1
|
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: 3] [Impact Index Per Article: 3.0] [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
|
2
|
Gautam M, Thapa G. Cytochrome P450-mediated estrogen catabolism therapeutic avenues in epilepsy. Acta Neurol Belg 2021; 121:603-612. [PMID: 32743748 DOI: 10.1007/s13760-020-01454-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 07/23/2020] [Indexed: 01/11/2023]
Abstract
Epilepsy is a neuropsychiatric disorder, which does not have any identifiable cause. However, experimental and clinical results have asserted that the sex hormone estrogen level and endocrine system function influence the seizure and epileptic episodes. There are available drugs to control epilepsy, which passes through the metabolism process. Cytochrome P-450 family 1 (CYP1A1) is a heme-containing mono-oxygenase that are induced several folds in most of the tissues and cells contributing to their differential expression, which regulates various metabolic processes upon administration of therapeutics. CYP1A1 gene family has been found to metabolize estrogen, a female sex hormone, which plays a central role in maintaining the health of brain altering the level of estrogen active neuropsychiatric disorder like epilepsy. Hence, in this article, we endeavor to provide an opinion of estrogen, its effects on epilepsy and catamenial epilepsy, their metabolism by CYP1A1 and new way forward to differential diagnosis and clinical management of epilepsy in future.
Collapse
Affiliation(s)
- Megha Gautam
- Department of Biological Science, Faculty of Science and Engineering, Health Research Institute, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Ganesh Thapa
- Department of Biological Science, Faculty of Science and Engineering, Health Research Institute, University of Limerick, Limerick, V94 T9PX, Ireland.
- Biohazards and Biosafety, Estates and Facilities, Trinity College of Dublin, The University of Dublin, College Green, Dublin 2, D02 PN40, Ireland.
| |
Collapse
|
3
|
Fanni D, Pinna F, Gerosa C, Paribello P, Carpiniello B, Faa G, Manchia M. Anatomical distribution and expression of CYP in humans: Neuropharmacological implications. Drug Dev Res 2021; 82:628-667. [PMID: 33533102 DOI: 10.1002/ddr.21778] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
The cytochrome P450 (CYP450) superfamily is responsible for the metabolism of most xenobiotics and pharmacological treatments generally used in clinical settings. Genetic factors as well as environmental determinants acting through fine epigenetic mechanisms modulate the expression of CYP over the lifespan (fetal vs. infancy vs. adult phases) and in diverse organs. In addition, pathological processes might alter the expression of CYP. In this selective review, we sought to summarize the evidence on the expression of CYP focusing on three specific aspects: (a) the anatomical distribution of the expression in body districts relevant in terms of drug pharmacokinetics (liver, gut, and kidney) and pharmacodynamics, focusing for the latter on the brain, since this is the target organ of psychopharmacological agents; (b) the patterns of expression during developmental phases; and (c) the expression of CYP450 enzymes during pathological processes such as cancer. We showed that CYP isoforms show distinct patterns of expression depending on the body district and the specific developmental phases. Of particular relevance for neuropsychopharmacology is the complex regulatory mechanisms that significantly modulate the complexity of the pharmacokinetic regulation, including the concentration of specific CYP isoforms in distinct areas of the brain, where they could greatly affect local substrate and metabolite concentrations of drugs.
Collapse
Affiliation(s)
- Daniela Fanni
- Unit of Anatomic Pathology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Anatomic Pathology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Federica Pinna
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Clara Gerosa
- Unit of Anatomic Pathology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Anatomic Pathology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Pasquale Paribello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Bernardo Carpiniello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Gavino Faa
- Unit of Anatomic Pathology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Anatomic Pathology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy.,Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
4
|
Han H, Mann A, Ekstein D, Eyal S. Breaking Bad: the Structure and Function of the Blood-Brain Barrier in Epilepsy. AAPS JOURNAL 2017; 19:973-988. [DOI: 10.1208/s12248-017-0096-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 04/28/2017] [Indexed: 12/27/2022]
|
5
|
Parmar D, Dhawan A, Dayal M, Seth PK. Immunochemical and Biochemical Evidence for Expression of Phenobarbital-and 3-Methylcholanthrene-Inducible Isoenzymes of Cytochrome P450 in Rat Brain. Int J Toxicol 2016. [DOI: 10.1080/109158198225883] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Expression of P450 1A1l 1A2 and 2 B1l 2B2 isoenzymes in rat brain was studied by Western blotting, using polyclonal antibodies raised against hepatic P450 1A1l 1A2 and 2B1l 2B2 isoenzymes. In addition, biochemical characterizations of the catalytic activities, pen toxyresorufin O-dealkylation (PROD) and ethoxyre-sorufin O-deethylation (EROD), selective for P450 2B1l 2B2 (PROD) and P450 1A1l 1A2 (EROD), were performed with rat brain microsomes. Control rat brain microsomes did not crossreact with either of the antibodies, whereas microsomes obtained from 3-methylcholanthrene (MC)-pretreated rats revealed significant immunoreactivity with anti-P450 1A1l 1A2. Similar results were observed with phenobarbital (PB)-pretreated rats, with the brain microsomes exhibiting significant immunoreactivity with anti-P450 2B1l 2B2. The induction in the P450 isoenzymes after PB or MC pretreatment was much less in the brain in comparison to the liver. Enzymatic studies indicated that the activities of PROD and EROD were induced in brain 3—4 fold by PB and MC pretreatment, respectively, and were almost completely inhibited on in vitro addition of anti-P450 2B1l 2B2 and 1A1l 1A2. These data demonstrate the expression of P4501A1l 1A2 and 2B1l 2B2 isoenzymes in the brain and indicate that, as in liver, these isoenzymes catalyze EROD and PROD, respectively, in the rat brain.
Collapse
Affiliation(s)
- Devendra Parmar
- Developmental Toxicology Section, Industrial Toxicology Research Centre, Lucknow, India
| | - Alok Dhawan
- Developmental Toxicology Section, Industrial Toxicology Research Centre, Lucknow, India
| | - Monika Dayal
- Developmental Toxicology Section, Industrial Toxicology Research Centre, Lucknow, India
| | - Prahlad K. Seth
- Developmental Toxicology Section, Industrial Toxicology Research Centre, Lucknow, India
| |
Collapse
|
6
|
Stamou M, Wu X, Kania-Korwel I, Lehmler HJ, Lein PJ. Cytochrome p450 mRNA expression in the rodent brain: species-, sex-, and region-dependent differences. Drug Metab Dispos 2014; 42:239-44. [PMID: 24255117 PMCID: PMC3912540 DOI: 10.1124/dmd.113.054239] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 11/19/2013] [Indexed: 01/22/2023] Open
Abstract
Cytochrome P450 (P450) enzymes play a critical role in the activation and detoxication of many neurotoxic chemicals. Although research has largely focused on P450-mediated metabolism in the liver, emerging evidence suggests that brain P450s influence neurotoxicity by modulating local metabolite levels. As a first step toward better understanding the relative role of brain P450s in determining neurotoxic outcome, we characterized mRNA expression of specific P450 isoforms in the rodent brain. Adult mice (male and female) and rats (male) were treated with vehicle, phenobarbital, or dexamethasone. Transcripts for CYP2B, CYP3A, CYP1A2, and the orphan CYP4X1 and CYP2S1 were quantified in the liver, hippocampus, cortex, and cerebellum by quantitative (real-time) polymerase chain reaction. These P450s were all detected in the liver with the exception of CYP4X1, which was detected in rat but not mouse liver. P450 expression profiles in the brain varied regionally. With the exception of the hippocampus, there were no sex differences in regional brain P450 expression profiles in mice; however, there were marked species differences. In the liver, phenobarbital induced CYP2B expression in both species. Dexamethasone induced hepatic CYP2B and CYP3A in mice but not rats. In contrast, brain P450s did not respond to these classic hepatic P450 inducers. Our findings demonstrate that P450 mRNA expression in the brain varies by region, regional brain P450 profiles vary between species, and their induction varies from that of hepatic P450s. These novel data will be useful for designing mechanistic studies to examine the relative role of P450-mediated brain metabolism in neurotoxicity.
