+ All Categories
Home > Documents > ABC transporters and cytochromes P450 in the human central nervous system: influence on brain...

ABC transporters and cytochromes P450 in the human central nervous system: influence on brain...

Date post: 08-Dec-2016
Category:
Upload: marie-anne
View: 212 times
Download: 0 times
Share this document with a friend
14
1. Introduction 2. CYP enzymes in the human brain 3. ABC transporters in the human brain 4. Regulation of ABC transporters and CYPs in the human brain 5. Drug metabolism and pharmacokinetic aspects 6. ABC transporters and CYP in neurodegenerative disorders 7. Conclusion 8. Expert opinion Review ABC transporters and cytochromes P450 in the human central nervous system: influence on brain pharmacokinetics and contribution to neurodegenerative disorders Fabien Dutheil, Aude Jacob, Sandrine Dauchy, Philippe Beaune, Jean-Michel Scherrmann, Xavier Decle `ves & Marie-Anne Loriot Universite´Paris Descartes, 45 rue des Saints-Pe`res, 75270 Paris Cedex 06, France Importance of the field: The identification of xenobiotic metabolizing enzymes (i.e., CYP) and transporters (i.e., ABC transporters) (XMET) in the human brain, including the BBB, raises the question whether these transporters and enzymes have specific functions in brain physiology, neuropharmacology and toxicology. Areas covered in this review: Relevant literature was identified using PubMed search articles published up to March 2010. Search terms included ‘ABC transporters and P450 or CYP’, ‘drug metabolism, effect and toxicity’ and ‘neurodegenerative disease (Alzheimer and Parkinson diseases)’ restricted to the field of ‘brain or human brain’. What the reader will gain: This review aims to provide a better understanding of XMET functions in the human brain and show their pharmacological importance for improving drug delivery and efficacy and also for managing their side effects. Finally, the impact of brain XMET activity during neurode- generative processes is discussed, giving an opportunity to identify new markers of human brain diseases. Take home message: During the last 2 decades, much evidence concerning the specific distribution patterns of XMET, their induction by xenobiotics and endobiotics and their genetic variations have made cerebral ABC trans- porters and CYP enzymes key elements in the way individual patients respond to centrally acting drugs. Keywords: ATP-binding cassette transporters, CYP enzymes, human brain, neurodegenerative diseases, xenobiotics Expert Opin. Drug Metab. Toxicol. (2010) 6(10):1161-1174 1. Introduction Xenobiotics are chemical compounds that are foreign to the body. They are found throughout our environment, in the air, water and our food. Exposure to them is unavoidable, whether it is deliberate (e.g., drugs) or involuntary (e.g., pollutants). Xenobiotics must be converted to their polar hydrophilic metabolites before they can be detoxified and eliminated from the body. The cytochrome P450 (CYP) enzymes and ATP-binding cassette (ABC) transporters form the main system for metabolizing and transporting xenobiotics (xenobiotic metabolizing enzymes and transporters; XMETs). The presence of these XMETs within the brain is important for the bioactivation of the xenobiotics and hence damage to cells in this target organ 10.1517/17425255.2010.510832 © 2010 Informa UK, Ltd. ISSN 1742-5255 1161 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by University of Queensland on 04/17/13 For personal use only.
Transcript

1. Introduction

2. CYP enzymes in the human

brain

3. ABC transporters in the human

brain

4. Regulation of ABC transporters

and CYPs in the human brain

5. Drug metabolism and

pharmacokinetic aspects

6. ABC transporters and CYP in

neurodegenerative disorders

7. Conclusion

8. Expert opinion

Review

ABC transporters andcytochromes P450 in the humancentral nervous system: influenceon brain pharmacokineticsand contribution toneurodegenerative disordersFabien Dutheil, Aude Jacob, Sandrine Dauchy, Philippe Beaune,Jean-Michel Scherrmann, Xavier Decleves & Marie-Anne Loriot††Universite Paris Descartes, 45 rue des Saints-Peres, 75270 Paris Cedex 06, France

Importance of the field: The identification of xenobiotic metabolizing

enzymes (i.e., CYP) and transporters (i.e., ABC transporters) (XMET) in the

human brain, including the BBB, raises the question whether these transporters

and enzymes have specific functions in brain physiology, neuropharmacology

and toxicology.

Areas covered in this review: Relevant literature was identified using PubMed

search articles published up to March 2010. Search terms included ‘ABC

transporters and P450 or CYP’, ‘drug metabolism, effect and toxicity’ and

‘neurodegenerative disease (Alzheimer and Parkinson diseases)’ restricted to

the field of ‘brain or human brain’.

What the reader will gain: This review aims to provide a better understanding

of XMET functions in the human brain and show their pharmacological

importance for improving drug delivery and efficacy and also for managing

their side effects. Finally, the impact of brain XMET activity during neurode-

generative processes is discussed, giving an opportunity to identify new

markers of human brain diseases.

Take home message: During the last 2 decades, much evidence concerning

the specific distribution patterns of XMET, their induction by xenobiotics

and endobiotics and their genetic variations have made cerebral ABC trans-

porters and CYP enzymes key elements in the way individual patients respond

to centrally acting drugs.

Keywords: ATP-binding cassette transporters, CYP enzymes, human brain,

neurodegenerative diseases, xenobiotics

Expert Opin. Drug Metab. Toxicol. (2010) 6(10):1161-1174

1. Introduction

Xenobiotics are chemical compounds that are foreign to the body. They are foundthroughout our environment, in the air, water and our food. Exposure to them isunavoidable, whether it is deliberate (e.g., drugs) or involuntary (e.g., pollutants).Xenobiotics must be converted to their polar hydrophilic metabolites before theycan be detoxified and eliminated from the body. The cytochrome P450 (CYP)enzymes and ATP-binding cassette (ABC) transporters form the main system formetabolizing and transporting xenobiotics (xenobiotic metabolizing enzymes andtransporters; XMETs). The presence of these XMETs within the brain is importantfor the bioactivation of the xenobiotics and hence damage to cells in this target organ

10.1517/17425255.2010.510832 © 2010 Informa UK, Ltd. ISSN 1742-5255 1161All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

for both centrally-acting drugs and environmental toxins thatcross the blood brain barrier (BBB) [1]. The lipophilic characterof several xenobiotics enables them to diffuse across BBB andenter the brain parenchyma, although the specific ABC trans-porters in the plasma membrane of brain microvessel endothe-lial cells may limit their brain distribution. The limited capacityof neurons to regenerate makes the brain extremely vulnerableto damage by toxic compounds. The expression and regulationof XMET is tissue-specific and cell type-specific, and the brainhas its own unique complement of them [2-4]. These XMETscan metabolize and transport a vast array of compounds, includ-ing centrally-acting drugs, neurotoxins, neurotransmitters andneurosteroids [5-7]. The identification of ABC transporters andCYPs in the brain, including the BBB, raises the questionwhether these transporters and enzymes have specific functionsin brain physiology, neuropharmacology and toxicology.Individuals respond differently to centrally-acting thera-

peutic drugs, and their response is not always predicted bythe concentration of drug in their blood plasma [8]. Drugsthat act on the central nervous system (CNS) may be metab-olized in situ within the brain, and alterations in the degree ofin situ metabolism may contribute to a great variability in theresponses of individuals to a drug. In the same way, ABCtransporters may limit the penetration of a CNS drug intothe brain and, therefore, its efficacy. The concentrations ofbrain ABC transporters and CYPs are determined by an indi-vidual’s genotype and also by their exposure to environmentalinducers and/or repressors.The brain cellular architecture is specific and complex com-

prising of the brain parenchyma with cell bodies of astrocytes,neurons, oligodendrocytes and microglia, and the BBB withbrain endothelial cells, foot astrocytes and pericytes. Thisreview focuses on the nature and distribution of the XMETin the human brain parenchyma and at the BBB. We examinethe recent developments in our understanding of the roleof these proteins in brain physiology, in the response to

xenobiotics (drugs, toxic compounds) and in the pathogenesisof neurodegenerative disorders.

2. CYP enzymes in the human brain

The first studies that established the existence of brain CYPswere done on rats in 1977. CYP monooxygenase activity wasfound in rat brain microsomes [9]. Two years later, Guengerichand Mason and Marietta et al. confirmed the catalytic capaci-ties and inducible properties of these enzymes in thebrain [10,11]. Although microsomal CYPs were present, theiractivity was only 2 -- 5% of the CYP activity in the liver, repre-senting ~ 30 pmol/ml of microsomal protein. Marietta et al.also demonstrated that cerebral CYPs were 3 -- 170 times lessactive than hepatic CYPs when acting on exogenous com-pounds such as meperidine, hexobarbital and benzo[a]pyr-ene [11]. However, Fishman et al. published new information,in 1980, on the role of mitochondrial CYP in cerebral homeo-stasis [12]. Estradiol-2-hydroxylase, also known as CYP1A1, wasvery active just before the preovulatory release of luteinizinghormone. This metabolic enhancement was measured in themicrosomal, mitochondrial and synaptosomal fractions [12].Walther et al. subsequently showed that there was a CYP inthe mitochondrial inner membrane whose activity was compa-rable to that of microsomal CYP [13]. Thus, the amount ofmicrosomal CYP in the rat brain is 6 -- 100 pmol/mg of totalproteins, while the amount of mitochondrial CYP is around75 -- 220 pmol/mg of total proteins [14-16]. The human cerebralcortex has a similar concentration of mitochondrial CYP,100 -- 300 pmol/mg of total proteins [16].

