Cholesterol potentiates ABCG2 activity in a heterologous expression system: improved in vitro model...

10
Cholesterol Potentiates ABCG2 Activity in a Heterologous Expression System: Improved in Vitro Model to Study Function of Human ABCG2 A ´ . Pa ´l, D. Me ´ hn, E ´ . Molna ´ r, S. Gedey, P. Me ´ sza ´ ros, T. Nagy, H. Glavinas, T. Jana ´ ky, O. von Richter, G. Ba ´ thori, L. Szente, and P. Krajcsi SOLVO Biotechnology, Central Hungarian Innovations Center, Budao ¨ rs, Hungary (A ´ .P., D.M., E ´ .M., S.G., P.M., T.N., H.G., G.B., P.K.); Department of Medical Chemistry, University of Szeged, Szeged, Hungary (T.J.); Division of Drug Metabolism and Pharmacokinetics, ALTANA Pharma AG, Konstanz, Germany (O.v.R.); CycloLab Cyclodextrin R. and D. Ltd., Budapest, Hungary (L.S.); and Rational Drug Design Laboratories, Budapest, Hungary (P.K.) Received January 3, 2007; accepted March 5, 2007 ABSTRACT ABCG2, a transporter of the ATP-binding cassette family, is known to play a prominent role in the absorption, distribution, metabolism, and excretion of xenobiotics. Drug-transporter in- teractions are commonly screened by high-throughput systems using transfected insect and/or human cell lines. The determi- nation of ABCG2-ATPase activity is one method to identify ABCG2 substrate and inhibitors. We demonstrate that the ATPase activities of the human ABCG2 transfected Sf9 cell membranes (MXR-Sf9) and ABCG2-overexpressing human cell membranes (MXR-M) differ. Variation due to disparity in the glycosylation level of the protein had no effect on the trans- porter. The influence of cholesterol on ABCG2-ATPase activity was investigated because the lipid compositions of insect and human cells are largely different from each other. Differences in cholesterol content, shown by cholesterol loading and deple- tion experiments, conferred the difference in stimulation of basal ABCG2-ATPase of the two cell membranes. Basal ABCG2-ATPase activity could be stimulated by sulfasalazine, prazosin, and topotecan, known substrates of ABCG2 in cho- lesterol-loaded MXR-Sf9 and MXR-M cell membranes. In con- trast, ABCG2-ATPase could not be stimulated in MXR-Sf9 or in cholesterol-depleted MXR-M membranes. Moreover, choles- terol loading significantly improved the drug transport into in- side-out membrane vesicles prepared from MXR-Sf9 cells. MXR-M and cholesterol-loaded MXR-Sf9 cell membranes dis- played similar ABCG2-ATPase activity and vesicular transport. Our study indicates an essential role of membrane cholesterol for the function of ABCG2. ABCG2 (also referred to as breast cancer resistance protein and MXR) is one of the most important efflux transporters in endothelial and epithelial cells, modulating absorption dis- tribution metabolism excretion properties of drugs and other xenobiotics (for review, see Mao and Unadkat, 2005). ABCG2 exhibits a broad substrate specificity because it transports hydrophobic, anionic, and cationic drugs (Mao and Unadkat, 2005). Therefore, it is widely believed that ABCG2 plays an important role in intestinal absorption (Polli et al., 2004) and secretion of xenobiotics and metabolites (Ebert et al., 2005), secretion of sulfate conjugates in the liver (Zamek-Gliszczyn- ski et al., 2006), and prevention of penetration of drugs into the brain (Breedveld et al., 2005). ABCG2 may play a pivotal role in placenta as a defensive barrier to drugs as well as reducing steroid levels in fetal tissues (Jonker et al., 2000). ABCG2 knockout mice models shed light on some special functions of ABCG2, such as secretion of xenobiotics into the milk (Jonker et al., 2005) and protection of stem cells from hypoxia-induced protoporphyrin accumulation and damage (Krishnamurthy and Schuetz, 2005). ABCG2 function is commonly studied using high-through- put cellular (Robey et al., 2004) or membrane-based assays (Ozvegy et al., 2001; Janvilisri et al., 2003) (for review, see Glavinas et al., 2004; http://www.solvo.com/). For most appli- cations, the transporter is expressed in insect cell lines (e.g., Sf9), taking advantage of the robust baculovirus insect cell This work was supported by Hungarian government Grants INFRA GVOP- 3.3.2.-2004-04 – 0001/3.0, ADME GVOP-3.1.1.-2004-05-0506/3.0, and Asbo ´th XTTPSRT1 and by European Grants EU-FP6 Biosim 005137 and EU FP6 Memtrans 518246. The authors do not have any financial or personal relationships to disclose that could potentially be perceived as influencing the described research. Article, publication date, and citation information can be found at http://jpet.aspetjournals.org. doi:10.1124/jpet.106.119289. ABBREVIATIONS: MXR, mitoxantrone resistance-associated protein; Sf9, Spodoptera frugiperda ovarian; WT, wild type; RAMEB, randomly methylated -cyclodextrin; cholesterol@RAMEB, cholesterol complex of RAMEB. 0022-3565/07/3213-1085–1094$20.00 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS Vol. 321, No. 3 Copyright © 2007 by The American Society for Pharmacology and Experimental Therapeutics 119289/3206609 JPET 321:1085–1094, 2007 Printed in U.S.A. 1085 at JETS L & S Journals 223003 on October 3, 2012 jpet.aspetjournals.org Downloaded from

Transcript of Cholesterol potentiates ABCG2 activity in a heterologous expression system: improved in vitro model...

Cholesterol Potentiates ABCG2 Activity in a HeterologousExpression System: Improved in Vitro Model to Study Functionof Human ABCG2

A. Pal, D. Mehn, E. Molnar, S. Gedey, P. Meszaros, T. Nagy, H. Glavinas, T. Janaky,O. von Richter, G. Bathori, L. Szente, and P. KrajcsiSOLVO Biotechnology, Central Hungarian Innovations Center, Budaors, Hungary (A.P., D.M., E.M., S.G., P.M., T.N., H.G., G.B.,P.K.); Department of Medical Chemistry, University of Szeged, Szeged, Hungary (T.J.); Division of Drug Metabolism andPharmacokinetics, ALTANA Pharma AG, Konstanz, Germany (O.v.R.); CycloLab Cyclodextrin R. and D. Ltd., Budapest,Hungary (L.S.); and Rational Drug Design Laboratories, Budapest, Hungary (P.K.)

Received January 3, 2007; accepted March 5, 2007

ABSTRACTABCG2, a transporter of the ATP-binding cassette family, isknown to play a prominent role in the absorption, distribution,metabolism, and excretion of xenobiotics. Drug-transporter in-teractions are commonly screened by high-throughput systemsusing transfected insect and/or human cell lines. The determi-nation of ABCG2-ATPase activity is one method to identifyABCG2 substrate and inhibitors. We demonstrate that theATPase activities of the human ABCG2 transfected Sf9 cellmembranes (MXR-Sf9) and ABCG2-overexpressing human cellmembranes (MXR-M) differ. Variation due to disparity in theglycosylation level of the protein had no effect on the trans-porter. The influence of cholesterol on ABCG2-ATPase activitywas investigated because the lipid compositions of insect andhuman cells are largely different from each other. Differences in

cholesterol content, shown by cholesterol loading and deple-tion experiments, conferred the difference in stimulation ofbasal ABCG2-ATPase of the two cell membranes. BasalABCG2-ATPase activity could be stimulated by sulfasalazine,prazosin, and topotecan, known substrates of ABCG2 in cho-lesterol-loaded MXR-Sf9 and MXR-M cell membranes. In con-trast, ABCG2-ATPase could not be stimulated in MXR-Sf9 or incholesterol-depleted MXR-M membranes. Moreover, choles-terol loading significantly improved the drug transport into in-side-out membrane vesicles prepared from MXR-Sf9 cells.MXR-M and cholesterol-loaded MXR-Sf9 cell membranes dis-played similar ABCG2-ATPase activity and vesicular transport.Our study indicates an essential role of membrane cholesterolfor the function of ABCG2.