Collapse
Affiliation(s)
- Marianna Stamou
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California (M.S., P.J.L.) and Department of Occupational and Environmental Health, College of Public Health, The University of Iowa, Iowa City, Iowa (X.W., I.K.-K., H.-J.L.)
| | | | | | | | | |
Collapse
|
7
|
Wu X, Kania-Korwel I, Chen H, Stamou M, Dammanahalli KJ, Duffel M, Lein PJ, Lehmler HJ. Metabolism of 2,2',3,3',6,6'-hexachlorobiphenyl (PCB 136) atropisomers in tissue slices from phenobarbital or dexamethasone-induced rats is sex-dependent. Xenobiotica 2013; 43:933-47. [PMID: 23581876 DOI: 10.3109/00498254.2013.785626] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
1. Chiral polychlorinated biphenyls (PCBs) such as PCB 136 enantioselectively sensitize the ryanodine receptor (RyR). In light of recent evidence that PCBs cause developmental neurotoxicity via RyR-dependent mechanisms, this suggests that enantioselective PCB metabolism may influence the developmental neurotoxicity of chiral PCBs. However, enantioselective disposition of PCBs has not been fully characterized. 2. The effect of sex and cytochrome P450 (P450) enzyme induction on the enantioselective metabolism of PCB 136 was studied using liver tissue slices prepared from naïve control (CTL), phenobarbital (PB; CYP2B inducer) or dexamethasone (DEX; CYP3A inducer) pretreated adult Sprague-Dawley rats. PCB 136 metabolism was also examined in hippocampal slices derived from untreated rat pups. 3. In liver tissue slices, hydroxylated PCB (OH-PCB) profiles depended on sex and inducer pretreatment, and OH-PCB levels followed the rank orders male > female and PB > DEX > CTL. In contrast, the enantiomeric enrichment of PCB 136 and its metabolites was independent of sex and inducer pretreatment. Only small amounts of PCB 136 partitioned into hippocampal tissue slices and no OH-PCB metabolites were detected. 4. Our results suggest that enantioselective metabolism, sex and induction status of P450 enzymes in the liver may modulate the neurotoxic outcomes of developmental exposure to chiral PCBs.
Collapse
Affiliation(s)
- Xianai Wu
- Department of Occupational and Environmental Health, College of Public Health, The University of Iowa , Iowa City, IA , USA
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Armagan G, Bojnik E, Turunc E, Kanit L, Gündüz Çinar O, Benyhe S, Borsodi A, Yalcin A. Kainic acid-induced changes in the opioid/nociceptin system and the stress/toxicity pathways in the rat hippocampus. Neurochem Int 2012; 60:555-64. [PMID: 22382076 DOI: 10.1016/j.neuint.2012.02.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 02/10/2012] [Accepted: 02/11/2012] [Indexed: 12/13/2022]
Abstract
Excitotoxicity is a contributing factor to the pathogenesis of acute or chronic neurodegenerative disease states. Kainic acid (KA) is an excitotoxic substance and the administration of it to rodents induces seizure activity (status epilepticus, SE) and leads to neurodegeneration. In this study the effect of KA-induced excitotoxicity on the G-protein activations and the gene expression levels of the opioid/nociceptin system receptors as MOPr, KOPr, DOPr, ORL-1, and PNOC (N/OFQ) were investigated, and the regulator effect of naloxone (Nal) on the gene expressions of the opioid system receptors against KA-induced seizures in the rat hippocampus was tested. In addition, the expression levels of stress-toxicity genes were assessed in the hippocampus following KA-induced excitotoxicity in order to determine the potential genetic targets which can be helpful for neuroprotective interventions. Our results indicate that the KA-induced excitotoxicity increased the mRNA levels of MOPr, DOPr, KOPr, PNOC, and ORL-1. However, G-protein activations of MOPr, DOPr, and KOPr remained relatively unchanged while both the potency and efficacy of N/OFQ were significantly increased. The PCR array data showed that KA-induced excitotoxicity altered the expression levels of genes in the cellular stress or toxicity pathways. Our data suggests that the induction of the opioid/nociceptin system may be involved in the cellular stress response following a neurodegenerative insult and that the genes modulated by the KA-treatment in the stress-toxicity pathways may be evaluated as targets of potential neuroprotective interventions.
Collapse
Affiliation(s)
- Guliz Armagan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, 35100 Bornova, Izmir, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Ghosh C, Gonzalez-Martinez J, Hossain M, Cucullo L, Fazio V, Janigro D, Marchi N. Pattern of P450 expression at the human blood-brain barrier: roles of epileptic condition and laminar flow. Epilepsia 2010; 51:1408-17. [PMID: 20074231 PMCID: PMC3386640 DOI: 10.1111/j.1528-1167.2009.02428.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE P450 enzymes (CYPs) play a major role in hepatic drug metabolism. It is unclear whether these enzymes are functionally expressed by the diseased human blood-brain barrier (BBB) and are involved in local drug metabolism or response. We have evaluated the cerebrovascular CYP expression and function, hypothesizing possible implication in drug-resistant epilepsy. METHODS CYP P450 transcript levels were assessed by cDNA microarray in primary endothelial cultures established from a cohort of brain resections (n = 12, drug-resistant epilepsy EPI-EC and aneurism domes ANE-EC). A human brain endothelial cell line (HBMEC) and non-brain endothelial cell line (HUVEC) were used as controls. The effect of exposure to shear stress on CYP expression was evaluated. Results were confirmed by Western blot and immunohistochemistry on brain specimens. Endothelial drug metabolism was assessed by high performance liquid chromatography (HPLC-UV). RESULTS cDNA microarray showed the presence of CYP enzymes in isolated human primary brain endothelial cells. Using EPI-EC and HBMEC we found that CYP mRNA levels were significantly affected by exposure to shear stress. CYP3A4 protein was overexpressed in EPI-EC (290 ± 30%) compared to HBMEC and further upregulated by shear stress exposure. CYP3A4 was increased in the vascular compartment at regions of reactive gliosis in the drug-resistant epileptic brain. Metabolism of carbamazepine was significantly elevated in EPI-EC compared to HBMEC. DISCUSSION These results support the hypothesis of local drug metabolism at the diseased BBB. The direct association between BBB CYP enzymes and the drug-resistant phenotype needs to be further investigated.
