15
Mol Divers (2014) 18:895–909 DOI 10.1007/s11030-014-9550-6 EXPERT OPINION Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management Sumit Jain · Vijay Rathod · Rameshwar Prajapati · Prajwal P. Nandekar · Abhay T. Sangamwar Received: 14 February 2014 / Accepted: 28 August 2014 / Published online: 12 September 2014 © Springer International Publishing Switzerland 2014 Abstract The translational failure between preclinical ani- mal models and clinical outcome has alarmed us to search for a new strategy in the treatment of Alzheimer’s disease (AD). Interlink between Pregnane X Receptor (PXR) and P-glycoprotein (Pgp) at the blood brain barrier (BBB) has raised hope toward a new disease modifying therapy in AD. Pgp is a major efflux transporter for beta amyloid (Aβ) at human BBB. A literature survey reveals diminished expres- sion of Pgp transporter at the BBB in AD patients. Pregnane X Receptor is a major transcriptional regulator of Pgp. Restora- tion of Pgp at the BBB enhances the elimination of the Aβ from brain alongside and inhibits the apical to basolateral movement of Aβ from the circulatory blood. This review concentrates on in vitro, in vivo, and in silico advancements on the study of the PXR in context to Pgp and discusses the substrate and inhibitor specificity between PXR and Pgp. Keywords Pregnane X Receptor (PXR) · Alzheimer’s disease · Amyloid beta (Aβ) · Blood brain barrier (BBB) · P-glycoprotein (Pgp) · Neuroinflammation · In silico Electronic supplementary material The online version of this article (doi:10.1007/s11030-014-9550-6) contains supplementary material, which is available to authorized users. Sumit Jain, Vijay Rathod and Rameshwar Prajapati have contributed equally to this study. S. Jain · V. Rathod · R. Prajapati · P. P. Nandekar · A. T. Sangamwar (B ) Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali 160062, Punjab, India e-mail: [email protected] Introduction Alzheimer’s disease (AD), discovered by Dr. Alois Alzheimer in 1907 [1], is a neurodegenerative disorder categorized by dementia owing to the deposition of extracellular amyloid plaques and intraneuronal neurofibrillary tangles. AD is now termed as the “disease of the twenty-first century.” Current statistics reveal the severity of AD, with over 36 million people suffering from this neurodegerative disorder world- wide. In the United States (U. S. A.), almost 5.4 million patients live with AD being the sixth leading cause of death among all ages, which climbs to fifth in the case of elderly patients (65 years or older) [2]. The death rate due to AD has shown a steep increment over the last decade in the U. S. A. (Fig. 1)[3]. The death rate due to AD is subsequently rising up to 39 %, while it declined tremendously for other diseases (Fig. 2)[2]. The expenditure share for the treatment of AD is about 170 billion annually in the U. S. A. [2]. Due to the rise in the number of cases over the time, AD is approach- ing an epidemic stature and thus demands effective treatment strategy. Currently, the strategies adopted for AD are basically categorized into symptomatic therapy or disease modify- ing therapy. Symptomatic therapy addresses cognitive and neuropsychiatric aspects of AD, and is prominently utilizing neurotransmitter modulators such as 5-hydroxytryptamine antagonists, acetyl cholinesterase (AChE) inhibitors, gamma amino butyric acid (GABA-B) antagonists, inverse GABA agonists, N-methyl-d-aspartate (NMDA) antagonists, cogni- tive enhancers, and L-type calcium channel blockers. Disease modifying therapy has added new therapeutic approaches, which mainly targets pathogenesis of AD, such as Aβ pep- tide vaccination, secretases inhibitors, cholesterol-lowering drugs, metal chelators, and anti-inflammatory agents (Table 1). Clinical benefits of these available pharmacologi- 123

Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

  • Upload
    abhay-t

  • View
    213

  • Download
    1

Embed Size (px)

Citation preview

Page 1: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

Mol Divers (2014) 18:895–909DOI 10.1007/s11030-014-9550-6

EXPERT OPINION

Pregnane X Receptor and P-glycoprotein: a connexionfor Alzheimer’s disease management

Sumit Jain · Vijay Rathod · Rameshwar Prajapati ·Prajwal P. Nandekar · Abhay T. Sangamwar

Received: 14 February 2014 / Accepted: 28 August 2014 / Published online: 12 September 2014© Springer International Publishing Switzerland 2014

Abstract The translational failure between preclinical ani-mal models and clinical outcome has alarmed us to searchfor a new strategy in the treatment of Alzheimer’s disease(AD). Interlink between Pregnane X Receptor (PXR) andP-glycoprotein (Pgp) at the blood brain barrier (BBB) hasraised hope toward a new disease modifying therapy in AD.Pgp is a major efflux transporter for beta amyloid (Aβ) athuman BBB. A literature survey reveals diminished expres-sion of Pgp transporter at the BBB in AD patients. Pregnane XReceptor is a major transcriptional regulator of Pgp. Restora-tion of Pgp at the BBB enhances the elimination of the Aβ

from brain alongside and inhibits the apical to basolateralmovement of Aβ from the circulatory blood. This reviewconcentrates on in vitro, in vivo, and in silico advancementson the study of the PXR in context to Pgp and discusses thesubstrate and inhibitor specificity between PXR and Pgp.

Keywords Pregnane X Receptor (PXR) · Alzheimer’sdisease · Amyloid beta (Aβ) · Blood brain barrier (BBB) ·P-glycoprotein (Pgp) · Neuroinflammation · In silico

Electronic supplementary material The online version of thisarticle (doi:10.1007/s11030-014-9550-6) contains supplementarymaterial, which is available to authorized users.

Sumit Jain, Vijay Rathod and Rameshwar Prajapati have contributedequally to this study.

S. Jain · V. Rathod · R. Prajapati · P. P. Nandekar ·A. T. Sangamwar (B)Department of Pharmacoinformatics, National Institute ofPharmaceutical Education and Research (NIPER),S.A.S. Nagar, Sector 67, Mohali 160062, Punjab, Indiae-mail: [email protected]

Introduction

Alzheimer’s disease (AD), discovered by Dr. Alois Alzheimerin 1907 [1], is a neurodegenerative disorder categorized bydementia owing to the deposition of extracellular amyloidplaques and intraneuronal neurofibrillary tangles. AD is nowtermed as the “disease of the twenty-first century.” Currentstatistics reveal the severity of AD, with over 36 millionpeople suffering from this neurodegerative disorder world-wide. In the United States (U. S. A.), almost 5.4 millionpatients live with AD being the sixth leading cause of deathamong all ages, which climbs to fifth in the case of elderlypatients (65 years or older) [2]. The death rate due to AD hasshown a steep increment over the last decade in the U. S. A.(Fig. 1) [3]. The death rate due to AD is subsequently risingup to 39 %, while it declined tremendously for other diseases(Fig. 2) [2]. The expenditure share for the treatment of ADis about 170 billion annually in the U. S. A. [2]. Due to therise in the number of cases over the time, AD is approach-ing an epidemic stature and thus demands effective treatmentstrategy.

Currently, the strategies adopted for AD are basicallycategorized into symptomatic therapy or disease modify-ing therapy. Symptomatic therapy addresses cognitive andneuropsychiatric aspects of AD, and is prominently utilizingneurotransmitter modulators such as 5-hydroxytryptamineantagonists, acetyl cholinesterase (AChE) inhibitors, gammaamino butyric acid (GABA-B) antagonists, inverse GABAagonists, N-methyl-d-aspartate (NMDA) antagonists, cogni-tive enhancers, and L-type calcium channel blockers. Diseasemodifying therapy has added new therapeutic approaches,which mainly targets pathogenesis of AD, such as Aβ pep-tide vaccination, secretases inhibitors, cholesterol-loweringdrugs, metal chelators, and anti-inflammatory agents(Table 1). Clinical benefits of these available pharmacologi-

123

Page 2: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

896 Mol Divers (2014) 18:895–909

Fig. 1 Death rate (%) for allages due to Alzheimer’s diseasein last decade in the U.S.A. [2]

Fig. 2 Change in death rate(%) among different causes ofdeath in the U.S.A. [3]. (Colorfigure Online)

cal treatments are indisputable although limited to temporaryand symptomatic support to cognitive functions. In addition,consistent failure of new therapeutic interventions in AD overthe last decade in Phase-II proof of concept and pivotal Phase-III clinical trials collectively supports the need for more reli-able and effective treatment strategies for AD [4,5].

Human Pregnane X Receptor (hPXR) is a member oforphan nuclear receptor subfamily known for wide varietyof substrates and act as the xenobiotic sensing receptor [6].Upon activation, hPXR enhances the expression of its targetprotein by transcriptional regulation at the different sites ofbody, mainly liver and BBB. Regulation of various trans-porters mainly Pgp through PXR has recently shown theeffective way of improving central nervous system (CNS)pharmacotherapy and management of Aβ burden in AD [7].

PXR is also involved in the turnover of Apolipoprotein E(ApoE), which facilitates the disposition of Aβ outside thebrain. hPXR, along with other nuclear receptors, shows sig-nificant anti-inflammatory response, which replicates its con-trol over inflammation intracerebrally (Fig. 3). Altogether,PXR is implicated as an emerging target for AD in manag-ing Aβ overload and related complications.

