+ All Categories
Home > Documents > UvA-DARE (Digital Academic Repository) Basolateral and ... · terminated by the addition of 20 vol...

UvA-DARE (Digital Academic Repository) Basolateral and ... · terminated by the addition of 20 vol...

Date post: 20-Sep-2019
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
18
UvA-DARE is a service provided by the library of the University of Amsterdam (http://dare.uva.nl) UvA-DARE (Digital Academic Repository) Basolateral and apical ABC transporters in liver and intestine de Waart, D.R. Link to publication Citation for published version (APA): de Waart, D. R. (2011). Basolateral and apical ABC transporters in liver and intestine. General rights It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible. Download date: 03 Jan 2020
Transcript

UvA-DARE is a service provided by the library of the University of Amsterdam (http://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

Basolateral and apical ABC transporters in liver and intestine

de Waart, D.R.

Link to publication

Citation for published version (APA):de Waart, D. R. (2011). Basolateral and apical ABC transporters in liver and intestine.

General rightsIt is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s),other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulationsIf you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, statingyour reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Askthe Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam,The Netherlands. You will be contacted as soon as possible.

Download date: 03 Jan 2020

113

Chapter 6

114

Oral availability of cefadroxil depends on ABCC3 and ABCC4.

Dirk R de Waart, Koen van de Wetering, Cindy Kunne, Suzanne Duijst, Coen C

Paulusma, and Ronald PJ Oude Elferink

Tytgat Institute, Academic Medical Center, Amsterdam, The Netherlands (D.R.d.W.,

C.K., S.D., C.C.P. and R.P.J.O.) and Division of Molecular Biology, The Netherlands

Cancer Institute, Amsterdam, The Netherlands (K. v.d. W.).

List of abbreviations

AUC, area under the curve; E217ßG, estradiol-17ß-glucuronide; H+-coupled peptide

transporter 1, PepT1;

114

115

Abstract

Some cephalosporins, like cefadroxil, are orally available. PepT1 mediates the transport of

cephalosporins across the apical membrane of enterocytes. It is not known which

mechanism(s) is (are) responsible for the subsequent transport of cephalosporins across the

basolateral membrane towards the circulation. We have tested whether ABCC3 and/or

ABCC4 are involved in the latter process. Transport experiments with plasma membrane

vesicles expressing these transporters were used to determine whether ABCC3 and ABCC4

can transport cephalosporins in vitro. The involvement of Abcc3 and Abcc4 in the transport

of cefadroxil from enterocytes was subsequently studied using intestinal explants from wild-

type, Abcc3-/-, Abcc4-/- and Abcc3-/-/Abcc4-/- mice in an Ussing chamber setup. Finally,

appearance of cefadroxil in portal blood was investigated in vivo after intra jejunal

administration of cefadroxil in wild-type, Abcc3-/-, Abcc4-/- and Abcc3-/-/Abcc4-/- mice.

ABCC3- and ABCC4-mediated transport of estradiol-17ß-glucuronide (E217ßG) was dose

dependently inhibited by cephalosporins in vesicular transport experiments. Furthermore,

transport of cefadroxil by ABCC3 and ABCC4 was saturable with Km values of 2.5±0.7 and

0.25±0.07 mM, respectively. Transport of cefadroxil from the apical to the basolateral side of

jejunal tissue explants was unchanged in Abcc3-/- but significantly reduced (approximately

two-fold) in Abcc4-/- and Abcc3-/-/Abcc4-/- when compared to wild type tissue. Upon

instillation of cefadroxil in the jejunum, portal- and peripheral blood concentrations were

similar in Abcc3-/- and Abcc4-/- but approximately two-fold reduced in Abcc3-/-/Abcc4-/-

compared to wild type mice. Our data demonstrate that intestinal absorption of cefadroxil

depends partly on ABCC3 and ABCC4.

Introduction

Infectious diseases are often treated with cephalosporins (468) which belong to the class of ß-

lactam antibiotics. Due to antibiotic resistance there is a continuous need for the development

of new cephalosporins (468). Knowledge of their pharmacokinetics is therefore of eminent

interest. One way of classifying cephalosporins is the route of administration: topical, oral or

parenteral. Oral administration is the most uncomplicated form of administration but requires

passage through the enterocytes. Since it is known that cephalosporins are ionized at

physiological pH and have very low lipid solubility, luminal enterocytic uptake was

considered to be transporter mediated (469). The H+-coupled peptide transporter 1 (PepT1)

was identified as the transporter involved (470). Similarly, cephalosporins can not passively

cross the basolateral membrane, implicating that transport over the basolateral membrane

must also be transporter mediated. A transporter known to mediate transport of

cephalosporins (like ceftriaxone, cefaperazone and cephalexin) is the efflux pump multidrug

