human multidrug and toxin extrusion protein 1 ...final approval and acceptance of this dissertation...

141
Human Multidrug and Toxin Extrusion Protein 1: Symmetry of substrate fluxes Item Type Electronic Dissertation; text Authors Dangprapai, Yodying Publisher The University of Arizona. Rights Copyright © is held by the author. Digital access to this material is made possible by the University Libraries, University of Arizona. Further transmission, reproduction or presentation (such as public display or performance) of protected items is prohibited except with permission of the author. Download date 08/09/2021 18:45:18 Link to Item http://hdl.handle.net/10150/145435

Upload: others

Post on 21-Aug-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

Human Multidrug and Toxin ExtrusionProtein 1: Symmetry of substrate fluxes

Item Type Electronic Dissertation; text

Authors Dangprapai, Yodying

Publisher The University of Arizona.

Rights Copyright © is held by the author. Digital access to this materialis made possible by the University Libraries, University of Arizona.Further transmission, reproduction or presentation (such aspublic display or performance) of protected items is prohibitedexcept with permission of the author.

Download date 08/09/2021 18:45:18

Link to Item http://hdl.handle.net/10150/145435

Page 2: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

1

HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1:

SYMMETRY OF SUBSTRATE FLUXES

by

Yodying Dangprapai

Copyright © Yodying Dangprapai 2011

A Dissertation Submitted to the Faculty of the

PHYSIOLOGICAL SCIENCES

GRADUATE INTERDISCIPLINARY PROGRAM

In Partial Fulfilment of the Requirements for the Degree of

DOCTOR OF PHILOSOPHY

In the Graduate College

THE UNIVERSITY OF ARIZONA

2011

Page 3: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

2

THE UNIVERSITY OF ARIZONA GRADUATE COLLEGE

As members of the Dissertation Committee, we certify that we have read the dissertation prepared by Yodying Dangprapai entitled Human Multidrug and Toxin Extrusion protein 1: Symmetry of substrate fluxes and recommend that it be accepted as fulfilling the dissertation requirement for the Degree of Doctor of Philosophy

Date: 6/17/2011 Stephen H. Wright

Date: 6/17/2011 William H. Dantzler

Date: 6/17/2011 Nicholas A. Delamere

Date: 6/17/2011 Heddwen L. Brooks Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation to the Graduate College. I hereby certify that I have read this dissertation prepared under my direction and recommend that it be accepted as fulfilling the dissertation requirement.

Date: 6/17/2011 Dissertation Director: Stephen H. Wright

Page 4: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

3

STATEMENT BY AUTHOR

This dissertation has been submitted in partial fulfillment of requirements for an advanced degree at the University of Arizona and is deposited in the University Library to be made available to borrowers under rules of the Library. Brief quotations from this dissertation are allowable without special permission, provided that accurate acknowledgment of source is made. Requests for permission for extended quotation from or reproduction of this manuscript in whole or in part may be granted by the copyright holder.

