• No results found

Transport of amino acids andgamma-aminobutyric acid analogues inMadin-Darby canine kidney cellsDan Li

N/A
N/A
Protected

Academic year: 2022

Share "Transport of amino acids andgamma-aminobutyric acid analogues inMadin-Darby canine kidney cellsDan Li"

Copied!
23
0
0

Loading.... (view fulltext now)

Full text

(1)

Transport of amino acids and

gamma-aminobutyric acid analogues in Madin-Darby canine kidney cells

Dan Li

Degree project in applied biotechnology, Master of Science (2 years), 2009 Examensarbete i tillämpad bioteknik 30 hp till masterexamen, 2009

Biology Education Centre, Uppsala University, and Department of Pharmaceutics and Analytical

Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen

(2)

 

Summary

Bioavailability of oral drugs absorbed across the human intestinal epithelium is facilitated by various transporters in intestine tract. Human proton-coupled amino acid transporter (hPAT1), as an imino acid carrier, shows the potential to deliver some orally administrated drugs.

The most popular in-vitro cell model for passive diffusion of lipophilic molecules is Caco-2 cell line, which is from human intestine cancer cells and grows slowly. The Madin-Darby canine kidney (MDCK) cell line is a common choice for studies of compound transport as well. These cells, from dog kidney, grow fast. They might be appropriate for hPAT1 studies to avoid the complicated interaction with other human transporters expressed in Caco-2 cells.

In this study, the MDCK cell line was investigated to figure out the possibility to set up a new in-vitro model in MDCK cells for drug screening. [

3

H]-proline uptake was done to characterize proton-coupled amino acid transport, and γ-aminobutyric acid (GABA) transport was measured via [

3

H]-GABA uptake under different conditions.

After that, transient transfection of hPAT1 into MDCK cells was conducted with transfection reagent Lipofectamin 2000. The transfection efficiency was measured as hPAT1 transport function by [

3

H]-proline uptake.

There was no proton-coupled amino acid transporter mediated transport characterized

in MDCK cells. A transporter facilitating GABA transport was tested in MDCK cells

and the K

m

value for GABA uptake via this transporter was measured as 2.2 ± 0.4

mM. Transient transfection was not successful under different conditions in this study.

(3)

Introduction

Bioavailability of oral drugs absorbed across the human intestinal epithelium is a big issue in drug delivery in biopharmaceutical research (Sarmento et al. 2007). The drug absorption is facilitated by various transporters located in the small intestine membrane (Nielsen & Brodin, 2003). Such transporters have a conformational change during the translocation process to recognize and transport the substrate molecules (Nelson & Cox, 2004).

S

ome drug substances are transported by one or more transporters and share the specificity with diet nutrients molecules entering the human body via transporters in the membrane (Nielsen & Brodin, 2003).

1.1. Carriers

1.1.1. Carrier transport mechanism

The substance movement across the cell membrane can be divided into two categories:

active transport and passive transport (Nelson & Cox, 2004). Active transport is activated by chemical energy like the energy released from ATP hydrolysis, and passive transport is a process not involving chemical energy. Passive transport includes simple diffusion, facilitated diffusion, filtration and osmosis (Saier, 2000).

Some small nonpolar molecules can just go across the cell membrane through the simple diffusion process, whereas for polar molecules, facilitated diffusion is mediated by transporters located on the lipid bilayer. Transporters that facilitate the translocation of smaller polar molecules, i.e. ions, at high speed are channels, and transporters where relative larger molecules, such as amino acids, are moving through at lower speed, are carriers (Fujiyoshi et al. 2002, Mueckler, 1994).

Most carriers are integral proteins with several transmembrane regions inside the lipid bilayer, responsible for the substance transport from the external to the internal environment, or from one aqueous compartment to another of an organism (Mueckler, 1994). The carrier undergoes a conformational change during the substrate translocation process, which can be divided into four steps: binding, translocation, release and recovery.

The transport across the lipid bilayer, can be classified into three general systems

according to the traffic of substrates: uniport, symport and antiport (Saier, 2000). A

uniport moves the substrate in the direction down the concentration gradient. A

symport transports the substrate as well as the second substrate simultaneously in the

same direction. An antiport transports the substrate with another substance

simultaneously in the opposite direction. Both a symport and an antiport can also be

grouped into cotransport system, which means the transporter can translocate more

than one substrate at a time (Nelson & Cox, 2004).

(4)

1.1.2. Carrier transport kinetics

The carrier transport can be interpreted as the chemical reaction catalyzed by the enzyme. The transporter plays as an enzyme and the translocated substrate does not have any chemical change (Mueckler, 1994). The transport process could be expressed as Eq. 1,

      T+ ↔ ⋅ ↔ +S T S T S       

Eq. (1) where T represents the transporter and S stands for the substrate (Jakubowski, 2008).

The initial velocity for the enzymes or transporters is described using Michaelis-Menten equation as Eq. 2,

max 0

[ ]

m

[ ]

V S

V K S

= ⋅

+

     

Eq. (2)

where V

0

is the initial velocity, [S] is the substrate concentration, V

max

is the maximal velocity at steady state when the tranporters are saturated and K

m

is an eqilibrium constant when V

0

is half of the V

max

, and is also called Michaelis-Menten constant (Pliška, 2003, Jakubowski, 2008). The relationship between velocity and the substrate concentration is shown below as the classical Michaelis-Menten curve (Fig. 1.1): the velocity increases with the substrate concentration, but the slope of the increase decreases to zero when all the transporters are satured and the velocity reaches V

max

and is constant. K

m

can be viewed as a measure of the substrate specificity for a given transporter, which is independent of the tranporter concentration. V

max

represents the capacity of the transporter (Nelson & Cox, 2004).