ABCG2 (also referred to as breast cancer resistance proteinand MXR) is one of the most important efflux transporters inendothelial and epithelial cells, modulating absorption dis-tribution metabolism excretion properties of drugs and otherxenobiotics (for review, see Mao and Unadkat, 2005). ABCG2exhibits a broad substrate specificity because it transportshydrophobic, anionic, and cationic drugs (Mao and Unadkat,2005). Therefore, it is widely believed that ABCG2 plays animportant role in intestinal absorption (Polli et al., 2004) and

secretion of xenobiotics and metabolites (Ebert et al., 2005),secretion of sulfate conjugates in the liver (Zamek-Gliszczyn-ski et al., 2006), and prevention of penetration of drugs intothe brain (Breedveld et al., 2005). ABCG2 may play a pivotalrole in placenta as a defensive barrier to drugs as well asreducing steroid levels in fetal tissues (Jonker et al., 2000).ABCG2 knockout mice models shed light on some specialfunctions of ABCG2, such as secretion of xenobiotics into themilk (Jonker et al., 2005) and protection of stem cells fromhypoxia-induced protoporphyrin accumulation and damage(Krishnamurthy and Schuetz, 2005).

ABCG2 function is commonly studied using high-through-put cellular (Robey et al., 2004) or membrane-based assays(Ozvegy et al., 2001; Janvilisri et al., 2003) (for review, seeGlavinas et al., 2004; http://www.solvo.com/). For most appli-cations, the transporter is expressed in insect cell lines (e.g.,Sf9), taking advantage of the robust baculovirus insect cell

This work was supported by Hungarian government Grants INFRA GVOP-3.3.2.-2004-04–0001/3.0, ADME GVOP-3.1.1.-2004-05-0506/3.0, and AsbothXTTPSRT1 and by European Grants EU-FP6 Biosim 005137 and EU FP6Memtrans 518246.

The authors do not have any financial or personal relationships to disclosethat could potentially be perceived as influencing the described research.

Article, publication date, and citation information can be found athttp://jpet.aspetjournals.org.

doi:10.1124/jpet.106.119289.

ABBREVIATIONS: MXR, mitoxantrone resistance-associated protein; Sf9, Spodoptera frugiperda ovarian; WT, wild type; RAMEB, randomlymethylated �-cyclodextrin; cholesterol@RAMEB, cholesterol complex of RAMEB.

0022-3565/07/3213-1085–1094$20.00THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS Vol. 321, No. 3Copyright © 2007 by The American Society for Pharmacology and Experimental Therapeutics 119289/3206609JPET 321:1085–1094, 2007 Printed in U.S.A.

1085

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from

system (Ozvegy et al., 2001). In contrast, membranes can beprepared from human cell lines selected for drug resistanceoverexpressing the transporter (Han and Zhang, 2004). Morerecently, it has turned out that during the selection process,the protein acquired a mutation in the position of amino acid482 (Honjo et al., 2001). The R482G/T mutants display asubstrate specificity different from the wild-type (WT) pro-tein (Honjo et al., 2001; Ozvegy et al., 2002). More strikingly,unlike the ATPase function of the R482G/T mutants, theATPase activity of the WT protein could not be stimulated byprazosin, a known ABCG2 substrate (Xiao et al., 2006), whenexpressed in insect cell membranes (Ozvegy et al., 2001;Ishikawa et al., 2003). We have shown that this nonrespon-siveness is not an inherent property of the transporter be-cause vanadate-sensitive ATPase activity of human cellmembranes expressing similar amounts of ABCG2 can bestimulated by substrate of the transporter (H. Glavinas, E.Kis, A. Pal, R. Kovacs, M. Jani, E. Vagi, E. Molnar, S.Banshagi, Z. Kele, T. Janaky, G. Bathori, O. von Richter,G.-J. Koomen, and P. Krajcsi, unpublished data). Likewise,ATPase activity of ABCG2-expressing membranes isolatedfrom Lactococcus lactis was shown to be stimulated by sim-ilar substrates (Janvilisri et al., 2003).

Because it is of pivotal importance that in vitro modelsclosely mimic the physiological phenotype, we have decidedto study the underlying differences between the ABCG2-ATPase when the transporter was expressed in Sf9 and inhuman cell membranes. We report here that differences inglycosylation of ABCG2 in the two expression systems do notaffect the ATPase activity of the transporter. In contrast, thelow-cholesterol content of the insect membranes profoundlyaffects drug-stimulated ATPase activity of the expressedABCG2 protein because cholesterol loading decreases thebasal ABCG2-ATPase activity in MXR SF9 membranes to adegree similar to the native, cholesterol-rich human MXR-Mmembranes. The modulation of ABCG2 activity by choles-terol may have implications on the localization of the ABCG2transporter in distinct compartments of the plasma mem-brane because two other multidrug resistance protein ABCtransporters ABCB1 (Bacso et al., 2004) and ABCC2 (Tietz etal., 2005) have been shown to localize in the cholesterol-richraft microdomains of cell membranes.

Materials and MethodsChemicals and Biochemicals. [3H]Methotrexate was pur-

chased from Moravek Biochemicals (Brea, CA). [3H]Estrone-3-sul-fate and [3H]prazosin were purchased from Perkin Elmer Life andAnalytical Sciences (Boston, MA). Topotecan was purchased fromLKT Laboratories (St. Paul, MN). The antibody against ABCG2 waspurchased from Abcam (Cambridge, UK). Recombinant baculovi-ruses encoding wild-type human ABCG2 were kind gifts from Prof.B. Sarkadi (National Medical Center, Budapest, Hungary). Ran-domly methylated-�-cyclodextrin (RAMEB) and cholesterol complexof RAMEB (cholesterol@RAMEB; Piel et al., 2006; cholesterol con-tent, 4.74%) was provided by Cyclolab (Cyclodextrin Research andDevelopment Laboratory, Budapest, Hungary). Ko134 and Ko143(Allen et al., 2002) were kind gifts of Prof. G. J. Koomen (NationalCancer Institute, Amsterdam, The Netherlands). Other reagentswere purchased from Sigma-Aldrich (St. Louis, MO) unless statedotherwise in the text.

Membrane Preparation. Human membrane vesicle prepara-tions (MXR-M) as well as membrane vesicle preparations obtainedfrom insect cells expressing ABCG2 (MXR-Sf9) were obtained from

SOLVO Biotechnology (Budapest, Hungary; http://www.solvo.com/).Both membranes are overexpressing the wild-type version ofABCG2. The insect membrane vesicle preparations were producedusing recombinant baculoviruses encoding ABCG2 (Ozvegy et al.,2002). Sf9 cells were cultured and infected with recombinant bacu-lovirus stocks as described earlier (Sarkadi et al., 1992). Purifiedmembrane vesicles from baculovirus-infected Sf9 cells were preparedessentially as described previously (Sarkadi et al., 1992). Membraneprotein content was determined using the BCA method (Pierce Bio-technology, Rockford, IL).

