Download ABC Pumps and Their Role in Active Drug Transport

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Secreted frizzled-related protein 1 wikipedia , lookup

Endogenous retrovirus wikipedia , lookup

Gene therapy of the human retina wikipedia , lookup

Expression vector wikipedia , lookup

Magnesium transporter wikipedia , lookup

Pharmaceutical industry wikipedia , lookup

Drug design wikipedia , lookup

Pharmacogenomics wikipedia , lookup

Drug discovery wikipedia , lookup

Clinical neurochemistry wikipedia , lookup

Transcript
Current Topics in Medicinal Chemistry, 2009, 9, 119-129
119
ABC Pumps and Their Role in Active Drug Transport
Nicola A. Colabufo*, Francesco Berardi, Marialessandra Contino, Mauro Niso and Roberto Perrone
Dipartimento Farmacochimico, Università degli Studi di Bari, via Orabona 4, 70125 Bari, Italy
Abstract: Pharmacokinetic limitations affect drug bioavailability determining the loss or the reduction of the
pharmacological effects. The Gastro Intestinal tract (GI) and the Blood Brain Barrier (BBB) are the most important
restrictive and selective physiological lines of defense of the organism. Although several parameters such as LogP, LogD
and Ka have been extensively employed for determining drug bioavailability, the active transports, present in these
biological barriers, play an important role for dosing and limiting cell drugs concentration. In particular, ATP Binding
Cassette (ABC) transporters are involved in the active transport both in GI and BBB. Their strategic activity and
biochemical and pharmacological role are herein treated.
Keywords: Blood brain barrier, ABC transporters, P-glycoprotein, MRPs, drug absorption and disposition.
INTRODUCTION
The therapeutic effect of many drugs is due to their
ability to cross cellular barriers for targeting the biological
sites. Usually it is accepted that lipophilic drugs can cross
these barriers in the absence of specific carriers because they
freely diffuse across the plasma membrane whereas
hydrophilic and charged compounds employ transporter
mechanisms to cross cell membranes. This is only a first
aspect of the question but another important aspect involves
the efflux of these drugs by active transporters; they
modulate the absorption and the accumulation of drugs and
toxic substances into the cells. In this paper we discuss the
critical aspects related to drug efflux by ABC transporters
considering the two major physiological barriers involved in
drug-transport ABC-mediated: the Gastro Intestinal tract
(GI) and the Blood Brain Barrier (BBB).
ACTIVE
TRACT
TRANSPORT
IN
Apical
1
2
Basolateral
GASTROINTESTINAL
Oral drug absorption from the intestine is a function of
different drug parameters: a) solubility in GI fluids; b)
metabolic stability; c) permeability [1,2]. Therefore, these
parameters modulate the oral bioavailability of drugs into the
systemic circulation. Drug absorption across the intestinal
membrane is due to passive and active absorption [3].
Passive absorption occurs through the cell membrane of
enterocytes (transcellular way) or through the tight junctions
between the enterocytes (paracellular way) as depicted in
Fig. (1).
Active absorption can be carrier-mediated or a facilitated
diffusion. Lipophilic molecules are absorbed through the
intestinal epithelium by passive diffusion whereas hydrophilic molecules by a carrier-mediated process. Some small
hydrophilic compounds are transported through the
paracellular junction [1]. As a first step transcellular
*Address correspondence to this author at the Dipartimento Farmacochimico, Università degli Studi di Bari, Via Orabona, 4, 70125, Bari, Italy;
Tel: +39-080-5442727; Fax: +39-080-5442231;
E-mail: [email protected]
1568-0266/09 $55.00+.00
365
Fig. (1). Transcellular (1) and paracellular (2) intestinal absorption.
absorption involves the crossing of the apical membrane
from lumen to blood. After transport across the cytosol, the
drug overcomes the basolateral membrane reaching the
blood. Usually, hydrophilic drugs employ the same carriers
of nutrients and micronutrients whereas lipophilic molecules
employ passive transcellular absorption [1,4] (Fig. (2)).
Active efflux pumps, belonging to ABC transporters such
as P-gp and MRP2, are localized on the apical membrane.
These transporters can extrude drugs back into the lumen [58] with the aim of dosing the absorption and the
accumulation of drugs and toxic substances into the cells and
of facilitating the intestinal clearance of compounds present
in the blood (Fig. (3)).
ABC transporters efflux unmodified drugs and/or drugs
metabolites as substrates (Fig. (4)) when these compounds
interact with cytochrome P450 (Phase I) and with
conjugating enzymes (Phase II). Intestinal CYP3A4, the
most important cytochrome isoform involved in first pass
© 2009 Bentham Science Publishers Ltd.
120 Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
Colabufo et al.
SHM
LM
HM
1
2
3
Apical
SHM
Small Hydrophilic Molecules
HM
LM
Lipophilic Molecules
HM
Hydrophilic Molecules
Basolateral
SHM
LM
HM
Fig. (2). Paracellular (1), facilitated diffusion (2); carrier-mediated (3) intestinal absorption.
Apical
ABC
trasporter
1
ABC
trasporter
2
Basolateral
Fig. (3). Intestinal absorption modulated by ABC transporters to regulate cell drug accumulation (1) and to facilitate intestinal clearance (2).
metabolism, regulates the systemic availability of many
orally administered drugs. It has been reported that in small
intestine P-gp and CYP3A4 are localized on the villus tip of
enterocytes [7,9-12] where they synergistically act. Each of
the two metabolic phases can transform the metabolized
drugs into efflux pumps substrates [13].
366
ATP-BINDING CASSETTE (ABC)
GASTROINTESTINAL TRACT
FAMILY
IN
ABC subfamilies involved in intestinal transport are the
MDR subfamily, including MDR1, MDR3, BSEP, and the
MRP subtype, including MRP1-9 [2]. Moreover, also BCRP
ABC Pumps and Their Role in Active Drug Transport
Apical
Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
1
121
2
ABC
trasporter
Metabolising
enzymes
Metabolising
enzymes
Basolateral
Fig. (4). Modified drug absorption (1) and elimination (2).
is considered to be involved in the intestinal transport
[14,15].
paclitaxel [24] although the relationship between MDR3 and
multidrug resistance has to be demonstrated yet.
ABCB SUBFAMILY
BSEP (Bile Salt Export Pump)
P-glycoprotein
ABCB1, also known as P-gp, displays a large spectrum
of transported substrates in particular lipophilic and cationic
compounds although P-gp potential substrates preferentially
are anticancer agents, antibiotics, antivirals, calcium channel
blockers, and immunosuppressive agents. At this list of
biologically active compounds food and vegetables should
also be added [3,16].