Pathophysiology of Alzheimer’s disease

Dementia is a vast term covering several diseases and disor-ders related to malfunctioning and death of nerve cells (neu-rons) in the brain, leading to impaired memory, thinking,behavior, mood orientation, aphasia, and way of living. ADis characterized as the most witnessed dementia especially

123

Page 3: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

Mol Divers (2014) 18:895–909 897

Tabl

e1

Lis

tof

the

ther

apie

sus

edin

the

trea

tmen

tof

Alz

heim

er’s

dise

ase

Sr.n

o.T

hera

pyM

echa

nism

Dru

gsR

efer

ence

sin

supp

ortin

gin

form

atio

n

1A

cety

l-ch

olin

este

rase

inhi

bito

rSe

lect

ivel

yin

hibi

tace

tylc

holin

este

rase

Tacr

ine,

Don

epez

il,R

ivas

tigm

ine

Gal

anta

min

e[1

]2

pept

ide

vacc

inat

ion

Red

uctio

nin

cere

bral

amyl

oid

burd

enPo

nezu

mab

(Pha

seII

Clin

ical

tria

l)[2

,3]

-Sec

reta

ses

inhi

bito

r(G

amm

aan

dbe

ta)

Cat

alys

esth

efir

stst

epO

fA

βpr

oduc

tion

Sem

agec

esta

tand

Ava

gace

stat

(Pha

seII

Clin

ical

tria

l)[4

]

4C

hole

ster

ollo

wer

ing

drug

Low

ers

chol

este

roll

evel

Ato

rvas

tatin

Sim

vast

atin

[5]

5M

etal

chel

ator

sD

ecre

ase

the

toxi

city

asso

ciat

edw

ithA

βpr

oduc

tion

Clio

quin

ol[6

]6

Ant

i-in

flam

mat

ory

agen

tsPl

aque

-ass

ocia

ted

infla

mm

atio

nre

spon

sibl

efo

rneu

rode

-ge

nera

tion

Ibup

rofe

n,In

dom

etha

cin,

Sulin

dac

[7]

7R

AG

Ein

hibi

tor

Inhi

biti

nflux

ofA

βin

toth

ebr

ain

Pfize

ris

curr

ently

test

ing

PF-0

4494

700,

RA

GE

inhi

bito

r,in

Phas

eII

tria

ls[8

]

8M

itoch

ondr

ials

tabi

lizer

Stab

ilize

sm

itoch

ondr

iaL

atre

pird

ine

(dim

ebon

)[9

]9

PPA

Rag

onis

tIn

hibi

tth

-am

yloi

d-st

imul

ated

expr

essi

onof

the

cyto

kine

gene

sin

terl

euki

n-6

and

tum

orne

cros

isfa

ctor

α

Tro

glita

zone

,C

iglit

azon

e,D

HA

,In

dom

etha

cin

and

Ibup

rofe

n[1

0]

10A

β-L

ower

ing

agen

tIn

hibi

tthe

β-a

myl

oid

Tere

nflub

il[1

1,12

]11

Plaq

uede

stab

ilise

rIn

hibi

tam

yloi

dde

posi

tion

Tra

mip

rosa

te[1

3]12

5H

ydro

xy-t

ript

amin

eIm

prov

esco

gniti

onan

dbe

havi

orSB

7424

57[1

4]13

GA

BA

anta

goni

stIn

hibi

tGA

BA

-αan

dG

AB

A-β

SGS-

742

[15]

14In

vers

eG

AB

Aag

onis

tIn

hibi

tGA

BA

-αan

dG

AB

A-β

AV

E-3

933

[16]

15N

MD

Aan

tago

nist

Inhi

bitN

MD

Are

cept

orM

eman

tine

and

Ner

amex

ane

[17,

18]

16C

ogni

tive

enha

ncer

(Nut

ropi

cen

hanc

er)

Faci

litat

ion

ofsy

napt

ictr

ansm

issi

onan

din

terh

emis

pher

ein

form

atio

ntr

ansf

erN

efira

ceta

man

dPi

race

tam

[19,

20]

17L

-typ

eca

lciu

mch

anne

lblo

cker

Blo

ckC

a2+in

flux

topr

even

tfr

eera

dica

lin

duce

dne

u-ro

nalc

elld

eath

ME

M10

03[2

1]

18A

4b2

nAch

Rm

odul

ator

Nic

otin

icre

cept

orm

odul

atio

nIs

pron

iclin

e,T

C66

83[2

2,23

]19

AM

PAki

neA

MPA

(α-a

min

o-3-

hydr

oxy-

5-m

ethy

l-4-

isox

azol

epro

pion

icac

id)

rece

ptor

mod

ulat

ion

CX

516

[24]

20Pa

rtia

lmA

ChR

agon

ist

Enh

ance

sm

usca

rini

cac

etyl

chol

ine

rece

ptor

activ

ityM

ilam

elin

e,Sa

bcom

elin

e,X

anim

elin

eN

GX

267

[25]

21A

dren

ergi

cB

lock

erB

lock

sad

rene

rgic

rece

ptor

Dih

ydro

ergo

toxi

ne[2

6]22

Dop

amin

ergi

cag

onis

tE

nhan

cedo

pam

iner

gic

activ

ityPi

ribe

dil

[27]

23PA

Fan

tago

nist

Inhi

bitp

late

leta

ctiv

atin

gfa

ctor

Gin

kgo

bilo

ba[2

8]

123

Page 4: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

898 Mol Divers (2014) 18:895–909

Fig. 3 PXR-mediated interplay of various efflux transporters at BBBand their role in Alzheimer’s disease pathophysiology. Ligand bind PXRhetrodimerize with RXR act on the DNA which generates mRNA tran-

scriptionally induces various efflux transporters at blood brain barrier(right). Regulation of various inflammatory mediators by PXR (left).(Color figure Online)

in elderly patients and proves lethal with weakened essen-tial body functions such as locomotion, eating, and swallow-ing. The neuropathological and clinical examination revealedthat AD is characterized by the presence of amyloid plaques,and neurofibrillary tangles with cerebral amyloid angiopathy(CAA) and glial response [8–10]. Amyloid plaques mainlyconsist of 40–42 amino acid residues long amyloid β-peptide(Aβ) derived from β-amyloid precursor protein (APP) aftera series of cleavage catalyzed by enzymes such as betaand gamma secretases [11]. Aβ mainly accumulates in theparenchyma of the brain including areas of cortex and hip-pocampus [12,13]. Genetic evidence suggests that the muta-tion in the APP encoding gene results into the overproductionof Aβ by the action of beta and gamma secretases [14–16]. Aβ

plaques, in association with the microglial response, lead tothe generation of several cytotoxic factors including inflam-matory cytokines, interferon gamma (INFγ), tissue necroticfactor alpha (TNFα), interleukin 1β (IL-1β) and interleukin6 (IL-6) and chemokines (CCL2) [17,18]. Toxic microenvi-ronment, Aβ plaques, and neurofibrillary tangles (tau) col-lectively contribute to AD and neuronal loss. Hence, muchof the focus is laid on the elimination or reduction of Aβ load

from the brain as a therapeutic approach in the managementof AD.

Mechanism of Pregnane X Receptor

PXR is also known as a Steroid X Receptor (SXR) [19].While it is located abundantly in the liver and intestine, it isalso found in other parts of the body, such as brain, kidney,and lungs. Other nuclear receptors co-expressed with PXRare Constitutive Androstane Receptor (CAR), Aryl Hydro-carbon Receptor (AhR), Liver X Receptor (LXR), FarnesoidX Receptor (FXR), and Vitamin D Receptor (VDR). PXRwas originally identified on the basis of its sequence homol-ogy with other nuclear receptors [20,21]. In 1997, a mousesequence appeared in sequence tag database maintained byWashington University, characterized as a novel NR LBD(nuclear receptor ligand binding domain) fragment, whichlater translated into the complete protein. A cDNA encrypt-ing the complete mouse protein was successively cloned, andthe receptor was named as PXR based upon its activation bya wide variety of natural and synthetic pregnanes. Human

123

Page 5: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

Mol Divers (2014) 18:895–909 899

Fig. 4 Mechanism of activity of PXR enhancing expression of efflux transporter in the various parts of the body. (Color figure Online)

PXR was cloned by three separate groups and, therefore, itis also referred as the Pregnane-Activated Receptor (PAR) orSXR, broadly coined as hPXR [22,23].

The activity of PXR is regulated by many of bio-moleculesand ligands namely, co-repressors, co-activators, induc-ers/agonists, inhibitors/antagonists, and endogenous ligands.In the absence of agonist, PXR complexes with the co-repressors such as NCoR1 and NCoR2 (nuclear receptor co-repressor 1 and 2), which checks the transcriptional potentialof PXR by the involvement of histone deacetylase. When anagonist binds to PXR, it induces conformational changes inthe PXR structure resulting in the breakage of co-repressor–PXR complex and formation of new co-activator–PXR com-plex, with co-activator such as SRC1 and SRC3 (steroidreceptor co-activator 1 and 3). These co-activator–PXR com-plexes possess intrinsic histone acetyltransferase activityresulting into chromatin rearrangement and, in turn, modifythe transcriptional activity of ligand bound PXR complex.The complex then combines with its heterodimer partnerRetinoid X Receptor-Alpha (RXRα), and heterodimeriza-tion acts on the DNA to produce mRNA for the transcrip-tional activation of the target genes for cytochrome P450enzymes (CYPs) and ATP binding cassette protein (ABC)efflux transporters, resulting in the over-expression of Pgpand other transporters (Fig. 4) [6,24].