ABCG2 Deglycosylation. Enzymatic deglycosylation was doneusing peptide-N-glycosidase F (Sigma-Aldrich). One microliter of theenzyme at 500 U/ml was added to 50 �l of membrane suspension (5mg/ml protein) in TMEP (50 mM Tris, 50 mM mannitol, 2 mMEGTA, 8 �g/ml aprotinin, 10 �g/ml leupeptin, 50 �g/ml phenylmeth-ylsulfonyl fluoride, and 2 mM dithiothreitol, pH 7.0) vigorouslymixed and incubated at 37°C for 10 min. Deglycosylation was de-tected by Western blotting.

Western Blotting. The proteins were separated using a 10%polyacrylamide gel and transferred to polyvinylidene difluoridemembrane (Immobilon-P; Millipore, Bedford, MA) at 350 mA in atransfer buffer composed of 25 mM Tris, 192 mM glycine, and 15%(v/v) methanol, pH 8.3. The membrane was treated with blockingbuffer (5% nonfat dry milk powder and 0.5% bovine serum albuminin phosphate-buffered saline with 0.05% Tween 20) for 2 h at roomtemperature. The membrane was then incubated with the primaryantibody, a mouse anti-ABCG2 monoclonal antibody BXP-21 (Ab-cam), diluted 1:1000 in blocking buffer for 2 h at room temperature.The membrane was washed for 3 � 10 min with phosphate-bufferedsaline/0.05% Tween 20 at room temperature. It was then incubatedwith the secondary antibody, anti-mouse IgG-HRP, a horseradishperoxidase-conjugated species-specific whole antibody (Sigma-Al-drich) diluted 1:5000 in blocking buffer for 1 h at room temperature.The membrane was subsequently washed as described above, andimmunoreactive bands were visualized with ECL Western BlottingDetection System (GE Healthcare, Buckinghamshire, UK).

ABCG2-ATPase Activity. ATPase activity was measured as de-scribed earlier (Sarkadi et al., 1992). In brief, membrane vesicles (20�g/well) were incubated in ATPase assay buffer (10 mM MgCl2, 40mM MOPS-Tris, pH 7.0, 50 mM KCl, 5 mM dithiothreitol, 0.1 mMEGTA, 4 mM sodium azide, 1 mM ouabain), 5 mM ATP, and variousconcentrations of test drugs for 40 min at 37°C. ATPase activitieswere determined as the difference of inorganic phosphate liberationmeasured with and without the presence of 1.2 mM sodium or-thovanadate (vanadate-sensitive ATPase activity). In the experi-ments presented in Fig. 5, the PREDEASY ABCG2-ATPase Kit(SB-MXR-HAM-PREDEASY-ATPase Kit; SOLVO Biotechnology,Szeged, Hungary) was used for the determination of ABCG2-ATPaseactivity according to the manufacturer’s suggestions.

Vesicular Transport Assay. Inside-out membrane vesicles wereincubated in the presence or absence of 4 mM ATP. For methotrexatevesicular transport, the measurements were carried out in 7.5 mMMgCl2, 40 mM MOPS-Tris, pH 7.0, 70 mM KCl at 37°C for 12 min.The transport was stopped by addition of cold wash buffer (40 mMMOPS-Tris, pH 7.0, 70 mM KCl).

For prazosin vesicular transport, 10 mM Tris-HCl, pH 7.4, 250mM sucrose, and 10 mM MgCl2 containing buffer was incubated at37°C for 20 min. The transport was stopped by addition of cold washbuffer (10 mM Tris-HCl, pH 7.4, 250 mM sucrose, and 100 mMNaCl).

For estrone-3-sulfate vesicular transport, 10 mM Tris-HCl, pH7.4, 250 mM sucrose, and 10 mM MgCl2 containing buffer wereincubated at 32°C for 1 min. The transport was stopped by additionof cold wash buffer (10 mM Tris-HCl, pH 7.4, 250 mM sucrose, and100 mM NaCl).

The incubation mix was then rapidly filtered through class B glassfiber filters (pore size, 0.1 �m). Filters were washed with 5 � 200 �lof ice-cold wash buffer, and radioactivity retained on the filter was

1086 Pal et al.

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from

measured by liquid scintillation counting. ATP-dependent transportwas calculated by subtracting the values obtained in the absence ofATP from those in the presence of ATP.

Determination of Cholesterol Content. The cholesterol con-tent of the membranes was determined using the cholesterol oxidasemethod (Contreras et al., 1992). Ten microliters of reaction mix (500mM MgCl2, 500 mM Tris buffer, 10 mM dithiothreitol, 100 mg TritonX-100, pH 7.4) was mixed with 10 �l of cholesterol oxidase enzyme at1 mg/ml (Roche Diagnostics, Basel, Switzerland) and 50 �l of mem-branes. The solution was incubated at 37°C for 30 min. The reactionwas stopped by adding 100 �l of a methanol/ethanol solution [50%(v/v)] and incubated at 0°C for an additional 30 min. After 5 min ofcentrifugation at 700g, 25 �l of the supernatant was analyzed byhigh-performance liquid chromatography. For chromatographic sep-aration, high-performance liquid chromatography (1100 Series set;Agilent Technologies, Santa Clara, CA) and C18 reverse-phasechromatographic column (3 �M, 100 � 2 mm Luna; Phenomenex,Torrance CA) were used. Analytes were separated using 1% (v/v)acetic acid/methanol as mobile phase at a flow rate of 0.3 ml/min. Theoxidized cholesterol was detected using a UV detector at 241 nm. Toquantitate the amount of cholesterol, external cholesterol calibrationstandards were used.

Cholesterol Loading and Depletion. For optimization ofcholesterol@RAMEB, concentrations used for cholesterol loading,MXR-Sf9, and MXR-M membrane preparations were diluted 10times in ATPase assay buffer containing cholesterol@RAMEB com-plex at concentrations indicated (Fig. 3, A and D). After a 1-h incu-bation at 37°C, membrane vesicles were washed with ATPase assaybuffer and suspended in the reaction medium. For optimization ofcholesterol loading time, the incubation was carried out in the pres-ence of 2 mM cholesterol@RAMEB at 37°C for time periods indicatedin Fig. 3C.

Cholesterol loading of MXR-Sf9 membranes used in Figs. 4 to 8was carried out for 30 min at 37°C in the presence of 1 mMcholesterol@RAMEB complex. In cholesterol depletion studies (Fig. 3B),MXR-M and MXR-Sf9 membranes were diluted 10 times in ATPaseassay buffer containing various concentrations of RAMEB and incu-bated for 30 min at 37°C. Membrane vesicles then were washed withATPase assay buffer and suspended in the reaction medium.

For cholesterol depletion of vesicles used in experiments shown inFigs. 4 and 7, MXR-M membranes were incubated in ATPase assaybuffer containing 8 mM RAMEB for 30 min at 37°C. Then, mem-brane vesicles were washed with ATPase assay buffer and sus-pended in the reaction medium. Control (nontreated) membraneswere incubated and washed similarly in ATPase reaction bufferwithout the presence of RAMEB or cholesterol@RAMEB complex.The cholesterol-loaded transporter-containing membrane prepara-tions are the proprietary technology of SOLVO Biotechnology.