P-gp is highly localized in kidney, adrenal gland, liver,
colon and lung, and also as basal level in prostate, skin,
spleen, heart, skeletal muscle, stomach and ovary and in
BBB [17,18]. This membrane apical localization plays a
critical role in the efflux of xenobiotics and toxins into the
intestinal lumen, bile, urine, and blood [7,11,12,18-20]. The
apical P-gp expression allows the efflux transport of several
anticancer drugs both from basolateral to apical and from
blood to lumen modulating their oral bioavailability.
MDR3
MDR3 has been firstly identified as a canalicular
phospholipid translocator [21,22] and the studies to investigate its role were carried out in human MDR3 gene
mutation experiments observing its involvement in pathologies that results in phospholipid secretion by the liver and
high serum -glutamyltransferase levels [23].
MDR3 protein effluxes several drugs such as digoxin,
and several antineoplastic agents such as vinblastine, and
367
BSEP, also known as Sister of P-gp (SP-gp) for its close
sequence homology with P-gp, was first cloned from porcine
liver [25]. BSEP mediates bile acid transport across the
canalicular membrane of hepatocytes downregulating
intracellular bile acid concentrations and regulating the
enterohepatic circulation of bile acids.
Although BSEP is a Sister P-gp protein, it is not involved
in the efflux of several chemotherapeutic agents that are
specific P-gp substrates. Consequently, BSEP in drug disposition is less involved than its close relative P-gp.
ABCC SUBFAMILY
MRP1
MRP1 (ABCC1) is localized in high levels in intestine,
brain, kidney, lung, and testis, and lower expression was
found in liver [26-29]. MRP1 is present in basolateral membranes of polarized epithelial cells, renal distal and collecting
tubules [30], and liver [31,32]. Although MRP1 involvement
in the intestinal drug disposition is not completely clarified,
its ability to confer resistance to antineoplastic drugs has
been demonstrated [33].
MRP1 protein effluxes several conjugated metabolites,
such as glutathione conjugates (leukotriene C4 and 2,4dinitrophenyl-S-glutathione), bilirubin glucuronides, estradiol-17--glucuronide, and dianionic bile salts [34-37]
indicating its involvement in detoxification of endogenous
metabolites.
122 Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
Colabufo et al.
MDR1 subfamily
Out
1
2
3
4
5
6
7
8
9
10
11
12
In
NH2
NBD 2
NBD 1
COOH
MRPs subfamily
NH2
Out
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
In
NBD 2
NBD 1
COOH
BCRP transporter
Out
1
2
3
4
5
6
In
NBD
COOH
NH2
Fig. (5). ABC transporters in GI tract.
MRP1 localization at basolateral level suggests that this
protein protects cells through the efflux of substrates into the
blood. Indeed, MRP1 is responsible for the elimination of
toxins and this action is more effective when it is coupled
with apical P-gp. Both MRP1 and P-gp expression was
higher in tumour tissues treated with chemotherapeutic
agents than in untreated tumours [38].
368
MRP2
MRP2 (ABCC2), originally identified in human liver as
the canalicular multispecific organic anion transporter
(cMOAT) [39], is highly expressed in liver, intestine, and
kidney tubules [6,40,41]. MRP2 is localized at the apical
membranes of these tissues [6,40,42,43]. This pump
ABC Pumps and Their Role in Active Drug Transport
Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
transports conjugated and unconjugated anionic compounds
into bile and the specific substrates of this pump are
equivalent to MRP1 substrates. Moreover, as reported for
BSEP, MRP2 mediates bile acid secretion from the
hepatocytes. Differing from BSEP, MRP2 transports only
sulphated bile salts such as taurochenodeoxycholate-3sulphate and taurolithocholate-3-sulphate, but not monoanionic bile salts such as taurocholate [44]. Considering the
transported substrates, MRP2 contributes to the detoxification by secreting metabolites into the bile. MRP2 protein
also transports potentially toxic or beneficial dietary
constituents, such as flavonoids [8,45].
It has been hypothesized that the MRP2 pump is coexpressed with enzymes involved in Phase II metabolism,
because of the fact that this pump transports several
conjugated compounds. Among Phase II metabolizing
enzymes, Glutathione-S-transferase catalyses the conjugation
of glutathione to a range of electrophilic compounds then
exported by MRP transporters. Glutathione-S-transferases
are reported in several isoforms (, μ, , , , and
microsomal). Human GST -isoform expression is reported
in duodenum and small intestine decreasing from proximal
to distal small intestine, and from small intestine to colon
[46]. Moreover, a co-expression of MRP2 with other Phase
II metabolizing enzymes such as UDP-glucuronosyltransferases (UGT) has been reported. These enzymes catalyse
the conjugation of glucuronic acid to nucleophilic CYP3A4.
Therefore, given the co-expression with Phase II metabolizing enzymes, is possible to assert that MRP2 plays a
synergistic role in intestinal drug detoxification and
excretion.
MRP3
The presence of MRP3 (ABCC3) has been reported in
liver, small intestine and colon and, differently from MRP2,
its expression is high in the ileum and colon [47]. MRP3 is
localized to the basolateral membrane [48] showing a similar
substrate specificity with MRP2 and BSEP, because it
effluxes bile salts [49,50]. The affinity for bile salts, and its
expression in the intestine suggests a possible involvement
of MRP3 in bile salt uptake.
MRP4
MRP4 (ABCC4) has been detected in jejunum, kidney,
brain, lung [51-54] and it is structurally more similar to P-gp
than to the other "typical" MRP transporters. MRP4 is
expressed on the apical membranes of human renal proximal
tubule cells [53] and basolaterally in tubuloacinar cells of the
prostate gland [55]. MRP4 transports folic acid, folinic acid
(leucovorin), methotrexate [56], cAMP, cGMP, estradiol-17-glucuronide, and bile acids [57-60] and its physiological
role regards prostaglandin-E cellular release. The apical
localization both in kidney and in the intestine suggests that
MRP4 is involved in biliary secretion of nucleoside phosphonate analogues and it limits their intestinal absorption
contributing to their poor oral bioavailability.
369
123
MRP5
MRP5 (ABCC5) is structurally similar to MRP4 and it is
reported basolaterally in colon, liver, kidney, skeletal muscle
[51,61] and brain [29]. MRP5 does not confer resistance to
antineoplastic drugs and in the meantime there is no
evidence of intestinal drug disposition MRP5-mediated.