Pregnane X Receptor and ABC transporters inAlzheimer’s disease

Over the last decade, researchers have been trying to estab-lish the role of ABC transporters in the pathophysiology ofAD and its complications [25]. ABC transporters are thesuperfamily of efflux transporters which utilize ATP to trans-port their substrates across the cell membrane and the BBB.ABC efflux transporters cover a wide variety of endoge-nous and exogenous substances as their substrates and areinvolved in the clearance of the toxic metabolites from differ-ent cells, tissues, and organelles. ABC transporters locatedaround the BBB create a barrier for many xenobiotics andalso clear their toxic metabolites from the brain [25]. Thetype of ABC transporters expressed in the vicinity of theBBB include Pgp (ABCB1/MDR1), BCRP (ABCG2), andvarious MRPs 1–5 (ABCC1-5) and CERP (ABCA1) (Table2) [26,27]. BBB accommodates PXR around blood capillar-ies that are involved in the regulation of the efflux transporters[28]. Efflux transporters that are transcriptionally regulatedby PXR include organic anion-transporting polypeptide iso-form 2 (SLCO1A4), bile salt export pump (ABCB11), andPgp [22,29,30]. The literature reveals that the Aβ is a sub-strate for various ABC transporters [31]. Pathophysiology ofAD is related to weak Pgp expression around BBB,which

123

Page 6: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

900 Mol Divers (2014) 18:895–909

Table 2 Various ABC transporters at blood brain barrier and their characteristics [25,65,66]

Name Synonyms Subfamilyclassification

Direction Location Substrate

Endobiotics Xenobiotics

P-glycoprotein Multidrugresistanceprotein(MDR1)

ABCB1 Basolateral toapical

Apical BBB choroidplexus (sub apical)

Amyloid beta Aβ),steroid hormones,cholesterol,prenylcystein-methylesters, IL-2, IL-4,IFN-γ and toxins

Vinblastine,cyclosporin A,digoxin,antidepressant,antipsychotic,antiepilepticdrugs

Multidrugresistance-associatedprotein

Canalicularmultispecificorganic aniontransportergene (CMOAT)

ABCC1-5 Basolateral Toapical andapical tobasolateral

Luminal and abluminalBBB choroid plexusbasolateral membrane,astrocytes, pericytes,neurons, and microglia

– Estradiolglucuronide

Breast cancerresistanceprotein

Mitoxantroneresistanceproteins(MXRP) andABCP

ABCG2 Basolateral toapical

Luminal BBB astrocytes,pericytes, neurons braincapillary endothelial cells

– Prazosin,mitoxantron

Cholesteroleffluxregulatoryprotein

– ABCA1-2 Basolateral toapical

Choroid plexus, neurons,astrocytes, microglia,oligodendrocytes, andbrain capillary endothelialcells

Amyloid beta (Aβ),cholesterol,phospholipid, highdensity lipoprotein

can be exploited for elimination of Aβ out of the brain bystimulating Pgp expression through PXR.

P-glycoprotein (Pgp) transporter

Pgp efflux transporter is well known among ABC trans-porters for its involvement in multidrug resistance and drug–drug interactions with many drugs. Various in vitro and invivo studies revealed that Pgp around the BBB regulates thetransport of endo-xenobiotics across the encephalon [32,33].Pgp also mediates the removal of toxins and reactive metabo-lites from the brain, since the high expression of Pgp isobserved in the luminal side of the endothelial cells [34].A similar study provided evidence that Pgp is involved in theclearance of Aβ from the brain into blood [35]. In anotherstudy, Pgp deficiency at the BBB increased Aβ deposition inan mdr1a/b double knockdown (Pgp null) mouse model. Fur-thermore, after administration of a Pgp inhibitor, the steeprise in Aβ level was observed in the brain interstitial fluid(ISF) [36]. There is an inverse relationship between Pgp andAβ in terms of expression. This validates the role of Pgp in theturnover of Aβ inside the intracerebral space. A circulatoryvessel with high Pgp expression showed no accumulation ofAβ, whereas Aβ deposition was detected in vessels with lowPgp expression. Recently, it has been observed that the Aβ

down regulates the Pgp expression in the vicinity of the BBB,

which diminishes the Aβ clearance from brain and exaggerateAD [37]. Circulating Aβ tends to decrease Pgp in the capil-laries which induces upregulation of Pgp in the vessels as anegative feedback mechanism to compensate for the loss ofPgp and enhances frequently generating Aβ clearance fromthe brain [38]. Later on, upregulated Pgp was lost resultingin the accumulation of Aβ in arteries, arterioles, capillaries,and vessels leading to CAA and finally into AD [38]. Theexpression of Pgp is regulated by various nuclear receptors,mainly PXR, and a variety of xenobiotics [39]. The PXR ago-nists pregnenolone-16α-carbonitrile (PCN) and dexametha-sone enhance the Pgp expression in the rat brain capillaries[28]. Further, in vitro and in vivo experiments using rifampinand hyperforin in the hPXR transgenic mice model revealedthat elevated Pgp levels in brain capillaries subsequentlytighten the BBB and restrict entry of methadone into the brain[40]. Similar results were obtained in a pig model utilizing pigbrain capillary endothelial cell expressing pig PXR (PgPXR).Rifampin, hyperforin, and PCN were used as PXR agonists.Human PXR ligands, rifampicin and hyperforin, have acti-vated PgPXR at BBB, and subsequently elevated Pgp expres-sion, whereas murine PXR ligand PCN was unable to stimu-late PgPXR [41]. Restoration of BBB Pgp has reduced intrac-erebral Aβ in male transgenic human APP-over-expressingmice using PCN-mediated PXR activation [7]. Collectively,upregulation of Pgp in the vicinity of the BBB can be usedas a novel approach toward the management of AD.

123

Page 7: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

Mol Divers (2014) 18:895–909 901

Table 3 Common PXR inhibitors and Pgp substrate and inhibitors[67–131]

PXR inhibitors Pgp inhibitors PXR inhibitors Pgp substrates

Coumestrol Y Camptothecin Y

Fucoxanthin Y Fluconazole Y

Itraconazole Y Itraconazole Y

Isosilybin Y Ketoconazole Y

Ketoconazole Y Leflunamide Y

Sesamin Y Stigmasterol Y

Stigmasterol Y

Y yes

PXR and Pgp: a cross-talk within similarities

Inducers/activators of PXR are increasingly investigated forcontrolling AD [7]. A plethora of data is published reportingnovel PXR activators, accountable for the regulation of Aβ.The accumulation of Aβ leads to inflammation at the BBB,which results in the worsening of AD. The activation of PXRis responsible for the induction of the transcription factor(TF), which in turn regulates various transporters, mainlyPgp. This suggests that PXR activators have a positive feed-back on expression of ABC transporters. Here, we reviewedmost of the potential PXR activators and their possible impacton expression of Pgp. Literature suggest that PXR activa-tors manifest favorable effect whereas, PXR inhibitors haveshown inverse effect on the AD. It has been found that theblockade of Pgp results in the increased level of the Aβ, fur-ther increasing the chances of inflammation in the brain, andultimately aggravates the AD condition. Tables 3 and 4 enlistthe drugs that act as activators and inhibitors of PXR. Thecritical review of activators and inhibitors of PXR (enlistedin Tables 3, 4) noticed that PXR inhibitors/activators may actas Pgp substrates/inhibitors.

Structural similarity between PXR and Pgp: in silicoinsights

Herein, we reviewed the structural details of PXR and Pgp fortheir structural similarities, particularly drug binding site(s).Both PXR and Pgp show substrate promiscuity, binding to adiverse class of chemically unrelated drugs. PXR has shownbinding promiscuity with substrates/activators as small asphenobarbital (232 Da) and as large as rifampicin (823 Da)and taxol (854 Da) [42]. Similarly, Pgp functions as anATP-driven efflux pump for substrates ranging from approx-imately 300 to 4,000 Da in mass, for example it allowsin a small molecule such as mitomycin (molecular weight334 Da) and a large drug molecule such as cyclosporine(molecular weight 1,202 Da) [43–45]. This suggests that both

Table 4 Common PXR activators and Pgp substrate and inhibitors[67–131]

PXRactivators

Pgpsubstrate

PXRactivators

Pgpinhibitor

St. John wart Y St. John wart Y

Rifampin Y Praeruptorin D Y

Paclitaxel Y 1-Piperoylpiperidine Y

Amprenavir Y Hyperforin Y

Carbamazepine Y Erlotinib Y

Corticosterone Y Clotrimazol Y

Dexamethasone Y Indomethacin Y

Phenobarbital Y Nifedipine Y

Vincristine Y Tamoxifen Y

Clotrimazole Y Schisandrin A Y

Verapamil Y Lovastatin Y

Lovastatin Y Mifepristone Y

Bisphenol A Y Pregnanedione Y

Cerivastatin Y 4-Hydroxtamoxifen Y

Clindamycin Y Hyperforin Y

Dicloxacillin Y Isradipine Y

Docetaxel Y Miconazole Y

Efavirenz Y Nicardipine Y

Endosulfan Y Progesterone Y

Erlotinib Y 17-OH-progesterone Y

Fluvastatin Y Santonin Y

Fenvalerate Y Crypterone acetate Y

Dehydroepiandrosterone Y DDT Y

17β-Estradiol Y Verapamil Y

Vitamin K2 Y Nicardipine Y

Atorvastatin Y Reserpin Y

Chlorpyrifos Y Atorvastatin Y

Warfarin Y Dihydrotestosterone Y

Cryptotanshinone Y Pentachlorophenol Y

Cypermethrin Y Zolpidem N

Y yes, N no

PXR and Pgp have wide range of substrate binding affinityand get activated by wide range of drug molecules.

hPXR has been extensively studied using X-ray crys-tallography. Eleven hPXR crystallographic structures areavailable in the Protein Data Bank (PDB). These structuresare available in different forms: apo form (without ligand)(PDB-ID: 1ILG) [46], in complex with co-crystallized lig-ands SR12813 (PDB-ID: 1ILH) [46] and hyperforin (PDB-ID: 1M13) [47], and in complex with SRC-1 peptide alongwith co-crystallized SR12813 (PDB-ID: 1NRL) [48]. Thecanonical structure of hPXR ligand binding domain (LBD)consists of seven α-helices, arranged in three layers sand-wiching the LBD. The three layers are arranged in asequence α 1, α 3, α 4, α 5, α8, and α 7, α10. Unlike othernuclear receptors, hPXR has five extended stranded antipar-

123

Page 8: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

902 Mol Divers (2014) 18:895–909

Fig. 5 Canonical structure of hPXR ligand-binding domain (LBD) andits structural topology showing arrangement of alpha helixes, beta sheet,and loops. (Color figure Online)

allel β-sheets (β 1, β 2, β 3, β 4, and β 1′); β1 and β 1′ that areunique to human PXR (Fig. 5) [42].