115

116

resistance associated protein 2 (Mrp2/Abcc2) (302, 471). ABCC2 is a broad-spectrum

transporter and can transport many different substrates including unconjugated anions,

glucuronate-, glutathione- and sulphate conjugates and furthermore, among others,

leukotriene C4 and methotrexate (472). ABCC2 has overlapping substrate specificity with its

close homologs ABCC3 and ABCC4 (110, 473). While ABCC2 resides in the apical

membrane of enterocytes (474), ABCC3 localizes to the basolateral membrane of epithelial

cells including enterocytes (475). The subcellular localization of ABCC4 is cell type

dependent: In prostate tubuloacinar cells, hepatocytes and choroid plexus epithelium ABCC4

is localized on the basolateral membrane, but in renal proximal tubule cells ABCC4 resides at

the apical membrane (476). It is not exactly clear where ABCC4 resides in the intestine: in

the colonic cell line HT29-CL19A the protein was localised to both the apical and basolateral

membrane, with a higher expression apically (477). However, data of Caco2 cells show

preferential basolateral expression (478). In the present study we explored whether ABCC3

and/or ABCC4 are able to transport cefadroxil in plasma membrane vesicles from cells

overxpressing the corresponding genes. In addition, we used Ussing chamber experiments and

intestinal uptake experiments with wild-type, Abcc3-/-, Abcc4-/- and Abcc3-/-/Abcc4-/- mice to

test whether Abcc3 and/or Abcc4 are involved in the basolateral transport of cefadroxil in

vivo.

Materials and methods

Materials

Acetonitrile (HPLC grade) was from Baker (Mallinckrodt Baker, Deventer, The Netherlands).

[3H]estradiol-17ß-glucuronide ([3H]E217ßG) was from Amersham (Amersham, Roosendaal,

The Netherlands). All other chemicals and reagents were from Sigma (Sigma-Aldrich,

Zwijndrecht, The Netherlands).

Animals

All animal experiments were approved by the Institutional Animal Care and Use Committee

(IUCAC) of the Academic Medical Center. Wild type and Abcc3-/- (479) mice were bred at

the Animal Institute of the Academic Medical Center. Abcc4-/- (232) and Abcc3-/-/Abcc4-/-

mice, generated by crossing Abcc3-/- and Abcc4-/- mice, were bred at the Netherlands Cancer

Institute (Amsterdam, The Netherlands). The animals used were 9 to 16 weeks old. All

animals were on a 99% FVB/N background and kept in a temperature-controlled environment

with a 12-h light/dark cycle. Standard diet (CRM; SDS diets, Germany) and water were

available ad libitum.

116

117

Methods

Preparation of membrane vesicles

The cDNA of the ABCC2 gene (480) was cloned into the pFastBac1 (Life Technologies,

Breda, The Netherlands) donor plasmid. This was followed by transformation of the donor

plasmid into DH10Bac (Life Technologies, Breda, The Netherlands) cells, which allowed for

transposition of the ABCC2 gene into bacmid DNA. ABCG2, ABCC1, ABCC3 and ABCC4-

recombinant baculovirus were a kind gift from Professor P. Borst (149, 181, 282, 481). Sf21

cells grown at 27°C were infected with ABCC1, ABCC2, ABCC3, ABCC4 or ABCG2-

recombinant baculovirus. Isolation of membrane vesicles was as described before (264). In

brief, cells were harvested 2 days (ABCC3), 3 days (ABCC1, ABCC2 and ABCG2) or 4 days

(ABCC4), respectively, postinfection and resuspended in ice-cold hypotonic buffer (250 mM

Mannitol/ 20 mM HEPES/Tris-buffered (pH 7.4)/2 mM EDTA) supplemented with protease

inhibitors (1 mM phenylmethylsulfonyl fluoride, 1 µg/ml aprotinin, 1 µg/ml leupeptin, 1

µg/ml pepstatin), and incubated for 30-60 min on ice. Cells were homogenized using a tight-

fitting Dounce homogenizer. The cell homogenate was layered on top of a discontinuous

sucrose gradient (i.e. 56%, 38% and 19% sucrose) and centrifuged for 3.2 h at 141000g

(SW28 rotor and a Beckman Optima L-90K centrifuge (Beckman, Mijdrecht, The

Netherlands)). The 19%-38% interface was collected, washed and resuspended in 250 mM

sucrose/ 20 mM HEPES/Tris-buffered pH 7.4. Membrane vesicles were obtained after

revesiculation of the preparation by passing it 30 times through a 27-gauge needle. Vesicles

were aliquoted and stored at –80°C until use. Protein content was determined using the BCA

method.

