific activity against classical inducers and MCP. The induction level of CYPs was comparatively lower in MCP exposed cells than cells exposed to classical inducers. Pre-exposure (12 h) of cells to classical inducers significantly added the MCP induced CYPs expression and activity. The findings were concurrent with protein ligand docking studies, which show a significant modulatory capacity of MCP by strong interaction with CYP regulators-CAR, PXR and AHR. Similarly, the known CYP inducers- 3-MC, CPA and ethanol have also shown significantly high docking scores with all the three studied CYP regulators. The expression of CYPs in neuronal and glial cells has suggested their possible association with the endogenous physiology of the brain. The findings also suggest the xenobiotic metabolizing capabilities of these cells against MCP, if received a pre-sensitization to trigger the xenobiotic metabolizing machinery. MCP induced CYP-specific activity in neuronal cells could help in explaining its effect on neurotransmission, as these CYPs are known to involve in the synthesis/transport of the neurotransmitters. The induction of CYPs in glial cells is also of significance as these cells are thought to be involved in protecting the neurons from environmental insults and safeguard them from toxicity. The data provide better understanding of the metabolizing capability of the human brain cells against xenobiotics.
Collapse
Affiliation(s)
- Vinay K. Tripathi
- In Vitro Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
- Department of Biotechnology, Integral University, Lucknow, India
| | - Vivek Kumar
- In Vitro Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
- Department of Biotechnology, Integral University, Lucknow, India
| | - Abhishek K. Singh
- In Vitro Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Mahendra P. Kashyap
- In Vitro Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Sadaf Jahan
- In Vitro Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Ankita Pandey
- In Vitro Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Sarfaraz Alam
- Metabolic and Structural Biology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Feroz Khan
- Metabolic and Structural Biology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Vinay K. Khanna
- In Vitro Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Sanjay Yadav
- In Vitro Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Mohtshim Lohani
- Department of Biotechnology, Integral University, Lucknow, India
| | - Aditya B. Pant
- In Vitro Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
- * E-mail:
| |
Collapse
|
35
|
Li L, Jia K, Zhou X, McCallum SE, Hough LB, Ding X. Impact of nicotine metabolism on nicotine's pharmacological effects and behavioral responses: insights from a Cyp2a(4/5)bgs-null mouse. J Pharmacol Exp Ther 2013; 347:746-54. [PMID: 24045421 DOI: 10.1124/jpet.113.208256] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nicotine metabolism is believed to affect not only nicotine's pharmacological effects but also nicotine addiction. As a key step toward testing this hypothesis, we have studied nicotine metabolism and nicotine's pharmacological and behavioral effects in a novel knockout mouse model [named Cyp2a(4/5)bgs-null] lacking a number of cytochrome P450 genes known to be or possibly involved in nicotine metabolism, including two Cyp2a and all Cyp2b genes. We found that, compared with wild-type mice, the Cyp2a(4/5)bgs-null mice showed >90% decreases in hepatic microsomal nicotine oxidase activity in vitro, and in rates of systemic nicotine clearance in vivo. Further comparisons of nicotine metabolism between Cyp2a(4/5)bgs-null and Cyp2a5-null mice revealed significant roles of both CYP2A5 and CYP2B enzymes in nicotine clearance. Compared with the behavioral responses in wild-type mice, the decreases in nicotine metabolism in the Cyp2a(4/5)bgs-null mice led to prolonged nicotine-induced acute pharmacological effects, in that null mice showed enhanced nicotine hypothermia and antinociception. Furthermore, we found that the Cyp2a(4/5)bgs-null mice developed a preference for nicotine in a conditioned place preference test, a commonly used test of nicotine's rewarding effects, at a nicotine dose that was 4-fold lower than what was required by wild-type mice. Thus, CYP2A/2B-catalyzed nicotine clearance affects nicotine's behavioral response as well as its acute pharmacological effects in mice. This result provides direct experimental support of the findings of pharmacogenetic studies that suggest linkage between rates of nicotine metabolism and smoking behavior in humans.