In contrast, no human Pgp crystal structure is yet avail-able. The crystallographic structures of Pgp available inPDB are those from different organisms with low reso-lutions. Sav1866 (Staphylococcus aureus transporter) andMsbA Pgp crystallographic structures were the initial struc-tures reported with better resolution compared to other avail-able structures [49,50]. The MsbA crystallographic structurewas reported in outward-open conformation. The Sav1866crystallographic structure was an outward-facing conforma-tion of Pgp obtained in the presence of ATP with NBDs inclose proximity. Other significant crystal structures of Pgpwere published by Aller et. al. include PDB IDs: 3G5U,3G60 and 3G61 [51]. Out of these crystallographic struc-

tures, 3G5U was reported with a resolution of 3.80 Å and87 % sequence identity with human Pgp. However, certainregistered errors in the TM3, TM4, and TM5 regions werereported in the murine Pgp crystallographic structure in therecently published C. elegans Pgp crystal structure [52].

Pgp is a trans-membrane single polypeptide that is struc-turally composed of two homologous parts; each homologcontains six trans-membrane (TM) segments followed bya consensus nucleotide-binding domain (NBD). The twohomologous parts are separated by an intracellular linkerregion of about 60 amino acid residues. Figure 6 shows thehomology model of human Pgp generated using C. eleganscrystal structure as template in our previous study [53]. TheTM segments of Pgp are the putative site for substrate recog-nition and the NBDs are the site for ATP binding/hydrolysis.

Although PXR and Pgp have different structures, theyshare some structural similarities in the LBD. The drug bind-ing domains of both the proteins are highly hydrophobicand share many substrates in common (Tables 3, 4). Thehydrophobic cavity volume in PXR ranges from 1,294 to1,544 Å3, depending on the substrate or co-activator bindingto PXR [54–56]. A cavity volume of 1,294 Å3 is reported inapo crystallographic structure [53]. PXR crystal structure incomplex with the cholesterol lowering compound SR12813showed the hydrophobic cavity volume of 1,280 Å3 [53]. Acavity volume of 1,344 Å3 was observed in PXR structure incomplex with SR12813 and SCR-1 co-activator [54], and acavity volume of 1,544 Å3 in co-crystallized structure withhyperforin [55]. This clearly indicates that the PXR LBDis highly flexible to accommodate compounds of varyingsize and molecular mass. However, the cavity volume of Pgp

Fig. 6 In house developed homology model of human P-gp and arrangement of TM segments. (Color figure Online)

123

Page 9: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

Mol Divers (2014) 18:895–909 903

Fig. 7 Electrostatic surfacepotential representation of PXRstructure. a Two possiblesubstrate gates (encircled)noticed in PXR crystal structureand the Leu residue controllingthe access of substrate throughthese gates. b The three loopsgoverning access to thehydrophobic cavity, substrateaccessibility depends on theflexibility of residues present onthese loops, Leu209 occupiesthe central position and isbelieved to be gating thesolvent-accessible channel.(Color figure Online)

varies dramatically during the course of transport cycle. Pgpcan accommodate two or more compounds simultaneouslywithin the internal cavity due to its large volume of ∼6,000Å3

[51,57]. The volume of this internal cavity is reduced gradu-ally as NBDs approach each other, ultimately reverting whenNBD dimerizes, exposing the drug binding region to extra-cellular space.

Access to the hydrophobic cavity is gated by a hydropho-bic channel in both PXR and Pgp. In PXR, the access tothe hydrophobic cavity is governed by loops joining α2 toβ1, α3 to β 1′, and α6 to β4. The access to the hydrophobiccavity depends on the flexibility of the residues present onthese loops, and in particular on the restructuring of α2 helixthat opens a solvent-accessible channel gated by Leu209(Fig. 7). This channel is over 9 Å in length and stretchesalong the loops joining α3 to β 1′ and α2 to β1, and is solvent-accessible, 3 Å wide [55].

However, in Pgp the access to the drug binding region israther bewildering. Three models for Pgp substrate transloca-tion are suggested: (1) pore, (2) flippase, and (3) hydrophobicvacuum cleaner [58,59]. The hydrophobic vacuum cleanermodel is most acceptable around scientific community, asrecent Pgp crystal structures show the presence of hydropho-bic portals in the TMD region nexus lipid bilayer. Aller etal. reported two portals that allow access to the entry ofhydrophobic molecules directly from the membrane [51].The portals are formed by TMs 4/6 and 10/12 (Fig. 8). Theportals formed by intertwining these TM helices are ∼9 Åwide at the widest point.

Figure 9 shows an electrostatic potential surface compar-ison of PXR and Pgp labeling the residues essential for sub-strate binding. The SR12813 ligand mostly interacts throughhydrophobic interactions at the active site of PXR, forminghydrophobic contacts with side chain of 11 amino acids liningthe PXR ligand-binding cavity (Fig. 9). Further, π -stackinginteraction between the aromatic indole ring of Trp299 and

O11 hydroxyl oxygen atom of SR12813, and H-bondinginteractions with Ser247 and His407 were also observed. Itis important to note that these interactions are not observedin the previous structure [42,54].

These structural similarities between PXR and Pgp alongwith the abundance of common ligands manifest beneficialeffect in the AD pathophysiology with respect to the effluxof intracerebral Aβ, a major cause of AD and its associatedneuroinflammation. Structural similarities mentioned abovereveal the reason behind the cross-talk between PXR and Pgpligands since both share hydrophobic binding site and suffi-cient degree of structural similarity resulting in the large num-ber of common ligands able to bind and activate both PXRand Pgp simultaneously. Further, PXR activation results inthe up-regulation of Pgp along with the BBB, which results ina synergistic effect in the efflux of intracerebral Aβ plaques.Exploiting this connexion between PXR and Pgp may proveeffective in AD management.

Role of PXR in neuroinflammation of Alzheimer’sdisease

Inflammation is the central component of AD pathophys-iology, which increases the neurodegeneration and deathof neuronal cells. Neuroinflammation is the successiveeffect of accumulation of a variety of substances in theperiphery of neurons, which includes Aβ neuritic plaques,tau-neurofibrillary tangles, neurofilament, oligodendrocytemyelin glycoprotein, extracellularly exposed DNA, andbyproduct of damaged neurons [60]. Deposition of Aβ andother proteins, such as tau and intraneuronal neurofibrillarytangles, results in increased expression of microglia and othermacrophages. Accumulation of microglia and macrophageswith Aβ and tau triggers various complementary and alter-native signaling cascades resulting in the generation of

123

Page 10: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

904 Mol Divers (2014) 18:895–909

Fig. 8 Electrostatic surfacepotential representation of P-gpstructure showing thehydrophobic gates noticed inP-gp structure. These gates arelocated in the membraneembedded region of P-gp andformed by TMs 4/6 and 10/12.(Color figure Online)

Fig. 9 Electrostatic surface potential representation of substrate bind-ing cavity of P-gp and PXR showing the arrangement of hydrophobicresidues in stick representation. a Hydrophobic substrate binding cavityof PXR showing the bound co-crystallized structure (orange) and thearrangement of hydrophobic residue that binds the substrates through

hydrophobic interactions. b, c Vertical sections of P-gp structure show-ing hydrophobic substrate binding region in P-gp and arrangement ofhydrophobic residues. Hydrophobic cavity of P-gp is relatively verylarge as compared to PXR. (Color figure Online)

pro-inflammatory mediators [61]. In addition to microglia,astrocytes also concentrate around Aβ deposits and neu-ritic plaques, which induce the production of the inflamma-tory regulator, such as NF-κB (nuclear factor-κB) and COX(cyclooxygenase). Furthermore, neurons themselves are the

producer of the neurotoxic milieu by enhancing inflamma-tory proteins. The inflammatory network comprises NF-κB,COX, β2-integrin, cytokines such as IL-1β and IL-6, TNFα,TGF β1, β2, β3 (transforming growth factor - β1, β2, β3), var-ious chemokines α (CXC), β (CC), γ (C), δ (CX3C), α1- ACT

123

Page 11: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

Mol Divers (2014) 18:895–909 905

(α1-antichymotrypsin), α2 -MAC (α2 - macroglobulin), C5b-9 MAC (membrane attack complex), M-CSF (macrophage-colony stimulating factor) prostaglandins, iNOS (induciblenitric oxide synthase), and oxygen-free radicals [62]. Further,chronic inflammation has a negative impact on efflux trans-porters mainly Pgp, drug metabolizing enzymes (DMEs),and their transcriptional regulatory machinery resulting inthe depletion of Pgp and cytochrome-P450 metabolizingenzymes. Collectively, these inflammatory mediators areresponsible for neurodegeneration and neuronal cell death.