Western blotting and protein analysis

Membrane vesicles were fractionated by 6% SDS-PAGE, blotted on nitrocellulose

membranes (Schleicher&Schuell, Dassel, Germany) which were blocked in phosphate-

buffered saline (PBS)/5% milk powder/0.05% Tween-20. The following antibodies were

used: anti-ABCG2; BXP-21 (78), anti-ABCC1; MRPr1 (482), anti-ABCC2; M2III6 (482),

anti-ABCC3; M3III21 (482) and anti-ABCC4; M4I10 (166) (all antibodies were kind gifts of

Dr. G.L. Scheffer (Free University Hospital, Amsterdam, The Netherlands)). Immune

complexes were visualized with horseradish-peroxidase-conjugated immunoglobulins

(Biorad, Veenendaal, The Netherlands) and detected using the ECL Western blot detection kit

(Amersham, Roosendaal, The Netherlands).

Vesicular transport assays

Vesicular transport studies were performed using the rapid filtration technique as described

(483). Briefly, 5-50 µg membrane vesicles were added to pre-warmed (37°C) reaction-

mixture containing [3H]estradiol-17ß-glucuronide or [3H]cefadroxil (0.05 and 0.2 µCi

117

118

respectively, in 250 mM sucrose, 20 mM HEPES/Tris (pH 7.4)/ 20 mM MgCl2/ 30 mM

creatine phosphate/ 1.2 mg creatine kinase per ml/ with or without ATP (8 mM). Uptake was

terminated by the addition of 20 vol of ice-cold 250 mM sucrose, 20 mM HEPES/Tris pH 7.4.

The suspension was immediately filtered through 0.2 µm nitrocellulose filters under vacuum.

The filters were washed three times and counted in a liquid scintillation counter.

Calculation of half-maximal inhibition

Transport of [3H]estradiol-17ß-glucuronide was plotted against cephalosporin concentration.

The program GraphPad using non-linear regression was used to calculate the concentration of

cephalosporin, at which halfmaximal inhibition was achieved.

Ussing chamber experiments and quantification of cefadroxil

Jejunal tissues from wild-type, Abcc3-/-, Abcc4-/- and Abcc3-/-/Abcc4-/- animals were mounted

in an Ussing chamber. The basolateral compartment contained 1.5 ml basolateral buffer which

consisted of Krebs/ bicarbonate, glucose (10 mM) and HEPES (10 mM, pH 7.4).

Furthermore, the apical compartment contained 1.5 ml apical buffer which consisted of

Krebs/ bicarbonate, mannitol (10 mM) and MES (10 mM, pH 6.0). Both buffers were

continuously gassed with carbogene. Temperature was set at 37 ± 1°C. After 5-10 min

equilibration the experiment was started by the addition of 100 µl cefadroxil containing 8 or

80 nmol (in apical buffer) at the apical side. Cefadroxil was quantified by reversed phase

High-performance liquid chromatography (HPLC) with UV detection (Dionex, Amsterdam,

The Netherlands). In brief, 100 µl sample was applied to a BDS Hypersil C18, 3 µm, 15 cm

HPLC column (Thermo scientific, Waltham, USA). Starting eluent consisted of 10%

acetonitrile/ 90% ammonium formate (20 mM), pH 3.5, followed by a linear gradient to 30%

acetonitrile in the same buffer in 20 min. Cefadroxil had a retention time of 14 min. Detection

of cefadroxil was done at 260 nm. Quantification of cefadroxil was done by using a

calibration curve of cefadroxil.

In vivo experiment: Jejunal administration of cefadroxil and portal blood collection

Mice were anesthetized with a combination of Hypnorm (VetaPharma, Leeds, UK; 11.8

mg/kg fluanisone and 0.37 mg/kg fentanyl citrate) and Valium (Centrafarm, Etten-Leur, The

Netherlands; 5.9 mg/kg Diazepam). Body temperature was maintained between 35°C and

37°C by keeping the mice on thermostatted heating pads. After induction of anaesthesia, the

vena porta was cannulated, followed by ligation of 10 cm of the middle part of the jejunum.