Fig. 1.1: Kinetics of compound transport across a cell membrane. For the Michaelis-Menten curve (●), the velocity for a saturable transport process is described as a function of the total concentration of substrate at one given transporter concentration. The linear regression (▲) represents a transport progressing by simple diffusion.

(5)

For simple diffusion process, small molecules go across the cell membrane freely in the direction dictated by the substrate concentration gradient (Nelson & Cox, 2004).

Simple diffusion does not use any carrier and its rate linearly depends on the solute concentration (Fig. 1.1) (Jakubowski, 2008).

1.2. Solute carrier family and amino acid transporters

The human small intestinal membrane is polarized, with an apical side facing the lumen and a basolateral side facing towards the interstitium and different transporters are located in the membrane. The solute carrier family (SLC family), a group of membrane transport protein, is responsible for the transport of nutrients across the human intestinal barrier. Some members of the SLC family also have substrate specificities for an administ ered oral drug, such as human proton-coupled di- or tri- peptide transporter (hPEPT1, SLC15A1), which has the affinity for di/tri peptides digested from diet protein (Nielsen & Brodin, 2003).

Most mammalian amino acid transporters are ion–independent and sodium coupled.

An imino acid carrier was demonstrated being located in the intact rat intestinal membrane and characterized as a proton mediated transporter (Rajendran et al. 1993).

Later it was named human proton-coupled amino acid transporter (hPAT1, SLC36A1) (Anderson et al. 2004).

The cDNA sequence of hPAT1 has been cloned from Caco-2 cells, encoding 476 amino acids (Chen et al. 2003). The protein expression was detected by an immunoassay at the apical side of the polarized small intestinal epithelium (Chen et al.

2003). hPAT1 is an amino acid transporter, facilitating the transport of zwitteronic amino acid, e.g. proline, glycine and alanine (Thwaites et al. 1995). It also has affinity for orally administrated compounds, such as D-serine and betaine (Anderson et al.

2004). In addition, it transports taurine, the neurotransmitter γ-aminobutyric acid (GABA) and several GABA analogues like nipecotic acid and guvacine (Huxtable et al. 1992, Thwaites et al. 2000).

GABA plays a key role as chief inhibitory neurotransmitter in the human nerve

system. GABA movement in the synaptic cleft is regulated by its reuptake through

GABA transporters (Fiorenzo et al. 2004). GABA transporters, facilitated by Na

+

and

Cl

-

, have been classified into four subtypes and all of them belong to solute carrier

family group SLC6. The four GABA transporters determined in human and rat are

GABA transporter (GAT)-1, betaine orγ-aminobutyric acid transporter (BGT)-1,

GAT-2 and GAT-3, respectively (Table 1). The cDNA sequence of BGT-1 was cloned

from MDCK cells and regulated by hypertonicity (Yamauchi et al. 1992).

(6)

Table 1 GABA transporters.

Systematic Nomenclature

Nomenclature by sourcea Protein expressionb

Localization in MDCK cellsb Rat and Human Mouse

SLC6A1 GAT-1 GAT1 Central nerve system Apical SLC6A11 GAT-3 GAT4 Central nerve system Basolateral

SLC6A12 BGT-1 GAT2 Central and Periphery Basolateral SLC6A13 GAT-2 GAT3a Central and Periphery Apical

a, Tomi et al. 2008

b, Christiansen, 2007.

The taurine transporter (TauT, SLC6A6) shares 61% amino acid identity with GAT-3 and GAT-2 and has a similar structure as GABA transporters (Borden, 1996). TauT could transport GABA with K

m

1.46 ± 0.47mM in a rat retinal capillary endothelial cell line (TR-iBRB2) (Tomi et al. 2008). Taurine functions as a nonperturbing osmolyte in retinal cells retaining the isotonicity in the extracellular hypertonic environment, mediated by the TauT and other transporters (Tomi et al. 2007).

1.3. Cell models

1.3.1. CaCo-2 cell model

In biopharmaceutical research, in-vitro assays are always used to investigate the drug delivery mechanisms across epithelia in cell lines (Thwaites et al. 1995, Thwaites et al. 2000). Transport of different drug substances transports within different cell lines is determined as different apparent permeability (P

app

) values (Irvine et al. 1999).

Caco-2 cells, derived from human epithelial colorectal adenocarcinoma cells, comprise the most popular in-vitro cell model for oral drug absorption and passive diffusion of lipophilic molecules across the human small intestinal barrier (Balmane et al. 2006). Cells from this immortalized heterogeneous cell line function morphologically and physiologically similarly as the human columnar cells.

Furthermore, the substance apparent permeability (P

app

) value measured in Caco-2 assays correlates well with values measured in the absorption from human intestine (Artursson & Karlsson, 1991).

Caco-2 cells need 21 days to grow and express protein before assays (Irvine et al.

1999). In addition, as Caco-2 is a natural cell line from human tissues, various transporters are expressed in the cells and the function of one transporter would be influenced by others (Larsen et al. 2008).