Inflammation enhances the neurodegeneration and neu-ronal death. In this aspect, NSAIDS are included in the ADpharmacotherapy. Activated PXR is known to reduce pro-inflammatory mediators in the intestinal bowel syndrome(IBD) by interfering with the NF-κB regulated inflammatorysignals [63]. PXR-mediated neuroprotection was observedin the case of the Neimann Pick C (NPC) disease utilizingallopregnanolone which activates PXR in vivo [64,65]. PXRis involved in the prominent cross-talk with PPAR, LXR, andRXRα which possess substantial anti-inflammatory responsein ligand-activated manner by functionally suppressing NF-κB inflammatory response in various AD experimental mod-els [19].

Hurdles for treating Alzheimer’s disease with PregnaneX Receptor

Various therapies are implicated in the treatment of AD, butnone could give a complete solution for the treatment ofAD due to their own limitations. A newly proposed ther-apy involves targeting the regulatory machinery of Pgp,which improved the AD pathophysiology by clearing Aβ bur-den intracerebrally. Regulatory machinery consists of PXRthat transcriptionally regulates the various DMEs and trans-porters in different parts of the body, prominently liver, intes-tine, and BBB. Recently, a study using a mouse AD modelhas shown that restoration of Pgp in the BBB by activat-ing PXR reduces the Aβ overload [7]. Thus, targeting Pgpvia PXR has a potential role in Aβ clearance from the brainand attenuates Aβ accumulation intracerebrally. This newapproach suggests a new therapeutic strategy in the AD. Fur-thermore, species-specific drug response is observed in thecase of PXR since enough species diversity exists among rat,mouse, rabbit, and human PXR where a drug shows differ-ent responses for PXR activation/deactivation. One drug mayactivate human PXR and no effect on rat PXR, and vice-versa.

Conclusions

Aβ accumulation accompanied with inflammation is one ofthe major contributing factors for AD-associated neurode-

generation and neuronal cell death leading to dementia andimpaired memory. Clearance of Aβ outside the brain emergesout as one of the therapeutic strategies to manage AD. PXR,as a nuclear receptor, shows promising results in transcrip-tional regulation of efflux transporters mainly Pgp at theBBB. Overexpression of the efflux transporters at the BBB ismediated by PXR, which reduces Aβ burden intracerebrally.Additionally, PXR controls ApoE and inflammation offeringa further advantage to its role in the management of AD.Collectively, we propose PXR as an attractive target for ADmanagement.

Acknowledgments This work was supported by the Departmentof Biotechnology (DBT), India.

Conflict of interest The author declares no competing financialinterest.

References

1. Graeber MB, Mehraein P (1999) Reanalysis of the first case ofAlzheimers disease. Eur Arch Psychiatry Clin Neurosci 249:S10–S13. doi:10.1007/PL00014167

2. Thies W, Bleiler L, Alzheimer’s Association (2013) 2013Alzheimer’s disease facts and figures. Alzheimers Dement 9:208–245. doi:10.1016/j.jalz.2013.02.003

3. Detailed Tables for the National Vital Statistics Report. Deaths:Final Data for 2010. National Center for Health Statistics. http://www.cdc.gov/nchs/data/dvs/deaths2010release.pdf

4. Aprahamian I, Stella F, Forlenza OV (2013) New treatment strate-gies for Alzheimer’s disease: is there a hope? Indian J Med Res138:449–460

5. Geerts H, Roberts P, Spiros A, Carr R (2013) A strategy for devel-oping new treatment paradigms for neuropsychiatric and neu-rocognitive symptoms in Alzheimers disease. Front Pharmacol4:1–10. doi:10.3389/fphar.2013.00047

6. Kliewer SA, Goodwin B, Willson TM (2002) The nuclear preg-nane X receptor: a key regulator of xenobiotic metabolism. EndocrRev 23:687–702. doi:10.1210/er.2001-0038

7. Hartz AMS, Miller DS, Bauer B (2010) Restoring blood–brainbarrier P-glycoprotein reduces brain amyloid-β in a mouse modelof Alzheimer’s disease. Mol Pharmacol 77:715–723. doi:10.1124/mol.109.061754

8. Schumacher U, Mollgard K (1997) The multidrug-resistanceP-glycoprotein (Pgp, MDR1) is an early marker of blood–brain barrier development in the microvessels of the developinghuman brain. Histochem Cell Biol 108:179–182. doi:10.1007/s004180050159

9. Braak H, Braak E (1997) Diagnostic criteria for neuropathologicassessment of Alzheimers disease. Neurobiol Aging 18:85–88.doi:10.1016/S0197-4580(97)00062-6

10. Kazee AM, Johnson EM (1998) Alzheimer’s disease pathologyin non-demented elderly. J Alzheimers Dis 1:81–89

11. Selkoe DJ, Wolfe MS (2007) Presenilin: running with scissorsin the membrane. Cell 131:215–221. doi:10.1016/j.cell.2007.10.012

12. D’Andrea MR, Nagele RG, Wang H-Y, Lee DHS (2002)Consistent immunohistochemical detection of intracellular β-amyloid42 in pyramidal neurons of Alzheimer’s diseaseentorhinal cortex. Neurosci Lett 333:163–166. doi:10.1016/S0304-3940(02)00875-3

123

Page 12: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

906 Mol Divers (2014) 18:895–909

13. Arnold SE, Hyman BT, Flory J, Damasio AR, Van Hoesen GW(1991) The topographical and neuroanatomical distribution ofneurofibrillary tangles and neuritic plaques in the cerebral cortexof patients with Alzheimer’s disease. Cereb Cortex 1:103–116.doi:10.1093/cercor/1.1.103

14. Kukar T, Murphy MP, Eriksen JL, Sagi SA, Weggen S, Smith TE,Ladd T, Khan MA, Kache R, Beard J (2005) Diverse compoundsmimic Alzheimer disease causing mutations by augmenting Aβ42production. Nat Med 11:545–550. doi:10.1038/nm1235

15. Hecimovic S, Wang J, Dolios G, Martinez M, Wang R, GoateAM (2004) Mutations in APP have independent effects on Aβ

and CTFγ generation. Neurobiol Dis 17:205–218. doi:10.1016/j.nbd.2004.04.018

16. Wakutani Y, Watanabe K, Adachi Y, Wada-Isoe K, Urakami K,Ninomiya H, Saido TC, Hashimoto T, Iwatsubo T, Nakashima K(2004) Novel amyloid precursor protein gene missense mutation(D678N) in probable familial Alzheimers disease. J Neurol Neu-rosurg Psychiatry 75:1039–1042. doi:10.1136/jnnp.2003.010611

17. Itagaki S, McGeer PL, Akiyama H, Zhu S, Selkoe D (1989)Relationship of microglia and astrocytes to amyloid deposits ofAlzheimer disease. J Neuroimmunol 24:173–182. doi:10.1016/0165-5728(89)90115-X

18. Meda L, Cassatella MA, Szendrei GI, Otvos L, Baron P, VillalbaM, Ferrari D, Rossi F (1995) Activation of microglial cells by β-amyloid protein and interferon-γ . Nature 374:647–650. doi:10.1038/374647a0

19. di Masi A, Marinis ED, Ascenzi P, Marino M (2009) Nuclearreceptors CAR and PXR: molecular, functional, and biomedicalaspects. Mol Asp Med 30:297–343. doi:10.1016/j.mam.2009.04.002

20. Rathod V, Jain S, Nandekar P, Sangamwar AT (2014) Humanpregnane X receptor: a novel target for anticancer drug develop-ment. Drug Discov Today 19:63–70. doi:10.1016/j.drudis.2013.08.009

21. Moore LB, Maglich JM, McKee DD, Wisely B, Willson TM,Kliewer SA, Lambert MH, Moore JT (2002) Pregnane X recep-tor (PXR), constitutive androstane receptor (CAR), and ben-zoate X receptor (BXR) define three pharmacologically dis-tinct classes of nuclear receptors. Mol Endocrinol 16:977–986

22. Kliewer SA, Moore JT, Wade L, Staudinger JL, WatsonMA, Jones SA, McKee DD, Oliver BB, Willson TM (1998)An orphan nuclear receptor activated by pregnanes defines anovel steroid signaling pathway. Cell 92:73–82. doi:10.1016/S0092-8674(00)80900-9

23. Kliewer SA, Goodwin B, Willson TM (2002) The nuclear preg-nane X receptor: a key regulator of xenobiotic metabolism. EndocrRev 23:687–702. doi:10.1210/er.2001-0038

24. Pondugula SR, Mani S (2013) Pregnane xenobiotic receptor incancer pathogenesis and therapeutic response. Cancer Lett 328:1–9. doi:10.1016/j.canlet.2012.08.030

25. Begley DJ (2004) ABC transporters and the blood–brain barrier.Curr Pharm Des 10:1295–1312. doi:10.2174/1381612043384844

26. Sun H, Dai H, Shaik N, Elmquist WF (2003) Drug efflux trans-porters in the CNS. Adv Drug Deliv Rev 55:83–105. doi:10.1016/S0169-409X(02)00172-2

27. Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Beg-ley DJ (2010) Structure and function of the blood brain barrier.Neurobiol Dis 37:13–25. doi:10.1016/j.nbd.2009.07.030

28. Bauer Br, Hartz AMS, Fricker G, Miller DS (2004) Pregnane Xreceptor up-regulation of P-glycoprotein expression and transportfunction at the blood–brain barrier. Mol Pharmacol 66:413–419

29. Geick A, Eichelbaum M, Burk O (2001) Nuclear recep-tor response elements mediate induction of intestinal MDR1by rifampin. J Biol Chem 276:14581–14587. doi:10.1074/jbc.M010173200

30. Lehmann JrM, McKee DD, Watson MA, Willson TM, Moore JT,Kliewer SA (1998) The human orphan nuclear receptor PXR isactivated by compounds that regulate CYP3A4 gene expressionand cause drug interactions. J Clin Invest 102:1016–1023. doi:10.1172/JCI3703