500 µl cefadroxil (5 µM; 2µCi) was subsequently injected into the ligated jejunum and portal

blood samples were collected after indicated time points. At the end of the experiment a

peripheral blood sample was taken by cardiac puncture. Mice were subsequently

anaesthetized and liver, the ligated jejunum, kidneys and gallbladder were collected. After

118

119

the addition of H2O2 (30%) to blood, jejunum, kidney, liver and jejunum radioactivity was

measured by liquid scintillation counting.

Statistical analyses

Statistical differences were determined by an unpaired Student’s t-test. All data were

expressed as means ± standard deviation.

Table 1: Cephalosporins used in the inhibition studies.

molecular weight oral available

Cephalexin 347.4 yes

Cephradine 349.4 yes

Cefadroxil 363.4 yes

Cefaclor 367.8 yes

Cefuroxime 424.4 yes

Cefoxitin 427.4 no

Cefamandole 462.5 no

Cefmetazole 471.5 no

Ceftriaxon 554.6 no

Cefoperazone 645.7 no

119

120

Figure 1: Inhibition of estradiol-17ß-glucuronide transport by cephalosporins. Cephalosporin inhibition of E217ßG (0.05 µCi; 1.0 µM) transport into, ABCC2, ABCC3 and ABCC4 containing Sf21 membrane vesicles. Shown are the measurements of the IC50 values for E217ßG (0.05 µCi; 1.0 µM) transport mediated by (A) ABCC2 (solid line), (B) ABCC3 (dashed line) and (C) ABCC4 (dotted line), inhibited by different concentrations of cefadroxil. (D) IC50 values for inhibition of E217ßG transport by cephalosporins. Transport mediated by ABCC2, ABCC3 and ABCC4 versus molecular weight of these cephalosporins.

120

121

Results

To study the contribution of ABCC3 and ABCC4 in the transport of cephalosporins, we

expressed the human proteins in Spodoptera Frugiperda 21 (Sf21) insect cells. ABCC2

protein was expressed as a positive control for cephalosporin transport. Transport activity was

determined by measuring uptake of substrates into isolated membrane vesicles. First,

ABCC2-, ABCC3- and ABCC4-dependent uptake of the model substrate E217ßG was studied

in the absence and presence of cefadroxil.

Figure 2: Vesicular transport assay. Transport of cefadroxil (0.2 µCi; 10µM) with (black bars) or without (white bars) ATP into mock-transduced, ABCC1, ABCC2, ABCC3, ABCG2 and ABCC4 expressing membrane vesicles. Incubation time is 6 min. Amount total protein used is 10 µg. Data represent the average ± SD. Shown is the result of one out of two independent experiments. Significance was tested using two-sided Student’s t-test: *P<0.05 for transport of ABCC1, ABCC2, ABCC3 and ABCC4 expressing membrane vesicles versus membrane vesicles from mock transduced cells.

121

122

Figure 3: Time- and concentration-dependent transport. Time-dependent transport of cefadroxil (0.2 µCi; 10 µM) into plasma membrane vesicles from Sf21 cells expressing ABCC1 (A), ABCC3 (B) and ABCC4 (C). Open squares (with dotted line) and open diamonds represent incubations with or without ATP, respectively. Filled triangles (with solid line) represent ATP dependent transport. Concentration-dependent transport of cefadroxil (0.2 µCi; 10 µM) into plasma membrane vesicles (open diamonds with solid line) from Sf21 cells expressing ABCC1 (D), ABCC3 (E) and ABCC4 (F). Shown are the average ± SD of ATP-dependent transport of a representative experiment with triplicate incubations performed 3-4 times.

122

123

Figure 4: Transport of cephadroxil across jejunal explants in Ussing chambers. Appearance of cefadroxil on the basolateral side of jejunal explants from Abcc3-/-, Abcc4-/- and Abcc3-/-/Abcc4-/- and wild type mice. Cefadroxil was applied to the apical compartment of Ussing chambers at a final concentration of 5 µM. At indicated time points samples were taken from the basolateral side and analyzed by HPLC. Shown is the average ± standard deviation of 3-4 experiments with triplicate incubations. ‡, p < 0.05 comparing appearance of cefadroxil using tissue from Abcc4-/- versus wild type mice for indicated time points. #, p < 0.05 comparing appearance of cefadroxil using tissue from Abcc3-/-/Abcc4-/- versus wild type mice for indicated time points.