Data Analysis. Assays were run in duplicates. Data are pre-sented as means � S.D. The potencies of drugs to alter ATPaseactivity were obtained from plots of the rate of ATP hydrolysis as afunction of logarithm of drug concentration by nonlinear regressionof the general, sigmoid dose-response equation:

v � Vmin �Vmax � Vmin

1 � 10�logEC50 � �A�� � nH(1)

where v is response (nanomoles of Pi per minute per milligram), Vmin

is minimal response, Vmax is maximal response, EC50 is ligand con-centration producing 50% of maximal response (efficacy), [A] is theactual test drug concentration, and Hill slope is the parameter char-acterizing the degree of cooperativity. EC50 is defined according tothe International Union of Pharmacology Committee (Neubig et al.,2003).

The transporters often have at least one high-affinity and onelow-affinity inhibitory binding site for the same drug. Therefore,bell-shaped response curves were frequently obtained. For these

types of curves, an equation that is a combination of two sigmoidresponses was used:

v � Dip �Vmin 1 � Dip

1 � 10�logEC501 � log�A�� � nH1�

Vmin 2 � Dip1 � 10�logEC502 � log�A�� � nH2

(2)

where v is response (nanomoles of Pi per minute per milligram),Vmin1 and Vmin2 are minimal responses, Dip is maximal response,EC501 and EC502 are ligand concentrations producing 50% of maxi-mal response (efficacy), [A] is the actual test drug concentration, andHill slope 1 and Hill slope 2 are the constants of cooperativity.

The Michaelis-Menten parameters of maximal velocity (Vmax) anddrug affinity (Km) were obtained from plots of the ATPase activity asa function of test drug concentration by nonlinear regression of thefollowing equation:

v �Vmax � �S�

km � �S�(3)

where v is enzyme activity (nanomoles of Pi per minute per milli-gram), Vmax is maximal ATPase activity (nanomoles of Pi per minuteper milligram), Km is the Michaelis-Menten constant for the testedsubstrate (nanomolar) and [S] substrate concentration (nanomolar).Equation 3 was used also for curve fitting on vesicular transportgraphs. For curve fitting, Vmax and Km slope calculations fromPRISM 3.0 software (GraphPad Software Inc., San Diego, CA) wereused.

ResultsKnown Substrates Show Different ATPase Profiles

in ABCG2-Expressing Insect and Human Membranes.To investigate the relevance of the heterologous baculovirus-insect cell expression system, the WT human ABCG2 proteinwas expressed in Sf9 cells. The membranes purified from theABCG2-overexpressing insect cells (MXR-Sf9 membranes)were studied along with membranes purified from ABCG2-overexpressing human cells (MXR-M membranes; http://www.solvo.com). Using prazosin, a known substrate of theWT human ABCG2 (Xiao et al., 2006), as well as sulfasala-zine, a substrate of the mouse breast cancer resistance pro-tein 1 (Zaher et al., 2006) and a substrate of the human protein(van der Heijden et al., 2004), a clear difference was seen inthe ATPase stimulation profile of ABCG2 expressed in thetwo membranes. Sulfasalazine and prazosin stimulated the

Fig. 1. Comparison of the ATPase activity profile of membranes preparedfrom ABCG2 transfected Sf9 insect cells (MXR-Sf9) and membranesprepared from ABCG2-overexpressing human cells (MXR-M). Fold-acti-vation is calculated as ratio of substrate-stimulated and basal vanadate-sensitive ATPase activity. Each column represents the mean of triplicatedeterminations.

Novel ABCG2 Model Membranes 1087

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from

ABCG2-ATPase activity (3.9- and 2.7-fold, respectively) whenexpressed in human cell membrane (Fig. 1). In contrast, in theSf9 system, neither sulfasalazine nor prazosin did significantlymodulate the ABCG2-ATPase activity (Fig. 1). The observeddifferences could not be attributed to differences in expression

levels because the proteins were overexpressed at similarlyhigh levels in the two systems (Fig. 2A).

The Difference in Glycosylation of ABCG2 in Sf9 ver-sus Human Membranes Does Not Confer the Differ-ence in ATPase Activation Profiles. The ABCG2 content

Fig. 2. Deglycosylation does not affect kinetics ofMXR-M ATPase. A, representative Western blotof ABCG2 in MXR-M and MXR-Sf9 membranes.Membrane fractions from ABCG2-overexpress-ing human cells were loaded as a reference forthe fully glycosylated protein (lane 1) and fullydeglycosylated protein after peptide-N-glycosi-dase (0.5 U) treatment (lane 2). Native MXR-Sf9membrane was loaded as control (lane 3). B,inhibition of basal ABCG2-ATPase activity ofdeglycosylated MXR-M membrane (�) and un-treated control (Œ) by the ABCG2-specific inhib-itor Ko134. C, activation of deglycosylatedMXR-M membrane (�) and untreated control(Œ) by estrone-3-sulfate. Data represent mean �S.D. of duplicates.

Fig. 3. Cholesterol content modulates ABCG2-ATPase activity in membrane preparations fromMXR-Sf9 and MXR-M. A, MXR-Sf9 (Œ) andMXR-M (f) membranes were loaded with cho-lesterol by treatment with cholesterol@RAMEBcomplex. Membrane cholesterol content was de-termined by the cholesterol oxidase method.Concentrations on x-axis represent the total cho-lesterol concentration in the incubation medium.B, cyclodextrin-mediated cholesterol depletion ofMXR-Sf9 (Œ) and MXR-M (f) membranes wascarried out by incubating the membranes for 30min at 37°C in the presence of RAMEB at con-centrations indicated in the figure. C and D,optimization of cholesterol loading. Basal (F)and sulfasalazine-stimulated (Œ) activities weremonitored after completing the loading proce-dure. For optimizing the incubation time (C),MXR-Sf9 membranes were incubated in thepresence of cholesterol@RAMEB (2 mM totalcholesterol in the incubation medium) for timesindicated in the figure. For optimization of cho-lesterol loading (D), MXR-Sf9 membranes wereincubated in the presence of various cholesterolconcentrations for 1 h. ABCG2-ATPase activitydata represent mean � S.D. of duplicate mea-surements.

1088 Pal et al.

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from

and glycosylation level of the membrane preparations wereanalyzed by Western blotting with BXP-21, an ABCG2-spe-cific antibody (Fig. 2A). MXR-M membranes (Fig. 2A, lane 1)and MXR-Sf9 membrane preparations (Fig. 2A, lane 3), bothoverexpressing ABCG2, displayed strong bands with appar-ent molecular masses of 75 to 80 and 60 to 65 kDa, respec-tively. ABCG2 is a glycosylated protein (Diop and Hrycyna,2005; Mohrmann et al., 2005), and it has been shown that inSf9 cells, the protein is severely underglycosylated becauseinhibition of glycosylation in a human cell line using tunica-mycin yielded nonglycosylated species with a mobility corre-sponding to a molecular mass of 60 to 65 kDa that is similarto the mobility of ABCG2 expressed in Sf9 membranes (Oz-vegy et al., 2001). However, in accordance with publisheddata (Ozvegy et al., 2001), tunicamycin did not yield completeunglycosylation (data not shown); therefore, we used peptide-N-glycosidase F to deglycosylate ABCG2 expressed in

MXR-M membranes. The enzymatic cleavage resulted in acomplete deglycosylation of ABCG2 (Fig. 2A, lane 2).