31. Vogelgesang S, Cascorbi I, Schroeder E, Pahnke J, Kroemer HK,Siegmund W, Kunert-Keil C, Walker LC, Warzok RW (2002)Deposition of Alzheimer’s [beta]-amyloid is inversely correlatedwith P-glycoprotein expression in the brains of elderly non-demented humans. Pharmacogenet Genomics 12:535–541

32. Cordon-Cardo C, O’Brien JP, Casals D, Rittman-Grauer L,Biedler JL, Melamed MR, Bertino JR (1989) Multidrug-resistancegene (P-glycoprotein) is expressed by endothelial cells at blood–brain barrier sites. Proc Natl Acad Sci USA 86:695–698

33. Potschka H, Fedrowitz M, Loscher W (2002) P-Glycoprotein-mediated efflux of phenobarbital, lamotrigine, and felba-mate at the blood brain barrier: evidence from microdialysisexperiments in rats. Neurosci Lett 327:173–176. doi:10.1016/S0304-3940(02)00423-8

34. Bendayan R, Ronaldson PT, Gingras D, Bendayan M (2006) Insitu localization of P-glycoprotein (ABCB1) in human and ratbrain. J Histochem Cytochem 54:1159–1167. doi:10.1369/jhc.5A6870.2006

35. van Assema Lubberink M, Bauer M, van der Flier WM, Schuit RC,Windhorst AD, Comans EFI, Hoetjes NJ, Tolboom N, Langer O(2012) Blood brain barrier P-glycoprotein function in Alzheimer’sdisease. Brain 135:181–189. doi:10.1093/brain/awr298

36. Cirrito JR, Deane R, Fagan AM, Spinner ML, Parsadanian M, FinnMB, Jiang H, Prior JL, Sagare A, Bales KR (2005) P-glycoproteindeficiency at the blood–brain barrier increases amyloid-β depo-sition in an Alzheimer disease mouse model. J Clin Invest115:3285–3290. doi:10.1172/JCI25247

37. Brenn A, Grube M, Peters M, Fischer A, Jedlitschky G, KroemerHK, Warzok RW, Vogelgesang S (2011) Beta-amyloid downreg-ulates MDR1-P-glycoprotein (Abcb1) expression at the blood–brain barrier in mice. Int J Alzheimer’s Dis 2011:1–6. doi:10.4061/2011/690121

38. Vogelgesang S, Warzok RW, Cascorbi I, Kunert-Keil C, SchroederE, Kroemer HK, Siegmund W, Walker LC, Pahnke J (2004) Therole of P-glycoprotein in cerebral amyloid angiopathy; impli-cations for the early pathogenesis of Alzheimers disease. CurrAlzheimer Res 1:121–125. doi:10.2174/1567205043332225

39. Breier A, Gibalova L, Seres M, Barancik M, Sulova Z (2013) NewInsight into P-glycoprotein as a drug target. Anti-Cancer AgentMed Chem 13:159–170. doi:10.2174/187152013804487380

40. Bauer Br, Yang X, Hartz AMS, Olson ER, Zhao R, Kalvass JC,Pollack GM, Miller DS (2006) In vivo activation of human preg-nane X receptor tightens the blood–brain barrier to methadonethrough P-glycoprotein up-regulation. Mol Pharmacol 70:1212–1219. doi:10.1124/mol.106.023796

41. Ott M, Fricker G, Bauer Br (2009) Pregnane X receptor (PXR) reg-ulates P-glycoprotein at the blood–brain barrier: functional sim-ilarities between pig and human PXR. J Pharmacol Exp Ther329:141–149. doi:10.1124/jpet.108.149690

42. Watkins RE, Davis-Searles PR, Lambert MH, Redinbo MR (2003)Coactivator binding promotes the specific interaction between lig-and and the pregnane X receptor. J Mol Biol 331:815–828. doi:10.1016/S0022-2836(03)00795-2

43. Synold TW, Dussault I, Forman BM (2001) The orphan nuclearreceptor SXR coordinately regulates drug metabolism and efflux.Nat Med 7:584–590. doi:10.1038/87912

44. Jones SA, Moore LB, Shenk JL, Wisely GB, Hamilton GA,McKee DD, Tomkinson NCO, LeCluyse EL, Lambert MH, Will-son TM (2000) The pregnane X receptor: a promiscuous xenobi-otic receptor that has diverged during evolution. Mol Endocrinol14:27–39. doi:10.4155/fmc.10.276

123

Page 13: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

Mol Divers (2014) 18:895–909 907

45. Tarcsay Á, Keseru GM (2011) Homology modeling and bindingsite assessment of the human P-glycoprotein. Future Med Chem3:297–307. doi:10.4155/fmc.10.276

46. Watkins RE, Wisely GB, Moore LB, Collins JL, Lambert MH,Williams SP, Redinbo MR (2001) The human nuclear xenobioticreceptor PXR: structural determinants of directed promiscuity.Science 292:2329–2333. doi:10.1126/science.1060762

47. Watkins RE, Maglich JM, Moore LB, Wisely GB, Noble SM,Davis-Searles PR, Redinbo MR (2003) 2.1 Å crystal structureof human PXR in complex with the St. John’s wort compoundhyperforin. Biochemistry 42:1430–1438. doi:10.1021/bi0268753

48. Watkins RE, Davis-Searles PR, Lambert MH, Redinbo MR (2003)Coactivator binding promotes the specific interaction between lig-and and the pregnane X receptor. J Mol Biol 331:815–828. doi:10.1016/S0022-2836(03)00795-2

49. Chang G, Roth CB (2001) Structure of MsbA from E. coli: ahomolog of the multidrug resistance ATP binding cassette (ABC)transporters. Science 293:1793–1800. doi:10.1126/science.293.5536.1793

50. Dawson RJP, Locher KP (2006) Structure of a bacterial multidrugABC transporter. Nature 443:180–185. doi:10.1038/nature05155

51. Aller SG, Yu J, Ward A, Weng Y, Chittaboina S, Zhuo R, HarrellPM, Trinh YT, Zhang Q, Urbatsch IL, Chang G (2009) Structureof P-glycoprotein reveals a molecular basis for poly-specific drugbinding. Science 323:1718–1722. doi:10.1126/science.1168750

52. Jin MS, Oldham ML, Zhang Q, Chen J (2012) Crystal structureof the multidrug transporter P-glycoprotein from Caenorhabditiselegans. Nature 490:566–569. doi:10.1038/nature11448

53. Prajapati R, Singh U, Patil A, Khomane KS, Bagul P, BansalAK, Sangamwar AT (2013) In silico model for P-glycoproteinsubstrate prediction: insights from molecular dynamics and invitro studies. J Comput Aided Mol Des 27:347–363. doi:10.1007/s10822-013-9650-x

54. Watkins RE, Davis-Searles PR, Lambert MH, Redinbo MR (2003)Coactivator binding promotes the specific interaction between lig-and and the pregnane X receptor. J Mol Biol 331:815–828. doi:10.1016/S0022-2836(03)00795-2

55. Watkins RE, Maglich JM, Moore LB, Wisely GB, Noble SM,Davis-Searles PR, Lambert MH, Kliewer SA, Redinbo MR (2003)2.1 Å crystal structure of human PXR in complex with the St.John’s wort compound hyperforin. Biochemistry 42:1430–1438.doi:10.1021/bi0268753

56. Watkins RE, Wisely GB, Moore LB, Collins JL, Lambert MH,Williams SP, Willson TM, Kliewer SA, Redinbo MR (2001) Thehuman nuclear xenobiotic receptor PXR: structural determinantsof directed promiscuity. Science 292:2329–2333. doi:10.1126/science.1060762

57. Loo TW, Bartlett MC, Clarke DM (2003) Substrate-induced con-formational changes in the transmembrane segments of humanP-glycoprotein. J Biol Chem 278:13603–13606. doi:10.1074/jbc.C300073200

58. Callaghan R, Ford RC, Kerr ID (2006) The translocation mecha-nism of P-glycoprotein. FEBS Lett 580:1056–1063. doi:10.1016/j.febslet.2005.11.083

59. Constantinides PP, Wasan KM (2007) Lipid formulation strate-gies for enhancing intestinal transport and absorption of P-glycoprotein (Pgp) substrate drugs: in vitro/in vivo case studies.J Pharm Sci 96:235–248. doi:10.1002/jps.20780

60. Aisen PS (1996) Inflammation and Alzheimer disease. Mol ChemNeuropathol 28:83–88. doi:10.1007/BF02815208

61. Leung R, Proitsi P, Simmons A, Lunnon K, Gntert A, Kronen-berg D, Pritchard M, Tsolaki M, Mecocci P, Kloszewska I (2013)Inflammatory proteins in plasma are associated with severity ofAlzheimers disease. PLoS One 8:649–671. doi:10.1371/journal.pone.0064971

62. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM,Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL (2000)Inflammation and Alzheimers disease. Neurobiol Aging 21:383–421. doi:10.1016/S0197-4580(00)00124-X

63. Shah YM, Ma X, Morimura K, Kim I, Gonzalez FJ (2007) Preg-nane X receptor activation ameliorates DSS-induced inflamma-tory bowel disease via inhibition of NF-kB target gene expres-sion. Am J Physiol Gastrointest Liver Physiol 292:G1114–G1122.doi:10.1152/ajpgi.00528.2006

64. Langmade SJ, Gale SE, Frolov A, Mohri I, Suzuki K, Mellon SH,Walkley SU, Covey DF, Schaffer JE, Ory DS (2006) PregnaneX receptor (PXR) activation: a mechanism for neuroprotection ina mouse model of Niemann–Pick C disease. Proc Natl Acad SciUSA 103:13807–13812. doi:10.1073/pnas.0606218103