MRP6
MRP6 (ABCC6) is highly expressed in kidney, liver,
duodenum, colon, brain, and salivary gland [29,62,63]. It
was hypothesized that MRP6 mediates the transport of
several natural cytotoxic agents that are also substrates for
MRP1-3, including etoposide, doxorubicin and cisplatin
[64].
MRP7-9
Recent reports indicate that MRP7 (ABCC7) is detected
in liver, kidney, colon, spleen, skin, stomach, and testis [65]
while MRP8 (ABCC8) is highly expressed in breast cancer,
and at moderate levels, in normal breast, testis, and, at low
levels, in liver, brain, and placenta [66]. Two variants of
MRP9 (ABCC9) have been identified, one subtype reported
in breast cancer, normal breast, and testis, and the other in
brain, skeletal muscle, and ovary [67]. Although MRPs7-9
are not functionally classified, their important roles in drug
resistance and drug efflux have been hypothesized.
ABCG SUBFAMILY
BCRP
BCRP (ABCG2) is an ABC half-transporter expressed in
breast, ovary, small intestine, colon, and liver [68,69]. BCRP
mediates secretory transport of drugs in polarized cancer cell
lines of breast, colon and stomach [70] and has a large effect
on the oral bioavailability of the anticancer drugs. Since the
in vivo data demonstrate that BCRP effluxes the same
substrates as P-gp [71], these findings suggest BCRP as halftransporter compared to P-gp in regulating drug absorption
and disposition. On the other hand, BCRP pump has six
transmembrane domains with respect to twelve transmembrane domains of P-gp.
ABC TRANSPORTERS IN GASTROINTESTINAL
TRACT AND IN PERIPHERAL REALATED ORGANS
In Fig. (6) ABC transporters are shown displaying for
each tract or organ with their basolateral or apical localization. In the enterocytes MRP1 and MRP3 are localized at
the basolateral level while MDR1, MRP2 and BCRP act at
the apical level. In the proximal tubular cells of kidney,
MDR1 and MRP2 are localized at the apical membrane.
Moreover, MDR1, MDR3, BSEP, BCRP and MRP2 are
present at the canalicular membranes and can secrete
compounds modified or unmodified, into the canaliculus and
ultimately into the bile.
124 Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
Colabufo et al.
A
Apical / Lumen
P-gp
MRP2
BCRP
Enterocyte
MRP3
MRP1
Basolateral / Blood
Intestinal villus
B
Apical
P-gp
MRP2
Proximal tubular cell
Basolateral
Nefron
370
ABC Pumps and Their Role in Active Drug Transport
C
Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
125
canalicular membrane
MDR1/P-gp
MRP1
MRP2
MDR3
Canaliculus
MRP3
BCRP
BSEP/SP-gp
Blood
Blood
Hepatocytes
Fig. (6). Localization of some ABC transporters in enterocytes (A), in proximal tubular cells (B) and in hepatocytes (C).
ACTIVE TRANSPORT IN THE BLOOD BRAIN
BARRIER
Drug availability into the brain is modulated by physical
barriers such as Blood Brain Barrier (BBB) and Blood
CerebroSpinal Fluid Barrier (BCSFB). BBB and BCSFB are
different in location, size, morphology, and function [72-74].
BBB regulates and preserves the brain acting as a physical
and metabolic barrier. The presence of tight junctions
(zonulae occludens) [75] between endothelial cells and the
lack of fenestrae and pinocytotic vesicles in the cerebral
arterioles, capillaries, and endothelium venules modulate
brain uptake. BCSFB is formed by the choroid plexus
epithelial cells which are connected by tight junctions,
whereas the capillaries are fenestrated.
BBB modulates molecules access to the brain by two
processes: 1) passive diffusion or 2) active transport [72].
The first process mediates lipid and small nonpolar
molecules access while the second process is mediated by
several membrane transporters modulating the influx or
efflux of various essential substrates (electrolytes, nucleosides, amino acids, and glucose). Although lipophilicity has
been considered a facilitating aspect to permeate the lipid
bilayer, several molecules and drugs display lower brain
permeability despite a favourable lipophylicity.
The molecules access trough BBB is performed by two
possible mechanisms: (i) carrier-mediated transport, (ii)
active efflux transport. Usually, CNS penetration of small
371
molecules is modulated by carriers and/or active transporters
[76,77] whereas large molecules permeation can be receptormediated. Carriers prevalently mediate the influx of
substrates from brain to blood, although some carriers can
modulate the opposite flux while the active transport
modulates the efflux from brain to blood.
Molecules access from brain to blood is a process
mediated by two transporter families including an energydependent and energy-independent transporters [74]. ABC
transporters are energy-dependent system and they are
prevalently localized at the luminal membrane. Solute
Carrier (SLC) transporters are energy-independent systems
and are localized at the abluminal membrane of the brain
capillary endothelial cells [78,79].
ATP-BINDING CASSETTE (ABC) FAMILY IN BBB
P-glycoprotein
High levels of P-gp are reported in the luminal membrane
of the endothelial cells constituting BBB, BCSFB, and
blood-testis barrier [76,80-82]. Such strategic localization
gives P-gp a physiologically crucial role. P-gp links drugs in
the core of the lipid bilayer and flips them into blood [83]. Pgp exerts a protective function in BBB; for example, recent
results reported a significant correlation between Parkinson’s
disorder induced by pesticides and ABCB1 gene polymorphism (C3435T). The mutation of this gene increases the
toxic effects of pesticides and of other toxic xenobiotics
126 Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
Colabufo et al.
usually effluxed by P-gp. In Alzheimer’s disease, the
expression of P-gp in blood vessels contributes to the
accumulation of -amyloid: the first step to diagnose this
pathology suggesting a P-gp involvement in Alzheimer’s
disease [84,85]. Moreover, the differences in P-gp expression and dislocation in the brain area could be correlated to
other diseases such as epilepsy. In this case, P-gp is present
both in endothelial cells and in perivascular astrocytes [86].
Furthermore, P-gp is highly expressed in many tumors [87].
MRPs
MRPs in particular are involved in the extrusion of
conjugated xenobiotics and the mostly representative
transporter of this family, MRP1, is expressed at the luminal
site of the brain capillaries. Evidence of the presence of
MRP3 and MRP5 proteins at the BBB is reported [88].