Ko134, a known specific inhibitor of ABCG2, fully inhibitedthe transporter-dependent vanadate-sensitive basal activityof the glycosylated and deglycosylated forms of the proteinwith equal potency (Fig. 2B). Likewise, no difference wasobserved in the estrone-3-sulfate-stimulated ATPase activa-tion profile of the fully glycosylated and deglycosylated forms(Fig. 2C). Therefore, we conclude that differences in the gly-cosylation of ABCG2 in the Sf9 and the human membranesdo not explain the observed differences in drug-stimulatedATPase activity of the protein.

Cholesterol Loading Potentiates Drug-InducedATPase Stimulation of ABCG2-Expressing MXR-Sf9Membranes. It has been known that lipid composition ofinsect and mammalian membranes significantly differ. Oneof the major difference is the cholesterol content of Sf9

Fig. 4. Cholesterol loading of MXR-Sf9membranes makes their ABCG2-ATPaseactivity profile similar to the ABCG2-AT-Pase activity profile of MXR-M mem-branes. MXR-Sf9 cells were loaded withcholesterol using cholesterol@RAMEB (1mM total cholesterol) for 30 min at 37°C.Cholesterol depletion of MXR-M mem-branes were carried out at 37°C for 30min using 8 mM RAMEB. The ABCG2-ATPase activation of cholesterol-loaded(Œ) and control (F) MXR-Sf9 membranes(A, C, and D) as well as cholesterol-de-pleted (Œ) and control (F) MXR-M mem-branes (B, D, and F) upon sulfasalazine(A and B), topotecan (C and D), and pra-zosin (E and D) treatment (40 min, 37°C)was determined. Data represent mean �S.D. of triplicates.

Novel ABCG2 Model Membranes 1089

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from

plasma membrane being approximately 5- to 10-fold lowerthan that of human plasma membrane (Gimpl et al., 1995).As expected, the cholesterol contents of the MXR-Sf9 andMXR-M vesicles containing ABCG2 protein were markedlydifferent, with MXR-Sf9 displaying an approximately 4- to5-fold lower cholesterol level [6.5 versus 28.99 �g/mgprotein (Fig. 3A) and 6.26 versus 23.68 �g/mg protein(Fig. 3B)]. Cholesterol loading of both membranes usingcholesterol@RAMEB treatment resulted in approximately a15-fold increase of cholesterol content in MXR-Sf9 vesiclesand 3-fold increase in MXR-M vesicles yielding 90 �g/mgprotein final cholesterol content in both membranes (Fig.3A). On the other hand, RAMEB treatment removed thecholesterol from both membranes very efficiently (Fig. 3B).

For further experiments, cholesterol loading of ABCG2-expressing Sf9 membranes has been optimized for ATPaseactivity with respect to exposure time (Fig. 3C) and loading

concentration of cholesterol (Fig. 3D). For experimentsshown in Figs. 4 to 7, 30 min and 1 mM cholesterol@RAMEBwere chosen as optimal time and concentration. The mem-brane cholesterol content after treatment is around 60 �g/mgprotein, approximately 2-fold greater than in the MXR-Mmembrane. We selected these values because under theseconditions the membrane cholesterol content was relativelyinsensitive for either parameter (Fig. 3, C and D); therefore,it allowed for reproducible production of cholesterol-loadedmembranes. For cholesterol depletion of MXR-M mem-branes, a 30-min treatment using 8 mM RAMEB was se-lected, yielding membrane preparations with an average cho-lesterol content of 3.5 �g/mg protein.

Differences in Cholesterol Content Confer the Dif-ference in ATPase Stimulation of ABCG2-ExpressingMXR-Sf9 and MXR-M Membranes. The ATPase activity ofWT ABCG2-expressing Sf9 membranes was refractory to

Fig. 5. Effect of activators and inhib-itors on basal (Œ) and sulfasalazine-stimulated (F) ABCG2-ATPase activ-ity. A, sulfasalazine. B, prazosin. C,topotecan. D, Hoechst 33342. E,Ko143. The ABCG2-ATPase activitymeasurements were carried out usingthe PREADEASY ATPase kit accord-ing to the manufacturer’s suggestions.Data represent mean � S.D. of dupli-cates.

1090 Pal et al.

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from

stimulation upon treatment with sulfasalazine (Fig. 4A).Cholesterol loading significantly potentiated sulfasalazine-induced activation of ABCG2-ATPase (Fig. 4A). In contrast,ABCG2 activity of MXR-M membranes was stimulated bysulfasalazine in a concentration-dependent manner (Fig. 4B)without requiring cholesterol treatment. Depletion of choles-terol of the MXR-M membranes by means of RAMEB treat-ment impaired the ABCG2-ATPase response (Fig. 4B). Boththe concentration dependence and the general biphasic shapeof the cholesterol-loaded MXR-Sf9 ABCG2-ATPase closelyresembled that of the MXR-M ATPase activation (Fig. 4, Aand B). Moreover, the low-cholesterol versions of both mem-branes (native MXR-Sf9 and cholesterol-depleted MXR-M)were also similar (Fig. 4, A and B) because ABCG2-ATPasewas not efficiently activated by sulfasalazine in either mem-brane. Likewise, topotecan and prazosin both significantlystimulated ABCG2-ATPase in cholesterol-loaded MXR-Sf9(Fig. 4, C and E) and MXR-M membranes (Fig. 4, D and F),whereas untreated MXR-Sf9 (Fig. 4, C and E) and cholester-ol-depleted MXR-M membranes (Fig. 4, D and F) were non-responsive. Kinetic data on the ABCG2-ATPase are summa-rized in Table 1. The data show good correlation betweenVmax values obtained with the cholesterol-loaded MXR-Sf9membranes and MXR-M membranes. However, the Km val-ues observed for the substrates were approximately 1.5 to 3.0times greater with the cholesterol-loaded MXR-Sf9 mem-branes.

Using the PREDEASY ABCG2-ATPase assay kit, choles-terol-loaded MXR-Sf9 membranes were tested for ATPaseactivation and inhibition. In the inhibition assay, the effect ofsubstrates and inhibitors on the sulfasalazine (10 �M)-stim-ulated cholesterol-loaded MXR-Sf9 ABCG2-ATPase weremonitored. All substrates, sulfasalazine, prazosin, and topo-tecan (Fig. 5, A–C), but Hoechst 33342 (Fig. 5D) stimulatedthe basal ATPase activity and inhibited the activatedATPase in a concentration-dependent manner. Hoechst33342, a known substrate of WT human ABCG2, however,displayed a profile similar to Ko134 (Fig. 5E), a known spe-cific inhibitor of ABCG2, because both compounds inhibitedthe basal as well as the stimulated ATPase activity ofABCG2.