1.3.2. MDCK cell model

Madin-Darby canine kidney (MDCK) cells have been shown as a common choice for

the study of compound transport in kidney epithelium (Irvine et al. 1999). Derived

from the

 

distal tubular part of dog kidney cells, MDCK cells grow fast, requiring only

3 days to be mature before assays (Irvine et al. 1999). Some transporters are located

(7)

in MCDK cell membranes, such as monocarboxylic acid transporter, peptide transporter, P-glycoprotein transporter and organic cation transporter (Volpe, 2008).

A test of transport of various antimicrobial agents has showed a series of similar P

app

in both Caco-2 and MDCK cell (Ranaldi et al. 1992). A stable transfected MDCK-hPepT1-V5&His clonal cell line could successfully express the transporter hPepT1 and this cell line was utilized to investigate the di- or tri- peptide uptake across the epithelium

 (

Herrera-Ruiz et al. 2004, Rajinder et al. 2005).

1.4. Aims

In this study, I aimed to investigate MDCK cell lines in terms of the characterization

and function of hPAT1. I would figure out if there was an expression of transporter,

such as BGT-1, that shared substrate specificity with hPAT1 in the cell line. After that,

if the result convinced me that MDCK could be an appropriate candidate to establish a

new in-vitro model, I aimed to transiently transfect MDCK cells with the plasmids

carrying the sequence encoding hPAT1 and make some characterization experiments

in the transfected cells as well.

(8)

2. Results

2.1. Characterization of proline transport

To investigate whether any proton-coupled amino acid transporter is present in the MDCK cell line, I conducted some basic uptake experiments using the common PAT1 substrate proline which was radioactively labeled. As PAT1 transports amino acids together with protons, the proline uptake was measured at different substrate concentration with different apical pH values (see “Apical uptake ” in “Material and Methods”) (Fig. 2.1). At both concentrations, the flux of proline uptake at physiological pH was higher than at lower pH, showing that the proline transport was not stimulated by the proton gradient across the apical side (proton gradient with the interior pH value at 7.4 and exterior pH value at 6.0 across the cell membrane).

A      B 

 

Fig. 2.1: Proline uptake in MDCK cells at apical pH values. A: proline concentration 23 nM; B: proline concentration 2.5 mM. Both of the uptakes were performed at pH 6 (white) and pH 7.4 (grey). Each point represents the mean ± S.E.M. of measurement (n = 3).

(9)

Fig. 2.2: Proline uptake rate in MDCK cells as a function of proline concentration. The uptake of proline was measured at apical pH 7.4 with different proline concentrations. A: proline concentrations (23 nM, 110 nM, 1 μM) and B: proline concentrations (1 μM, 10 μM, 100 μM) C: all proline

concentrations (23 nM, 110 nM, 1 μM, 10 μM, 100 μM, 0.99 mM, 9.9 mM, 99 mM) almost increased by magnitude. The points represent the mean ± S.E.M. of measurement (n = 3).

As the proline transport activity at pH 7.4 was relatively higher than pH 6.0 (Fig. 2.1), I measured the proline uptake at apical pH 7.4 as a function of proline concentration.

The substrate concentration for the experiment was from 23 nM to 99 mM. The proline uptake increased linearly with the substrate concentration instead of following Michaelis–Menten kinetics, suggesting that the proline uptake in MDCK cells was mediated by diffusion rather than by the amino acid transporter (Fig. 2.2).

2.2. Characterization of GABA transport

As hPAT1 transports GABA and GABA analogues(Larsen et al. 2008), other transporters expressed in MDCK cells should be paid attention to if they are capable of transporting these compounds as well. BGT-1 has been shown to be expressed naturally in MDCK cells and located in membranes on the basolateral side of cells (Yamauchi et al. 1992). Since BGT-1 is a member of the SLC6 family with sodium

A B

C

 

(10)

dependency and proton independency, the basolateral GABA uptake in MDCK cells was measured in a buffer with and without sodium at different pH values (Fig. 2.3) (See “Basolateral uptake” in “Material and Methods”).

Fig. 2.3: Basolateral GABA uptake rate in MDCK cells at different pH values. GABA uptake was measured at pH 7.4 and pH 6 with 28 nM GABA. Both conditions were performed in the regular buffer or sodium free buffer. Each point represents the mean ± S.E.M. of measurement (n = 3).

The data suggest that the flux of GABA uptake in MDCK cells at the basolateral side was almost totally inhibited in the absence of sodium at both pH values. GABA uptake at physiological pH (pH 7.4) was higher than the one at lower pH (pH 6.0).

Therefore pH 7.4 was chosen to measure GABA uptake for later experiments.

To further study the kinetics and determine the K

m

for GABA uptake by the transporter, basolateral GABA uptake at different substrate concentration was measured in buffers with and without sodium (Fig. 2.4). In the presence of sodium, the GABA uptake rate fitted the Michaelis-Menten Equation with the K

m

2.2 ± 0.4 mM, whereas in the absence of sodium, the uptake increased linearly with GABA concentration.

Fig. 2.4: Basolateral GABA uptake in MDCK cells as a function of GABA concentration. The GABA uptake with sodium (▲) and without sodium (▼) at pH 7.4 is shown. The Km for GABA uptake in the

(11)

presence of sodium (▲) was calculated as 2.2 ± 0.4 mM. The points represent the mean ± S.E.M. of measurement (n = 3).