Collapse
Affiliation(s)
- Chaitali Ghosh
- Department of Cerebrovascular Research, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Neurological Surgery, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Cell Biology, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
| | | | - Mohammed Hossain
- Department of Cerebrovascular Research, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Neurological Surgery, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Cell Biology, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
| | - Luca Cucullo
- Department of Cerebrovascular Research, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Neurological Surgery, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Cell Biology, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
| | - Vincent Fazio
- Department of Cerebrovascular Research, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Neurological Surgery, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Cell Biology, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
| | - Damir Janigro
- Department of Cerebrovascular Research, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Neurological Surgery, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Cell Biology, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Molecular Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
| | - Nicola Marchi
- Department of Cerebrovascular Research, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Neurological Surgery, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
- Department of Cell Biology, Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A
| |
Collapse
|
10
|
Meyer RP, Gehlhaus M. A role for CYP in the drug–hormone crosstalk of the brain. Expert Opin Drug Metab Toxicol 2010; 6:675-87. [DOI: 10.1517/17425251003680791] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Kapoor N, Pant AB, Dhawan A, Dwievedi UN, Seth PK, Parmar D. Differences in the expression and inducibility of cytochrome P450 2B isoenzymes in cultured rat brain neuronal and glial cells. Mol Cell Biochem 2007; 305:199-207. [PMID: 17646928 DOI: 10.1007/s11010-007-9544-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Accepted: 06/21/2007] [Indexed: 10/23/2022]
Abstract
Studies initiated to investigate the distribution of cytochrome P450 2B (CYP2B) isoenzymes in rat brain cells revealed significant activity of CYP2B-dependent 7-pentoxyresorufin-O-dealkylase (PROD) in microsomes prepared from both, cultured rat brain neuronal and glial cells. Neuronal cells exhibited 2-fold higher activity of PROD than the glial cells. RT-PCR and immunocytochemical studies demonstrated significant constitutive mRNA and protein expression of CYP2B in cultured neuronal and glial cells. Induction studies with phenobarbital (PB), a known CYP2B inducer, revealed significant concentration dependent increase in the activity of PROD in cultured brain cells with glial cells exhibiting greater magnitude of induction than the neuronal cells. This difference in the increase in enzyme activity was also observed with RT-PCR and immunocytochemical studies indicating differences in the induction of CYP2B1 and 2B2 mRNA as well as protein expression in the cultured brain cells. Furthermore, a greater magnitude of induction was observed in CYP2B2 than CYP2B1 in the brain cells. Our data indicating differences in the expression and sensitivity of the CYP2B isoenzymes in cultured rat brain cells will help in identifying and distinguishing xenobiotic metabolizing capability of these cells and understanding the vulnerability of the specific cell types toward neurotoxins.
Collapse
Affiliation(s)
- Nidhi Kapoor
- Biochemistry Department, Lucknow University, University Road, Lucknow, India
| | | | | | | | | | | |
Collapse
|
12
|
Meyer RP, Hagemeyer CE, Knoth R, Kaufmann MR, Volk B. Anti-epileptic drug phenytoin enhances androgen metabolism and androgen receptor expression in murine hippocampus. J Neurochem 2006; 96:460-72. [PMID: 16336225 DOI: 10.1111/j.1471-4159.2005.03555.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Epilepsy is very often related to strong impairment of neuronal networks, particularly in the hippocampus. Previous studies of brain tissue have demonstrated that long-term administration of the anti-epileptic drug (AED) phenytoin leads to enhanced metabolism of testosterone mediated by cytochrome P450 (CYP) isoforms. Thus, we speculate that AEDs affect androgen signalling in the hippocampus. In the present study, we investigated how the AED phenytoin influences the levels of testosterone, 17beta-oestradiol, and androgen receptor (AR) in the hippocampus of male C57Bl/6J mice. Phenytoin administration led to a 61.24% decreased hippocampal testosterone level as compared with controls, while serum levels were slightly enhanced. 17beta-Oestradiol serum level was elevated 2.6-fold. Concomitantly, the testosterone metabolizing CYP isoforms CYP3A11 and CYP19 (aromatase) have been found to be induced 2.4- and 4.2-fold, respectively. CYP3A-mediated depletion of testosterone-forming 2beta-, and 6beta-hydroxytestosterone was significantly enhanced. Additionally, AR expression was increased 2-fold (mRNA) and 1.8-fold (protein), predominantly in the CA1 region. AR was shown to concentrate in nuclei of CA1 pyramidal neurons. We conclude that phenytoin affects testosterone metabolism via induction of CYP isoforms. The increased metabolism of testosterone leading to augmented androgen metabolite formation most likely led to enhanced expression of CYP19 and AR in hippocampus. Phenytoin obviously modulates the androgen signalling in the hippocampus.
Collapse
Affiliation(s)
- Ralf Peter Meyer
- Pathologisches Institut, Abt. Neuropathologie, Neurozentrum, Universitätsklinik Freiburg, Freiburg, Germany.
| | | | | | | | | |
Collapse
|
13
|
Zobel A, Wellmer J, Schulze-Rauschenbach S, Pfeiffer U, Schnell S, Elger C, Maier W. Impairment of inhibitory control of the hypothalamic pituitary adrenocortical system in epilepsy. Eur Arch Psychiatry Clin Neurosci 2004; 254:303-11. [PMID: 15365705 DOI: 10.1007/s00406-004-0499-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2003] [Accepted: 02/20/2004] [Indexed: 10/26/2022]
Abstract
Excess comorbidity between depression and epilepsy proposes common pathophysiological patterns in both disorders. Neuroendocrine abnormalities were often observed in depression as well as in epilepsy. Lack of inhibitory control of the hypothalamic pituitary adrenocortical (HPA) system is a core feature of depression; main relay stations of this system are located in the amygdala and hippocampus, which are key regions for both disorders. Therefore we explored the feedback mechanism of the HPA system in epilepsy. In order to control for the impact of depression we focused on epilepsies without depression. We compared patients with epilepsy (subdivided by medication with or without hepatic enzyme inducing antiepileptic medication) with 16 healthy controls and 16 patients with unipolar major depression but without epilepsy. We observed a lack of inhibitory control of the HPA system in patients with epilepsy, also in the absence of enzyme inducing medication. An impact of the temporal lobe location of the epileptic focus could not be observed. Thus, epilepsies share with depression the deficiencies in the feedback mechanism of the HPA system, proposing common pathophysiological features of up to now unknown nature.