CONCLUSIONS
The active transporters play a crucial role in drug
bioavailability and disposition in several compartments such
as BBB, BCSFB and GI. The study and the characterization
of ABC transporters give perspectives and a new address in
drugs discovery. These findings should be addressed by the
medicinal chemist in the plan of new drugs optimizing their
pharmacodynamic properties by strategies consistent with
pharmacokinetic requirements for overcoming biological
barriers. Therefore, more predictive biological assays are
needed to assess the interaction of drugs with active transporters in order to limit the discrepancies between the
encouraging pharmacodynamic in vitro profile and the
results found in the in vivo assays because of the involvement of active transporters.
REFERENCES
SLC FAMILY IN BBB
[1]
The SoLute Carrier (SLC) family comprises 43
subfamilies on the basis of their sequences and recently,
three novel families (SLC44-SLC46) have been added. SLC
transporters move the solutes employing as a driving force
their electrochemical gradient [78]. Some SLC transporters
are biochemically classified as coupled transporters,
exchangers and as passive transporters.
Each transporter displays distinct pharmacological roles
and transports specific substrates, such as amino acids,
oligopeptides, sugars, monocarboxylic acids, organic cations,
anions, phosphates, nucleosides, metals and water-soluble
vitamins.
Among the SLCs, SLC19, SLC28, SLC29, SLC7A,
SLC3A and SLC31A modulate chemotherapeutic treatment
because these transporters sensitize tumor cells to anticancer
drugs.
Polymorphisms of SLC transporters are an important tool
to predict the therapeutic response and toxicity of
antineoplastic agents and for the survival of patients after
chemotherapy.
Some Organic Anion Transporters (OAT1 and OAT3)
were monitored on the brush border of the choroids plexus
and in an in vitro BBB model [89,90]. Among organic
anions transported by OATs p-aminohippurate, dicarboxylates and -lactam antibiotics are reported [79].
Also some Organic Cation Transporters (OCT2 in
particular) are expressed in the brain [76,91]. They modulate
neurotransmitters in neurons rather than at the BBB. A new
family of OCTNs has been also found, that displays
significant homology to the OTC and OAT families. Indeed,
the first study classified OCTNs as organic cation transporters while recent findings demonstrated their physiological function as sodium ion-dependent transporters for
carnitine involved in fatty acids metabolism. Finally,
monocarboxylic acid transporters (MTC), detected at both
luminal and abluminal membranes of the brain capillaries,
are involved in the transport of lactic and pyruvic acids [92].
In particular MCT1 subtype contributes to the regulation of
energy substrates in the brain parenchyma at BBB level [93].
372
[2]
[3]
[4]
[5]
[6]
[7]
[8]
[9]
[10]
[11]
[12]
[13]
[14]
Hayashi, M.; Tomita, M.; Awzu, S. Transcellular and paracellular
contribution to transport process in the colorectal route. Adv. Drug
Deliv. Rev. 1997, 28, 191-204.
Chan, L. M.; Lowes, S.; Hirst, B. H. The ABCs of drug transport in
intestine and liver: efflux proteins limiting drug absorption and
bioavailability. Eur. J. Pharm. Sci. 2004, 21, 25-51.
Hunter, J.; Hirst, B. H. Intestinal secretion of drugs. The role of Pglycoprotein and related drug efflux systems in limiting oral drug
absorption. Adv. Drug. Del. 1997, 25, 129-157.
Montalto, M.; Veneto, G.; Cuoco, L.; Cammarota, G.; Tursi, A.;
Papa, A.; Fedeli, G.; Gasbarrini, G. Intestinal permeability. Recenti
Prog. Med. 1997, 88, 140-147.
Evers, R.; Kool, M.; van Deemter, L.; Janssen, H.; Calafat, J.;
Oomen, L. C.; Paulusma, C. C.; Oude Elferink, R. P.; Baas, F.;
Schinkel, A. H.; Borst, P. Drug export activity of the human
canalicular multispecific organic anion transporter in polarized
kidney MDCK cells expressing cMOAT (MRP2) cDNA. J. Clin.
Invest. 1998, 101, 1310-1319.
Fromm, M. F.; Kauffmann, H. M.; Fritz, P.; Burk, O.; Kroemer, H.
K.; Warzok, R. W.; Eichelbaum, M.; Siegmund, W.; Schrenk, D.
The effect of rifampin treatment on intestinal expression of human
MRP transporters. Am. J. Pathol. 2000, 157, 1575-1580.
Hunter, J.; Jepson, M.; Tsuruo, T.; Simmons, N. L.; Hirst, B. H.
Functional expression of P-glycoprotein in apical membranes of
human intestinal Caco-2 cells. Kinetics of vinblastine secretion and
interaction with modulators. J. Biol. Chem. 1993, 268, 1499114997.
Walgren, R. A.; Karnaky, K. J.; Lindenmayer, G. E.; Walle, T.;
Efflux of dietary flavonoid quercetin 4’--glucoside across human
intestinal Caco-2 cell monolayers by apical multidrug resistanceassociated protein-2a. J. Pharmacol. Exp. Ther. 2000, 294, 830836.
Canaparo, R.; Finnström, N.; Serpe, L.; Nordmark, A.; Muntoni,
E.; Eandi, M.; Rane, A.; Zara, G. P. Expression of CYP3A
isoforms and P-glycoprotein in human stomach, jejunum and
ileum. Clin. Exp. Pharmacol. Physiol. 2007, 34, 1138-1144.
Chan, L. M.; Cooper, A. E.; Dudley, A. L.; Ford, D.; Hirst, B. H. Pglycoprotein potentiates CYP3A4-mediated drug disappearance
during Caco-2 intestinal secretory detoxification. J. Drug Target
2004, 12, 405-413.
Hunter, J.; Hirst, B. H.; Simmons, N. L. Drug absorption limited by
P-glycoprotein-mediated secretory drug transport in human
intestinal epithelial Caco-2 cell layers. Pharm. Res. 1993, 10, 743749.
Terao, T.; Hisanaga, E.; Sai, Y.; Tamai, I.; Tsuji, A. Active
secretion of drugs from the small intestinal epithelium in rats by Pglycoprotein functioning as an absorption barrier. J. Pharm.
Pharmacol. 1996, 48, 1083-1089.
Keppler, D.; Cui, Y.; König, J.; Leier, I.; Nies, A. Export pumps for
anionic conjugates encoded by MRP genes. Adv. Enzyme Regul.
1999, 39, 237-246.