Collapse
Affiliation(s)
- Lei Li
- Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York at Albany (L.L., K.J., X.Z., X.D.) and Center for Neuropharmacology and Neuroscience, Albany Medical College (S.E.M., L.B.H.), Albany, New York
| | | | | | | | | | | |
Collapse
|
36
|
Ethanol self-administration and nicotine treatment induce brain levels of CYP2B6 and CYP2E1 in African green monkeys. Neuropharmacology 2013; 72:74-81. [DOI: 10.1016/j.neuropharm.2013.04.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/09/2013] [Accepted: 04/10/2013] [Indexed: 11/20/2022]
|
37
|
Abstract
Cytochrome P450 enzymes (CYPs) metabolize many drugs that act on the central nervous system (CNS), such as antidepressants and antipsychotics; drugs of abuse; endogenous neurochemicals, such as serotonin and dopamine; neurotoxins; and carcinogens. This takes place primarily in the liver, but metabolism can also occur in extrahepatic organs, including the brain. This is important for CNS-acting drugs, as variation in brain CYP-mediated metabolism may be a contributing factor when plasma levels do not predict drug response. This review summarizes the characterization of CYPs in the brain, using examples from the CYP2 subfamily, and discusses sources of variation in brain CYP levels and metabolism. Some recent experiments are described that demonstrate how changes in brain CYP metabolism can influence drug response, toxicity and drug-induced behaviours. Advancing knowledge of brain CYP-mediated metabolism may help us understand why patients respond differently to drugs used in psychiatry and predict risk for psychiatric disorders, including neurodegenerative diseases and substance abuse.
Collapse
Affiliation(s)
| | - Rachel F. Tyndale
- Correspondence to: R.F. Tyndale, Department of Pharmacology and Toxicology, 1 King’s College Circle, Toronto ON M5S 1A8;
| |
Collapse
|
38
|
Zhou K, Khokhar JY, Zhao B, Tyndale RF. First demonstration that brain CYP2D-mediated opiate metabolic activation alters analgesia in vivo. Biochem Pharmacol 2013; 85:1848-55. [PMID: 23623752 DOI: 10.1016/j.bcp.2013.04.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 04/13/2013] [Accepted: 04/16/2013] [Indexed: 12/13/2022]
Abstract
The response to centrally acting drugs is highly variable between individuals and does not always correlate with plasma drug levels. Drug-metabolizing CYP enzymes in the brain may contribute to this variability by affecting local drug and metabolite concentrations. CYP2D metabolizes codeine to the active morphine metabolite. We investigated the effect of inhibiting brain, and not liver, CYP2D activity on codeine-induced analgesia. Rats received intracerebroventricular injections of CYP2D inhibitors (20 μg propranolol or 40 μg propafenone) or vehicle controls. Compared to vehicle-pretreated rats, inhibitor-pretreated rats had: (a) lower analgesia in the tail-flick test (p<0.05) and lower areas under the analgesia-time curve (p<0.02) within the first hour after 30 mg/kg subcutaneous codeine, (b) lower morphine concentrations and morphine to codeine ratios in the brain (p<0.02 and p<0.05, respectively), but not in plasma (p>0.6 and p>0.7, respectively), tested at 30 min after 30 mg/kg subcutaneous codeine, and (c) lower morphine formation from codeine ex vivo by brain membranes (p<0.04), but not by liver microsomes (p>0.9). Analgesia trended toward a correlation with brain morphine concentrations (p=0.07) and correlated with brain morphine to codeine ratios (p<0.005), but not with plasma morphine concentrations (p>0.8) or plasma morphine to codeine ratios (p>0.8). Our findings suggest that brain CYP2D affects brain morphine levels after peripheral codeine administration, and may thereby alter codeine's therapeutic efficacy, side-effect profile and abuse liability. Brain CYPs are highly variable due to genetics, environmental factors and age, and may therefore contribute to interindividual variation in the response to centrally acting drugs.