Unlike the liver, the cytoarchitectural organization and cellfunctions in the brain are extremely variable and the concen-trations of CYPs in certain specific neurons can be as high asor higher than those in hepatocytes [17]. These brain enzymesare unlikely to contribute to overall drug metabolism, butthey may contribute to variations in the drug response ofindividuals through local in situ metabolism [18]. Brain CYPsare enzymatically active in vitro, and we and others haverecently identified the cofactors and coenzymes needed forthem to be active in vivo in their immediate environment [2,4].The presence of the enzymes, CYP reductase and ferredoxinreductase, which are required for a functional enzymaticsystem [16,19] supports the existence of active enzymes, as previ-ously documented in human and rodent brains by measuringmicrosomal monooxygenase activities and enzyme inhibitionstudies [4]. Moreover, a recent study has assessed the importanceof brain CYP activity in the analgesic action of µ-opioids [20].Conroy et al. produced a mutant mouse with reduced brainneuron-specific P450 activity. These mice showed much lowermorphine anti-nociception than controls. Pharmacologicalinhibition of brain P450 arachidonate epoxygenases also blocksmorphine anti-nociception in mice and rats [20]. Furthermore,ABC transporters, particularly ABCB1 (MDR1, P-gp), werefound in the human BBB 20 years ago [21]. Almost 1000original articles have been published on it since their discovery,

Article highlights.

. Functional xenobiotic metabolizing enzymes andtransporters (XMET) including ATP-binding cassettetransporters and P450 enzymes are present in humanbrain both in the parenchyma and the blood brainbarrier (BBB) according to a specific pattern.

. Their activities towards endogenous and exogenoussubstrates have pharmacological andtoxicological consequences.

. A better understanding of their role will allowdeveloping more efficient centrally active drugs andmodulating metabolic pathways involved inneurodegenerative diseases.

. Further studies are required to elucidate the role ofXMET and the molecular mechanisms modulating theirexpression and activity and to identify the metaboliteend products biologically active in the human brain.

This box summarizes key points contained in the article.

ABC transporters and cytochromes P450 in the human central nervous system

1162 Expert Opin. Drug Metab. Toxicol. (2010) 6(10)

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

but there have been few studies on the drug-metabolizing con-tent of the BBB. We recently determined the gene expressionprofiles of the CYPs and ABC transporters at the humanBBB and in the brain parenchyma [3].

2.1 CYP in the human brain parenchymaA total of 20 isoforms of human CYP (CYP1A1, 1A2, 1B1,2B6, 2C8, 2D6, 2E1, 3A4, 3A5, 8A1, 11A1, 11B1, 11B2,17A1, 19A1, 21A2, 26A1, 26B1, 27B1 and 46A1) havebeen identified in the brain. But only a limited number ofthem have been extensively studied: CYP1A1, 1A2, 2B6,2D6, 2E1 and 46A1. Most of them are widely distributedin the cortex, cerebellum, basal ganglia, hippocampus, sub-stantia nigra, medulla oblongata, pons and so on (Figure 1),but the reported data vary greatly. The discrepancies betweenthe studies are probably due to the different techniques usedto identify the CYPs. Their mRNAs or cDNA have beenidentified by real-time PCR, qualitative RT-PCR, northernblotting, Southern blotting, RNA dot blot, FISH and in situhybridization. The CYP proteins have been detected by west-ern blotting or immunostaining. The conflicting resultsbetween studies are partly due to problems of primer or anti-body specificity and sensitivity because of the great sequencehomology between CYPs [22]. We recently examined the24 genes encoding all the known human CYPs of families 1,2 and 3 and CYP46A1 using highly specific primers thathad been tested and validated in several tissues to avoid anyrisk of nonspecific amplification [23]. We found a wide varietyof xenobiotic-metabolizing enzymes in the human brain anddemonstrated their apparent selective distribution in specificparts of the brain. The major transcripts corresponding toCYP46A1, 2J2, 2U1, 1B1, 2E1 and 2D6 were present in allstructures of the human brain, but the concentrations ofmRNAs encoding each of them were unequally distributedin the different brain regions [2]. CYP transcripts had beendetected previously in whole human brain extracts [24], buttheir distribution and the presence of the corresponding pro-teins was unknown or poorly documented [24]. Our studies onCYP proteins confirmed the presence of extensively studiedCYPs such as CYP1B1 and 2D6, and we used specific anti-bodies to determine the distribution patterns of CYP2J2,CYP2U1 and CYP46A1 in the cells at different parts ofthe human brain. The main CYPs (CYP1B1, 2D6, 2J2,2U1 and 46A1) were found in the mitochondrial and micro-somal fractions of CYP proteins in the human frontal lobe,hippocampus, substantia nigra and cerebellum, but the distri-butions of these five isoforms in specific cell types all differed.CYP46A1 protein was present only in neuronal cells such asthe pyramidal cells of the frontal cortex, the CA1 -- 4 pyrami-dal cells and the granule cell of the dentate gyrus of the hippo-campus, and in cerebellar Purkinje cells. CYP1B1 protein wasmainly detected in microglial cells and to a lesser extent in theastrocytes of these four locations. Immunohistochemical stud-ies showed CYP2D6, 2J2 and 2U1 in both neuronal cells andastrocytes, but the amounts varied with the brain region.

CYP2J2 and 2U1 proteins were predominantly found in theastrocytes surrounding the cerebral blood vessels of the frontallobe and cerebellum, and these two isoforms were presentin both neurons and astrocytes of the hippocampus. Wealso identified other CYP isoforms, such as CYP2E1 andCYP2B6, in human cerebral cells. CYP2E1 protein wasdetected in specific cells at several brain locations [25]. Thisenzyme was mainly found in the pyramidal neurons and glialcells of the frontal cortex, in the pyramidal cells CA2 andCA3 and in the granular cells of the hippocampus dentategyrus [25]. CYP2B6 is also widely distributed throughout thebrain but its basal concentration is generally low [26]. Thisenzyme is present in both the neurons and glial cells of theneocortex layer I, including the astrocytes surrounding cere-bral blood vessels, where it colocalizes with glial fibrillaryacidic protein [25]. All these data are summarized in Figure 2A.

2.2 CYP in the human brain microvesselsThe BBB is composed of three main cellular elements, themicrovessel endothelial cells sealed by tight junctions, theend-feet of the astrocytes sheathing the microvessels andthe pericytes that share the basal membrane with endothelialcells (Figure 2B). They form a dynamic neurovascular unitthat is the first line of defense for the brain against unwantedcompounds. The entry of many compounds into the brain,including numerous commercial drugs, is also restricted byABC transporters; their presence in the BBB is detailed later.The BBB is not only a physical barrier but also a metabolicbarrier because of the drug-metabolizing enzymes, especiallythe CYPs, in the endothelial cells [27]. Several functionalCYPs have been found in isolated rodent brain interfaces [28].However, little is known about the CYPs at the human BBB.We recently used highly specific primers to detect the mRNAof 23 CYP isoforms from families 1, 2 and 3 in isolatedhuman brain microvessels. CYP1B1 and 2U1 mRNA werethe main CYPs in the BBB [3]. There was 14 times moreCYP1B1 mRNA in the microvessels than in the cerebral cor-tex, and the corresponding protein was also detected. In con-trast, the CYP2U1 gene was similarly expressed in isolatedmicrovessels and in the brain cortex. The CYP2D6,CYP2E1, CYP2J2 and CYP2R1 genes were alsoweakly expressed.

3. ABC transporters in the human brain

The human ABC transporter superfamily is a ubiquitous groupof 48 membrane proteins organized into seven subfamilies.These ABC transporters use the energy from ATP hydrolysisto transport endogenous and exogenous compounds acrosscell membranes. They are widely distributed in normal tissuesand play a key role in protecting the entire body against toxinsand xenobiotics by preventing their penetration (into the brain,testis or placenta) or excreting them (via the liver, kidney, gas-trointestinal tract) [21,29]. ABC transporters are also stronglyimplicated in the development of the multi-drug resistance

Dutheil, Jacob, Dauchy, Beaune, Scherrmann, Decleves & Loriot

Expert Opin. Drug Metab. Toxicol. (2010) 6(10) 1163

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

200

Hippocampus

Cerebral cortex

Medulla oblongataPons

Substantia nigra

Corpus callosum

Temporal lobeNucleus accumbens

Putamen

Brain

Insula

Frontal lobe

Caudate nucleusCerebellum

150

100

50

0

-50

-100

-150

-200

0.61A1

2C8

2B6

1B1

2D6

46A1

2E1

2U12J2

0.4

0.2

0

-0.2

-0.4

-0.6

-1 -0.5 0

P[1]

P[2

]

0.5 1

-200 -100 0

T[1] (40%)

T[2

] (29

%)

100 200

4

321

A.