 

Fig. 2.5: Basolateral GABA uptake rate in MDCK cells as a function of GABA concentrations. The GABA uptake at isotonic condition 350 mOsmo/L (■) and hypertonic condition 500 mOsmo/L (▲) at pH 7.4 is shown. The Km for GABA uptake at isotonic condition was calculated as 2.8 ± 0.7 mM and 2.0 ± 0.3 mM for hypertonic condition. The points represent the mean ± S.E.M. of measurement (n = 9).

As BGT-1 activity in MDCK cells is up regulated by hypertonicity (Yamauchi et al.

1992), I conducted the basolateral GABA uptake at different GABA concentrations in buffers with osmolarity 350 mOsmo/L (isotonic) and 500 mOsmo/L (hypertonic) (Fig.

2.5). Both curves followed Michaelis-Menten kinetics with K

m

and V

max

summarized in Table 2. This suggests that the GABA transporter activity was stimulated or the protein expression increased in MDCK cells membranes under hypertonic conditions, contributing to 3.5 times larger V

max

, whereas the K

m

was not influenced.

Table 2 Km and Vmax of basolateral GABA uptake in MDCK cells.

osmolarity condition Km (mM) Vmax (pmol·cm-2·min-1) isononic (350 mOsmo/L) 2.0 ± 0.3 160.0 ± 15.5 hypertonic (500 mOsmo/L) 2.8 ± 0.7 572.2 ± 33.7

Another GABA uptake was designed to investigate the effect of BGT-1 and TauT (Fig.

2.6). Some compounds, serving as ligands to BGT-1 or TauT or not, were selected to

compete with GABA. Substrates affinities of these two transporters are shown in

Table 3. [

3

H]-GABA uptake was inhibited by taurine to 60% of that without inhibitor

and only 10% of that without inhibitor in bovine brain capillaries and immortalized

mouse brain capillary endothelial cell line (TM-BBB) cells, respectively (Takanaga et

al. 2001, Zhang et al. 1998).

(12)

10 

Fig. 2.6: Basolateral GABA uptake in MDCK cells inhibited by different compounds. The GABA concentration was 28 nM (trace amount) with blank (Trace), 20 mM taurine (Tau), 20 mM guvacine (Guv), 20 mM nipeotic acid (Nip), 20 mM beta-alanine (β-ala), 20 mM alpha-alanine (α-ala), 20 mM leucine (Leu) and 20 mM betaine (Bet). All measurements were made in isotonic buffer 350 mOsmo/L (black) and hypertonic buffer 500 mOsmo/L (grey) at pH 7.4. The points represent the mean ± S.E.M.

of measurement (n = 9).

Table 3 Substrate specificity of BGT-1 and TauT transporter taurine guvacine nipecotic

acid

β-alanine α-alanine leucine betaine

BGT-1 ++a, e ++ a, e ++e + a, d - f - f ++d

TauT ++c ++b ++b ++c -b -c -c

a, “++” reduced transporter activity to < 40% of that without inhibitor; t; “+” reduced transporter activity to 40% - 70% of that without inhibitor and “-” reduced transporter activity to > 70% of that without inhibitor.

b, Source: TauT expressed Xenopus oocytes (Anderson et al. 2004).

c, Source: rat retinal capillary endothelial cell line (Tomi et al. 2008).

d, Source:BGT-1 expressed Xenopus oocytes (Matskevitch et al.1999).

e,Source: mouse brain capillary endothelial cell line (Takanaga et al. 2001).

f, Source: bovine brain capillary endothelial cells (Zhang & Liu, 1998).

As shown in Fig.2.6, the transporter activity with blank (without any inhibitor) was

defined as 100%. The function of transporter could be inhibited partially by taurine,

guvacine, nipecotic acid and betaine to < 40% residual activity compare with the

blank, and the inhibition was similar both in isotonic and hypertonic conditions.

(13)

2.3.

Attempts at establishment of a stable cell line MDCK-hPAT1

I attempted to transiently transfect a plasmid encoding hPAT1 into MDCK cells.

Lipofectamin 2000 was chosen as the transfection reagent and attempts were made between 5×10

4

to 1.2×10

5

cells/mL, with 1 μg DNA per 3 mL Lipofectamine 2000 and approximately 0.4 - 1.3 ng DNA per 10

2

cells (see “Transient transfection” in

“Material and Methods”). The transfection efficiency was tested as hPAT1 activity by

the uptake of [

3

H]-proline for all the cells. The uptake of water control group, where

pCP was replaced with water and transfected into cells, was used to see how much the

uptake would be stimulated if the transfection succeeded. However, for all the

conditions I tried, the uptake of the transfected cells did not show any apparent

increase compared with water-control group, and sometimes this uptake decreased

because of the presence of the dead cell killed by Lipofectamine 2000. The

transfection condition with 9×10

4

cells/mL and 400 ng DNA was measured as the

optimum condition, resulting in only 7% higher uptake compared with the control

group. Transfection then was also attempted in human embryonic kidney 293 (HEK

293) cells, but Lipofectamine 2000 seemed toxic and killed some cells because the

cell monolayer was hardly formed.