Urquhart, B. L.; Ware, J. A.; Tirona, R. G.; Ho, R. H.; Leake, B. F.;
Schwarz, U. I.; Zaher, H.; Palandra, J.; Gregor, J. C.; Dresser, G.
ABC Pumps and Their Role in Active Drug Transport
[15]
[16]
[17]
[18]
[19]
[20]
[21]
[22]
[23]
[24]
[25]
[26]
[27]
[28]
[29]
[30]
[31]
Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
K.; Kim, R. B. Breast cancer resistance protein (ABCG2) and drug
disposition: intestinal expression, polymorphisms and sulfasalazine
as an in vivo probe. Pharmacogenet. Genomics 2008, 18, 439-448.
Zamber, C. P.; Lamba, J. K.; Yasuda, K.; Farnum, J.; Thummel,
K.; Schuetz, J. D.; Schuetz, E. G. Natural allelic variants of breast
cancer resistance protein (BCRP) and their relationship to BCRP
expression in human intestine. Pharmacogenetics 2003, 13, 19-28.
Evans, A. M. Influence of dietary components on the
gastrointestinal metabolism and transport of drugs. Ther. Drug
Monit. 2000, 22, 131-136.
Fojo, A. T.; Ueda, K.; Slamon, D. J.; Poplack, D. G.; Gottesman,
M. M.; Pastan, I. Expression of a multidrug-resistance gene in
human tumors and tissues. Proc. Natl. Acad. Sci. USA 1987, 84,
265-269.
Gatmaitan, Z. C.; Arias, I. M. Structure and function of P-glycoprotein in normal liver and small intestine. Adv. Pharmacol. 1993,
24, 77-97.
Thiebaut, F.; Tsuruo, T.; Hamada, H.; Gottesman, M. M.; Pastan,
I.; Willingham, M. C. Cellular localisation of the multidrugresistance gene product P-glycoprotein in normal human tissues.
Proc. Natl. Acad. Sci. USA 1987, 84, 7735-7738.
Tsuji, A.; Tamai, I. Carrier-mediated intestinal transport of drugs.
Pharm. Res. 1996, 13, 963-977.
Smit, J. J. M.; Schinkel, A. H.; Oude Elferink, R. P. J.; Groen, A.
K.; Wagenaar, E.; Van Deemter, L.; Mol, C. A. A. M.; Ottenhoff,
R.; Van Der Lugt, N. M. T.; Van Roon, M. A.; Van Der Valk, M.
A.; Offerhaus, G. J. A.; Berns, A. J. M.; Borst, P. Homozygous
disruption of the murine mdr2 P-glycoprotein gene leads to a
complete absence of phospholipid from bile and to liver disease.
Cell 1993, 75, 451-462.
Smith, A. J.; Timmermans-Hereijgers, J. L. P. M.; Roelofsen, B.;
Wirtz, K. W. A.; Van Blitterswijk, W. J.; Smit, J. J. M.; Schinkel,
A. H.; Borst, P. The human MDR3 P-glycoprotein promotes
translocation of phosphatidylcholine through the plasma membrane
of fibroblasts from transgenic mice. FEBS Lett. 1994, 354, 263266.
De Vree, J. M.; Jacquemin, E.; Sturm, E.; Cresteil, D.; Bosma, P.
J.; Aten, J.; Deleuze, J. F.; Desrochers, M.; Burdelski, M.; Bernard,
O.; Oude Elferink, R. P.; Hadchouel, M. Mutations in the MDR3
gene cause progressive familial intrahepatic cholestasis. Proc. Natl.
Acad. Sci. USA 1998, 95, 282-287.
Smith, A. J.; Van Helvoort, A.; Van Meer, G.; Szabo, K.; Welker,
E.; Szakacs, G.; Váradi, A.; Sarkadi, B.; Borst, P. MDR3 Pglycoprotein, a phosphatidylcholine translocase, transports several
cytotoxic drugs and directly interacts with drugs as judged by
interference with nucleotide trapping. J. Biol. Chem. 2000, 275,
23530-23539.
Childs, S.; Yeh, R. L.; Georges, E.; Ling, V. Identification of a
sister gene to P-glycoprotein. Cancer Res. 1995, 55, 2029-2034.
Cherrington, N. J.; Hartley, D. P.; Li, N.; Johnson, D. R.; Klaassen,
C. D. Organ distribution of multidrug resistance proteins 1, 2, and 3
(Mrp1, 2, and 3) mRNA and hepatic induction of Mrp3 by
constitutive androstane receptor activators in rats. J. Pharmacol.
Exp. Ther. 2002, 300, 97-104.
Flens, M. J.; Zaman, G. J.; Van Der Valk, P.; Izquierdo, M. A.;
Schroeijers, A. B.; Scheffer, G. L.; Van Der Groep, P.; De Haas,
M.; Meijer, C. J.; Scheper, R. J. Tissue distribution of the multidrug
resistance protein. Am. J. Pathol. 1996, 148, 1237-1247.
Zaman, G. J.; Versantvoort, C. H.; Smit, J. J.; Eijdems, E. W.; De
Haas, M.; Smith, A. J.; Broxterman, H. J.; Mulder, N. H.; De Vries,
E. G.; Baas, F.; Borst, P. Analysis of the expression of MRP, the
gene for a new putative transmembrane drug transporter, in human
multidrug resistant lung cancer cell lines. Cancer Res. 1993, 53,
1747-1750.
Gazzin, S.; Strazielle, N.; Schmitt, C.; Fevre-Montange, M.;
Ostrow, J. D.; Tiribelli, C.; Ghersi-Egea, J. F. Differential
expression of the multidrug resistance-related proteins ABCb1 and
ABCc1 between blood-brain interfaces. J. Comp. Neurol. 2008,
510, 497-507.
Peng, K. C.; Cluzeaud, F.; Bens, M.; Van Huyen, J. P.; Wioland,
M. A.; Lacave, R.; Vandewalle, A. Tissue and cell distribution of
the multidrug resistance-associated protein (MRP) in mouse
intestine and kidney. J. Histochem. Cytochem. 1999, 47, 757-768.
Mayer, R.; Kartenbeck, J.; Büchler, M.; Jedlitschky, G.; Leier, I.;
Keppler, D. Expression of the MRP gene-encoded conjugate export
pump in liver and its selective absence from the canalicular
373
[32]
[33]
[34]
[35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
[46]
[47]
[48]
127
membrane in transport-deficient mutant hepatocytes. J. Cell. Biol.