B.

Figure 1. CYP expression in the different human encephalic locations. The separation of the total human brain, cerebellum

and 16 human brain regions (A) and the contribution of variables (B) were obtained by using multivariate analysis. Each brain

region is represented by different color points. The stars indicated the total cerebral cortex and the different cortical lobes

(occipital, parietal, frontal, temporal). In the center of the figure, the numbers were used to represent the following regions:

1 = occipital lobe; 2 = parietal lobe; 3 = postcentral gyrus; 4 = paracentral gyrus. Quantitative analysis of P450 transcripts was

performed and the normalized matrix of P450 expression was imported into MATLAB (Mathworks, Natick, MA, USA). The

data are recalculated into a number of PCs where the first PC (T1 axis) represents the largest variance in the data and the

second PC (T2 axis) less variance. In part A of the figure, the inspection of the PCs reveals that the two first PCs (T1 and T2)

accounted for ~ 69% of the variance of independent variables, 40 and 29%, respectively. In part B of the figure, the data show

that the T1 contribution (horizontal axis) was dominated by CYP46A1 (0.9) and CYP2D6 (0.4), whereas the T2 contribution

(vertical axis) was mainly explained by CYP 2C8, CYP1A1 and CYP2B6. The pictures show the major CYP isoforms found both at

mRNA (real-time PCR) and protein levels (western blotting).Reproduced with permission from [2].

PC: Principal component; PCA: Principal component analysis.

ABC transporters and cytochromes P450 in the human central nervous system

1164 Expert Opin. Drug Metab. Toxicol. (2010) 6(10)

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

Astrocyte foot Basal membrane

Pericyte

Endothelialcell

CYP2J2CYP2U1

CYP1B1

Lumen Tight junction

ABCB1

ABCG2

B.

Astrocyte

Neuron

Microglial cells

CYP46A1CYP2D6CYP2E1

CYP2D6CYP1B1CYP2J2CYP2U1

CYP1B1

ABCC4

CYP2U1CYP2D6

ABCC5 CYP2J2, CYP2E1, CYP2R1

Pericyte Pericyte

Pericyte Pericyte Pericyte

A.

Figure 2. The main subcellular localizations of xenobiotic-metabolizing enzymes and ABC transporters in the human brain

parenchyma (A) and at the BBB (B). A. Immunohistochemistry studies revealed that CYP2J2, 2U1 and 2D6 were expressed in

both neurons and astrocytes whereas CYP2E1 was exclusively expressed in neurons. The main CYP expressed in neurons was

the CYP46A1. Moreover, CYP1B1 was expressed in astrocytes. B. ABCB1, ABCG2, ABCC4 and ABCC5 are the main ABC

transporters expressed at the human BBB. CYP1B1 was highly detected in human brain microvessels at both mRNA and

protein levels. Other CYPs (CYP2U1, CYP2D6, CYP2J2, CYP2E1, CYP2R1) were detected in human brain microvessels at mRNA

level (in italics). CYP2J2 and 2U1 proteins were predominantly found in the astrocyte foot processes surrounding the cerebral

blood vessels.

Dutheil, Jacob, Dauchy, Beaune, Scherrmann, Decleves & Loriot

Expert Opin. Drug Metab. Toxicol. (2010) 6(10) 1165

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

(MDR) phenotype in cancer cells [30]. This review focuses onthe expression and function of ABC transporters in the humanBBB, even if some ABC transporters are also expressed inastrocytes [31,32] and pericytes [32].The first ABC transporter detected at the human BBB was

ABCB1 (MDR1, P-gp), which was initially discovered in1976 [33,49]. ABCB1 is principally present at the luminalside of the brain capillaries [32,34], but has also been found inboth the luminal and abluminal membranes of endothelialcells [32]. The ABCC (MDR associated-protein) family mem-bers are also present at the human BBB, but their expressionand functional activity are still a matter for discussion. WhileABCC4 and ABCC5 have been firmly located at the luminalside of the BBB [35], the cellular and subcellular distributionsof ABCC1 mRNA and protein are still unclear. Most studieshave not detected ABCC1 in the human brain or at thehuman BBB [36,37]. But, two studies reported findingABCC1 at the luminal side of human brain capillary endothe-lial cells [34,35]. Other ABCC transporters are not present atthe human BBB (ABCC2) or the concentrations of theirmRNAs are very low (ABCC2, ABCC3) [34,35]. Recently, weshowed that ABCC5 transcripts were the most abundantABCC mRNAs in isolated human brain microvessels [3].ABCG2 (breast cancer resistance protein), a more recently

discovered ABC transporter, is found in capillary and venousendothelium cells of many normal human tissues [38], andespecially on the luminal side of the human BBB [36]. Whilethe presence of some ABC transporters in the human BBBremains controversial, ABCB1, ABCC4 -- 5 and ABCG2 areprobably the main ABC transporters at the human BBB,where they act as the gatekeepers of the brain (Figure 2B).Their expression vary during the development of the humanCNS [39] and in disorders such as epilepsy. The genes encod-ing ABCB1, ABCC2 and ABCC5 are all overexpressed in thebrain microvessels endothelial cells of epileptic humans,whereas ABCC1 is not [40]. Thus, they are involved in bothphysiological and pathological situations.Some recent studies have focused on the relative abundance

of ABC transporters in the human brain and the BBB. A com-parative analysis of the expressions of the ABCG2, ABCB1 andABCC1 genes in homogenates of human non-malignant braintissue and human glioblastoma multiform tumors by real-timequantitative PCR showed that the expressions of ABCG2and ABCC1 were similar, higher than the expression ofABCB1 [41]. A recent study on human cerebral microvesselsisolated from patients suffering from epilepsy or glioma estab-lished the expression profiles of the genes encoding the mainABC transporters at the human BBB. Analyses of mRNA indi-cated that ABCG2 was seven times more expressed thanABCB1 in human brain microvessels, whereas ABCC4 andABCC5 were much less expressed [3]. This was also true for por-cine brain endothelium, where ABCB1 levels determined bynorthern blot analysis were lower than those of ABCG2 [42].As the relative expression of ABC transporters varies

between species [43], it is difficult to find a good animal model

with which to predict the permeability of drugs across thehuman BBB. We need to know the relative expression ofthe main ABC transporters in the human BBB in order tobetter understand the pharmacoresistance and pharmacoki-netics of CNS drugs. These data will help improve drug deliv-ery to the CNS. Additional studies are required to assess therelative amounts of ABC transporter proteins at the humanBBB to confirm that the gene expression profile agrees withthe protein profile so as to better elucidate the implicationof ABC transporters at the BBB.

4. Regulation of ABC transporters and CYPsin the human brain

The amounts of CYPs in the brain can be regulated differentlyfrom those of hepatic CYPs. The concentration of CYP2B6 inthe brain regions of smokers and alcoholics is abnormally high,particularly in the cerebellar Purkinje cells, granular cell layerand hippocampal pyramidal neurons [25]. Like CYP2B6, theconcentration of CYP2D6 protein in the brains of alcoholicsis greater than in those of non-alcoholics. It is particularlyhigh in the putamen, globus pallidus and substantia nigra,despite the fact that it is not elevated in the liver [26].CYP2E1 expression is induced by ethanol and nicotine [25].Smokers have significantly more CYP2E1 protein in severalbrain regions than do non-smokers [26]. The CYP2E1 synthesisis increased when human cultured neuroblastoma cells aretreated with nicotine [25]. Pollutants such as 2,3,7,8-tetrachloro-dibenzo-p-dioxin (or dioxin), an aromatic halogen hydrocar-bon, and b-naphtoflavone are also involved in the inductionof CYP1A1 and 1A2 synthesis in the CNS [44,45].

Transcription factors (TFs) such as nuclear receptors (NRs)and PAS/ARNT family members such as the aryl hydrocar-bon receptor (AhR) are involved in the regulation of ABCtransporter and CYP expression in peripheral tissues [46]. Weshowed that LXRb, RXRb, PPAR-d, RXRa, PPAR-a andPPAR-g are, in the order of appearance, the main TFs inthe human brain that have a location-specific action [2] inline with an earlier Japanese study [47] suggesting the in situregulation of CYP synthesis.