(14)

12 

3. Discussion

3.1. Proline uptake in MDCK

According to the first two proline uptake experiments, [

3

H]-proline uptake in MDCK cell was not stimulated by a proton gradient across the cell membrane and the uptake increased linearly with the proline concentration. It was reported that the uptake of proline was two times larger at higher pH than lower pH (Røigaard-Petersen et al.

1987). It can be concluded that there does not appear to be any proton-coupled amino acid transporter in these MDCK cells.

3.2. GABA uptake in MDCK

A BGT-1 encoding sequence has been cloned from MDCK, showing the presence of BGT-1 in MDCK cells (Yamauchi et al. 1992). The GABA transport activity was characterized kinetically by [

3

H]-GABA uptake. The calculated K

m

value 2.2 ± 0.4 mM here was not in the same range as reported previously; K

m

for GABA uptake in MDCK cells was 120 μM and 93 μM in occytes (Yamauchi et al. 1992). This indicated that there might be another transporter with GABA affinity in MDCK cells.

As the K

m

value for [3H]-GABA uptake via TauT was reported as 1.5 mM, in a rat retinal capillary endothelial cell line (TR-iBRB2) (Tomi et al. 2008), TauT might have an influence on this GABA transport in MDCK cells.

3.3. Transient transfection

The transfection efficiency was tested as hPAT1 activity by [

3

H]-proline uptake. There was one limitation for this approach as it could not be concluded that the lack of the protein function resulted from the failure of the transfection or from the problem of protein expression after the plasmid entering cells. mRNA assay would an appropriate method to clarify this puzzle. Neomycin resistance selection was used for stable transefection. As the initial proline uptake assay showed that the transient transfection did not work well, neomycin selection was not conducted for this transfection attempts.

The transient transfection should be tried with other transfection reagents in the future.

A more direct assay for transfection efficiency is needed. The alternative is virus-mediated stable transfection to establish the new cell line MDCK-hPAT1.

Xenopus oocytes expressing hPAT1 would give an in-vitro model to get as much

kinetical data as possible.

(15)

4. Materials and Methods

4.1. Cell cultures and plasmid

MDCK cells were obtained from Lundbeck A/S (Taastrup, DK) and the cell culture medium, Dulbecco’s Modified Eagle Medium (DMEM), was from Life Technologies (Høje Taastrup, DK). MDCK cells were cultured in flasks and passaged in DMEM supplemented with 0.1 mM non-essential amino acid (NEAA), 10% Fetal Bovine Serum (FBS), 0.1 mg/mL penicillin and 0.1 mg/mL streptomycin, maintained at 37 ℃ in a humidified atmosphere of 95% air and 5% CO

2

. Cell culture medium supplements were from Life Technologies (Høje Taastrup, DK).

Plasmids are described in Table 4.

Table 4 Plasmids.

plasmid parental plasmid relevant properties source pCDNA3.1(+)a cloning vector; NeoR Invitrogen

pCP pCM NeoR, hPAT1

Synthesized by Bioneer A/S, Hørsholm, DK

a, 5.4 kbp vector for high levels of expression in mammalian cells; multicloning site after cytomegalovirus immediate early promoter, encodes neomycin resistance.

4.2. [

3

H]-proline and [

3

H]-GABA uptake assays

4.2.1. Apical uptake

The MDCK cells were split into a 24-well plate and grown for three days until 100%

confluency. For each well, the medium was discarded and the cells were incubated

with 1 mL Hank’s balanced salt solution (HBSS, from GIBCO, Invitrogen, Paislay,

UK) supplemented with 10 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

(HEPES) and 0.05% bovine serum albumin (BSA), pH 7.4 at 37 ℃ for more than 10

min at 90 revolutions per minute (RPM) so that the pH value of the interior

environment of the cell would be kept at 7.4. The buffer was discarded and then 1 mL

labeling mix was added to each well with radioactivity 1 μCi/mL. The labeling mix

consisted of HBSS buffer supplemented with radioactive substrates. γ-[2,3-

3

H(N)]-

aminobutyric acid (GABA, 35 Ci/mmol), D-[1-

14

C]-mannitol (56.5 Ci/mmol) and L-

[2,3,4,5-

3

H]-proline (75 Ci/mmol) were from PerkinElmer (Boston, MA, USA). For

[

3

H]-proline uptake, the radioactive proline were dissolved in HBSS supplemented

with 10 mM 2-(N-morpholino)ethanesulfonic acid (MES) and 0.05% BSA, pH 6.0 or

7.4. The first resulted in a proton gradient with the interior pH value at 7.4 and

exterior pH value at 6.0 across the cell membrane and the second buffer caused no pH

gradient across the membrane. For [

3

H]-GABA uptake, the isotope labeled GABA and

other non-labeled competitive substrates were dissolved in modified HBSS (with or

(16)

14 

without 137 mM sodium chloride) supplemented with 10 mM HEPES and 0.05%

BSA, pH 7.4. For both situations, [

14

C]-mannitol (1 μCi/mL) was added to the labeling mix to correct for artifact due to entry of the radioactive solution into minor spaces among cells, also called residual volume, which would be counted as [

3

H]-proline or [

3

H]-GABA uptake. As mannitol does not pass across the cell membrane, its presence could be used to estimate the residual volume, so that the radioactivity of the [

3

H]-proline or [

3

H]-GABA left in the residual volume could be calculated and subtracted.