Cholesterol Loading Significantly Enhances ABCG2-Mediated Vesicular Transport into MXR-Sf9 Inside-Out Vesicles. The membrane assay system allows for trans-port studies as the transported substrates accumulate in theinside-out vesicles in an ATP-dependent manner. To studywhether cholesterol loading affects transport kinetics ofABCG2-expressing MXR-Sf9 vesicles, we monitored thetransport of a known ABCG2 substrate, methotrexate, com-monly used in VT studies (Fig. 6A). To evaluate the choles-terol effect, Vmax and Km have been calculated. Cholesterolloading dramatically increased maximal velocity of the trans-port (2144 versus 367 pmol/mg/min) without affecting Km

(1068 versus 935 �M). We also studied prazosin transport.Prazosin, although a known ABCG2 substrate (Xiao et al.,2006), is not commonly used in VT studies. Indeed, littleATP-dependent transport (85 pmol/mg/min) was seen in theMXR-Sf9 vesicles (Fig. 6B). On the contrary, a very signifi-cant transport was seen in the cholesterol-loaded MXR-Sf9vesicles with a maximal velocity of 702 pmol/mg/min(Fig. 6B).

Effects of Cholesterol on ATPase and VT Measure-ments Correlate. To study the effect of membrane choles-terol levels on ABCG2-ATPase activity and vesicular trans-port, estrone-3-sulfate was used as a substrate (Fig. 7).Cholesterol loading increased Vmax of transport of estrone-3-sulfate by more than 20-fold (3408 versus 122.7 pmol/mg/min) with relatively little change in Km (14.11 versus 27.58�M) (Fig. 7A). In the MXR-M membranes, a similar effect ofmembrane cholesterol level was observed because cholesteroldepletion decreased Vmax by approximately 5-fold (455.5 ver-sus 99.02 pmol/mg/min) with little change in Km (5.49 versus7.89 �M) (Fig. 7B). The increased transport rate was paral-leled in the ATPase assay because low-cholesterol mem-branes (MXR-Sf9 and cholesterol-depleted MXR-M) dis-played an impaired activation upon estrone-3-sulfatetreatment (Fig. 7, C and D). Moreover, the Km and Vmax

values for estrone-3-sulfate-induced ABCG2-ATPase activityshowed good correlation between the cholesterol-loadedMXR-Sf9 and MXR-M membranes (Table 1). The correlationbetween membrane cholesterol content of MXR-Sf9 vesiclesloaded with different concentrations of cholesterol@RAMEBcomplex and initial rate of [3H]estrone-3-sulfate (1 �M)transport was excellent (Fig. 8). The correlation betweenmembrane cholesterol content and initial rate of sulfasala-zine (10 �M)-induced ATPase activity was reasonably good(Fig. 8).

Fig. 6. Effect of cholesterol loading on vesicular transport activity ofABCG2 containing MXR-Sf9 membranes. Methotrexate (A) and prazosin(B) uptake of cholesterol-loaded (Œ) and control (F) inside-out membranevesicles was carried out at 37°C for 12 and 20 min, respectively. Theamount of radiolabeled compounds retained by the inside-out vesicleswas measured at various drug concentrations as indicated in the figure.The data were calculated as a difference of measured vesicular uptake inthe presence and absence of ATP. Data represent mean � S.D. of dupli-cates.

Novel ABCG2 Model Membranes 1091

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from

DiscussionABCG2 protein is one of the most important multidrug

transporters involved in drug absorption distribution metab-olism excretion. Many assays used to study the effects of drugtransporters are based on membranes purified from Sf9 in-sect cells into which the respective transporter gene is intro-duced by means of baculoviral infection. Utilization of anyheterologous expression systems should warrant thoroughcorrelation and validation studies. However, few studies ad-dress directly this correlation. One way of assay validation iscross-validation with other assays. For ABCB1, daunomycinEC50 values in the MDR1-Sf9 ATPase assay showed an ac-ceptable correlation with the reversal of daunomycin resis-tance in the human cell line (2 and 5 �M, respectively) (forreview, see Litman et al., 2001). The correlation of transportwas even better for the ABCC1/MRP1-mediated LTC4 trans-port (Km in HeLa-MRP1 and MRP1-Sf9 cells, 97 versus 67nM; Leier et al., 1994; Gao et al., 1996, respectively) and ratAbcb11/Bsep-mediated taurocholate transport in the Sf9 sys-tem and rat canalicular membranes (Km in rat liver canalic-

ular membranes and BsepSf9 membranes were 2.1 versus5.0 �M) (Stieger et al., 2000).

There was good correlation between the ATP dependenceof the ABCG2-mediated methotrexate transport and sub-strate specificity of transport inhibition studies in MXR-Sf9and MXR-M vesicles (Glavinas et al., unpublished data).However, there was no correlation in drug-stimulatedABCG2-ATPase profiles in plasma membrane preparationfrom Sf9 cells transfected with ABCG2 and plasma mem-branes from human cells overexpressing the transporter(Fig. 1) (Glavinas et al., unpublished data). We hypothesizedthat either the altered glycosylation and/or the differentmembrane composition of Sf9 and of human plasma mem-

Fig. 7. Effect of cholesterol on ABCG2vesicular transport and ABCG2-ATPase correlates in both the MXR-Sf9 and MXR-M membranes. Estrone-3-sulfate transport was carried out at32°C for 1 min and monitored by mea-suring the amount of membrane-asso-ciated [3H]estrone-3-sulfate (A andB). In estrone-3-sulfate-stimulatedABCG2-ATPase activity measure-ments, membranes were incubatedfor 40 min at 37°C with estrone-3-sulfate at concentrations indicated inthe figure (C and D). Cholesterol-loaded (F) and control (Œ) MXR-Sf9membranes (A and C) and cholesterol-depleted (Œ) and control (F) MXR-Mmembranes (B and C) were studied.Data represent mean � S.D. of dupli-cates.

TABLE 1Comparison of ABCG2-ATPase kinetics of sulfasalazine, estrone-3-sulfate, and topotecan obtained in cholesterol-loaded MXR-Sf9 andnative MXR-M membranes

Test Drug

MXR-Sf9 CholesterolLoaded MXR-M

Vmax* Km† Vmax* Km

Sulfasalazine 26.30 2.46 21.18 0.76Estrone-3-sulfate 35.58 26.42 38.87 15.12Topotecan 5.64 18.06 5.83 8.54

* nanomoles of Pi per milligram of protein per minute.† Micromolar.

Fig. 8. Correlation of membrane cholesterol content and ABCG2 activityin MXR-Sf9 membranes. Cholesterol-loaded membranes were preparedby the treatment of MXR-Sf9 with various concentrations of choles-terol@RAMEB. The cholesterol concentration of cholesterol@RAMEB-treated membranes is shown in the x-axis. Transport rate of estrone-3-sulfate (1 �M) measured at 32°C for 1 min and sulfasalazine (10 �M)-stimulated ATPase activity measured at 37°C for 40 min is shown in they-axis. Data are plotted as mean � S.D. of duplicates.