The cerebral distributions of several major regulation path-ways involving the AhR, NR1H2/LXRb or NR1C/PPARsubfamilies correlate well with those of the correspondingCYP target genes. High concentrations of AhR transcriptshave been found in the dura mater, which also has largeamounts of CYP1A1, CYP2S1 and CYP1B1, which can beinduced in an AhR-dependent manner [2]. The epoxygenaseCYP2J2 synthesizes EETs and other metabolites that activateboth NR1C1/PPAR-a and NR1C3/PPAR-g . Finally, recentstudies have clearly demonstrated that PPAR and LXR areNRs that are activated by fatty acid and cholesterol derivates,respectively, and control the expression of a range of genesinvolved in lipid metabolism and inflammation [48].

The pregnane X receptor (PXR) induces the expression ofABCB1, CYP3A4 and CYP2C9, the constitutive androstane

ABC transporters and cytochromes P450 in the human central nervous system

1166 Expert Opin. Drug Metab. Toxicol. (2010) 6(10)

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

receptor (CAR) the synthesis of CYP2B6, while the AhRmainly regulates the expression of the CYP1A and CYP1Bgenes [46]. Although PXR, CAR and AhR have been found inthe rodent BBB, the findings remain controversial [50,51]. Thereis little evidence for the presence of TFs that can stimulate thesynthesis of ABC transporters and CYP in the human BBB.Significant amounts of AhR have been found in isolatedhuman brain microvessels [3] and in human cerebral microves-sel endothelial cells (hCMEC/D3). These hCMEC/D3 cellsare a promising in vitro model of the human BBB [52]. In con-trast, PXR and CAR were barely detected in both human brainmicrovessels and hCMEC/D3 cells [3,52,53].

The majority of studies examining XMET regulationfocused at the transcriptional levels. However, some studiespointed out the implication of epigenetic modifications inXMET expression. For example, Shafaati et al. have shown ade-repression of CYP46A1 expression following a treatmentwith an histone deacetylase inhibitor [54]. RegardingABCB1, in vitro studies showed that epigenetic modulationssuch as modifications of methylation or acetylation statusmay lead to modulation of ABCB1 expression [55]. To date,epigenetic regulation of CYPs and ABC transporters withinthe human brain remains unknown.

5. Drug metabolism and pharmacokineticaspects

The BBB containing ABCB1, ABCG2, ABCC4 andABCC5 at the luminal membrane of the cerebral endothelialcells may be considered as the first line of defense againstthe entry of their substrates, limiting their distribution inthe CNS and thus their effects on the CNS (Table 1). Oncea drug has been able to reach the intracellular space, drug-metabolizing enzymes may be considered either as a seconddetoxifying barrier or as a CYP-based toxicological orpharmacological activation. In this review, we focus on theeffect of drug-metabolizing enzymes in the human brain asthey may increase or decrease efficacy and/or toxicity ofCNS-targeted drugs.

CYPs generally metabolize drugs to form hydrophilicmetabolites that are readily excreted from the body. However,the production of such metabolites in the brain could result intheir prolonged presence and slower clearance from the brain.Thus, cerebral metabolism could modulate the pharmacolog-ical response and explain, in part, the variability in responseto centrally psychoactive drugs due to differences in brainCYP-mediated metabolism between individuals [18]. Indeed,there is often a poor correlation between the plasma concentra-tions of neuroleptics and antidepressants and their therapeuticeffects. Hence, in situ metabolism may modulate the responseto these drugs. Brain microsomes slowly metabolize drugs suchas debrisoquine, sparteine and dextrometorphan, probablydue to CYP2D6 activity [56,57]. Because CYP2D6 metabolizesa wide range of centrally-acting drugs such as analgesics,anti-dementia drugs, b-blockers, tricyclic antidepressants,

anti-psychotics, monoamine oxidase-inhibitors and vasodilata-tors [58], it could be important to better understand thecontribution of this isoform to drug metabolism in the brain.

Minor drug metabolic pathways could also produce signif-icant pharmacological or toxicological responses, particularlyif they occur at the site of action. For example, a minor met-abolic pathway for codeine results in the formation of mor-phine that is the O-demethylated metabolite produced byCYP2D6. Chen et al. showed that at least the initial analgesiceffects of codeine are due to morphine produced in the brain,not in the liver [59]. Others have shown that this synthesisoccurs in the cerebral regions containing the µ-opioid recep-tors [60]. Several publications have found no link between theside effects of newer antidepressants, or the clinical responseto them and the CYP2D6 genotype [61,62], even thoughthe plasma concentrations of these drugs are influenced bythe same genotypes [61].

Because CYPs are present in the CNS and the brain is thetarget of centrally-acting drugs, their concentrations may beparticularly important for determining the response of anindividual to centrally-acting substances [22].

6. ABC transporters and CYP inneurodegenerative disorders

The identification of high concentrations of XMET in thebrain strongly suggests that the environment influences cere-bral functions. Several publications have provided new signif-icant insights into the distribution of the ABC transportersand CYP involved in the transport and metabolism of bothexogenous and endogenous substances. Previous reportshave provided evidence for the biochemical basis of neurode-generative disorders. This review examines the role of ABCtransporters and CYP in Parkinson’s and Alzheimer’s diseases.

6.1 Parkinson’s disease, CYP2D6 and ABCB1The etiology of Parkinson’s disease (PD) is usually multi-factorial, involving environmental factors and their interac-tion with susceptible genes [63]. People who have beenexposed to 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine(MPTP) develop Parkinsonism due to the inhibition of mito-chondrial function in dopaminergic neurons [64]. MPTPbecomes active when it is metabolized to 1-methyl-4-phenyl-pyridinium (MPP+), which has a chemical structure similar tothat of the herbicide paraquat.

While environmental chemicals can increase the risk ofPD, host factors that influence their uptake, metabolismand distribution in the body may modulate the risk to agiven individual. Many substances can induce PD. MPTP,an analog of meperidine (or pethidine) which induces PDin humans and primates, is metabolized to MPP+ by themonoamine oxidase B in astrocytes [65]. MPTP is themain toxic compound used to generate a model of PD.This neurotoxic chemical selectively destroys substantianigra cells by inhibiting neurone mitochondrial respiration.

Dutheil, Jacob, Dauchy, Beaune, Scherrmann, Decleves & Loriot

Expert Opin. Drug Metab. Toxicol. (2010) 6(10) 1167

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

CYP2D6 catalyzes the N-demethylatio of MPTP [66], whichdecreases its neurotoxic effect. Thus, CYP2D6 helps protectagainst environmental stress. In contrast, organophosphatepesticides such as parathion and chlorpyrifos are metabolizedby CYP2D6 to metabolites that can be toxic for theCNS [67].Epidemiological studies have shown that exposure to pesti-

cides may be associated with PD [68]. Organochlorines tend toaccumulate in lipid-rich organs such as the brain [69], and postmortem studies found higher concentrations of dieldrin andlindane in PD brains than in control brains [70]. Several orga-nochlorines including DDT and cyclodienes (e.g., heptachlor,endosulfan, dieldrin) have selective effects on striatal dopami-nergic neurons [71]. Dieldrin is more toxic for dopaminergiccells than for non-dopaminergic cells [72] and depletes braindopamine by increasing the production of the dopamineand vesicular monoamine transporters in the presynaptic ter-minals of dopaminergic neurons in the striatum of mice [73].Dieldrin also causes apoptotic cell death, mitochondrial dys-function and protein aggregation [74], and induces oxidativedamage in the mouse nigrostriatal dopamine system [75].Elbaz et al. studied French farmers exposed to pesticidesand showed that those with a poor metabolizer CYP2D6

phenotype were at an increased risk of PD (odds ratio: 4.7;95% CI 1.3 -- 17.4) [6].

In summary, it seems likely that the metabolism of pesticidesby brain CYPs is linked to the increased risk of PD, particularlyduring professional exposure, and possibly to the susceptibilityof an individual to PD.

The ABCB1 gene contains > 40 single nucleotide polymor-phisms, some of them linked to changes in the expressionand/or function of this ABC transporter [76]. Many studieshave investigated the relationship between ABCB1 polymor-phisms and the risk of developing PD, but only two studieshave detected a direct association to date [77-80]. Therefore,ABCB1 variants are now thought to rather act indirectly as asusceptibility factor or modulator of the risk of PD in con-junction with exposure to known risk factors of PD such aspesticides [68]. Three studies have underlined the increasedrisk of developing PD for carriers of variant C3435T orG2677[A,T] ABCB1 alleles and have been exposed topesticides [77,80,81].

Bartels et al. conducted two in vivo studies using [11C]-verapamil PET to assess the functional ABCB1 status at theBBB of patients suffering from PD. They found thatABCB1 function was not altered in the early stages of PD,

Table 1. List of substrates and modulators of CYPs and ABC transporters in human brain.