The plate was incubated at 37 ℃ for uptake at 90 RPM. After the desired uptake time, the labeling mix was discarded and the cells were washed twice with 1 mL ice cold HBSS. After that, 200 μL 0.1% Triton-100 was added to each well and the plate was shaken at 37 ℃ at least 10 min for the cell detachment. All the contents of each well were transferred to a transparent counting vial with 2 mL Ultima Gold scintillations liquid (PerkinElmer, Boston, MA, USA). 20 μL labeling mix were taken out and mixed with 2 mL scintillation fluid to measure the isotope standard radioactivity. All the vials were vortexed for 10 seconds before counting.

4.2.2. Basolateral uptake

The MDCK cells were split into a 12-well transwell plate and grown for three days until 100% confluency (Fig. 4.1). The monolayer could not be seen by naked eyes when seeded on the transwell support material. If the monolayer was formed, there should be an apparent transepithelial resistance (TEER) of the monolayer, which was assessed by a tissue resistance measurement chamber (Endohm) at room temperature with a voltmeter (EVOM) (World Precision Instrument, Sarasota, FL, USA). For each well, the medium was discarded. 0.5 mL and 1 mL HBSS supplemented with 10 mM HEPES and 0.05% BSA (pH 7.4) was added to the upper well and lower well respectively. The cells were incubated at 37 ℃ for more than 10 min at 90 RPM.

Fig. 4.1: 12-well transwell plate. A:one 12-well late and B: one well of a transwell plate. The insert divides each well into upper well and lower well. The bottom of the insert is a support material where the cells grow.

For both [

3

H]-proline and [

3

H]-GABA, the labeling mix used in basolateral uptake

(17)

was the same as used in apical uptake. The modified HBSS with or without sodium was made according to the recipe in Table 5.

Table 5 Recipes for 500 mL modified HBSS with or without sodium.

chemical formula

HBSS with sodium HBSS without sodium concentration (mM) amount (mg) concentration (mM) amount (mg)

CaCl2·2H2O 1.3 92.5 1.3 92.5

KCl 5.4 200.0 5.4 200.0

KH2PO4 0.4 30.0 0.4 30.0

MgCl2·6H2O 0.5 50.0 0.5 50.0

MgSO4·7H2O 0.4 50.0 0.4 50.0

NaCl 137 4000 --- ---

Na2HPO4·7H2O 0.34 45.0 --- ---

C5H14ClNO --- --- 137.5 9600

K2HPO4 --- --- 0.34 30.0

After the incubation, the buffer in the lower well was discarded. 1 mL labeling mix was added only into the lower well and the timer started. The plate was incubated at 37 ℃, 90 RPM. The reaction was stopped by removal of the labeling mix in the lower well and washing with 1 mL ice cold HBSS. After that, the upper well and the lower well were washed with 0.5 mL and 1.0 mL ice cold HBSS twice, respectively.

The support material of the insert in each well was cut down and transferred to a counting tube with 2 mL Ultima Gold scintillations liquid. 20 μL labeling mix were taken out and mixed with 2 mL scintillation fluid as donor samples. All the vials were vortexed for 10 seconds before counting.

4.3. Transient transfection

The day before transfection, cells were trypsinized and plated in a 24-well plate at the concentration of 1.0×10

5

cell/mL, resulting in 90% confluency on the following day.

Plasmid pCP was transiently transfected into MDCK cells using Lipofectamin 2000 (LF2000) according to the protocol of the manufacturer (Invitrogen). The ratio of DNA: LF2000 was kept at 1 μg: 3 mL constantly. For each well in 24-well plate, LF2000 and DNA were separately diluted into 50 μL normal medium without serum, being incubated for 5 min at room temperature. Then the diluted LF2000 was taken out and put into the diluted DNA (total volume = 100 μL for each well), and kept at room temperature for 20 min. After that the DNA - LF2000 complex was added directly to each well. The cells were incubated at 37 ℃ with 5% CO

2

for a total of 24 h. Different transfection conditions are shown in Table 6. Other two groups, water and plasmid pCDNA3.1(+), instead of plasmid pCP, were transfected into cells as control.

The efficiency of transient transfection was measured by the function of hPAT1.

Control groups also were tested by hPAT1 assay, assumed to be the baseline to

measure the efficiency of hPAT1 transfected cells. As hPAT1 could transport proline

across the cell membrane, apical [

3

H]-proline uptake was done for all transfected cells

(18)

16 

(see “Apical uptake” in “Material and Methods”).

Table 6 Conditions for transient transfection in MDCK cells.

No. cell density (cells/mL) amount of DNA (ng)

1 5.0×104 400

2 6.0×104 800

3 7.0×104 400

4 9.0×104 400

5 9.0×104 800

6 1.2×105 800

4.4. Osmolarity measurement

The osmolarity of the buffers used in the uptake and inhibition test was adjusted with mannitol (C

6

H

8

(OH)

6

, a chemical frequently used as osmolyte), and the value was measured by freezing point measurements using an Osmomat 030 (Gonotec, GmbH).

The Osmomat was calibrated with Gonotec Calibration Solution, NaCl in H

2

O (0.300 Osmol/kg).

4.5. Data analysis

Values were calculated as mean±S.E.M. unless otherwise stated. The statistical tests

were performed in GraphPad Prism, GraphPad software v.5.01.