A concentration-dependent inhibition of E217ßG uptake by cefadroxil was observed in

membranes expressing ABCC2 (Fig. 1A), ABCC3 (Fig. 1B) and ABCC4 (Fig. 1C), albeit

with very different IC50’s. The same inhibition studies were performed for nine other

cephalosporins (table 1). For all cephalosporins the concentration at which E217ßG uptake

was inhibited by 50% (IC50) was determined and plotted as a function of the molecular weight

of the cephalosporins (Fig. 1D). For all three ABC transporters a near linear, inverse

correlation between IC50-values and molecular weight of the cephalosporins was observed.

The orally-available cephalosporins (see Table 1) have the lowest molecular weight,

indicating that molecular weight/bulkiness of the substrate is an important determinant for

oral availability. Moreover, the orally available cephalosporins show the largest difference

with regard to IC50-values of cephalosporins towards transport of E217ßG mediated by

ABCC4 versus ABCC2 and ABCC3, respectively. Competitive inhibition is an indication that

the inhibitor may be a transported substrate. Cefadroxil is an orally prescribed cephalosporin

that is commercially available in a radiolabelled form allowing its use in direct transport

experiments. We, therefore, used cefadroxil as a model substrate for orally prescribed

cephalosporins, in vesicular transport experiments. ABCG2 did not transport cefadroxil.

However, membrane vesicles containing ABCC1, ABCC2, ABCC3 and ABCC4 all showed

ATP-dependent uptake of cefadroxil (Fig. 2). In line with the inhibition experiments,

transport of cefadroxil mediated by ABCC4 was three- to six-fold higher compared to

transport by ABCC2 and ABCC3 (Fig. 2). Furthermore, cefadroxil transport was time

dependent (Fig. 3A-C) and saturable (Fig. 3D-E) for ABCC1, ABCC3 and ABCC4 with Km

values of 3.9±0.6, 2.5±0.7 and 0.25±0.07 mM, respectively.

We subsequently tested whether there could be a role for Abcc3 and/or Abcc4 in vivo in the

transport of cefadroxil across the basolateral membrane of the enterocyte in Ussing chamber

experiments. No difference was found in transport of cefadroxil from the apical to the

123

124

basolateral compartment using jejunal tissue from wild type and Abcc3-/- mice. Transport of

cefadroxil, (5 µM), was reduced to about 50% of normal levels, when jejunal tissue of Abcc4-

/- and Abcc3-/-/Abcc4-/- mice were used (Fig. 4). Similar results were found with 50 µM

cefadroxil (data not shown). Finally, in intestinal loop experiments we found that injection of

cefadroxil in the ligated jejunum resulted in the time-dependent appearance of cefadroxil in

portal blood of wild type mice (Fig. 5). The appearance of cefadroxil in portal blood of Abcc3-

/- and Abcc4-/- mice was however not different from wild type mice, the latter result being in

contrast to the Ussing chamber data. Interestingly, the appearance of cefadroxil in portal

blood of Abcc3-/-/Abcc4-/- mice was reduced. Similar results were found for peripheral blood:

A significantly lower concentration of cefadroxil was found in Abcc3-/-/Abcc4-/- mice in

comparison with wild type mice. There was no difference in the cefadroxil tissue content of

intestine, kidney and liver nor in its urinary excretion between the mice strains tested.

However, an increased amount of cefadroxil was noticed in the very low (less than 0.4% of

dose) biliary content in Abcc3-/-/Abcc4-/- versus wild type mice (Fig. 5D).

Figure 5: In vivo uptake of cefadroxil by the jejunum. Appearance of cefadroxil in portal (closed symbols) and peripheral blood (open symbols) of Abcc3-/- versus wild type mice (A), Abcc4-/- versus wild type mice (B) and Abcc3-/-/Abcc4-/- versus wild type mice (C) after injection of 500 µl cefadroxil (5 µM; 2µCi) into ligated jejunum. Portal blood was collected after indicated time points, peripheral blood after the last portal blood sample was drawn. Appearance of cefadroxil in bile of Abcc3-/-/Abcc4-/- versus wild type mice (D) after injection of 500 µl cefadroxil (5 µM; 2µCi) into ligated jejunum. Data represent average ± SD of at least 5 mice. Significance was tested using two-sided Student’s t-test: *P<0.05 for Abcc3-/-/Abcc4-/- versus wild type mice.