CYP

ABC

CNS compounds as substrates Modulators

transporters Xenobiotics Endogenous compounds Inducers Inhibitors

CYP1B1 Caffeine, polycyclic aromatichydrocarbons

Arachidonic acid, retinoicacid, melatonin, estradiol,prostanoids

b-Naphtophlafone,dioxin,3-methylcholanthren

NA

CYP2D6 Codeine, tramadol, clozapine,imipramine, haloperidol, ecstasy

Progesterone Dexamethasone,rifampicin

Bupropion, fluoxetine,paroxetine, amiodarone

CYP2E1 Alcohols, halothane,procarcinogens

Fatty acids Ethanol, isoniazid Disulfiram

CYP2J2 Ebastine, astemizole,terfenadine

Arachidonic acid NA Terfenadine

CYP2U1 NA Long-chain fatty acids,arachidonic acid

NA NA

CYP46A1 NA Cholesterol NA NA

ABCB1 Opioids (morphine, methadone,oxycodone, fentanyl)Antidepressants (amitriptyline,fluoxetine, paroxetine, sertraline)Antipsychotics (amisulpride,risperidone), antiepileptics

NA Rifampin,dexamethasone

Verapamil, cyclosporine A,amlodipine, quinidine

ABCG2 Sulfate conjugates Estrone-3-sulfate,17b-estradiol

NA Imatinib

ABCC4 NA E217bG, PGE2,DHEA sulfate

NA Celecoxib, diclofenac

ABCC5 NA NA Probenecid,sulfinpyrazone

From [94-98].

ABC: ATP-binding cassette; NA: Not available.

ABC transporters and cytochromes P450 in the human central nervous system

1168 Expert Opin. Drug Metab. Toxicol. (2010) 6(10)

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

but its function was decreased in specific brain regions ofthose with advanced PD, implying that the BBB had becomeless protective [82].

Thus, ABCB1 may be a factor in the susceptibility of PDpatients and also explains their ongoing neurodegeneration.

6.2 Alzheimer’s disease and ABC transportersAlzheimer’s disease (AD) is characterized by the accumulationand deposition of amyloid b (Ab) peptides in the brain paren-chyma and cerebral vessels. This leads to senile plaques andcerebral amyloid angiopathy (CAA) that is toxic to both neu-rons and cerebral endothelial cells [83,84]. An imbalancebetween the production of Ab peptides and their clearancefrom the brain or the aberrant trafficking of Ab peptidesacross the BBB are the two main hypotheses advanced toexplain the accumulation of Ab in the brain and the develop-ment of CAA in AD patients [85]. Ab peptides are transportedacross the BBB by receptor-mediated transcytosis: low-density lipoprotein receptor-related protein-1 is involved inthe efflux of Ab peptides from the brain, whereas the receptorfor advanced glycation end products is involved in shuttlingAb from the circulation into the brain [85,86]. Two ABC trans-porters, ABCB1 and ABCG2, have also recently been linkedto the transport of Ab peptides at the BBB. Thus,ABCB1 transports Ab peptides in vitro [87,88]. In vivo studieson Abcb1-null mice and wild-type mice showed thatABCB1 prevented the deposition of Ab in the brains of thewild-type mice as compared to the Abcb1-null mice [89].Vogelgesang et al. found no significant association betweenABCB1 polymorphisms in humans and the risk of AD, butthey highlighted an inverse correlation between Ab depositionrates and the cerebrovascular ABCB1 expression [90]. Theyalso established that the cerebral blood vessels of non-demented elderly subjects had a low ABCB1 content butabundant CAA [91]. However, it is not clear whether the lossof ABCB1 is the cause or consequence of Ab deposition.Xiong et al. showed the amount of ABCG2 in the brains ofAD/CAA patients was higher than in age-matched controls.They also established that the brains of Abcg2-null mice accu-mulated significantly more Ab peptides than did those ofwild-type mice [92]. The implication of both ABCB1 andABCG2 in preventing Ab from entering the brain was alsoconfirmed in vitro using a human brain endothelial cell line,hCMEC/D3 [93].

The above data thus strongly suggest that the ABC trans-porters, ABCB1 and ABCG2, act to protect the brain by pre-venting Ab deposition, and hence against the developmentof AD.

7. Conclusion

There is still great interest in ABC transporters and CYPenzymes because of their significant implication in the trans-port and metabolism of substances by the brain. Their activityand their subcellular and cellular distributions in the brain

parenchyma and at the BBB underline their importance formaintaining the physiology and homeostasis of the brain.Their specific distribution patterns, their induction by xeno-biotics and endobiotics and their genetic variations makeABC transporters and CYP enzymes key elements in theway individual patients respond to centrally acting drugs. Italso suggests that ABC transporters and CYPs are implicatedin the development of neurodegenerative disorders. Thus, abetter understanding of XMET functions in the human brainis of major pharmacological importance because it may pro-vide a useful tool for improving drug delivery and efficacyand also for managing their side effects. It is also an opportu-nity to identify new markers of human brain diseases. Last, abetter understanding of cerebral ABC transporters and CYPenzymes could contribute greatly to the development andoptimization of new therapeutic strategies.

8. Expert opinion

The metabolism and transport of xenobiotics in the humanbrain are a very attractive field of research because of theirimpact on the variations in individual response to drugs actingon the CNS, and susceptibility to psychiatric and neurode-generative disorders. The activities of the XMET towardsendogenous and exogenous compounds in the brain showthe importance of these proteins in brain physiology, neuro-pharmacology, development and diseases. A xenobiotic mustcross the BBB before it can reach its targets within the brainparenchyma. It is equally important to characterize the BBBand the brain parenchyma in terms of their XMET distribu-tion patterns. It is now clear that XMETs are present at theBBB and in the brain parenchyma and several recent studieshave clearly shown that the subcellular and cellular patternsof XMET distribution in these two brain structures differ dra-matically. It is also essential to identify their substrates and themetabolites that are produced in situ in order to better under-stand the pharmacological and toxicological effects of xenobi-otics. The levels of CYPs and ABC transporters in the brainare probably too low to influence the overall pharmacokinet-ics of the drugs. Nevertheless, the presence of a functionalXMET in the brain could partly explain why the differentresponses of individuals to centrally acting drugs do notalways correlate with the plasma drug concentrations. Altera-tions in brain XMET levels through genetic variation orthrough induction by xenobiotics (including drugs, nicotineor ethanol) would result in variations in local metabolism,which may contribute to the inter-individual variations seenin efficacy, drug--drug or drug--endobiotic interactions, andside effects of drugs that enter and act on the CNS. Further-more, it is probably crucial to assess the XMET activity inthe human BBB for a better understanding of the pharma-coresistance and the pharmacokinetics of drugs in order toimprove drug delivery. We, therefore, need to identify theenvironmental and genetic factors that modulate the activityof cerebral XMET in order to predict a xenobiotic response.

Dutheil, Jacob, Dauchy, Beaune, Scherrmann, Decleves & Loriot

Expert Opin. Drug Metab. Toxicol. (2010) 6(10) 1169

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

We believe that CNS drug development should use specificin vitro and in vivo models to determine how new chemicalentities transported by the BBB and metabolized by thebrain. However, the complexity of cerebral structure andphysiology plus the multiple functions of the BBB makeit difficult to develop such reliable models for predictingthe metabolism and transport of xenobiotics that targetthe brain.The transcriptional and post-transcriptional regulation of

XMET activity in the brain may involve molecular mecha-nisms that differ from those occurring in the liver and otherextra-hepatic tissues. Thus, the classical inducers of XMETspecifically activate TFs (PXR, CAR) found at low levels inthe brain. The regulatory pathways probably depend notonly on the presence or the absence of TFs, but also on thecellular environment provided by biochemical mediators andtransduction signaling specific to brain cells. The recent map-ping studies on the human brain identified the major isoformsof CYPs and ABC transporters that can be potential targetsfor identifying new drugs or significant modulators of theireffects. Thus, the pharmacological inhibition or induction ofbrain CYP or ABC transporter activities can block or mediatedrug effects. A quantitative picture of XMET in the brainregions, cells and subcellular organelles will help establishtheir relative contributions to the response to xenobiotics.The cellular and regional distribution of XMET may beresponsible for neuroprotective or neurotoxic properties,depending on the biological activities of the metabolic endproducts generated by these enzymes. The location of CYPenzymes in the mitochondria could reflect a specific brainfunction for these proteins. XMET may be involved in theneuroprotection or toxic effect of neurotoxins that causeneurodegenerative disease, especially in some brain regions

where XMET concentrations are high. The involvement ofmany XMETs in the metabolism of endogenous compoundsand xenobiotics provides the opportunity to develop pharma-cological tools for modulating the biochemical pathwaysimplicated in psychiatric or neurodegenerative disorders. Forinstance, the lipid mediators produced by the activity ofCYPs are important endogenous regulators of cell prolifera-tion, differentiation, oxidative stress, neuroinflammation andapoptosis. In addition, CYP epoxygenases, which catalyzethe formation of vasoactive compounds during the arachi-donic acid cascade, may actively contribute to the regulationof the cerebral blood flow. Interestingly, while endobioticmolecules can influence the oxidation of xenobiotics byCYP enzymes, the endobiotic--xenobiotic interactions maybe of great importance for establishing the pharmacologicaland toxicological effects of xenobiotics on the brain. Finally,we need to understand how the XMET functions in orderto improve drug efficacy and explain their side effects. Suchinformation may also identify new markers of human braindiseases. The tools and concepts needed to document the rolesof CYP enzymes and ABC transporters in brain physiologyand the pathology and treatment of neurological disordersare now available.