Collapse
Affiliation(s)
- Astrid Zobel
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
14
|
Mulero-Navarro S, Santiago-Josefat B, Pozo-Guisado E, Merino JM, Fernandez-Salguero PM. Down-regulation of CYP1A2 induction during the maturation of mouse cerebellar granule cells in culture: role of nitric oxide accumulation. Eur J Neurosci 2003; 18:2265-72. [PMID: 14622187 DOI: 10.1046/j.1460-9568.2003.02972.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Nitric oxide (NO) is responsible for cytochrome P450 (CYP450) loss during isolation and cytokine treatment of primary rat hepatocytes. As P450s mediate the metabolism of toxic chemicals, their inhibition could compromise the cells competence to eliminate toxins, a condition potentially relevant in neurological diseases involving constitutive activation of nitric oxide synthase (NOS) and NO over-production. Here, we have investigated the correlation between NO accumulation and CYP1A2 down-regulation during maturation of mouse cerebellar granule cells (CGC). As neurons matured in culture, the inducible levels of CYP1A2 protein and catalytic activity decreased to almost undetectable values. In parallel, a significant increase in NO concentration was observed. Neuronal NOS remained constitutively active during maturation, thus contributing to NO accumulation. The NOS inhibitor l-NAME, restored CYP1A2 catalytic activity up to 9 days in vitro, supporting a role for NO in the inhibition process. Maturation was also followed by increased NMDA receptor activity and intracellular Ca2+ concentration. We suggest that maintained NOS activity during CGC maturation could lead to NO accumulation and to decreased CYP1A2 inducibility. Increased NMDA receptor activity and Ca2+ entry could contribute to this process. Thus, neurodegeneration could diminish the induction of specific P450s and impair the metabolism of foreign and/or endogenous chemicals in the CNS.
Collapse
Affiliation(s)
- Sonia Mulero-Navarro
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, 06071-Badajoz, Spain
| | | | | | | | | |
Collapse
|
15
|
Parmar D, Yadav S, Dayal M, Johri A, Dhawan A, Seth PK. Effect of lindane on hepatic and brain cytochrome P450s and influence of P450 modulation in lindane induced neurotoxicity. Food Chem Toxicol 2003; 41:1077-87. [PMID: 12842176 DOI: 10.1016/s0278-6915(03)00045-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Oral administration of lindane (2.5, 5, 10 and 15 mg/kg, body weight) for 5 days was found to produce a dose-dependent increase in the activity of P450 dependent 7-ethoxyresorufin-O-deethylase (EROD), 7-pentoxyresorufin-O-dealkylase (PROD) and N-nitrosodimethylamine demethylase (NDMA-d) in rat brain and liver. A significant increase in the hepatic and brain P450 monooxygenases was also observed when the duration of exposure of low dose (2.5 mg/kg) of lindane was increased from 5 days to 15 or 21 days. As observed with different doses, the magnitude of induction in the activity of P450 monooxygenases was several fold higher in liver microsomes when compared with the brain. Western blotting studies have indicated that the increase in the P450 enzymes could be due to the increase in the expression of P450 1A1/1A2, 2B1/2B2 and 2E1 isoenzymes. In vitro studies using organic inhibitors specific for individual P450 isoenzymes and antibody inhibition experiments have further demonstrated that the increase in the activity of PROD, EROD and NDMA-d are due to the increase in the levels of P450 2B1/2B2, 1A1/1A2 and 2E1 isoenzymes, respectively. Induction studies have further shown that while pretreatment of 3-methylcholanthrene (MC), an inducer of P4501A1/1A2, did not produce any significant effect in the incidence of lindane induced convulsions, pretreatment with phenobarbital (PB), an inducer of P450 2B1/2B2 or ethanol, an inducer of P450 2E1 catalysed reactions, significantly increased the incidence of lindane induced convulsions. Similarly, when the P450-mediated metabolism of lindane was blocked by cobalt chloride incidence of convulsions was increased in animals treated with lindane indicating that lindane per se or its metabolites formed by PB or ethanol inducible P450 isoenzymes are involved in its neurobehavioral toxicity.
Collapse
Affiliation(s)
- D Parmar
- Developmental Toxicology Division, Industrial Toxicology Research Centre, PO Box 80, M.G. Marg, -226 001, U.P, Lucknow, India.
| | | | | | | | | | | |
Collapse
|
16
|
Meyer RP, Podvinec M, Meyer UA. Cytochrome P450 CYP1A1 accumulates in the cytosol of kidney and brain and is activated by heme. Mol Pharmacol 2002; 62:1061-7. [PMID: 12391268 DOI: 10.1124/mol.62.5.1061] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cytochrome P450 CYP1A1 is expressed in most tissues. In brain and kidney, its function remains unclear because its enzymatic activity is barely measurable. Here, we report on the localization of CYP1A1 in the cytosol of kidney and brain, as revealed by immunoblotting with anti-CYP1A1 antibodies and by 7-ethoxyresorufin deethylation (EROD). Hematin (8 microM) added in vitro to cytosol increased the EROD-activity 10-fold in brain olfactory bulb and 7-fold in kidney, presumably by reconstitution of apocytochrome. Succinylacetone, an inhibitor of heme biosynthesis, increased the ratio of cytosolic to microsomal EROD activity of transiently expressed CYP1A1 in COS-1 cells from 1:1 to nearly 6:1. This indicates a strong decrease of microsomal activity with increasing succinylacetone concentration. CYP1A1 activities correlated with CYP1A1 protein assessed by immunoblotting. We conclude that the availability of heme is a limiting factor of P450 function in extrahepatic tissue. Our data further suggest that reduced availability of heme limits the incorporation of P450s into brain endoplasmic reticulum. These observations are important when assessing the function of P450s in extrahepatic tissue.
Collapse
Affiliation(s)
- Ralf Peter Meyer
- Division of Pharmacology/Neurobiology, Biozentrum of the University of Basel, Basel, Switzerland.
| | | | | |
Collapse
|
17
|
Seegers U, Potschka H, Löscher W. Lack of effects of prolonged treatment with phenobarbital or phenytoin on the expression of P-glycoprotein in various rat brain regions. Eur J Pharmacol 2002; 451:149-55. [PMID: 12231384 DOI: 10.1016/s0014-2999(02)02235-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
P-glycoprotein is an ATP-dependent drug transport protein that is predominantly found in the apical membranes of various epithelial cell types in the body, including the blood luminar membrane of the brain capillary endothelial cells that make up the blood-brain barrier. Increased P-glycoprotein expression in the blood-brain barrier has been described in epileptogenic brain tissue of patients with pharmacoresistant epilepsy, suggesting that overexpression of P-glycoprotein may be involved in multidrug resistance of epilepsy. The mechanisms underlying the overexpression of P-glycoprotein in brain tissue of epileptic patients are not clear. Two antiepileptic drugs, phenobarbital and phenytoin, have been reported to up-regulate P-glycoprotein in cell cultures, so that chronic treatment with antiepileptic drugs may enhance P-glycoprotein expression in the blood-brain barrier. To directly address this possibility, we treated rats with phenobarbital or phenytoin over 11 days and subsequently determined expression of P-glycoprotein by immunohistochemistry in endothelium and parenchyma of several brain regions, including regions of the temporal lobe, which is often involved in pharmacoresistant types of epilepsy. Except for a moderate increase in the intensity of P-glycoprotein expression in the piriform/parietal cortex and cerebellum of phenobarbital-treated rats, no significant P-glycoprotein increases were seen after prolonged treatment with phenobarbital or phenytoin in any brain region examined. In view of recent findings that seizures lead to a transient induction of P-glycoprotein in the brain of rats, it seems reasonable to suggest that the overexpression of P-glycoprotein in brain regions of patients with intractable epilepsy is a consequence of uncontrolled seizures rather than of chronic treatment with antiepileptic drugs.