1995, 131, 137-150.
Roelofsen, H.; Vos, T. A.; Schippers, I. J.; Kuipers, F.; Koning, H.;
Moshage, H.; Jansen, P. L.; Müller, M. Increased levels of the
multidrug resistance protein in lateral membranes of proliferating
hepatocyte-derived cells. Gastroenterology 1997, 112, 511-521.
Multidrug resistance: biological and pharmaceutical advances in
antitumour treatment. Review book, Research Signpost, 2008,
Editor Colabufo, N. A.
Jedlitschky, G.; Leier, I.; Buchholz, U.; Barnouin, K.; Kurz, G.;
Keppler, D. Transport of glutathione, glucuronate, and sulfate
conjugates by the MRP gene-encoded conjugate export pump.
Cancer Res. 1996, 56, 988-994.
Jedlitschky, G.; Leier, I.; Buchholz, U.; Hummel-Eisenbeiss, J.;
Burchell, B.; Keppler, D. ATP-dependent transport of bilirubin
glucuronides by the multidrug resistance protein MRP1 and its
hepatocyte canalicular isoform MRP2A. Biochem. J. 1997, 327,
305-310.
Leier, I.; Hummel-Eisenbeiss, J.; Cui, Y.; Keppler, D. ATPdependent para-aminohippurate transport by apical multidrug
resistance protein MRP2. Kidney Int. 2000, 57, 1636-1642.
Stride, B. D.; Grant, C. E.; Loe, D. W.; Hipfner, D. R.; Cole, S. P.;
Deeley, R. G. Pharmacological characterization of the murine and
human orthologs of multidrug-resistance protein in transfected
human embryonic kidney cells. Mol. Pharmacol. 1997, 52, 344353.
Tada, Y.; Wada, M.; Migita, T.; Nagayama, J.; Hinoshita, E.;
Mochida, Y.; Maehara, Y.; Tsuneyoshi, M.; Kuwano, M.; Naito, S.
Increased expression of multidrug resistance-associated proteins in
bladder cancer during clinical course and drug resistance to
doxorubicin. Int. J. Cancer 2002, 98, 630-635.
Paulusma, C. C.; Bosma, P. J.; Zaman, G. J.; Bakker, C. T.; Otter,
M.; Scheffer, G. L.; Scheper, R. J.; Borst, P.; Oude Elferink, R. P.
Congenital jaundice in rats with a mutation in a multidrug
resistance-associated protein gene. Science 1996, 271, 1126-1128.
Schaub, T. P.; Kartenbeck, J.; König, J.; Vogel, O.; Witzgall, R.;
Kriz, W.; Keppler, D.; Expression of the conjugate export pump
encoded by the mrp2 gene in the apical membrane of kidney
proximal tubules. J. Am. Soc. Nephrol. 1997, 8, 1213-1221.
Scheffer, G. L.; Kool, M.; De Haas, M.; De Vree, J. M.;
Pijnenborg, A. C.; Bosman, D. K.; Elferink, R. P.; Van Der Valk,
P.; Borst, P.; Scheper, R. J. Tissue distribution and induction of
human multidrug resistant protein 3. Lab. Invest. 2002, 82, 193201.
Büchler, M.; König, J.; Brom, M.; Kartenbeck, J.; Spring, H.;
Horie, T.; Keppler, D. cDNA cloning of the hepatocyte canalicular
isoform of the multidrug resistance protein, cMrp, reveals a novel
conjugate export pump deficient in hyperbilirubinemic mutant rats.
J. Biol. Chem. 1996, 271, 15091-15098.
Schaub, T. P.; Kartenbeck, J.; König, J.; Spring, H.; Dorsam, J.;
Staehler, G.; Storkel, S.; Thon, W. F.; Keppler, D. Expression of
the MRP2 gene-encoded conjugate export pump in human kidney
proximal tubules and in renal cell carcinoma. J. Am. Soc. Nephrol.
1999, 10, 1159-1169.
Akita, H.; Suzuki, H.; Ito, K.; Kinoshita, S.; Sato, N.; Takikawa,
H.; Sugiyama, Y. Characterization of bile acid transport mediated
by multidrug resistance associated protein 2 and bile salt export
pump. Biochim. Biophys. Acta 2001, 1511, 7-16.
Vaidyanathan, J. B.; Walle, T. Transport and metabolism of the tea
flavonoid ()-epicatechin by the human intestinal cell line Caco-2.
Pharm. Res. 2001, 18, 1420-1425.
Coles, B. F.; Chen, G.; Kadlubar, F. F.; Radominska-Pandya, A.
Interindividual variation and organ-specific patterns of glutathione
S-transferase alpha, mu, and pi expression in gastrointestinal tract
mucosa of normal individuals. Arch. Biochem. Biophys. 2002, 403,
270-276.
Rost, D.; Mahner, S.; Sugiyama, Y.; Stremmel, W. Expression and
localization of the multidrug resistance-associated protein 3 in rat
small and large intestine. Am. J. Physiol. Gastrointest. Liver
Physiol. 2002, 282, G720-G726.
Kool, M.; Van Der Linden, M.; De Haas, M.; Scheffer, G. L.; De
Vree, J. M.; Smith, A. J.; Jansen, G.; Peters, G. J.; Ponne, N.;
Scheper, R. J.; Elferink, R. P.; Baas, F.; Borst, P. MRP3 an organic
anion transporter able to transport anti-cancer drugs. Proc. Natl.
Acad. Sci. USA 1999, 96, 6914-6919.
128 Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
[49]
[50]
[51]
[52]
[53]
[54]
[55]
[56]
[57]
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
Colabufo et al.
Hirohashi, T.; Suzuki, H.; Takikawa, H.; Sugiyama, Y. ATPdependent transport of bile salts by rat multidrug resistanceassociated protein 3 (Mrp3). J. Biol. Chem. 2000, 275, 2905-2910.
Zeng, H.; Liu, G.; Rea, P. A.; Kruh, G. D. Transport of
amphipathic anions by human multidrug resistance protein 3.
Cancer Res. 2000, 60, 4779-4784.
Kool, M.; De Haas, M.; Scheffer, G. L.; Scheper, R. J.; Van Eijk,
M. J.; Juijn, J. A.; Baas, F.; Borst, P. Analysis of expression of
cMOAT (MRP2), MRP3, MRP4, and MRP5, homologues of the
multidrug resistance-associated protein gene (MRP1), in human
cancer cell lines. Cancer Res. 1997, 57, 3537-3547.