Acknowledgements

F Dutheil and A Jacob contributed equally to the work.X Decleves English text was edited by O Parkes. X Declevesand M-A Loriot contributed equally to this review.

Declaration of interest

This work was supported by a grant from Servier Technology.

ABC transporters and cytochromes P450 in the human central nervous system

1170 Expert Opin. Drug Metab. Toxicol. (2010) 6(10)

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Guengerich FP, Okazaki O, Seto Y,

et al. Radical cation intermediates in

N-dealkylation reactions. Xenobiotica

1995;25:689-709

2. Dutheil F, Dauchy S, Diry M,

et al. Xenobiotic-metabolizing enzymes

and transporters in the normal human

brain: regional and cellular mapping

as a basis for putative roles in cerebral

function. Drug Metab Dispos

2009;37:1528-38.. Provides novel information on the

localization and the nature of CYP

enzymes in the human brain.

3. Dauchy S, Dutheil F, Weaver RJ, et al.

ABC transporters, cytochromes P450

and their main transcription factors:

expression at the human blood--brain

barrier. J Neurochem 2008;107:1518-28.. First demonstration of gene expression

profile of ABC and CYP in the

human BBB.

4. Miksys S, Tyndale RF. Brain

drug-metabolizing cytochrome

P450 enzymes are active in vivo,

demonstrated by mechanism-based

enzyme inhibition.

Neuropsychopharmacology

2009;34:634-40.. Original approach to demonstrate for

the first time that both the constitutive

and induced forms of this enzyme are

active in situ in the rat brain.

5. Hiroi T, Kishimoto W, Chow T, et al.

Progesterone oxidation by cytochrome

P450 2D isoforms in the brain.

Endocrinology 2001;142:3901-8

6. Elbaz A, Levecque C, Clavel J, et al.

CYP2D6 polymorphism, pesticide

exposure, and Parkinson’s disease.

Ann Neurol 2004;55:430-4. Case--control study of Parkinson’s

disease demonstrating a strong gene

environment interaction with a higher

susceptibility to pesticide’s effect

among poor metabolizers for CYP2D6.

7. Seliskar M, Rozman D. Mammalian

cytochromes P450 -- importance of tissue

specificity. Biochim Biophys Acta

2007;1770:458-66

8. Michels R, Marzuk PM. Progress

in psychiatry (1). N Engl J Med

1993;329:552-60

9. Cohn JA, Alvares AP, Kappas A. On the

occurrence of cytochrome P-450 and aryl

hydrocarbon hydroxylase activity in rat

brain. J Exp Med 1977;145:1607-11

10. Guengerich FP, Mason PS.

Immunological comparison of hepatic

and extrahepatic cytochromes P-450.

Mol Pharmacol 1979;15:154-64

11. Marietta MP, Vesell ES, Hartman RD,

et al. Characterization of cytochrome

P-450-dependent aminopyrine

N-demethylase in rat brain:

comparison with hepatic aminopyrine

N-demethylation. J Pharmacol Exp Ther

1979;208:271-9

12. Fishman J, Norton BI, Krey L.

2-Hydroxylation of estrogens in

the brain participates in the initiation of

the preovulatory LH surge in the rat.

Biochem Biophys Res Commun

1980;93:471-7

13. Walther B, Ghersi-Egea JF, Minn A,

et al. Subcellular distribution of

cytochrome P-450 in the brain.

Brain Res 1986;375:338-44

14. Ghersi-Egea JF, Walther B, Minn A,

et al. Quantitative measurement of

cerebral cytochrome P-450 by second

derivative spectrophotometry.

J Neurosci Methods 1987;20:261-9

15. Bhagwat SV, Boyd MR, Ravindranath V.

Rat brain cytochrome P450.

Reassessment of monooxygenase activities

and cytochrome P450 levels.

Drug Metab Dispos 1995;23:651-4

16. Bhagwat SV, Boyd MR, Ravindranath V.

Multiple forms of cytochrome P450 and

associated monooxygenase activities in

human brain mitochondria.

Biochem Pharmacol 2000;59:573-82. First demonstration in human brain

mitochondria of the presence of

multiple forms of P450 belonging to

the 1A, 2B, and 2E families.

17. 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

18. Britto MR, Wedlund PJ. Cytochrome

P-450 in the brain. Potential

evolutionary and therapeutic relevance

of localization of drug-metabolizing

enzymes. Drug Metab Dispos

1992;20:446-50

19. Lavandera JV, Batlle AM, Buzaleh AM.

Metabolization of porphyrinogenic agents

in brain: involvement of the phase I drug

metabolizing system. A comparative

study in liver and kidney.

Cell Mol Neurobiol 2007;27:717-29

20. Conroy JL, Fang C, Gu J, et al. Opioids

activate brain analgesic circuits through

cytochrome P450/epoxygenase signaling.

Nat Neurosci 2010;13:284-6.. Very nice study demonstrating the

importance of brain cytochrome P450

activity in mu opioid analgesic action

with brain neuron-specific reductions

in P450 activity in a mouse model.

21. Thiebaut F, Tsuruo T, Hamada H,

et al. Cellular localization of the

multidrug-resistance gene product

P-glycoprotein in normal human tissues.

Proc Natl Acad Sci USA 1987;84:7735-8

22. Miksys SL, Tyndale RF.

Drug-metabolizing cytochrome P450s

in the brain. J Psychiatry Neurosci

2002;27:406-15

23. Bieche I, Narjoz C, Asselah T, et al.

Reverse transcriptase-PCR quantification

of mRNA levels from cytochrome

(CYP)1, CYP2 and CYP3 families in

22 different human tissues.

Pharmacogenet Genomics

2007;17:731-42.. Original study describing the

expression profile of the 23 CYP

mRNAs of CYP1, CTP2 and CYP3

families in 22 different human tissues

for a better understanding of the

physiological functions of these CYPs.

24. Nishimura M, Yaguti H, Yoshitsugu H,

et al. Tissue distribution of mRNA

expression of human cytochrome P450

isoforms assessed by high-sensitivity

real-time reverse transcription PCR.

Yakugaku Zasshi 2003;123:369-75

25. Howard LA, Miksys S, Hoffmann E,

et al. Brain CYP2E1 is induced by

nicotine and ethanol in rat and is

higher in smokers and alcoholics.

Br J Pharmacol 2003;138:1376-86

26. Miksys S, Tyndale RF. The unique

regulation of brain cytochrome P450 2

Dutheil, Jacob, Dauchy, Beaune, Scherrmann, Decleves & Loriot

Expert Opin. Drug Metab. Toxicol. (2010) 6(10) 1171

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

(CYP2) family enzymes by drugs and

genetics. Drug Metab Rev

2004;36:313-33

27. el-Bacha RS, Minn A. Drug metabolizing

enzymes in cerebrovascular endothelial

cells afford a metabolic protection to the

brain. Cell Mol Biol (Noisy-le-grand)

1999;45:15-23

28. Granberg L, Ostergren A, Brandt I, et al.

CYP1A1 and CYP1B1 in blood--brain

interfaces: CYP1A1-dependent

bioactivation of 7,12-dimethylbenz(a)

anthracene in endothelial cells.

Drug Metab Dispos 2003;31:259-65

29. Dean M, Rzhetsky A, Allikmets R.

The human ATP-binding cassette (ABC)

transporter superfamily. Genome Res

2001;11:1156-66

30. Gottesman MM, Fojo T, Bates SE.

Multidrug resistance in cancer: role

of ATP-dependent transporters.

Nat Rev Cancer 2002;2:48-58

31. Decleves X, Fajac A, Lehmann-Che J,

et al. Molecular and functional

MDR1-Pgp and MRPs expression in

human glioblastoma multiforme cell

lines. Int J Cancer 2002;98:173-80

32. Bendayan R, Ronaldson PT, Gingras D,

et al. In situ localization of

P-glycoprotein (ABCB1) in human

and rat brain. J Histochem Cytochem

2006;54:1159-67

33. Juliano RL, Ling V. A surface

glycoprotein modulating drug

permeability in Chinese hamster ovary

cell mutants. Biochim Biophys Acta

1976;455:152-62

34. Gazzin S, Strazielle N, Schmitt C, et al.

Differential expression of the multidrug

resistance-related proteins ABCb1 and

ABCc1 between blood--brain interfaces.