(19)

5. Acknowledgements

Firstly, I would like to show the respect to my supervisor Carsten Uhd Nielson supporting me this opportunity to experiencing in the biopharmaceutical research, which was brand new for me. The study and life in Copenhagen was really impressive.

Thanks to the associate professor Birger Brodin Larsen, for the help of my research supervision and thesis revision.

Thanks to the PhD student Sidsel Ullerup Frølund, for her assistance for lots of problems happening during my daily work and for her passion and cleverness as a nice workmate.

Thanks to the technician staff Bettina Dinitzen, Maria Læssøe Pedersen and Birgitte Eltong, for their professional advice and generous help.

Thanks to my beloved family and parents, to my supportive friends in China, Sweden, Denmark and all over the world, for the excellent moments we shared with each other.

Commutation without barriers;

Science without borders;

And Love, without end.

(20)

18 

6. References

Ahn J., Mundigl O., Muth T.R., Rudnick G., Caplan M.J. 1996. Polarized expression of GABA transporters in Madin-Darby canine kidney cells and cultured hippocampal neurons. J Biol Chem., 271: 6917-6924.

Anderson C.M., Grenade D.S., Boll M., Foltz M., Wake K.A., Kennedy D.J., Munck L.K., Miyauchi S., Taylor P.M., Campbell F.C., Munck B.G., Daniel H., Ganapathy V., Thwaites D.T. 2004. H+/amino acid transporter 1 (PAT1) is the imino acid carrier:

An intestinal nutrient/drug transporter in human and rat. Gastroenterology., 127:

1410-1422.

Artursson P. and Karlsson J. 1991. Correlation between oral drug absorption in humans and apparent drug permeability coefficients in human intestinal epithelial (Caco-2) cells. Biochem Biophys Res Commun., 175: 880-885

Balimane P.V., Han Y.H., Chong S. 2006. Current industrial practices of assessing permeability and P-glycoprotein interaction. AAPS J., 13: 1-13

Bhardwaj R.K., Herrera-Ruiz D., Sinko P. J., Gudmundsson O.S., and Knipp G. 2005.

Delineation of Human Peptide Transporter 1 (hPepT1)- Mediated Uptake and Transport of Substrates with Varying Transporter Affinities Utilizing Stably Transfected hPepT1/Madin-Darby Canine Kidney Clones and Caco-2 Cells. J Pharmacol Exp Ther., 314: 1093–1100.

Borden LA. 1996. GABA transporter heterogeneity: pharmacology and cellular localization. Neurochem Int., 29: 335-356.

Chen Z., Fei Y.J., Anderson C.M.H., Wake K.A., Miyauchi S., Huang W., Thwaites D.T., Ganapathy V. 2003. Structure, function and immunolocalization of a

proton-coupled amino acid transporter (hPAT1) in the human intestinal cell line Caco-2. J. Physiol., 546: 349–361.

Conti F., Minelli A., Melone M. 2004. GABA transporters in the mammalian cerebral cortex: localization, development and pathological implications, Brain Res. Brain Res.

Rev., 45: 196–212.

Christiansen B., Meinild A.K., Jensen A.A., Braüner-Osborne H. 2007. Cloning and characterization of a functional human gamma-aminobutyric acid (GABA) transporter, human GAT-2. J Biol Chem., 282: 19331-19341

Fujiyoshi Y., Mitsuoka K., de Groot B.L., Philippsen A., Grubmüller H., Agre P., Engel, A. 2002. Structure and function of water channels. Curr. Opin. Struct. Biol., 12:

509–515.

(21)

Gether U., Andersen P.H., Larsson O.M., Schousboe A. 2006. Neurotransmitter transporters: molecular function of important drug targets. Trends Pharmacol Sci., 27:

375-383.

Herrera-Ruiz D., Faria T.N., Bhardwaj R.K., Timoszyk J., Gudmundsson O.S., Moench P., Wall D.A., Smith R.L., Knipp G.T. 2004. A novel hPepT1 stably

transfected cell line: establishing a correlation between expression and function. Mol Pharm., 1: 136-144.

Huxtable R.J. 1992. Physiological actions of taurine. Physiol. Rev., 72: 101–163.

Invitrogen manual. 2008, pCDNA3.1

TM

(+) and pCDNA3.1

TM

(-), Catalog nos.

V790-20 and V795-20, Version J, Invitrogen, Paislay, UK.

Irvine J.D., Takahashi L., Lockhart K., Cheong J., Tolan J.W., Selick H.E., Grove J.R.

1999. MDCK (Madin-Darby canine kidney) cells: A tool for membrane permeability screening. J Pharm Sci., 88: 28-33.

Jakubowski H. 2008, Biochemistry Online: An Approach Based on Chemical Logic.

[WWW document]. URL

http://employees.csbsju.edu/hjakubowski/classes/ch331/bcintro/default.html. Date read 2009-07-02.

Larsen M., Larsen B.B., Frølund B., Nielsen C.U. 2008. Transport of amino acids and GABA analogues via the human proton-coupled amino acid transporter, hPAT1:

characterization of conditions for affinity and transport experiments in Caco-2 cells.

Eur J Pharm Sci., 35: 86-95.

Liang R., Fei Y.J., Prasad P.D., Ramamoorthy S., Han H, Yang-Feng T.L., Hediger M.A., Ganapathy V, Leibach F.H. 1995. Human intestinal H

+

/peptide cotransporter:

cloning, functional expression, and chromosomal localization. J. Biol. Chem., 270:

6456–6463.