65. Wolf A, Bauer B, Hartz AMS (2012) ABC transporters and theAlzheimer’s disease enigma. Front Psychiatry 3:1–14. doi:10.3389/fpsyt.2012.00054

66. Löscher W, Potschka H (2005) Drug resistance in brain diseasesand the role of drug efflux transporters. Nat Rev Neurosci 6:591–602. doi:10.1038/nrn1728

67. Enokizono J, Kusuhara H, Sugiyama Y (2007) Effect of breastcancer resistance protein (Bcrp/Abcg2) on the disposition of phy-toestrogens. Mol Pharmacol 72:967–975. doi:10.1124/mol.107.034751

68. Sharom FJ (2008) ABC multidrug transporters: structure, func-tion and role in chemoresistance. Pharmacogenomics 9:105–127.doi:10.2217/14622416.9.1.105

69. Eid SY, El-Readi MZ, Wink M (2012) arotenoids reverse mul-tidrug resistance in cancer cells by interfering with ABC-transporters. Phytomedicine 19:977–987. doi:10.1016/j.phymed.2012.05.010

70. E-j Wang, Lew K, Casciano CN, Clement RP, Johnson WW (2002)Interaction of common azole antifungals with P glycoprotein.Antimicrob Agents Chem 46:160–165. doi:10.1128/AAC.46.1.160-165.2002

71. Maitrejean M, Comte G, Barron D, El Kirat K, Di PietroA (2000) The flavanolignan silybin and its hemisyntheticderivatives, a novel series of potential modulators of P-glycoprotein. Bioorg Med Chem Lett 10:157–160. doi:10.1016/S0960-894X(99)00636-8

72. Kis E, Nagy T, Jani M, Molnár E, Jánossy J, Ujhellyi O, Német K,Herédi-Szabó K, Krajcsi P (2009) Leflunomide and its metaboliteA771726 are high affinity substrates of BCRP: implications fordrug resistance. Ann Rheum Dis 68:1201–1207. doi:10.1136/ard.2007.086264

73. Nabekura T, Yamaki T, Ueno K, Kitagawa S (2008) Inhibition ofP-glycoprotein and multidrug resistance protein 1 by dietary phy-tochemicals. Cancer Chemother Pharmacol 62:867–873. doi:10.1007/s00280-007-0676-4

74. El-Readi MZ, Hamdan D, Farrag N, El-Shazly A, Wink M (2010)Inhibition of P-glycoprotein activity by limonin and other sec-ondary metabolites from Citrus species in human colon andleukaemia cell lines. Eur J Pharmacol 626:139–145. doi:10.1016/j.ejphar.2009.09.040

75. von Moltke LL, Weemhoff JL, Perloff MD, Hesse LM, HarmatzJS, Roth-Schechter BF, Greenblatt DJ (2002) Effect of zolpidemon human cytochrome P450 activity, and on transport mediatedby P-glycoprotein. Biopharm Drug Dispos 23:361–367. doi:10.1002/bdd.329

76. Hennessy M, Kelleher D, Spiers JP, Barry M, Kavanagh P, BackD, Mulcahy F, Feely J (2002) St John’s Wort increases expressionof P-glycoprotein: implications for drug interactions. Br J ClinPharmacol 53:75–82. doi:10.1046/j.0306-5251.2001.01516.x

77. Perloff MD, Von Moltke LL, Stormer E, Shader RI, Greenblatt DJ(2001) Saint John’s wort: an in vitro analysis of P-glycoprotein

123

Page 14: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

908 Mol Divers (2014) 18:895–909

induction due to extended exposure. Br J Pharmacol 134:1601–1608. doi:10.1038/sj.bjp.0704399

78. Ott M, Huls M, Cornelius M, Fricker G (2010) St. John’swort constituents modulate P-glycoprotein transport activity atthe blood–brain barrier. Pharm Res 27:811–822. doi:10.1007/s11095-010-0074-1

79. Fromm MF (2004) Importance of P-glycoprotein at blood–tissuebarriers. Trends Pharmacol Sci 25:423–429. doi:10.1016/j.tips.2004.06.002

80. Shen X, Chen G, Zhu G, Fong W-F (2006) (±)-3′-O,4′-O-dicynnamoyl-cis-khellactone, a derivative of (±)-praeruptorin A,reverses P-glycoprotein mediated multidrug resistance in cancercells. Bioorg Med Chem Lett 14:7138–7145. doi:10.1016/j.bmc.2006.06.066

81. Sparreboom A, Van Asperen J, Mayer U, Schinkel AH, Smit JW,Meijer DKF, Borst P, Nooijen WJ, Beijnen JH, Van Tellingen O(1997) Limited oral bioavailability and active epithelial excretionof paclitaxel (Taxol) caused by P-glycoprotein in the intestine.Proc Natl Acad Sci India A/B 94:2031–2035

82. Bhardwaj RK, Glaeser H, Becquemont L, Klotz U, Gupta SK,Fromm MF (2002) Piperine, a major constituent of black pepper,inhibits human P-glycoprotein and CYP3A4. J Pharmacol ExpTher 302:645–650. doi:10.1124/jpet.102.034728

83. Polli JW, Jarrett JL, Studenberg SD, Humphreys JE, DennisSW, Brouwer KR, Woolley JL (1999) Role of P-glycoproteinon the CNS disposition of amprenavir (141W94), an HIVprotease inhibitor. Pharm Res 16:1206–1212. doi:10.1023/A:1018941328702

84. Wang E-j, Barecki-Roach M, Johnson WW (2004) Quantitativecharacterization of direct P-glycoprotein inhibition by St John’swort constituents hypericin and hyperforin. J Pharm Pharmacol56:123–128. doi:10.1211/0022357022395

85. Potschka H, Fedrowitz M, Loscher W (2001) P-glycoproteinand multidrug resistance-associated protein are involved inthe regulation of extracellular levels of the major antiepilep-tic drug carbamazepine in the brain. Neuroreport 12:3557–3560

86. Noguchi K, Kawahara H, Kaji A, Katayama K, Mitsuhashi J,Sugimoto Y (2009) Substrate-dependent bidirectional modulationof P-glycoprotein-mediated drug resistance by erlotinib. CancerSci 100:1701–1707. doi:10.1111/j.1349-7006.2009.01213.x

87. Ueda K, Okamura N, Hirai M, Tanigawara Y, Saeki T, KiokaN, Komano T, Hori R (1992) Human P-glycoprotein transportscortisol, aldosterone, and dexamethasone, but not progesterone. JBiol Chem 267:24248–24252

88. Yasuda K, Lan L-b, Sanglard D, Furuya K, Schuetz JD, SchuetzEG (2002) Interaction of cytochrome P450 3A inhibitors with P-glycoprotein. J Pharmacol Exp Ther 303:323–332. doi:10.1124/jpet.102.037549

89. Perloff MD, Stormer E, von Moltke LL, Greenblatt DJ (2003)Rapid assessment of P-glycoprotein inhibition and induction invitro. Pharm Res 20:1177–1183. doi:10.1023/A:1025092829696

90. Potschka H, Fedrowitz M, Loscher W (2002) P-glycoprotein-mediated efflux of phenobarbital, lamotrigine, and felba-mate at the blood–barrier: evidence from microdialysis exper-iments in rats. Neurosci Lett 327:173–176. doi:10.1016/S0304-3940(02)00423-8

91. Robert J, Jarry C (2003) Multidrug resistance reversal agents. JMed Chem 46:4805–4817. doi:10.1021/jm030183a

92. Relling MV (1996) Are the major effects of P-glycoprotein mod-ulators due to altered pharmacokinetics of anticancer drugs? TherDrug Monit 18:350–356

93. Wasserman L, Aviram R, Levavi H, Ovadia J, Shneyuor Y, FrischA, Blau O, Beery E, Novogrodsky A, Nordenberg J (1992) Acell line with unusual characteristics from an ovarian carcinoma

patient: modulation of sensitivity to antitumour drugs. Eur J Can-cer 28:22–27. doi:10.1016/0959-8049(92)90376-D

94. Schuetz EG, Beck WT, Schuetz JD (1996) Modulators and sub-strates of P-glycoprotein and cytochrome P4503A coordinatelyup-regulate these proteins in human colon carcinoma cells. MolPharmacol 49:311–318

95. Xin HW, Wu XC, Li Q, Yu AR, Zhu M, Shen Y, Su D, Xiong L(2007) Effects of Schisandra sphenanthera extract on the phar-macokinetics of tacrolimus in healthy volunteers. Br J Clin Phar-macol 64:469–475. doi:10.1111/j.1365-2125.2007.02922.x

96. Kim RB (2002) Drugs as P-glycoprotein substrates, inhibitors,and inducers. Drug Metab Rev 34:47–54. doi:10.1081/DMR-120001389

97. Dimitroulakos J, Yeger H (1996) HMG-CoA reductase mediatesthe biological effects of retinoic acid on human neuroblastomacells: lovastatin specifically targets P-glycoprotein-expressingcells. Nat Med 2:326–333. doi:10.1038/nm0396-326

98. Li D-Q, Wang Z-B, Bai J, Zhao J, Wang Y, Hu K, Du Y-H (2004)Reversal of multidrug resistance in drug-resistant human gastriccancer cell line SGC7901/VCR by antiprogestin drug mifepris-tone. World J Gastroenterol 10:1722–1725

99. Balakrishnan B, Henare K, Thorstensen EB, Ponnampalam AP,Mitchell MD (2010) Transfer of bisphenol A across the humanplacenta. Am J Obstet Gynacol 202:393–397. doi:10.1016/j.ajog.2010.01.025