Collapse
Affiliation(s)
- Ulrike Seegers
- Department of Pharmacology, Toxicology and Pharmacy, School of Veterinary Medicine, Bünteweg 17 Building 218, D-30559 Hannover, Germany
| | | | | |
Collapse
|
18
|
Abstract
During infection or inflammation, the expression of cytochrome P450 and its dependent biotransformation pathways are modified. This results in a change in the capacity of the liver to handle drugs and in alterations in the production and elimination of endogenous substances throughout the body. The majority of the CYP isoforms are modified at pre-translational steps in protein synthesis, and, in most cases, cytokines are involved as mediators of the response. Recent information suggests that inflammatory responses that are localized to the CNS cause a loss of CYP within the brain. This is accompanied by a parallel down-regulation of CYP in peripheral organs that is mediated by a signaling pathway between the brain and periphery. This review covers the loss that occurs in the major mammalian CYP families in response to infection/inflammation and the mediator pathways that are key to this response.
Collapse
Affiliation(s)
- K W Renton
- Department of Pharmacology, Sir Charles Tupper Medical Building, Dalhousie University Halifax, Nova Scotia, Canada B3H 4H7.
| |
Collapse
|
19
|
Meyer RP, Hagemeyer CE, Knoth R, Kurz G, Volk B. Oxidative hydrolysis of scoparone by cytochrome p450 CYP2C29 reveals a novel metabolite. Biochem Biophys Res Commun 2001; 285:32-9. [PMID: 11437368 DOI: 10.1006/bbrc.2001.5111] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Regioselective 7-demethylation of scoparone is regularly employed as an indicator of phenobarbital-like induction of rat liver cytochrome P450 isoform CYP2B1, e.g., by the antiepileptic drug phenytoin. After induction with phenobarbital and phenytoin, a new reaction sequence catalyzed by Cyp2c29 was identified in mouse liver microsomes. Cyp2c29-dependent 6-demethylation of scoparone resulted in the formation of isoscopoletin, an intermediate which is susceptible to further oxidation. This subsequent oxidation was also catalyzed by Cyp2c29 with a K(m) of 30,31 microM and a V(max) of 3,41 microM/min x microM P450, and resulted in the formation of the new metabolite 3-[4-methoxy-p-(3,6)-benzoquinone]-2-propenoate. This novel metabolite is the product of two consecutive oxidation reactions, proceeding over isoscopoletin to a putative lactone which is accessible to immediate hydrolysis, due to the onium character of the ring oxygen. This opening of the lactone ring corresponds to an oxidative hydrolysis. Differential oxidation of scoparone can be used as a sensitive indicator for distinguishing between different cytochrome P450 isoforms.
Collapse
Affiliation(s)
- R P Meyer
- Department of Neuropathology, University of Freiburg, Freiburg, D-79106, Germany.
| | | | | | | | | |
Collapse
|
20
|
Diwan BA, Henneman JR, Nims RW. Enhancement of N-Nitrosodiethylamine-lnitiated Hepatocarcinogenesis by Phenytoin in Male F344/NCr Rats at a Dose Causing Maximal Induction of CYP2B. Int J Toxicol 2001. [DOI: 10.1177/109158180102000205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The effect of the clinically important anticonvulsant phenytoin (DPH) on hepatocarcinogenesis of male F344/NCr rats initiated with a single i.p. dose of N-nitrosodiethylamine (75 mg/kg b.w.) was studied. Beginning 2 weeks post-initiation, the rats received control diet or diet containing 500 or 1500 ppm DPH or 500 ppm phenobarbital. At 52 weeks age, the incidences (and multiplicities, in units of tumors per tumor-bearing rat) of hepatocellular adenomas were 0%, 17% (1 ± 0), 42% (1.8 ± 0.8), or 67% (2.5 ± 1.9) in rats exposed to N-nitrosodiethylamine alone, or the carcinogen followed by 500 ppm DPH, 1500 ppm DPH, or 500 ppm phenobarbital, respectively. Between 53 and 79 weeks of age, 39% of rats receiving N-nitrosodiethylamine alone developed multiple (1.5 ± 0.8) hepatocellular adenomas. A similar incidence (41%) occurred in the rats administered the carcinogen followed by 500 ppm DPH. The incidence of hepatocellular adenomas (88% and 89%) was significantly greater in rats exposed to N-nitrosodiethylamine followed by 1500 ppm DPH or 500 ppm phenobarbital, respectively. Multiplicities of hepatocellular adenomas were significantly greater than the control value in rats fed 1500 ppm DPH or 500 ppm phenobarbital (5.9 ± 4.8 and 10.1 ± 6.7, respectively), but not in the rats receiving 500 ppm DPH (2.3 ± 1.6). No rats exposed to N-nitrosodiethylamine alone or the carcinogen followed by 500 ppm DPH developed hepatocellular carcinomas, while hepatocellular carcinomas occurred in 29% or 67% of the rats given 1500 ppm DPH or 500 ppm phenobarbital, respectively, following initiation. Increases in hepatic CYP2B-mediated benzyloxyresorufin O-dealkylation activity in rats exposed to 500 and 1500 ppm DPH for 2 or 23 weeks were ∼50% and ∼100%, respectively, of the maximal induction caused by 500 ppm phenobarbital. Thus, in the rat model, DPH enhanced N-nitrosodiethyl-amine-initiated hepatocarcinogenesis when administered at a dose causing maximal CYP2B induction.
Collapse
Affiliation(s)
- Bhalchandra A. Diwan
- Intramural Research Support Program, SAIC Frederick, NCI–Frederick Cancer Research and Development Center, Frederick, Maryland, USA
| | - John R. Henneman
- Intramural Research Support Program, SAIC Frederick, NCI–Frederick Cancer Research and Development Center, Frederick, Maryland, USA
| | - Raymond W. Nims
- Laboratory of Comparative Carcinogenesis, National Cancer Institute, Frederick Cancer Research and Development Center, Frederick, Maryland, USA
| |
Collapse
|
21
|
Meyer RP, Knoth R, Schiltz E, Volk B. Possible function of astrocyte cytochrome P450 in control of xenobiotic phenytoin in the brain: in vitro studies on murine astrocyte primary cultures. Exp Neurol 2001; 167:376-84. [PMID: 11161626 DOI: 10.1006/exnr.2000.7553] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
[4-(14)C]Phenytoin underwent a rapid cellular uptake by diffusion within 5 min when applied in a concentration of 10 microM to mouse brain astrocyte cultures. Subsequently, a slow linear increase of intracellular radioactivity indicated metabolic trapping of the drug, with final concentrations reaching 144 pmol phenytoin/mg protein in the astrocytes. Phenytoin levels from 1 to 10 microM decreased cell viability by 15%. The action of cytochrome P450 present in astrocytes in concentrations of 16-17 pmol P450/mg protein could explain these slight cytotoxic effects by generating intermediate metabolites of phenytoin. In contrast, concentrations of 50 microM strongly inhibited cell proliferation. A Cyp2c29 immunorelated P450 isoform was expressed in nearly all astrocytes in culture. Intracellular [4-(14)C]phenytoin was degraded to its major metabolites dihydrodiol, p-HPPH, and m-HPPH through a P450-dependent reaction with a specific activity of 0.66 pmol/min x mg protein, or 0.12 pmol/min x mg protein as measured in cell homogenates. These data underscore the importance of astrocytes as brain cells active in the detoxification of foreign substrates, but also in their toxification due to reactive metabolites generated during these metabolic processes. After diffusionary influx of drugs and other xenobiotics, the astrocyte P450 monooxygenases perform an essential role in the mediation of toxicity most frequently encountered in highly vulnerable neurons.