J Comp Neurol 2008;510:497-507

35. Nies AT, Jedlitschky G, Konig J, et al.

Expression and immunolocalization of

the multidrug resistance proteins,

MRP1-MRP6 (ABCC1-ABCC6), in

human brain. Neuroscience

2004;129:349-60.. Nice article showing the cellular

immunolocalization of MRP in the

human brain including BBB.

36. Cooray HC, Blackmore CG, Maskell L,

et al. Localisation of breast cancer

resistance protein in microvessel

endothelium of human brain.

Neuroreport 2002;13:2059-63

37. Sisodiya SM, Martinian L, Scheffer GL,

et al. Vascular colocalization of

P-glycoprotein, multidrug-resistance

associated protein 1, breast cancer

resistance protein and major vault

protein in human epileptogenic

pathologies. Neuropathol Appl Neurobiol

2006;32:51-63

38. Maliepaard M, Scheffer GL, Faneyte IF,

et al. Subcellular localization and

distribution of the breast cancer

resistance protein transporter in

normal human tissues. Cancer Res

2001;61:3458-64

39. Daood M, Tsai C, Ahdab-Barmada M,

et al. ABC transporter (P-gp/ABCB1,

MRP1/ABCC1, BCRP/ABCG2)

expression in the developing human

CNS. Neuropediatrics 2008;39:211-18.. Nice study showing the developmental

expression of ABC transporters

in humans.

40. Dombrowski SM, Desai SY, Marroni M,

et al. Overexpression of multiple drug

resistance genes in endothelial cells from

patients with refractory epilepsy.

Epilepsia 2001;42:1501-6

41. Zhang W, Mojsilovic-Petrovic J,

Andrade MF, et al. The expression and

functional characterization of ABCG2 in

brain endothelial cells and vessels.

FASEB J 2003;17:2085-7

42. Eisenblatter T, Huwel S, Galla HJ.

Characterisation of the brain multidrug

resistance protein (BMDP/ABCG2/

BCRP) expressed at the blood--brain

barrier. Brain Res 2003;971:221-31

43. Warren MS, Zerangue N, Woodford K,

et al. Comparative gene expression

profiles of ABC transporters in brain

microvessel endothelial cells and brain in

five species including human.

Pharmacol Res 2009;59:404-13.. First interspecies comparison of gene

expression profile of ABC transporters.

44. Huang P, Rannug A, Ahlbom E, et al.

Effect of 2,3,7,8-tetrachlorodibenzo-p-

dioxin on the expression of cytochrome

P450 1A1, the aryl hydrocarbon

receptor, and the aryl hydrocarbon

receptor nuclear translocator in rat brain

and pituitary. Toxicol Appl Pharmacol

2000;169:159-67

45. Iba MM, Storch A, Ghosal A, et al.

Constitutive and inducible levels of

CYP1A1 and CYP1A2 in rat cerebral

cortex and cerebellum. Arch Toxicol

2003;77:547-54

46. Xu C, Li CY, Kong AN. Induction

of phase I, II and III drug

metabolism/transport by xenobiotics.

Arch Pharm Res 2005;28:249-68

47. Nishimura M, Naito S, Yokoi T.

Tissue-specific mRNA expression

profiles of human nuclear receptor

subfamilies. Drug Metab Pharmacokinet

2004;19:135-49

48. Bensinger SJ, Tontonoz P. Integration

of metabolism and inflammation by

lipid-activated nuclear receptors. Nature

2008;454:470-7

49. Cordon-Cardo C, O’Brien JP, Casals D,

et al. Multidrug-resistance gene

(P-glycoprotein) is expressed by endothelial

cells at blood--brain barrier sites. Proc Natl

Acad Sci USA 1989;86:695-8

50. Akanuma S, Hori S, Ohtsuki S, et al.

Expression of nuclear receptor

mRNA and liver X receptor-mediated

regulation of ABC transporter A1 at rat

blood--brain barrier. Neurochem Int

2008;52:669-74.. An exhaustive study showing the

expression of transcription factors

regulating ABC transporters and CYPs

in the rat BBB.

51. Bauer B, Hartz AM, Fricker G, et al.

Pregnane X receptor up-regulation of

P-glycoprotein expression and transport

function at the blood--brain barrier.

Mol Pharmacol 2004;66:413-19.. First study showing the functional

expression of PXR regulating P-gp

expression in rat brain microvessels.

52. Dauchy S, Miller F, Couraud PO, et al.

Expression and transcriptional regulation

of ABC transporters and cytochromes

P450 in hCMEC/D3 human cerebral

microvascular endothelial cells.

Biochem Pharmacol 2009;77:897-909. An exhaustive characterization of the

hCMEC/D3 cell line in terms of ABC

and CYP.

53. Sasame HA, Ames MM, Nelson SD.

Cytochrome P-450 and NADPH

cytochrome c reductase in rat brain:

formation of catechols and reactive

catechol metabolites. Biochem Biophys

Res Commun 1977;78:919-26

54. Shafaati M, O’Driscoll R, Bjorkhem I,

et al. Transcriptional regulation of

cholesterol 24-hydroxylase by histone

ABC transporters and cytochromes P450 in the human central nervous system

1172 Expert Opin. Drug Metab. Toxicol. (2010) 6(10)

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

deacetylase inhibitors. Biochem Biophys

Res Commun 2009;378:689-94.. This work demonstrates a marked

time-dependent derepression of the

expression of CYP46A1, in response to

treatment with the potent histone

deacetylase (HDAC) inhibitor.

55. Baker EK, El-Osta A. Epigenetic

regulation of multidrug resistance 1 gene

expression: profiling CpG methylation

status using bisulphite sequencing.

Methods Mol Biol 2010;596:183-98

56. Tyndale RF, Sunahara R, Inaba T, et al.

Neuronal cytochrome P450IID1

(debrisoquine/sparteine-type): potent

inhibition of activity by (-)-cocaine and

nucleotide sequence identity to human

hepatic P450 gene CYP2D6.

Mol Pharmacol 1991;40:63-8

57. Tyndale RF, Li Y, Li NY, et al.

Characterization of cytochrome P-450

2D1 activity in rat brain: high-affinity

kinetics for dextromethorphan.

Drug Metab Dispos 1999;27:924-30

58. Zanger UM, Raimundo S,

Eichelbaum M. Cytochrome

P450 2D6: overview and update on

pharmacology, genetics, biochemistry.

Naunyn Schmiedebergs Arch Pharmacol

2004;369:23-37

59. Chen ZR, Irvine RJ, Bochner F, et al.

Morphine formation from codeine in rat

brain: a possible mechanism of codeine

analgesia. Life Sci 1990;46:1067-74

60. Eckardt K, Li S, Ammon S, et al.

Same incidence of adverse drug events

after codeine administration irrespective

of the genetically determined differences

in morphine formation. Pain

1998;76:27-33

61. Grasmader K, Verwohlt PL, Rietschel M,

et al. Impact of polymorphisms of

cytochrome-P450 isoenzymes 2C9,

2C19 and 2D6 on plasma concentrations

and clinical effects of antidepressants in a

naturalistic clinical setting. Eur J

Clin Pharmacol 2004;60:329-36

62. Peters EJ, Slager SL, Kraft JB, et al.

Pharmacokinetic genes do not influence

response or tolerance to citalopram

in the STAR*D sample. PLoS One

2008;3:e1872

63. Klein C, Schlossmacher MG.

The genetics of Parkinson disease:

implications for neurological care.

Nat Clin Pract Neurol 2006;2:136-46

64. Langston JW, Langston EB, Irwin I.

MPTP-induced parkinsonism in human

and non-human primates -- clinical and

experimental aspects. Acta Neurol

Scand Suppl 1984;100:49-54

65. Brooks WJ, Jarvis MF, Wagner GC.

Astrocytes as a primary locus for the

conversion MPTP into MPP+.

J Neural Transm 1989;76:1-12

66. Gilham DE, Cairns W, Paine MJ, et al.

Metabolism of MPTP by cytochrome

P4502D6 and the demonstration of

2D6 mRNA in human foetal and adult

brain by in situ hybridization.

Xenobiotica 1997;27:111-25

67. Sams C, Mason HJ, Rawbone R. Evidence

for the activation of organophosphate

pesticides by cytochromes P450 3A4 and

2D6 in human liver microsomes.

Toxicol Lett 2000;116:217-21

68. Brown TP, Rumsby PC, Capleton AC,

et al. Pesticides and Parkinson’s

disease -- is there a link?