100. Ichikawa-Haraguchi M, Sumizawa T, Yoshimura A, Furukawa T,Hiramoto S, Sugita M (1993) Progesterone and its metabolites: thepotent inhibitors of the transporting activity of P-glycoprotein inthe adrenal gland. Biochim Biophys Acta 1158:201–208. doi:10.1016/0304-4165(93)90016-2

101. Kivisto KT, Zukunft J, Hofmann U, Niemi M, Rekersbrink S,Schneider S, Luippold G, Schwab M, Eichelbaum M, Fromm MF(2004) Characterisation of cerivastatin as a P-glycoprotein sub-strate: studies in P-glycoprotein-expressing cell monolayers andmdr1a/b knock-out mice. Naunyn Schmiedebergs Arch Pharma-col 370:124–130. doi:10.1007/s00210-004-0948-z

102. Bekaii-Saab TS, Perloff MD, Weemhoff JL, GreenblattDJ, von Moltke LL (2004) Interactions of tamoxifen,N-desmethyltamoxifen and 4-hydroxytamoxifen with P-glycoprotein and CYP3A. Biopharm Drug Dispos 25:283–289.doi:10.1002/bdd.411

103. Konings WN, Poelarends GJ (2002) Bacterial multidrug resis-tance mediated by a homologue of the human multidrug trans-porter P-glycoprotein. IUBMB Life 53:213–218. doi:10.1080/15216540212646

104. Susanto M, Benet LZ (2002) Can the enhanced renal clear-ance of antibiotics in cystic fibrosis patients be explained by P-glycoprotein transport? Pharm Res 19:457–462. doi:10.1023/A:1015191511817

105. X-f Zhou, Zhang L, Tseng E, Scott-Ramsay E, Schentag JJ,Coburn RA, Morris ME (2005) New 4-aryl-1, 4-dihydropyridinesand 4-arylpyridines as P-glycoprotein inhibitors. Drug Metab Dis-pos 33:321–328. doi:10.1124/dmd.104.00208

106. Wils P, Phung-Ba Vr, Warnery A, Lechardeur D, Raeissi S,Hidalgo IJ, Scherman D (1994) Polarized transport of docetaxeland vinblastine mediated by P-glycoprotein in human intestinalepithelial cell monolayers. Biochem Pharmacol 48:1528–1530.doi:10.1016/0006-2952(94)90580-0

107. Aungst BJ (1999) P-glycoprotein, secretory transport, and otherbarriers to the oral delivery of anti-HIV drugs. Adv Drug DelivRev 39:105–116. doi:10.1016/S0169-409X(99)00022-8

108. Lentz K, Polli J, Wring S, Humphreys J, Polli J (2000) Influence ofpassive permeability on apparent P-glycoprotein kinetics. PharmRes 17:1456–1460. doi:10.1023/a:1007692622216

123

Page 15: Pregnane X Receptor and P-glycoprotein: a connexion for Alzheimer’s disease management

Mol Divers (2014) 18:895–909 909

109. Bain LJ, Leblanc GA (1996) Interaction of structurally diversepesticides with the human MDR1 gene product P-glycoprotein.Toxicol Appl Pharm 141:288–298. doi:10.1006/taap.1996.0286

110. Barnes KM, Dickstein B, Cutler GB, Fojo T, Bates SE(1996) Steroid transport, accumulation, and antagonism of P-glycoprotein in multidrug-resistant cells. Biochemistry 35:4820–4827. doi:10.1021/bi952380k

111. Kodaira H, Kusuhara H, Ushiki J, Fuse E, Sugiyama Y(2010) Kinetic analysis of the cooperation of P-glycoprotein (P-gp/Abcb1) and breast cancer resistance protein (Bcrp/Abcg2) inlimiting the brain and testis penetration of erlotinib, flavopiridol,and mitoxantrone. J Pharmacol Exp Ther 333:788–796. doi:10.1124/jpet.109.162321

112. Holtzman CW, Wiggins BS, Spinler SA (2006) Role ofP-glycoprotein in statin drug interactions. Pharmacotherapy26:1601–1607. doi:10.1592/phco.26.11.1601

113. Najar IA, Sachin BS, Sharma SC, Satti NK, Suri KA, Johri RK(2010) Modulation of P-glycoprotein ATPase activity by somephytoconstituents. Phytother Res 24:454–458. doi:10.1002/ptr.2951

114. Sreeramulu K, Liu R, Sharom FJ (2007) Interaction of insecticideswith mammalian P-glycoprotein and their effect on its transportfunction. Biochim Biophys Acta 1768:1750–1757. doi:10.1016/j.bbamem.2007.04.001

115. Frohlich M, Albermann N, Sauer A, Walter-Sack I, Haefeli WE,Weiss J (2004) In vitro and ex vivo evidence for modulationof P-glycoprotein activity by progestins. Biochem Pharmacol68:2409–2416. doi:10.1016/j.bcp.2004.08.026

116. Shabbir A, DiStasio S, Zhao J, Cardozo CP, Wolff MS, CaplanAJ (2005) Differential effects of the organochlorine pesticideDDT and its metabolite p,p′-DDE on p-glycoprotein activity andexpression. Toxicol Appl Pharm 203:91–98. doi:10.1016/j.taap.2004.07.011

117. Gosland M, Tsuboi C, Hoffman T, Goodin S, Vore M (1993)17β-Estradiol glucuronide: an inducer of cholestasis and a physi-ological substrate for the multidrug resistance transporter. CancerRes 53:5382–5385

118. Giacomini KM, Huang S-M, Tweedie DJ, Benet LZ, BrouwerKLR, Chu X, Dahlin A, Evers R, Fischer V, Hillgren KM (2010)Membrane transporters in drug development. Nat Rev Drug Dis-cov 9:215–236. doi:10.1038/nrd3028

119. Katoh M, Nakajima M, Yamazaki H, Yokoi T (2000)Inhibitory potencies of 1,4-dihydropyridine calcium antago-nists to P-glycoprotein-mediated transport: comparison with theeffects on CYP3A4. Pharm Res 17:1189–1197. doi:10.1023/A:1007568811691

120. Wu X, Whitfield LR, Stewart BH (2000) Atorvastatin transportin the Caco-2 cell model: contributions of P-glycoprotein and theproton-monocarboxylic acid co-transporter. Pharm Res 17:209–215. doi:10.1023/A:1007525616017

121. E-j Wang, Lew K, Barecki M, Casciano CN, Clement RP, John-son WW (2001) Quantitative distinctions of active site molecular

recognition by P-glycoprotein and cytochrome P450 3A4. ChemRes Toxicol 14:1596–1603. doi:10.1021/tx010125x

122. Lanning CL (1996) Chlorpyrifos oxon interacts with the mam-malian multidrug resistance protein, P-glycoprotein. J ToxicolEnviron Health 47:95–407. doi:10.1080/009841096161726

123. Boyd RA, Stern RH, Stewart BH, Wu X, Reyner EL, Zegarac EA,Randinitis EJ, Whitfield L (2000) Atorvastatin coadministrationmay increase digoxin concentrations by inhibition of intestinalP-glycoprotein mediated secretion. J Clin Pharmacol 40:91–98.doi:10.1177/009127000004000112

124. Wadelius M, Sorlin K, Wallerman O, Karlsson J, Yue QY, Mag-nusson PKE, Wadelius C (2003) Warfarin sensitivity related toCYP2C9, CYP3A5, ABCB1 (MDR1) and other factors. Pharma-cogenomics J 4:40–48. doi:10.1038/sj.tpj.6500220

125. Zhang J, Huang M, Guan S, Bi H-C, Pan Y, Duan W, Chan SY,Chen X, Hong Y-H, Bian J-S (2006) A mechanistic study of theintestinal absorption of cryptotanshinone, the major active con-stituent of Salvia miltiorrhiza. J Pharmacol Exp Ther 317:1285–1294. doi:10.1124/jpet.105.10070

126. Buss DS, McCaffery AR, Callaghan A (2002) Evidence for p-glycoprotein modification of insecticide toxicity in mosquitoes ofthe Culex pipiens complex. Med Vet Entomol 16:218–222. doi:10.1046/j.1365-2915.2002.00365.x

127. Eufemia NA, Epel D (2000) Induction of the multixenobi-otic defense mechanism (MXR), P-glycoprotein, in the mus-sel Mytilus californianus as a general cellular response toenvironmental stresses. Aquat Toxicol 49:89–100. doi:10.1016/S0166-445X(99)00068-5

128. Fedoruk MN, Gimenez-Bonafe P, Guns ES, Mayer LD, NelsonCC (2004) P-glycoprotein increases the efflux of the androgendihydrotestosterone and reduces androgen responsive gene activ-ity in prostate tumor cells. Prostate 59:77–90. doi:10.1002/pros.10354

129. Wang EJ, Casciano CN, Clement RP, Johnson WW (2001)HMG-CoA reductase inhibitors (statins) characterized as directinhibitors of P-glycoprotein. Pharm Res 18:800–806. doi:10.1023/A:1011036428972

130. Takano M, Hasegawa R, Fukuda T, Yumoto R, Nagai J, MurakamiT (1998) Interaction with P-glycoprotein and transport of ery-thromycin, midazolam and ketoconazole in Caco-2 cells. Eur JPharmcol 358:289–294. doi:10.1016/S0014-2999(98)00607-4

131. El-Readi MZ, Hamdan D, Farrag N, El-Shazly A, Wink M (2010)Inhibition of P-glycoprotein activity by limonin and other sec-ondary metabolites from Citrus species in human colon andleukaemia cell lines. Eur J Pharmcol 626:139–145. doi:10.1016/j.ejphar.2009.09.040

123