Collapse
Affiliation(s)
- R P Meyer
- Department of Neuropathology, Neurozentrum, University of Freiburg, Breisacher Strasse 64, D-79106 Freiburg, Germany.
| | | | | | | |
Collapse
|
22
|
Miksys S, Hoffmann E, Tyndale RF. Regional and cellular induction of nicotine-metabolizing CYP2B1 in rat brain by chronic nicotine treatment. Biochem Pharmacol 2000; 59:1501-11. [PMID: 10799646 DOI: 10.1016/s0006-2952(00)00281-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
In the rat, nicotine is metabolized to cotinine primarily by hepatic cytochrome P450 (CYP) 2B1. This enzyme is also found in other organs such as the lung and the brain. Hepatic nicotine metabolism is unaltered after nicotine exposure; however, nicotine may regulate CYP2B1 in other tissues. We hypothesized that nicotine induces its own metabolism in brain by increasing CYP2B1. Male rats were treated with nicotine (0.0, 0.1, 0.3, or 1.0 mg base/kg in saline) s.c. daily for 7 days. CYP2B1 mRNA and protein were assayed in the brain and liver by reverse transcriptase-polymerase chain reaction (RT-PCR), immunoblotting, and immunocytochemistry. In control rats, CYP2B1 mRNA and protein expression were brain region- and cell-specific. CYP2B1 was not induced in the liver, but CYP2B1 mRNA and protein showed dose-dependent, region- and cell-specific patterns of induction across brain regions. At 1.0 mg nicotine/kg, the largest increase in protein was in the brain stem (5.8-fold, P < 0.05) with a corresponding increase in CYP2B1 mRNA (7.6-fold, P < 0.05). Induction of CYP2B1 was also observed in the frontal cortex, striatum, and olfactory tubercle. Immunocytochemistry showed that induction was restricted principally to neurons. These data indicate that nicotine may alter its own metabolism in the brain through transcriptional regulation, perhaps contributing to central tolerance to the effects of nicotine. CYP2B1 and its human homologue CYP2B6 also activate tobacco smoke procarcinogens such as NNK [4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone]. Highly localized increases in CYP2B could result in increased mutagenesis. These data suggest roles for nicotine-induced CYP2B in central metabolic tolerance, nicotine-induced neurotoxicity, neuroplasticity, and carcinogenesis.
Collapse
Affiliation(s)
- S Miksys
- Centre for Addiction and Mental Health and Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
23
|
Abstract
When host defence mechanisms are stimulated there is a concomitant decrease in cytochrome P450 based drug biotransformation and elimination. This has resulted in a number of clinically important unwanted drug responses in patients with infections or inflammatory responses. The loss in cytochrome P450 is predominantly an effect at the level of the gene expression and the majority of enzyme forms examined to date are involved. Although the effect occurs predominantly in the liver it has been recently shown that inflammatory responses in the brain also cause a loss of the same enzyme forms in that organ. The loss of cytochrome P450 in the brain in response to localised inflammation is accompanied by a similar loss in the liver. The decrease of cytochrome P450 and its dependent drug biotransformation is of concern whenever drugs are used in patients with infections or disease states with an inflammatory component.
Collapse
Affiliation(s)
- K W Renton
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
24
|
Renton KW, Dibb S, Levatte TL. Lipopolysaccharide evokes the modulation of brain cytochrome P4501A in the rat. Brain Res 1999; 842:139-47. [PMID: 10526104 DOI: 10.1016/s0006-8993(99)01773-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cytochrome P450 enzyme system is a multigene family of enzymes that is modulated in the liver during systemic inflammatory responses or during infection Several forms of the enzyme are expressed in discrete areas of the brain and likely play a critical role in the metabolism of drugs and endogenous chemicals in the central nervous system (CNS). Even though the brain responds to inflammation in a manner different from most tissues, we examined the possible modification of a major cytochrome P450 form (CYP1A) in the brain during inflammation confined to that organ. Total brain CYP1A activity, as measured by ethoxyresorufin dealkylase (EROD), was downregulated 24 and 48 h following the administration of a single dose of lipopolysaccharide (LPS). Regionally, a similar effect was determined in the cortex, hippocampus and the mid-brain but the activity in the cerebellum was unaffected. The examination of coronal brain sections using an antibody directed against CYP1A indicated that the enzyme was distributed in discrete cells of the hippocampus, thalamus and cortex and in the tanycytes surrounding the third ventricle. In each of these areas, the immunoreactivity was diminished in animals receiving LPS as compared to saline-treated animals. LPS also evoked the expression of the small molecular weight heat shock protein hsp27 throughout the brain indicating the development of an inflammatory response. These studies indicate that inflammation localized to the CNS causes an alteration in the levels and activity of a major cytochrome P450 form in the brain. This could have implications to the metabolism or activation of drugs and endogenous chemicals in the CNS during a disease state that features an inflammatory component.
Collapse
Affiliation(s)
- K W Renton
- Department of Pharmacology, Dalhousie University Halifax, Sir Charles Tupper Medical Bldg., Halifax, Nova Scotia, Canada.
| | | | | |
Collapse
|
25
|
Rosenbrock H, Hagemeyer CE, Singeç I, Knoth R, Volk B. Testosterone metabolism in rat brain is differentially enhanced by phenytoin-inducible cytochrome P450 isoforms. J Neuroendocrinol 1999; 11:597-604. [PMID: 10447797 DOI: 10.1046/j.1365-2826.1999.00371.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many cytochrome P450 (P450) isoforms are selectively inducible by xenobiotics, e.g. pharmaceuticals like the anti-epileptic drug phenytoin. Some of these P450 enzymes are involved in the metabolism of gonadal hormones and are of great importance, especially in early brain development. In this study, the hydroxylation of testosterone by rat brain microsomes from control and phenytoin-induced animals was examined by use of high performance liquid chromatography (HPLC) provided with a photodiode array detector (PDA). In control rats, testosterone is converted by cytochrome(s) P450 to 6alpha-hydroxytestosterone (OHT) as the main metabolite and 6beta-OHT as well as androstenedione as minor metabolites. After phenytoin treatment, brain microsomes showed a strong increase of testosterone metabolism to 2alpha-, 6beta-, 16alpha-, 16beta-OHT and androstenedione, whereby 16alpha-OHT was the main degradation product. These metabolites indicated the action of isoforms of the P450 subfamilies CYP2B, CYP2C and CYP3A. Inhibition experiments with antibodies against CYP2B1/2 and with the CYP2B specific inhibitor orphenadrine indicated the occurrence of members of this subfamily which are known to catalyse the oxidation of testosterone to 16alpha-OHT, 16beta-OHT and androstenedione. Western blots revealed the phenytoin-inducible expression of CYP2B1 and the constitutive expression of CYP3A. The latter is involved in the 6beta-hydroxylation of testosterone which was found correspondingly in control microsomes. Distinct CYP2C isoforms involved in the hydroxylation of testosterone in phenytoin-induced microsomes are not yet identified. The highly increased testosterone metabolism by phenytoin-dependent induction of specific cytochrome P450 isoforms in adult rat brain illustrates the potential influence of exogenous substances on internal regulative and metabolic pathways in the brain.