1092 Pal et al.

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from

brane vesicles could be responsible for the differences ob-served in the activation profile of ABCG2-ATPase. Deglyco-sylation of human WT ABCG2 expressed in human MXR-Mmembranes did not affect the ATPase activity (Fig. 2, B andC). Therefore, the effect of cholesterol on the ABCG2-ATPaseactivation was studied because insect cells harbor much lesscholesterol in their membrane than human cells (Gimpl etal., 1995). Indeed, cholesterol loading specifically improvedthe rate of drug-stimulated ABCG2-ATPase and maximalvelocity of transport for the substrates studied in vesiculartransport experiments (Figs. 3–8). Cholesterol loading ofMXR-Sf9 membranes makes their ATPase (Table 1; Figs. 4and 7, C and D) and transport properties (Fig. 7, A and B)similar to the MXR-M membranes that contain high levelsof endogenous cholesterol. Therefore, cholesterol-loadedABCG2-overexpressing insect cell membranes are suitablemodels to study ABCG2 function. Our results have two majorimplications on data generated earlier, using the native,low-cholesterol MXR-Sf9 membranes. Cholesterol loadingmainly affected enzyme activity with relatively little effect onthe affinity to ABCG2. Therefore, vesicular transport datagenerated in the past using plasma membranes preparedfrom insect cells with the aim of identifying ABCG2 sub-strates as well as substrate specificities are not affected.However, ABCG2-ATPase data using the MXR-Sf9 systemmay have given false negatives especially for high-affinitysubstrates. This supports the notion that cholesterol-loadedMXR-Sf9 membranes are the test systems of choice to studydrug-ABCG2 interactions at high throughput.

Importantly, the effect of cholesterol loading inhibited thetransport activity of endogenous calcium ATPase in the Sf9membranes (data not shown). No significant change wasobserved in permeability of membranes for the substratesused in the vesicular transport studies (data not shown).Therefore, the observed potentiating effect of cholesterol ontransport rate by ABCG2 is not a nonspecific event due toincreased vesicular size or decreased passive permeability ofsubstrates.

A few studies addressing the effect of cholesterol on ABCtransporters have been published so far (Garrigues et al.,2002; Arima et al., 2004). ABCB1 is the only ABC transporterwhere the effect of cholesterol on transporter activity hasbeen investigated in detail (Garrigues et al., 2002). Likewise,to our observations (data not shown), most studies havereported an elevation of basal ABCB1 ATPase activity (Wanget al., 2000; Garrigues et al., 2002; Gayet et al., 2005). Onestudy has attributed this increase in ABCB1 ATPase activityto ABCB1-mediated cholesterol flipping (Garrigues et al.,2002). In some studies, cholesterol depletion resulted in anincreased stimulation of ABCB1 ATPase activity upon sub-strate treatment (Garrigues et al., 2002) or increased trans-port rate (Wang et al., 2000; Gayet et al., 2005). It has beensuggested that cholesterol is an ABCB1 substrate, a conclu-sion challenged lately (Le Goff et al., 2006).

The mechanism of the stimulating effect of cholesterol onABCG2-function is not clear. The elevated ABCG2-ATPasebaseline levels may imply that cholesterol is an ABCG2 sub-strate. This would not be unprecedented in the ABCG familybecause other members, ABCG1 and ABCG4 (Wang et al.,2004), as well as the ABCG5/ABCG8 dimer (Yu et al., 2002)have been implicated in cholesterol transport. However, wehave not observed any signs of inhibition of ABCG2 transport

exerted by cholesterol in our studies, questioning the sub-strate nature of cholesterol. Alternatively, cholesterol mayact as an allosteric regulator for ABCG2-function. Furtherstudies are required to elucidate the mechanism of choles-terol-mediated potentiation of ABCG2-ATPase activity andvesicular transport.

Cholesterol forms separate domains (rafts or caveolae) inmammalian membranes. Interestingly, several reports foundmultidrug transporters localized in the raft/caveolar regions(Bacso et al., 2004; Tietz et al., 2005). One may speculate thatthe cholesterol sensitivity of the ABCG2 protein might berelated to a raft/caveolar localization of the transporter.

Acknowledgments

The expert help of Judit Janossy in reviewing and preparation ofthe manuscript is acknowledged.

ReferencesAllen JD, van Loevezijn A, Lakhai JM, van der Valk M, van Tellingen O, Reid G,

Schellens JH, Koomen GJ, and Schinkel AH (2002) Potent and specific inhibitionof the breast cancer resistance protein multidrug transporter in vitro and in mouseintestine by a novel analogue of fumitremorgin C. Mol Cancer Ther 1:417–425.

Arima H, Yunomae K, Morikawa T, Hirayama F, and Uekama K (2004) Contributionof cholesterol and phospholipids to inhibitory effect of dimethyl-beta-cyclodextrinon efflux function of P-glycoprotein and multidrug resistance-associated protein 2in vinblastine-resistant Caco-2 cell monolayers. Pharm Res 21:625–634.

Bacso Z, Nagy H, Goda K, Bene L, Fenyvesi F, Matko J, and Szabo G (2004) Raft andcytoskeleton associations of an ABC transporter: P-glycoprotein. Cytometry A61:105–116.

Breedveld P, Pluim D, Cipriani G, Wielinga P, van Tellingen O, Schinkel AH, andSchellens JH (2005) The effect of Bcrp1 (Abcg2) on the in vivo pharmacokineticsand brain penetration of imatinib mesylate (Gleevec): implications for the use ofbreast cancer resistance protein and P-glycoprotein inhibitors to enable the brainpenetration of imatinib in patients. Cancer Res 65:2577–2582.

Contreras JA, Castro M, Bocos C, Herrera E, and Lasuncion MA (1992) Combinationof an enzymatic method and HPLC for the quantitation of cholesterol in culturedcells. J Lipid Res 33:931–936.

Diop NK and Hrycyna CA (2005) N-Linked glycosylation of the human ABC trans-porter ABCG2 on asparagine 596 is not essential for expression, transport activity,or trafficking to the plasma membrane. Biochemistry 44:5420–5429.

Ebert B, Seidel A, and Lampen A (2005) Identification of BCRP as transporter ofbenzopyrene conjugates metabolically formed in Caco-2 cells and its induction byAh-receptor agonists. Carcinogenesis 26:1754–1763.

Gao M, Loe DW, Grant CE, Cole SP, and Deeley RG (1996) Reconstitution ofATP-dependent leukotriene C4 transport by Co-expression of both half-moleculesof human multidrug resistance protein in insect cells. J Biol Chem 271:27782–27787.

Garrigues A, Escargueil AE, and Orlowski S (2002) The multidrug transporter,P-glycoprotein, actively mediates cholesterol redistribution in the cell membrane.Proc Natl Acad Sci USA 99:10347–10352.

Gayet L, Dayan G, Barakat S, Labialle S, Michaud M, Cogne S, Mazane A, ColemanAW, Rigal D, and Baggetto LG (2005) Control of P-glycoprotein activity by mem-brane cholesterol amounts and their relation to multidrug resistance in humanCEM leukemia cells. Biochemistry 44:4499–4509.

Gimpl G, Klein U, Reilander H, and Fahrenholz F (1995) Expression of the humanoxytocin receptor in baculovirus-infected insect cells: high-affinity binding is in-duced by a cholesterol-cyclodextrin complex. Biochemistry 34:13794–13801.

Glavinas H, Krajcsi P, Cserepes J, and Sarkadi B (2004) The role of ABC transport-ers in drug resistance, metabolism and toxicity. Curr Drug Deliv 1:27–42.

Han B and Zhang JT (2004) Multidrug resistance in cancer chemotherapy andxenobiotic protection mediated by the half ATP-binding cassette transporterABCG2. Curr Med Chem Anticancer Agents 4:31–42.

Honjo Y, Hrycyna CA, Yan QW, Medina-Perez WY, Robey RW, van de Laar A,Litman T, Dean M, and Bates SE (2001) Acquired mutations in the MXR/BCRP/ABCP gene alter substrate specificity in MXR/BCRP/ABCP-overexpressing cells.Cancer Res 61:6635–6639.