124

125

Discussion

Orally prescribed cephalosporins are efficiently taken up in the gut. At the luminal

side of the small intestine the dipeptide transporter PepT1 is involved in the import of

cephalosporin. At present the transporter(s) involved in the translocation of

cephalosporins from the enterocytes into blood is (are) still unknown. ABCC2

transports several cephalosporins like ceftriaxone, cefaperazone and cephalexin (302,

471). Because of its apical expression, ABCC2 can not be responsible for the

transport of oral cephalosporins into blood. As ABCC3 and ABCC4 are related

transporters we tested their ability to mediate transport of one of the orally prescribed

cephalosporins, namely cefadroxil. In this article we show that both ABCC3 and

ABCC4 mediate transport of cefadroxil albeit with different kinetics: of the ABC

transporters under study ABCC4 mediates transport with the highest affinity (Fig. 2

and 3). This is in line with an earlier report showing that the non-oral available

cephalosporins ceftizoxime, cefazolin, cefotaxime and cefmetazole are substrates of

ABCC4 (484). In this paper the Km values of 18 and 81 µM were stated for MRP4-

mediated transport of respectively ceftizoxime and cefazolin (484). Since cefadroxil is

a smaller compound than both these cephalosporins the Km value of 246 µM is higher

as expected. This is based on our findings that an inverse correlation exists between

molecular weights of cephalosporins and IC50 values (Fig. 1D) and a relation between

Km and IC50 values. Our in vivo experiments with wild type, Abcc3-/- and Abcc4-/-

single knockout mice showed no difference in transport of cefadroxil from luminal to

serosal side (Fig 5). We have two explanations for this result: one explanation is that

murine Abcc3 and Abcc4, unlike their human orthologues used in the vesicular

transport experiments, are unable to mediate transport of cefadroxil (Fig. 2 and 3).

This explanation is unlikely as we observed in Ussing chamber experiments that

transport was reduced in intestinal explants from Abcc4-/- mice compared to wild type

mice. Another explanation is that these transporters can compensate for the loss of

each other. Therefore, we extended the experiments with Abcc3-/-/Abcc4-/- mice. The

Abcc3-/-/Abcc4-/- mice had reduced levels of cefadroxil in their portal blood (Fig. 5).

As similar results were found in Ussing chamber experiments using jejunal explants

from Abcc3-/-/Abcc4-/- and wild type mice, conclude that both Abcc3 and Abcc4

transport cefadroxil and when one transporter is lacking the other can fully

compensate. Human ABCC4 transports cefadroxil at higher rates than ABCC3 in

vesicular transport experiments. Expression of murine Abcc3 is higher than murine

125

126

Abcc4 in the gut. This might be the reason that although Abcc3 transports cefadroxil

only at moderate rates it can still compensate for the loss of Abcc4. As the difference

in expression level between Abcc3/ABCC3 and Abcc4/ABCC4 in jejunum is similar

in mouse and man we speculate that also in the human situation ABCC3 and ABCC4

are involved in transport of cefadroxil from enterocyte to blood (485, 486).

The potential involvement of Abcc4 in the basolateral transport of cefadroxil is

important since literature data are not consistent about the cellular localization of

Abcc4. In the human colonic cell line HT29-CL19A, Li et al. found Abcc4 in both the

apical and basolateral membrane, with a higher expression apically (477). In Caco2

cells Ming and Thakker detected Abcc4 mainly in basolateral membrane (478). Our

data that intestinal Abcc4 influences cefadroxil uptake supports the notion that Abcc4

is present at the basolateral mebrane of the enterocytes. Our functional data suggest

that Abcc4 is present at the basolateral membrane of enterocytes and confirm the data

obtained in Caco2 cells by Ming and Thakker (478).

Abcc3-/-/Abcc4-/- mice still show a considerable amount of transport of cefadroxil over

the basolateral membrane of enterocytes. Theoretically, Abcc1 could be responsible

for the residual transport of cefadroxil as we found that human ABCC1 is able to

mediate transport of cefadroxil (Fig 2 and 3). However, Abcc1 is expressed in the

small intestine, mainly in the crypts (86, 189) which does not colocalize with PepT1,

which is abundantly present in the villus tip with decreasing levels towards the villus

base. The localization of Abcc1 therefore argues against a role of Abcc1 in the

basolateral efflux of cefadroxil in enterocytes. We can, however, not fully exclude

that there is a role of Abcc1 in the efflux of cefadroxil as this transporter protects

against the intestinal toxicity evoked by methotrexate (86). This means that Abcc1 is

pumping methotrexate out of the cell at a physiological relevant speed. The same

might be the case for Abcc1 mediated transport of cefadroxil into blood from the

enterocyte, especially in the absence of Abcc3 and Abcc4 in the used murine model:

Abcc3-/-/Abcc4-/- mice.