Collapse
Affiliation(s)
- H Rosenbrock
- Institute of Pathology, Department of Neuropathology, Neurocentre, University of Freiburg, Freiburg, Germany.
| | | | | | | | | |
Collapse
|
26
|
Dayal M, Parmar D, Dhawan A, Dwivedi UN, Doehmer J, Seth PK. Induction of rat brain and liver cytochrome P450 1A1/1A2 and 2B1/2B2 isoenzymes by deltamethrin. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 1999; 7:169-178. [PMID: 21781923 DOI: 10.1016/s1382-6689(99)00011-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/1998] [Revised: 02/10/1999] [Accepted: 02/22/1999] [Indexed: 05/31/2023]
Abstract
Deltamethrin, an α-cyano type II pyrethroid, administered orally (5, 10 and 15 mg/kg body weight for 7 consecutive days or at 5 mg/kg for further 15 and 21 days) to young albino Wistar rats (approximately 8 weeks old) produced a dose- and time-dependent increase in the activity of cytochrome P450-dependent 7-ethoxyresorufin-O-dealkylase (EROD) and 7-pentoxyresorufin-O-dealkylase (PROD) in rat liver and brain. However, significant induction in the enzyme activities was observed at higher doses or prolonged exposure. The magnitude of induction in rat liver microsomes was less at 15 mg/kg dose as compared to 10 mg/kg dose. Western blot analysis revealed a similar dose-related and time-dependent increase in the expression of P450 2B1/2B2 and 1A1 isoenzymes as indicated by the increased cross-reactivity of liver microsomes isolated from deltamethrin-treated animals with anti-P450 2B1/2B2 and 1A1. Inhibition of EROD and PROD observed after in vitro addition of anti-P450 2B1/2B2 and 1A1/1A2 or organic inhibitors, metyrapone and α-naphthoflavone, to the brain and liver microsomes of deltamethrin-pretreated animals (5 mg/kg×21 days), further provided support that the induction observed in the EROD and PROD activity in brain is due to the increased expression of P450 2B1/2B2 and 1A1/1A2, while, in the liver, isoenzymes other than these are also involved.
Collapse
Affiliation(s)
- M Dayal
- Developmental Toxicology Division, Industrial Toxicology Research Centre, P.O. Box 80, M.G. Marg, Lucknow-226001, UP, India; Department of Biochemistry, University of Lucknow, Lucknow-226007, UP, India
| | | | | | | | | | | |
Collapse
|
27
|
Attal-Khémis S, Dalmeyda V, Morfin R. Change of 7alpha-hydroxy-dehydroepiandrosterone levels in serum of mice treated by cytochrome P450-modifying agents. Life Sci 1998; 63:1543-53. [PMID: 9808065 DOI: 10.1016/s0024-3205(98)00421-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Dehydroepiandrosterone (DHEA) is 7alpha-hydroxylated in liver, brain and other organs of murine and in other species. Several works suggest that the 7alpha-hydroxy-DHEA produced may be one of the native antiglucocorticoids, and compounds modifying its production may prove useful in investigation of 7alpha-hydroxy-DHEA production and effects. After treatment of mice with dexamethasone, phenobarbital, trilostane, melatonin or metyrapone, we have used gas chromatography-mass spectrometry with negative ion detection for measurement of 7alpha-hydroxy-DHEA levels in serum of control and treated animals. The 7alpha-hydroxylating rates of liver and brain microsomes from the same animals were also measured. Results showed that serum levels of 7alpha-hydroxy-DHEA were significantly increased after treatment by all compounds except metyrapone. Significantly increased 7alpha-hydroxy-DHEA levels were directly related with significantly increased 7alpha-hydroxylation yields in liver and not in brain. In contrast, metyrapone decreased 7alpha-hydroxylation in liver and brain. These findings indicate that in brain and in liver, different enzyme systems may be responsible for production of 7alpha-hydroxy-DHEA and that treatment-induced modifications of circulating 7alpha-hydroxy-DHEA levels are mainly due to change of 7alpha-hydroxylating rates in liver.
Collapse
Affiliation(s)
- S Attal-Khémis
- Laboratoire de Biotechnologie, Conservatoire National des Arts et Métiers, Paris, France
| | | | | |
Collapse
|
28
|
Stanley LA, Copp AJ, Pope J, Rolls S, Smelt V, Perry VH, Sim E. Immunochemical detection of arylamine N-acetyltransferase during mouse embryonic development and in adult mouse brain. TERATOLOGY 1998; 58:174-82. [PMID: 9839355 DOI: 10.1002/(sici)1096-9926(199811)58:5<174::aid-tera3>3.0.co;2-q] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Arylamine N-acetyltransferases (NATs) are important in susceptibility to xenobiotic-induced disorders (e.g., drug-induced autoimmune disease, bladder cancer), but their role in endogenous metabolism is yet to be elucidated. The discovery that human NAT1 acts upon p-aminobenzoylgluatamate (p-ABG) to generate p-acetamidobenzoylglutamate (p-AABG), a major urinary metabolite of folic acid, suggests that human NAT1 may play a role in folic acid metabolism and hence in the normal development of the neural tube. In this study we examined the distribution of NAT in neuronal tissue from adult mice and embryos. Immunohistochemical staining of the adult mouse cerebellum revealed NAT2 (the mouse homologue of human NAT1) expression in the cell bodies and dendrites of Purkinje cells and in the neuroglia of the molecular layer. In embryos, NAT2 was detected in developing neuronal tissue on days 9.5, 11.5, and 13.5. It was expressed intensely in the nerual tube around the time of closure. The level of expression subsequently declined in the neuroepithelium but increased in glial cells. In addition, NAT2 was detected in the developing heart and gut. These findings demonstrate that the embryo itself expresses an enzyme which is involved in the metabolism of folic acid, so that the role played by both mother and embryo must be considered when examining the role of folic acid in embryonic development. These findings imply that polymorphisms in NAT genes could play a role in determining susceptibility to neural tube defects (NTD) and orofacial clefting, developmental disorders which can be prevented by dietary administration of folic acid.