Collapse
Affiliation(s)
- Kaidi Zhou
- Departments of Pharmacology & Toxicology and Psychiatry, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | | | | | | |
Collapse
|
39
|
Genetic variability of drug-metabolizing enzymes: the dual impact on psychiatric therapy and regulation of brain function. Mol Psychiatry 2013; 18:273-87. [PMID: 22565785 DOI: 10.1038/mp.2012.42] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Polymorphic drug-metabolizing enzymes (DMEs) are responsible for the metabolism of the majority of psychotropic drugs. By explaining a large portion of variability in individual drug metabolism, pharmacogenetics offers a diagnostic tool in the burgeoning era of personalized medicine. This review updates existing evidence on the influence of pharmacogenetic variants on drug exposure and discusses the rationale for genetic testing in the clinical context. Dose adjustments based on pharmacogenetic knowledge are the first step to translate pharmacogenetics into clinical practice. However, also clinical factors, such as the consequences on toxicity and therapeutic failure, must be considered to provide clinical recommendations and assess the cost-effectiveness of pharmacogenetic treatment strategies. DME polymorphisms are relevant not only for clinical pharmacology and practice but also for research in psychiatry and neuroscience. Several DMEs, above all the cytochrome P (CYP) enzymes, are expressed in the brain, where they may contribute to the local biochemical homeostasis. Of particular interest is the possibility of DMEs playing a physiological role through their action on endogenous substrates, which may underlie the reported associations between genetic polymorphisms and cognitive function, personality and vulnerability to mental disorders. Neuroimaging studies have recently presented evidence of an effect of the CYP2D6 polymorphism on basic brain function. This review summarizes evidence on the effect of DME polymorphisms on brain function that adds to the well-known effects of DME polymorphisms on pharmacokinetics in explaining the range of phenotypes that are relevant to psychiatric practice.
Collapse
|
40
|
The neuroprotective enzyme CYP2D6 increases in the brain with age and is lower in Parkinson's disease patients. Neurobiol Aging 2012; 33:2160-71. [DOI: 10.1016/j.neurobiolaging.2011.08.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 07/18/2011] [Accepted: 08/21/2011] [Indexed: 11/22/2022]
|
41
|
Khokhar JY, Tyndale RF. Rat Brain CYP2B-Enzymatic Activation of Chlorpyrifos to the Oxon Mediates Cholinergic Neurotoxicity. Toxicol Sci 2012; 126:325-35. [DOI: 10.1093/toxsci/kfs029] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
42
|
Ferguson CS, Tyndale RF. Cytochrome P450 enzymes in the brain: emerging evidence of biological significance. Trends Pharmacol Sci 2011; 32:708-14. [PMID: 21975165 DOI: 10.1016/j.tips.2011.08.005] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/25/2011] [Accepted: 08/29/2011] [Indexed: 10/17/2022]
Abstract
Cytochrome P450 (CYP) enzymes are responsible for the metabolism of many exogenous and endogenous compounds. CYPs are abundant in the liver and are also expressed in many extra-hepatic tissues including the brain. Although total CYP levels in the brain are much lower than in the liver, brain CYPs are concentrated near drug targets in specific regions and cell types, and can potentially have a considerable impact on local metabolism. Individual differences in brain CYP metabolism, due to inducers, inhibitors or genetic variation, can influence sensitivity and response to centrally acting drugs. Brain CYPs may also play a role in modulating brain activity, behavior, susceptibility to central nervous system diseases and treatment outcomes. This review highlights recent progress that has been made in understanding the functional significance of CYPs in the brain.
Collapse
Affiliation(s)
- Charmaine S Ferguson
- Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, Canada
| | | |
Collapse
|