Oral administration is a preferable route of administration of drugs. In general, the

oral available cephalosporins are low molecular weight molecules. Kato et al. showed

a correlation between size and biliary excretion for several cephalosporins (471). This

can be partially caused by a size-dependent affinity for the transporter mainly

responsible for the biliary excretion, Abcc2/ABCC2 (471). In line with data of Kato et

al., we found a size dependent inhibition by cephalosporins of ABCC2-mediated

126

127

transport of E217ßG (Fig 1D). Also, a size-dependent inhibition by cephalosporins

was found by us for ABCC3- and ABCC4-mediated transport of E217ßG (Fig 1D),

but the slope for ABCC2 is steeper than for ABCC4 and similar for ABCC3. This

could indicate that the influence of size is relatively big for ABCC2 (which prevents

net uptake) and relatively small for ABCC4 (which stimulates net uptake).

Interestingly, an inverse selectivity of PepT1 was shown to be dependent on

molecular weight as well, with small cephalosporins being preferentially transported.

Hence, the combined selectivity of the uptake transporter Pept1 and the efflux

transporter ABCC2 may determine the typical pharmacokinetic behaviour of

cephalosporins.

In conclusion, the data presented in this paper demonstrate that murine intestinal

uptake depends partly on Abcc3 and Abcc4. We therefore speculate that in the human

situation oral availability involves, at least partly, uptake via ABCC3 and ABCC4.

127

128

Reference List

Assem M, Schuetz EG, Leggas M, Sun D, Yasuda K, Reid G, Zelcer N, Adachi M, Strom S, Evans RM, Moore DD, Borst P and Schuetz JD (2004) Interactions between hepatic Mrp4 and Sult2a as revealed by the constitutive androstane receptor and Mrp4 knockout mice. J Biol Chem 279:22250-22257. Bakos E, Evers R, Sinko E, Varadi A, Borst P and Sarkadi B (2000) Interactions of the human multidrug resistance proteins MRP1 and MRP2 with organic anions. Mol Pharmacol 57:760-768. Borst P, de WC and van de WK (2007) Multidrug resistance-associated proteins 3, 4, and 5. Pflugers Arch 453:661-673. Breedveld P, Zelcer N, Pluim D, Sonmezer O, Tibben MM, Beijnen JH, Schinkel AH, van TO, Borst P and Schellens JH (2004) Mechanism of the pharmacokinetic interaction between methotrexate and benzimidazoles: potential role for breast cancer resistance protein in clinical drug-drug interactions. Cancer Res 64:5804-5811. Ci L, Kusuhara H, Adachi M, Schuetz JD, Takeuchi K and Sugiyama Y (2007) Involvement of MRP4 (ABCC4) in the luminal efflux of ceftizoxime and cefazolin in the kidney. Mol Pharmacol 71:1591-1597.

de Waart DR, Paulusma CC, Kunne C and Oude Elferink RP (2006) Multidrug resistance associated protein 2 mediates transport of prostaglandin E2. Liver Int 26:362-368.

Heijn M, Oude Elferink RP and Jansen PL (1992) ATP-dependent multispecific organic anion transport system in rat erythrocyte membrane vesicles. Am J Physiol 262:C104-C110.

Kato S, Ito K, Kato Y, Wakayama T, Kubo Y, Iseki S and Tsuji A (2009) Involvement of multidrug resistance-associated protein 1 in intestinal toxicity of methotrexate. Pharm Res 26:1467-1476. Kato Y, Takahara S, Kato S, Kubo Y, Sai Y, Tamai I, Yabuuchi H and Tsuji A (2008) Involvement of multidrug resistance-associated protein 2 (Abcc2) in molecular weight-dependent biliary excretion of beta-lactam antibiotics. Drug Metab Dispos 36:1088-1096. Leggas M, Adachi M, Scheffer GL, Sun D, Wielinga P, Du G, Mercer KE, Zhuang Y, Panetta JC, Johnston B, Scheper RJ, Stewart CF and Schuetz JD (2004) Mrp4 confers resistance to topotecan and protects the brain from chemotherapy. Mol Cell Biol 24:7612-7621. Li C, Krishnamurthy PC, Penmatsa H, Marrs KL, Wang XQ, Zaccolo M, Jalink K, Li M, Nelson DJ, Schuetz JD and Naren AP (2007) Spatiotemporal coupling of cAMP transporter to CFTR chloride channel function in the gut epithelia. Cell 131:940-951. Livermore DM (2009) Has the era of untreatable infections arrived? J Antimicrob Chemother 64 Suppl 1:i29-i36. Maher JM, Slitt AL, Cherrington NJ, Cheng X and Klaassen CD (2005) Tissue distribution and hepatic and renal ontogeny of the multidrug resistance-associated protein (Mrp) family in mice. Drug Metab Dispos 33:947-955. Maliepaard M, Scheffer GL, Faneyte IF, van Gastelen MA, Pijnenborg AC, Schinkel AH, van d, V, Scheper RJ and Schellens JH (2001) Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues. Cancer Res 61:3458-3464. Ming X and Thakker DR (2010) Role of basolateral efflux transporter MRP4 in the intestinal absorption of the antiviral drug adefovir dipivoxil. Biochem Pharmacol 79:455-462. Mottino AD, Hoffman T, Jennes L and Vore M (2000) Expression and localization of multidrug resistant protein mrp2 in rat small intestine. J Pharmacol Exp Ther 293:717-723.