Ishikawa T, Kasamatsu S, Hagiwara Y, Mitomo H, Kato R, and Sumino Y (2003)Expression and functional characterization of human ABC transporter ABCG2variants in insect cells. Drug Metab Pharmacokinet 18:194–202.

Janvilisri T, Venter H, Shahi S, Reuter G, Balakrishnan L, and van Veen HW (2003)Sterol transport by the human breast cancer resistance protein (ABCG2) ex-pressed in Lactococcus lactis. J Biol Chem 278:20645–20651.

Jonker JW, Merino G, Musters S, van Herwaarden AE, Bolscher E, Wagenaar E,Mesman E, Dale TC, and Schinkel AH (2005) The breast cancer resistance proteinBCRP (ABCG2) concentrates drugs and carcinogenic xenotoxins into milk. NatMed 11:127–129.

Jonker JW, Smit JW, Brinkhuis RF, Maliepaard M, Beijnen JH, Schellens JH, andSchinkel AH (2000) Role of breast cancer resistance protein in the bioavailabilityand fetal penetration of topotecan. J Natl Cancer Inst 92:1651–1656.

Krishnamurthy P and Schuetz JD (2005) The ABC transporter Abcg2/Bcrp: role inhypoxia mediated survival. Biometals 18:349–358.

Le Goff W, Settle M, Greene DJ, Morton RE, and Smith JD (2006) Reevaluation of

Novel ABCG2 Model Membranes 1093

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from

the role of the multidrug-resistant P-glycoprotein in cellular cholesterol homeosta-sis. J Lipid Res 47:51–58.

Leier I, Jedlitschky G, Buchholz U, and Keppler D (1994) Characterization of theATP-dependent leukotriene C4 export carrier in mastocytoma cells. Eur J Biochem220:599–606.

Litman T, Druley TE, Stein WD, and Bates SE (2001) From MDR to MXR: newunderstanding of multidrug resistance systems, their properties and clinical sig-nificance. Cell Mol Life Sci 58:931–959.

Mao Q and Unadkat JD (2005) Role of the breast cancer resistance protein (ABCG2)in drug transport. AAPS J 7:E118–133.

Mohrmann K, van Eijndhoven MA, Schinkel AH, and Schellens JH (2005) Absenceof N-linked glycosylation does not affect plasma membrane localization of breastcancer resistance protein (BCRP/ABCG2). Cancer Chemother Pharmacol 56:344–350.

Neubig RR, Spedding M, Kenakin T, and Christopoulos A (2003) International Unionof Pharmacology Committee on Receptor Nomenclature and Drug Classification:XXXVIII. Update on terms and symbols in quantitative pharmacology. PharmacolRev 55:597–606.

Ozvegy C, Litman T, Szakacs G, Nagy Z, Bates S, Varadi A, and Sarkadi B (2001)Functional characterization of the human multidrug transporter, ABCG2, ex-pressed in insect cells. Biochem Biophys Res Commun 285:111–117.

Ozvegy C, Varadi A, and Sarkadi B (2002) Characterization of drug transport, ATPhydrolysis, and nucleotide trapping by the human ABCG2 multidrug transporter:modulation of substrate specificity by a point mutation. J Biol Chem 277:47980–47990.

Piel G, Piette M, Barillaro V, Castagne D, Evrard B, and Delattre L (2006) Beta-methasone-in-cyclodextrin-in-liposome: the effect of cyclodextrins on encapsula-tion efficiency and release kinetics. Int J Pharm 312:75–82.

Polli JW, Baughman TM, Humphreys JE, Jordan KH, Mote AL, Webster LO,Barnaby RJ, Vitulli G, Bertolotti L, Read KD, et al. (2004) The systemic exposureof an N-methyl-D-aspartate receptor antagonist is limited in mice by the P-glycoprotein and breast cancer resistance protein efflux transporters. Drug MetabDispos 32:722–726.

Robey RW, Steadman K, Polgar O, Morisaki K, Blayney M, Mistry P, and Bates SE(2004) Pheophorbide a is a specific probe for ABCG2 function and inhibition.Cancer Res 64:1242–1246.

Sarkadi B, Price EM, Boucher RC, Germann UA, and Scarborough GA (1992)

Expression of the human multidrug resistance cDNA in insect cells generates ahigh activity drug-stimulated membrane ATPase. J Biol Chem 267:4854–4858.

Stieger B, Fattinger K, Madon J, Kullak-Ublick GA, and Meier PJ (2000) Drug- andestrogen-induced cholestasis through inhibition of the hepatocellular bile saltexport pump (Bsep) of rat liver. Gastroenterology 118:422–430.

Tietz P, Jefferson J, Pagano R, and Larusso NF (2005) Membrane microdomains inhepatocytes: potential target areas for proteins involved in canalicular bile secre-tion. J Lipid Res 46:1426–1432.

van der Heijden J, de Jong MC, Dijkmans BA, Lems WF, Oerlemans R, KathmannI, Schalkwijk CG, Scheffer GL, Scheper RJ, and Jansen G (2004) Development ofsulfasalazine resistance in human T cells induces expression of the multidrugresistance transporter ABCG2 (BCRP) and augmented production of TNFalpha.Ann Rheum Dis 63:138–143.

Wang E, Casciano CN, Clement RP, and Johnson WW (2000) Cholesterol interactionwith the daunorubicin binding site of P-glycoprotein. Biochem Biophys Res Com-mun 276:909–916.

Wang N, Lan D, Chen W, Matsuura F, and Tall AR (2004) ATP-binding cassettetransporters G1 and G4 mediate cellular cholesterol efflux to high-density lipopro-teins. Proc Natl Acad Sci USA 101:9774–9779.

Xiao Y, Davidson R, Smith A, Pereira D, Zhao S, Soglia J, Gebhard D, de Morais S,and Duignan DB (2006) A 96-well efflux assay to identify ABCG2 substrates usinga stably transfected MDCK II cell line. Mol Pharm 3:45–54.

Yu L, Hammer RE, Li-Hawkins J, Von Bergmann K, Lutjohann D, Cohen JC, andHobbs HH (2002) Disruption of Abcg5 and Abcg8 in mice reveals their crucial rolein biliary cholesterol secretion. Proc Natl Acad Sci USA 99:16237–16242.

Zaher H, Khan AA, Palandra J, Brayman TG, Yu L, and Ware JA (2006) Breastcancer resistance protein (Bcrp/abcg2) is a major determinant of sulfasalazineabsorption and elimination in the mouse. Mol Pharm 3:55–61.

Zamek-Gliszczynski MJ, Nezasa K, Tian X, Kalvass JC, Patel NJ, Raub TJ, andBrouwer KL (2006) The important role of Bcrp (Abcg2) in the biliary excretion ofsulfate and glucuronide metabolites of acetaminophen, 4-methylumbelliferone,and harmol in mice. Mol Pharmacol 70:2127–2133.

Address correspondence to: Dr. Peter Krajcsi, SOLVO Biotechnology, Cen-tral Hungarian Innovations Center, Gyar u. 2., H-2040 Budaors, Hungary.E-mail: [email protected]

1094 Pal et al.

at JET

S L &

S Journals 223003 on O

ctober 3, 2012jpet.aspetjournals.org

Dow

nloaded from