Environ Health Perspect

2006;114:156-64

69. Tebourbi O, Driss MR, Sakly M, et al.

Metabolism of DDT in different tissues

of young rats. J Environ Sci Health B

2006;41:167-76

70. Corrigan FM, Wienburg CL, Shore RF,

et al. Organochlorine insecticides in

substantia nigra in Parkinson’s disease.

J Toxicol Environ Health A

2000;59:229-34

71. Kirby ML, Barlow RL, Bloomquist JR.

Neurotoxicity of the organochlorine

insecticide heptachlor to murine striatal

dopaminergic pathways. Toxicol Sci

2001;61:100-6

72. Kitazawa M, Anantharam V,

Kanthasamy AG. Dieldrin-induced

oxidative stress and neurochemical

changes contribute to apoptopic cell

death in dopaminergic cells. Free Radic

Biol Med 2001;31:1473-85

73. Miller GW, Erickson JD, Perez JT, et al.

Immunochemical analysis of vesicular

monoamine transporter (VMAT2)

protein in Parkinson’s disease.

Exp Neurol 1999;156:138-48

74. Kanthasamy AG, Kitazawa M,

Kanthasamy A, et al. Dieldrin-induced

neurotoxicity: relevance to Parkinson’s

disease pathogenesis. Neurotoxicology

2005;26:701-19

75. Hatcher JM, Richardson JR, Guillot TS,

et al. Dieldrin exposure induces oxidative

damage in the mouse nigrostriatal

dopamine system. Exp Neurol

2007;204:619-30

76. Cascorbi I, Gerloff T, Johne A, et al.

Frequency of single nucleotide

polymorphisms in the P-glycoprotein

drug transporter MDR1 gene in white

subjects. Clin Pharmacol Ther

2001;69:169-74

77. Drozdzik M, Bialecka M, Mysliwiec K,

et al. Polymorphism in the P-glycoprotein

drug transporter MDR1 gene: a possible

link between environmental and genetic

factors in Parkinson’s disease.

Pharmacogenetics 2003;13:259-63

78. Lee CG, Tang K, Cheung YB, et al.

MDR1, the blood--brain barrier

transporter, is associated with Parkinson’s

disease in ethnic Chinese. J Med Genet

2004;41:e60

79. Westerlund M, Belin AC, Anvret A, et al.

Association of a polymorphism in the

ABCB1 gene with Parkinson’s disease.

Parkinsonism Relat Disord 2009;15:422-4

80. Zschiedrich K, Konig IR,

Bruggemann N, et al. MDR1 variants

and risk of Parkinson disease. Association

with pesticide exposure? J Neurol

2009;256:115-20

81. Dutheil F, Beaune P, Tzourio C, et al.

Interaction between ABCB1 and

professional exposure to organochlorine

insecticides in Parkinson disease.

Arch Neurol 2010;67:739-45

82. Bartels AL, Kortekaas R, Bart J, et al.

Blood--brain barrier P-glycoprotein

function decreases in specific brain

regions with aging: a possible role in

progressive neurodegeneration.

Neurobiol Aging 2009;30:1818-24

83. Selkoe DJ. Alzheimer’s disease results

from the cerebral accumulation and

cytotoxicity of amyloid beta-protein.

J Alzheimers Dis 2001;3:75-80

84. Weller RO, Nicoll JA. Cerebral amyloid

angiopathy: pathogenesis and effects on

the ageing and Alzheimer brain.

Neurol Res 2003;25:611-16

85. Shibata M, Yamada S, Kumar SR,

et al. Clearance of Alzheimer’s amyloid-ss

(1-40) peptide from brain by LDL

receptor-related protein-1 at the

Dutheil, Jacob, Dauchy, Beaune, Scherrmann, Decleves & Loriot

Expert Opin. Drug Metab. Toxicol. (2010) 6(10) 1173

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.

blood--brain barrier. J Clin Invest

2000;106:1489-99

86. Deane R, Du Yan S, Submamaryan RK,

et al. RAGE mediates amyloid-beta

peptide transport across the blood--brain

barrier and accumulation in brain.

Nat Med 2003;9:907-13

87. Lam FC, Liu R, Lu P, et al. Beta-amyloid

efflux mediated by p-glycoprotein.

J Neurochem 2001;76:1121-8

88. Kuhnke D, Jedlitschky G, Grube M,

et al. MDR1-P-glycoprotein (ABCB1)

mediates transport of Alzheimer’s

amyloid-beta peptides -- implications for

the mechanisms of Abeta clearance at the

blood--brain barrier. Brain Pathol

2007;17:347-53

89. Cirrito JR, Deane R, Fagan AM, et al.

P-glycoprotein deficiency at the

blood--brain barrier increases amyloid-beta

deposition in an Alzheimer disease mouse

model. J Clin Invest 2005;115:3285-90. This study is a nice demonstration

of P-gp-dependent transport of

beta-amyloid at the mouse BBB.

90. Vogelgesang S, Cascorbi I, Schroeder E,

et al. Deposition of Alzheimer’s

beta-amyloid is inversely correlated

with P-glycoprotein expression in the

brains of elderly non-demented humans.

Pharmacogenetics 2002;12:535-41

91. Vogelgesang S, Warzok RW, Cascorbi I,

et al. The role of P-glycoprotein in

cerebral amyloid angiopathy; implications

for the early pathogenesis of Alzheimer’s

disease. Curr Alzheimer Res 2004;1:121-5

92. Xiong H, Callaghan D, Jones A, et al.

ABCG2 is upregulated in Alzheimer’s

brain with cerebral amyloid angiopathy

and may act as a gatekeeper at the

blood-brain barrier for Abeta(1-40)

peptides. J Neurosci 2009;29:5463-75

93. Tai LM, Loughlin AJ, Male DK, et al.

P-glycoprotein and breast cancer resistance

protein restrict apical-to-basolateral

permeability of human brain endothelium

to amyloid-beta. J Cereb Blood

Flow Metab 2009;29:1079-83

94. Dutheil F, Beaune P, Loriot MA.

Xenobiotic metabolizing enzymes in the

central nervous system: contribution of

cytochrome P450 enzymes in normal and

pathological human brain. Biochimie

2008;90:426-36. Recent review on the role of CYP

enzymes in the endogenous and

xenobiotic metabolism in human brain

and their implication in

pathological brain.

95. Giacomini KM, Huang SM,

Tweedie DJ, et al. Membrane

transporters in drug development.

Nat Rev Drug Discov 2010;9:215-36.. Nice review on the impact of

transporters in drug development as a

basis for FDA regulatory.

96. Robey RW, To KK, Polgar O, et al.

ABCG2: a perspective. Adv Drug

Deliv Rev 2009;61:3-13.. A nice exhaustive review on BCRP.

97. Shimada T, Fujii-Kuriyama Y.

Metabolic activation of polycyclic

aromatic hydrocarbons to carcinogens

by cytochromes P450 1A1 and 1B1.

Cancer Sci 2004;95:1-6

98. Vasiliou V, Gonzalez FJ. Role of

CYP1B1 in glaucoma. Annu Rev

Pharmacol Toxicol 2008;48:333-58

AffiliationFabien Dutheil1,2, Aude Jacob1,3,

Sandrine Dauchy1,3, Philippe Beaune2,5,6,

Jean-Michel Scherrmann3,6,

Xavier Decleves1,3,4 & Marie-Anne Loriot†1,2,5

†Author for correspondence1Universite Paris Descartes,

45 rue des Saints-Peres,

75270 Paris Cedex 06, France

E-mail: [email protected] National de la Sante et de la

Recherche Medicale,

INSERM UMR-S775,

Universite Paris Descartes,

45 rue des Saints-Peres,

75270 Paris Cedex 06, France3Institut National de la Sante et de la

Recherche Medicale,

Centre National de la Recherche

Scientifique,

INSERM U705,

CNRS UMR 8206,

Neuropsychopharmacologie des addictions,

Faculte de Pharmacie,

Paris, France4Assistance-Publique Hopitaux de Paris,

Hotel-Dieu,

Service de Pharmacie-

Pharmacologie-Toxicologie,

1, place du Parvis Notre-Dame,

75181 Paris Cedex 4, France5Assistance-Publique Hopitaux de Paris,

Hopital Europeen Georges Pompidou,

Service de Biochimie,

Pharmacogenetique et Oncologie Moleculaire,

20 rue Leblanc,

75908 Paris Cedex 15, France6Professor,

Universite Paris Descartes,

45 rue des Saints-Peres,

75270 Paris Cedex 06, France

ABC transporters and cytochromes P450 in the human central nervous system

1174 Expert Opin. Drug Metab. Toxicol. (2010) 6(10)

Exp

ert O

pin.

Dru

g M

etab

. Tox

icol

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

ity o

f Q

ueen

slan

d on

04/

17/1

3Fo

r pe

rson

al u

se o

nly.


Recommended