Taipalensuu, J.; Tornblom, H.; Lindberg, G.; Einarsson, C.;
Sjoqvist, F.; Melhus, H.; Garberg, P.; Sjostrom, B.; Lundgren, B.;
Artursson, P. Correlation of gene expression of ten drug efflux
proteins of the ATP-binding cassette transporter family in normal
human jejunum and in human intestinal epithelial Caco-2 cell
monolayers. J. Pharmacol. Exp. Ther. 2001, 299, 164-170.
Van Aubel, R. A.; Smeets, P. H.; Peters, J. G.; Bindels, R. J.;
Russel, F. G. The MRP4/ABCC4 gene encodes a novel apical
organic anion transporter in human kidney proximal tubules:
putative efflux pump for urinary cAMP and cGMP. J. Am. Soc.
Nephrol. 2002, 13, 595-603.
Zhang, Y.; Han, H.; Elmquist, W. F.; Miller, D. W. Expression of
various multidrug resistance-associated protein (MRP) homologues
in brain microvessel endothelial cells. Brain Res. 2000, 876, 148153.
Lee, K.; Klein-Szanto, A. J.; Kruh, G. D. Analysis of the MRP4
drug resistance profile in transfected NIH3T3 cells. J. Natl. Cancer
Inst. 2000, 92, 1934-1940.
Chen, Z. S.; Lee, K.; Walther, S.; Raftogianis, R. B.; Kuwano, M.;
Zeng, H.; Kruh, G. D. Analysis of methotrexate and folate transport
by multidrug resistance protein 4 (ABCC4): MRP4 is a component
of the methotrexate efflux system. Cancer Res. 2002, 62, 31443150.
Chen, Z. S.; Lee, K.; Kruh, G. D. Transport of cyclic nucleotides
and estradiol 17--d-glucuronide by multidrug resistance protein 4.
Resistance to 6-mercaptopurine and 6-thioguanine. J. Biol. Chem.
2001, 276, 33747-33754.
Lai, L.; Tan, T. M. Role of glutathione in the multidrug resistance
protein 4 (MRP4/ABCC4)-mediated efflux of cAMP and resistance
to purine analogues. Biochem. J. 2002, 361, 497-503.
Van Aubel, R. A.; Smeets, P. H.; Peters, J. G.; Bindels, R. J.;
Russel, F. G. The MRP4/ABCC4 gene encodes a novel apical
organic anion transporter in human kidney proximal tubules:
putative efflux pump for urinary cAMP and cGMP. J. Am. Soc.
Nephrol. 2002, 13, 595-603.
Zelcer, N.; Reid, G.; Wielinga, P.; Kuil, A.; van der Heijden, I.;
Schuetz, J. D.; Borst, P. Steroid and bile acid conjugates are
substrates of human multidrug-resistance protein (MRP) 4 (ATPbinding cassette C4). Biochem. J. 2003, 371, 361-367.
McAleer, M. A.; Breen, M. A.; White, N. L.; Matthews, N.
pABC11 (also known as MOAT-C and MRP5), a member of the
ABC family of proteins, has anion transporter activity but does not
confer multidrug resistance when overexpressed in human
embryonic kidney 293 cells. J. Biol. Chem. 1999, 274, 2354123548.
Kool, M.; Van Der Linden, M.; De Haas, M.; Baas, F.; Borst, P.
Expression of human MRP6. a homologue of the multidrug
resistance protein gene MRP1, in tissues and cancer cells. Cancer
Res. 1999, 59, 175-182.
Scheffer, G. L.; Hu, X.; Pijnenborg, A. C.; Wijnholds, J.; Bergen,
A. A.; Scheper, R. J. MRP6 (ABCC6) detection in normal human
tissues and tumors. Lab. Invest. 2002, 82, 515-518.
Belinsky, M. G.; Chen, Z. S.; Shchaveleva, I.; Zeng, H.; Kruh, G.
D. Characterization of the drug resistance and transport properties
of multidrug resistance protein 6 (MRP6, ABCC6). Cancer Res.
2002, 62, 6172-6177.
Hopper, E.; Belinsky, M. G.; Zeng, H.; Tosolini, A.; Testa, J. R.;
Kruh, G. D. Analysis of the structure and expression pattern of
MRP7 (ABCC10), a new member of the MRP subfamily. Cancer
Lett. 2001, 162, 181-191.
Bera, T. K.; Lee, S.; Salvatore, G.; Lee, B.; Pastan, I. MRP8, a new
member of ABC transporter superfamily, identified by EST
database mining and gene prediction program, is highly expressed
in breast cancer. Mol. Med. 2001, 7, 509-516.
374
[67]
[68]
[69]
[70]
[71]
[72]
[73]
[74]
[75]
[76]
[77]
[78]
[79]
[80]
[81]
[82]
[83]
[84]
[85]
[86]
Bera, T. K.; Iavarone, C.; Kumar, V.; Lee, S.; Lee, B.; Pastan, I.
MRP9, an unusual truncated member of the ABC transporter
superfamily, is highly expressed in breast cancer. Proc. Natl. Acad.
Sci. USA 2002, 99, 6997-7002.
Allikmets, R.; Schriml, L.; Hutchinson, A.; Romano-Spica, V.;
Dean, M. A human placenta-specific ATP-binding cassette gene
(ABCP) on chromosome 4q22 that is involved in multidrug
resistance. Cancer Res. 1998, 58, 5337-5339.
Maliepaard, M.; Scheffer, G. L.; Faneyte, I. F.; Van Gastelen, M.
A.; Pijnenborg, A. C. L. M.; Schinkel, A. H.; Van De Vijver, M. J.;
Scheper, R. J.; Schellens, J. H. M. Subcellular localisation and
distribution of the breast cancer resistance protein transporter in
normal human tissues. Cancer Res. 2001, 61, 3458-3464.
Ross, D. D.; Yang, W.; Abruzzo, L. V.; Dalton, W. S.; Schneider,
E.; Lage, H.; Dietel, M.; Greenberger, L.; Cole, S. P. C.; Doyle, L.
A. Atypical multidrug resistance: breast cancer resistance protein
messenger RNA expression in mitoxantrone-selected cell lines. J.
Natl. Cancer Inst. 1999, 91, 429-433.
Allen, J.; Brinkhuis, R.; Wijnholds, J.; Schinkel, A. The mouse
Bcrp1/Mxr/Abcp gene: amplification and overexpression in cell
lines selected for resistance to topotecan, mitoxantrone, or
doxorubicin. Cancer Res. 1999, 59, 4237-4241.