Collapse
Affiliation(s)
- L A Stanley
- Department of Pharmaceutical Sciences, De Montfort University, Leicester, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
29
|
Ibach B, Appel K, Gebicke-Haerter P, Meyer RP, Friedberg T, Knoth R, Volk B. Effect of phenytoin on cytochrome P450 2B mRNA expression in primary rat astrocyte cultures. J Neurosci Res 1998; 54:402-11. [PMID: 9819145 DOI: 10.1002/(sici)1097-4547(19981101)54:3<402::aid-jnr11>3.0.co;2-k] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Studies on cytochrome P450 2B (CYP2B) in the brain have essentially been focused on protein characterization and regional distribution. Due to the high sequence homology between the closely related CYP2B1 and 2B2 isoforms and the low amounts of the corresponding mRNAs few efforts have been made to analyze the expression, regulation, and inducibility of these P450 genes in a specific cell type. In the present study, we investigated CYP2B mRNA expression in primary rat astrocyte cultures under the influence of the anti-epileptic drug phenytoin, which is known to be a CYP2B inducing agent in liver. In situ hybridization with a digoxigenin (DIG)-labeled cRNA probe demonstrated that 30-40% of the astrocytes strongly expressed a CYP2B mRNA-specific signal within the first week of cultivation. With increasing age (> 14 days) a greater percentage of cells (>90%) expressed mRNA for P450 2B. However, the level of transcriptional activity was substantially lower than in younger cultures. To discriminate between the 2B1 and 2B2 isoforms the reverse transcription/polymerase chain reaction (RT/PCR) procedures were proved for rat hepatic mRNA as a control assay. Subsequently, the application of this method on cultured astrocytes confirmed that these brain cells may express CYP2B1 mRNA. CYP2B2 mRNA could not be detected in astrocyte cultures at any age examined. Phenytoin led to the down regulation of CYP2B1 mRNA, which contrasts with the drug inducing effect on hepatic CYP2B1 and 2B2 levels. After 4 hr of exposure of phenytoin to the astrocytes no amplification product could be detected at all. Phenytoin did not induce either CYP2B1 or 2B2 expression.
Collapse
Affiliation(s)
- B Ibach
- Department of Neuropathology, Neurocenter, University of Freiburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
30
|
LaBella FS, Stein D, Queen G. Occupation of the cytochrome P450 substrate pocket by diverse compounds at general anesthesia concentrations. Eur J Pharmacol 1998; 358:177-85. [PMID: 9808268 DOI: 10.1016/s0014-2999(98)00596-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Each of a diverse array of compounds, at concentrations reported to effect general anesthesia, when added to liver microsomes, forms a complex with cytochromes P450 to generate, with reference to a cuvette containing microsomes only, a characteristic absorbance-difference spectrum. This spectrum results from a change in the electron-spin state of the heme iron atom induced upon entry by the anesthetic molecule into the enzyme catalytic pocket. The difference spectrum, representing the anesthetic-P450 complex, is characteristic of substances that are substrates for the enzyme. For the group of compounds as a whole, the magnitudes of the absorbance-difference spectra vary only about twofold, although the anesthetic potencies vary by several orders of magnitude. The dissociation constants (Ks), calculated from absorbance data and representing affinities of the anesthetics for P450, agree closely with the respective EC50 (concentration that effects anesthesia in 50% of individuals) values, and with the respective Ki (concentration that inhibits P450 catalytic activities half-maximally) values reported by us previously. The absorbance complex resulting from the occupation of the catalytic pocket by endogenous substrates, androstenedione and arachidonic acid, is inhibited, competitively, by anesthetics. Occupation of and perturbation of the heme catalytic pocket by anesthetic, as monitored by the absorbance-difference spectrum, is rapidly reversible. The presumed in vivo consequences of perturbation by general anesthetics of heme proteins is suppression of the generation of chemical signals that determine cell sensitivity and response.
Collapse
Affiliation(s)
- F S LaBella
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | | | | |
Collapse
|
31
|
Cok I, Wang-Buhler JL, Kedzierski MM, Miranda CL, Yang YH, Buhler DR. Expression of CYP2M1, CYP2K1, and CYP3A27 in brain, blood, small intestine, and other tissues of rainbow trout. Biochem Biophys Res Commun 1998; 244:790-5. [PMID: 9535744 DOI: 10.1006/bbrc.1998.8273] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Expression of five constitutive forms of cytochrome P450 [(LMC1 (CYP2M1), LMC2 (CYP2K1), LMC3, LMC4, and LMC5 (CYP3A27)] in selected tissues from sexually immature 2-year old female and male rainbow trout (Oncorhynchus mykiss) were examined at the translational level by Western blot using polyclonal antibodies raised in rabbits against those purified trout hepatic P450s. Tissues examined were from brain, liver, muscle, blood, head kidney, trunk kidney, upper intestine, stomach, heart, and gonad (ovary or testis). The results showed that the liver was the major organ for expression of all the trout P450s studied. Trunk kidney was the secondary expression site except for LMC5. Selective translational expression of these P450 isoforms or similar proteins was observed for LCM1 and LMC5 in brain; for LMC2 and LMC5 in female upper intestine; and for LMC2 in blood plasma of the fish studied under the experimental and sampling conditions.
Collapse
Affiliation(s)
- I Cok
- Department of Agricultural Chemistry and Marine/Freshwater Biomedical Sciences Center, Oregon State University, Corvallis 97331, USA
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Birth defects cause a myriad of societal problems and place tremendous anguish on the affected individual and his or her family. Current estimates categorize about 3% of all newborn infants as having some form of birth defect or congenital anomaly. As more precise means of detecting subtle anomalies become available this estimate, no doubt, will increase. Even though birth defects have been observed in newborns throughout history, our knowledge about the causes and mechanisms through which these defects are manifested is limited. For example, it has been estimated that around 20% of all birth defects are due to gene mutations, 5-10% to chromosomal abnormalities, and another 5-10% to exposure to a known teratogenic agent or maternal factor [D.A. Beckman, R.L. Brent, Mechanisms of teratogenesis. Ann. Rev. Pharmacol. Toxicol. 24 (1984) 483-500; K. Nelson, L.B. Holmes Malformations due to presumed spontaneous mutations in newborn infants, N. Engl. J. Med. 320 (1989) 19-23.]. Together, these percentages account for only 30-40%, leaving the etiology of more than half of all human birth defects unexplained. It has been speculated that environmental factors account for no more than one-tenth of all congenital anomalies [D.A. Beckman, R.L. Brent, Mechanisms of teratogenesis, Ann. Rev. Pharmacol. Toxicol. 24 (1984) 483-500]. Furthermore, since there is no evidence in humans that the exposure of an individual to any mutagen measurably increases the risk of congenital anomalies in his or her offspring' [J.F. Crow, C. Denniston, Mutation in human populations, Adv. Human Genet. 14 (1985) 59-121; J.M. Friedman, J.E. Polifka, Teratogenic Effects of Drugs: A Resource for Clinicians (TERIS). The John Hopkins University Press, Baltimore, 1994], the mutagenic activity of environmental agents and drugs as a factor in teratogenesis has been given very little attention. Epigenetic activity has also been given only limited consideration as a mechanism for teratogenesis. As new molecular methods are developed for assessing processes associated with teratogenesis, especially those with a genetic or an epigenetic basis, additional environmental factors may be identified. These are especially important because they are potentially preventable. This paper examines the relationships between chemicals identified as human teratogens (agents that cause birth defects) and their mutagenic activity as evaluated in one or more of the established short-term bioassays currently used to measure such damage. Those agents lacking mutagenic activity but with published evidence that they may otherwise alter the expressions or regulate interactions of the genetic material, i.e. exhibit epigenetic activity, have likewise been identified. The information used in making these comparisons comes from the published literature as well as from unpublished data of the U.S. National Toxicology Program (NTP).
Collapse
Affiliation(s)
- J B Bishop
- Laboratory of Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | | | | |
Collapse
|