Matskevitch I., Wagner C.A., Stegen C., Bröer S., Noll B., Risler T., Kwon H.M., Handler J.S., Waldegger S., Busch A.E., Lang F. 1999. Functional characterization of the Betaine/gamma-aminobutyric acid transporter BGT-1 expressed in Xenopus oocytes. J Biol Chem., 274: 16709-16716.

Metzner L., Kottra G., Neubert K., Daniel H., Brandsch M. 2005. Serotonin, L-tryptophan, and tryptamine are effective inhibitors of the amino acid transport system PAT1. FASEB J., 19: 1468-1473.

Mueckler M. 1994. Facilitative glucose transporters. Eur. J. Biochem., 219:713-725.

(22)

20 

Nelson D.L. and Cox M.M. 2004, Lehninger Principles of biochemistry, 4

th

edition, W.

H. Freeman, New York, USA.

Nielsen C.U., Brodin B. 2003. Di/tri-peptide transporters as drug delivery targets:

regulation of transport under physiological and patho-physiological conditions. Curr Drug Targets., 4: 373-388

Pliška V.K. 2001. Substrate binding to enzymes. [WWW document]. URL

http://mrw.interscience.wiley.com/emrw/9780470015902/els/article/a0000862/current /html. Date read 2009-07-02.

Ranaldi G., Islam K., Sambuy Y. 1992. Epithelial cells in culture as a model for the intestinal transport of antimicrobial agents. Antimicrob Agents Chemother., 36:

1374-1381.

Røigaard-Petersen H., Jacobsen C., Iqbal Sheikh M.1987. H

+

-L-proline cotransport by vesicles from pars convoluta of rabbit proximal tubule. Am J Physiol., 253: F15-20.

Saier MH Jr. 2000. A functional-phylogenetic classification system for transmembrane solute transporters. Microbiol Mol Biol Rev., 64: 354-411

Sarmento B., Ribeiro A., Veiga F., Ferreira D., Neufeld R. 2007. Oral bioavailability of insulin contained in polysaccharide nanoparticles. Biomacromolecules. 8:

3054-3060.

Takanaga H., Ohtsuki S., Hosoya Ki., Terasaki T. 2001. GAT2/BGT-1 as a system responsible for the transport of gamma-aminobutyric acid at the mouse blood-brain barrier. J Cereb Blood Flow Metab., 21: 1232-1239.

Thwaites D.T., McEwan G.T.A., Simmons N.L. 1995. The role of the proton electrochemical gradient in the transepithelial absorption of amino acids by human intestinal Caco-2 cell monolayers. J. Membr. Biol., 145: 245–256.

Thwaites D.T., Basterfield L., McCleave P.M.J., Carter S.M., Simmons N.L. 2000.

Gamma-aminobutyric acid (GABA) transport across human intestinal epithelial (Caco-2) cell monolayers. Br. J. Pharmacol., 129: 457–464.

Tomi M., Terayama T., Isobe T., Egami F., Morito A., Kurachi M., Ohtsuki S., Kang Y.S., Terasaki T., Hosoya K. 2007. Function and regulation of taurine transport at the inner blood-retinal barrier. Microvasc Res., 73: 100-106.

Tomi M., Tajima A., Tachikawa M., Hosoya K. 2008. Function of taurine transporter

(Slc6a6/TauT) as a GABA transporting protein and its relevance to GABA transport in

(23)

rat retinal capillary endothelial cells. Biochim Biophys Acta., 1778: 2138-2142.

Volpe D.A. 2008. Variability in Caco-2 and MDCK cell-based intestinal permeability assays. J Pharm Sci., 97: 712-725

Yamauchi A., Uchida S., Kwon H.M., Preston A.S., Robey R.B., Garcia-Perez A., Burg M.B., Handler J.S. 1992. Cloning of a Na(+)- and Cl(-)-dependent betaine transporter that is regulated by hypertonicity. J Biol Chem., 267: 649-52.

Zhang Y., Liu G.Q. 1998. Sodium and chloride-dependent high and low-affinity takes

of GABA by brain capillary endothelial cells. Brain Res., 808: 1-7.

References

Related documents

Herein, we have modified and explored the choline cation in amino acid ILs while generating a novel series of less toxic green amino acid ILs with reduced viscosity and high CO 2

The central hypothesis was that mammalian target of rapamycin (mTOR) signaling regulates placental amino acid transporters in the human placenta in response to nutrient

Keywords: amino acids, fetal growth restriction, human, mammalian target of rapamycin, membrane transporters, metabolism, placenta, pregnancy, system A, system L, taurine

Characterization of Amino Acid tRNA Ligases using the

indicus cells in nutrient-rich growth media delayed cell separation, it is plausible that when these multi-cell chains are transferred back to nutritionally poor medium, they

Focus was also placed on functional aspects of the GABA signaling system in the adult zebrafish brain, and specifically on the effects of stress on GABAA receptor subunits

To evaluate the involvement of the SLC6A15 amino acid transporter in the appetite-suppressing effects of leucine, we first monitored consumption of standard lab chow for four days

amino acid tRNA ligase; analytical ultracentrifugation (AUC); antennapedia; Antp gene; binding site; circular dichroism; conformational change; HMG-CoA lyase; homeodomain;