Ueno, M. Molecular anatomy of the brain endothelial barrier: an
overview of the distributional features. Curr. Med. Chem. 2007, 14,
1199-1206.
Pardridge, W. M. Blood-brain barrier delivery. Drug Discov. Today
2007, 12, 54-61.
Hammarlund-Udenaes, M.; Fridén, M.; Syvänen, S.; Gupta, A. On
the rate and extent of drug delivery to the brain. Pharm. Res. 2008,
25, 1737-1750.
Mitic, L. L.; Van Itallie, C. M.; Anderson, J. M. Molecular
physiology and pathophysiology of tight junctions I. Tight junction
structure and function: lessons from mutant animals and proteins.
Am. J. Physiol. Gastrointest. Liver Physiol. 2000, 279, G250-254.
Tamai, I.; Tsuji, A. Transporter-mediated permeation of drugs
across the blood-brain barrier. J. Pharm. Sci. 2000, 89, 1371-1388.
Kusuhara, H.; Sugiyama, Y. Efflux transport systems for drugs at
the blood-brain barrier and blood-cerebrospinal fluid barrier (Part
1). Drug Discov. Today 2001, 6, 150-156.
Kusuhara, H.; Sugiyama, Y. Active efflux across the blood-brain
barrier: Role of the solute carrier family. J. Am. Soc. Exp.
NeuroTherapeutics 2005, 2, 73-85.
Ohtsuki, S.; Terasaki, T. Contribution of carrier-mediated transport
systems to the blood-brain barrier as a supporting and protecting
interface for the brain; importance for CNS drug discovery and
development. Pharm. Res. 2007, 24, 1745-1758.
Ebinger, M.; Uhr, M. ABC drug transporter at the blood-brain
barrier. Effects on drug metabolism and drug response. Eur. Arch.
Psychiatry Clin. Neurosci. 2006, 256, 294-298.
Cordon-Cardo, C.; O’Brien, J. P.; Casals, D.; Rittman-Grauer, L.;
Biedler, J. L.; Melamed, M. R.; Bertino, J. R. Multidrug-resistance
gene (P-glycoprotein) is expressed by endothelial cells at bloodbrain barrier sites. Proc. Natl. Acad. Sci USA 1989, 86, 695-698.
Wijnholds, J.; de Lange, E. C.; Scheffer, G. L.; van den Berg, D. J.;
Mol, C. A.; van der Valk, M. A.; Schinkel, A. H.; Scheper, R. J.;
Breimer, D. D.; Borst, P. Multidrug resistance protein 1 protects
the choroid plexus epithelium and contributes to the blood–
cerebrospinal fluid barrier. J. Clin. Invest. 2000, 105, 279-285.
Fromm, M. F. P-glycoprotein: a defense mechanism limiting oral
bioavailability and CNS accumulation of drugs. Int. J. Clin.
Pharmacol. Ther. 2000, 38, 69-74.
Vogelgesang, S.; Warzok, R. W.; Cascorbi, I.; Kunert-Keil, C.;
Schroeder, E.; Kroemer, H. K.; Siegmund, W.; Walker, L. C.;
Pahnke, J. The role of P-glycoprotein in cerebral amyloid
angiopathy; implications for the early pathogenesis of Alzheimer's
disease. Curr. Alzheimer Res. 2004, 1, 121-125.
Kuhnke, D.; Jedlitschky, G.; Grube, M.; Krohn, M.; Jucker, M.;
Mosyagin, I.; Cascorbi, I.; Walker, L. C.; Kroemer, H. K.; Warzok,
R. W.; Vogelgesang, S. MDR1-P-Glycoprotein (ABCB1) Mediates
Transport of Alzheimer's amyloid- peptides-implications for the
mechanisms of A clearance at the blood-brain barrier. Brain
Pathol. 2007, 17, 347-353.
Marroni, M.; Marchi, N.; Cucullo, L.; Abbott, N. J.; Signorelli, K.;
Janigro, D. Vascular and parenchymal mechanisms in multiple
drug resistance: a lesson from human epilepsy. Curr. Drug Targets
2003, 4, 297-304.
ABC Pumps and Their Role in Active Drug Transport
Current Topics in Medicinal Chemistry, 2009, Vol. 9, No. 2
[87]
toxin: its involvement in neurotransmitter metabolite clearance
from the brain. J. Neurochem. 2002, 83, 57-66.
Tamai, I.; Yabuuchi, H.; Nezu, J.; Sai, Y.; Oku, A.; Shimane, M.;
Tsuji, A. Cloning and characterization of a novel human pHdependent organic cation transporter, OCTN1. FEBS Lett. 1997,
419, 107-111.
Garcia, C. K.; Goldstein, J. L.; Pathak, R. K.; Anderson, R. G.;
Brown, M. S. Molecular characterization of a membrane
transporter for lactate, pyruvate, and other monocarboxylates:
implications for the Cori cycle. Cell 1994, 76, 865-873.
Gerhart, D. Z.; Enerson, B. E.; Zhdankina, O. Y.; Leino, R. L.;
Drewes, L. R. Expression of monocarboxylate transporter MCT1
by brain endothelium and glia in adult and suckling rats. Am. J.
Physiol. 1997, 273, E207-13.
[88]
[89]
[90]
Leonard, G. D.; Fojo, T.; Bates, S. E. The role of ABC transporters
in clinical practice. Oncologist 2003, 8, 411-424.
Yousif, S.; Marie-Claire, C.; Roux, F.; Scherrmann, J. M.;
Declèves, X. Expression of drug transporters at the blood-brain
barrier using an optimized isolated rat brain microvessel strategy.
Brain Res. 2007, 1134, 1-11.
Mori, S.; Ohtsuki, S.; Takanaga, H.; Kikkawa, T.; Kang, Y. S.;
Terasaki, T. Organic anion transporter 3 is involved in the brain-toblood efflux transport of thiopurine nucleobase analogs. J.
Neurochem. 2004, 90, 931-41.
Ohtsuki, S.; Asaba, H.; Takanaga, H.; Deguchi, T.; Hosoya, K.;
Otagiri, M.; Terasaki, T. Role of blood-brain barrier organic anion
transporter 3 (OAT3) in the efflux of indoxyl sulfate, a uremic
Received December 3, 2008
Accepted January 21, 2009
375
[91]
[92]
[93]
129