128

129

Nakashima E, Tsuji A, Mizuo H and Yamana T (1984) Kinetics and mechanism of in vitro uptake of amino-beta-lactam antibiotics by rat small intestine and relation to the intact-peptide transport system. Biochem Pharmacol 33:3345-3352. Oude Elferink RP and de WR (2007) Transporters in the intestine limiting drug and toxin absorption. J Physiol Biochem 63:75-81. Oude Elferink RP and Jansen PL (1994) The role of the canalicular multispecific organic anion transporter in the disposal of endo- and xenobiotics. Pharmacol Ther 64:77-97. Paulusma CC, Kool M, Bosma PJ, Scheffer GL, ter Borg F, Scheper RJ, Tytgat GN, Borst P, Baas F and Oude Elferink RP (1997) A mutation in the human canalicular multispecific organic anion transporter gene causes the Dubin-Johnson syndrome. Hepatology 25:1539-1542.

Peng KC, Cluzeaud F, Bens M, Van Huyen JP, Wioland MA, Lacave R and Vandewalle A (1999) Tissue and cell distribution of the multidrug resistance-associated protein (MRP) in mouse intestine and kidney. J Histochem Cytochem 47:757-768. Rius M, Hummel-Eisenbeiss J and Keppler D (2008) ATP-dependent transport of leukotrienes B4 and C4 by the multidrug resistance protein ABCC4 (MRP4). J Pharmacol Exp Ther 324:86-94. Russel FG, Koenderink JB and Masereeuw R (2008) Multidrug resistance protein 4 (MRP4/ABCC4): a versatile efflux transporter for drugs and signalling molecules. Trends Pharmacol Sci 29:200-207. Scheffer GL, Kool M, de Haas M, de Vree JM, Pijnenborg AC, Bosman DK, Elferink RP, van d, V, Borst P and Scheper RJ (2002) Tissue distribution and induction of human multidrug resistant protein 3. Lab Invest 82:193-201.

Scheffer GL, Kool M, Heijn M, De HM, Pijnenborg AC, Wijnholds J, van HA, de Jong MC, Hooijberg JH, Mol CA, van der LM, de Vree JM, van d, V, Elferink RP, Borst P and Scheper RJ (2000) Specific detection of multidrug resistance proteins MRP1, MRP2, MRP3, MRP5, and MDR3 P-glycoprotein with a panel of monoclonal antibodies. Cancer Res 60:5269-5277. Taipalensuu J, Tornblom H, Lindberg G, Einarsson C, Sjoqvist F, Melhus H, Garberg P, Sjostrom B, Lundgren B and Artursson P (2001) 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 299:164-170. Tsuji A, Nakashima E, Kagami I and Yamana T (1981) Intestinal absorption mechanism of amphoteric beta-lactam antibiotics I: Comparative absorption and evidence for saturable transport of amino-beta-lactam antibiotics by in situ rat small intestine. J Pharm Sci 70:768-772. van Aubel RA, Smeets PH, Peters JG, Bindels RJ and Russel FG (2002) 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 13:595-603. Zelcer N, Saeki T, Reid G, Beijnen JH and Borst P (2001) Characterization of drug transport by the human multidrug resistance protein 3 (ABCC3). J Biol Chem 276:46400-46407. Zelcer N, van de WK, de WR, Scheffer GL, Marschall HU, Wielinga PR, Kuil A, Kunne C, Smith A, van d, V, Wijnholds J, Elferink RO and Borst P (2006) Mice lacking Mrp3 (Abcc3) have normal bile salt transport, but altered hepatic transport of endogenous glucuronides. J Hepatol 44:768-775.

129


Recommended