SIGNED: Yodying Dangprapai

Page 5: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

4

ACKNOWLEDGEMENTS

To Dr. Wattana Watanapa (Department of Physiology, Faculty of Medicine Siriraj hospital, Mahidol University, Bangkok, Thailand) who introduced me to the Physiological Sciences (PS) program at the University of Arizona. She also introduced me to Dr. William H. Dantzler who facilitated my admission to the PS program. To all faculty members who shared their knowledge and scientific skills during the years of coursework and research. Particularly, I would like to thank my committee members for the qualifying examination, including Dr. William H. Dantzler, Dr. Janis M. Burt, Dr. Heddwen L. Brooks, Dr. Alexander M. Simon, and Dr. Stephen H. Wright for your patience and kindness. To friends and colleagues in Tucson. Ryan; I would graduate earlier if I did not take your advice (I know that I asked for it), but working with you was so much fun. Taka; I wish you could have stayed here longer. Bethzaida; I had a good time sharing (political) opinions with you. Xiaohong; It was amazing how you cloned all those fifty-something mutants in no time (It would take me forever). I appreciate your help, including many trips to Costco. Theresa; thank you for keeping our lab organized. I know doing that can drive one crazy, so I wish you a peaceful retirement. Mark; although I prefer a nice and quiet lab, I have to (unwillingly) admit that our lab is too quiet when you are not around. Also, I would like to thank Kris for her kindness and Amrit for his assistance in Dr. Delamere’s lab. To friends back in Thailand for ongoing encouragement, especially to Dr. Suthammarak, Dr. Sewatanon, and Dr. Srisuma for all the phone conversations we shared. Additionally, I would like to thank Mr. Wiggins for being a generous landlord. You are very brave to let me take care of your house and your puppy. Susie is such wonderful company. To the Thai community in the University of Arizona for all the delicious lunches and dinners we shared. Importantly, I would like to express my gratitude to Dr. Supapan Seraphin (Faculty of Engineering, University of Arizona) for her kindness and her wisdom that kept me moving forward during a difficult time. “��������ก���” To the Department of Physiology, Faculty of Medicine Siriraj hospital and to the PS GIDP program, especially Holly A. Lopez, for this invaluable opportunity, for the financial support and for all administrative assistance. To my family for your trust in me, all along, and last but not least, ‘Thank you very much, Dr. Wright!’ Yodying Dangprapai June 2011

Page 6: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

5

TABLE OF CONTENTS

LIST OF FIGURES.................................................................................................8

LIST OF TABLES.................................................................................................10

LIST OF ACRONYMS.........................................................................................11

ABSTRACT...........................................................................................................12

CHAPTER 1: INTRODUCTION

1.1 Organic cations and the human body...........................................................13

1.2 Organic cations and the kidney....................................................................15

1.3 A cellular model of renal organic cation secretion......................................15

1.4 Study of organic cation transport in the kidney...........................................16

1.5 Cloning of the basolateral organic cation transporter..................................18

1.6 hOCT2 as a major contributor for renal basolateral OC uptake.................19

1.7 A molecular identity for the apical organic cation efflux transporter..........21

1.8 The Multidrug And Toxin Extrusion (MATE) transporters........................24

1.9 Cloning of MATE proteins in humans and other mammals........................25

1.10 The physiological fingerprint of MATE-mediated transport.....................26

1.11 Structure of MATE transporters................................................................27

1.12 Kinetics and Selectivity of MATEs...........................................................28

1.13 Kinetics and Selectivity of MATE-mediated transport:

uptake vs. efflux.........................................................................................28

Page 7: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

6

TABLE OF CONTENTS – Continued

CHAPTER 2: MATERIALS AND METHODS

2.1 Chemicals...................................................................................................31

2.2 Cloning of the hMATE1 gene....................................................................31

2.3 Expression of hMATE1 in CHO Flp-In cells.............................................31

2.4 Measurement of [3H]MPP uptake (for time course and kinetic studies)

into CHO hMATE1 cells............................................................................32

2.5 Measurement of MPP efflux from CHO hMATE1 cells............................33

2.6 Manipulation of intracellular pH................................................................35

2.7 Measurement of CHO cell volume.............................................................36

2.8 Statistics......................................................................................................37

CHAPTER 3: SYMMETRY OF INTERACTION FOR H+ AND hMATE1

3.1 Background.................................................................................................38

3.2 Cis-inhibition by extracellular H+ of hMATE1-mediated OC uptake........40

3.3 Characterization of hMATE1-mediated OC efflux....................................43

3.4 Interaction of intracellular H+ with the cytoplasmic face of hMATE1......48

3.5 Interaction between H+ and hMATE1: relevance to renal OC secretion....51

CHAPTER 4: AN APPLICATION OF THE EFFLUX PROTOCOL TO THE

STUDY OF hMATE1-MEDIATED OC INFLUX

4.1 Background................................................................................................56

4.2 Trans-stimulation of hMATE1-mediated OC efflux by MPP...................57

4.3 Trans-stimulation of hMATE1-mediated OC efflux by TEMA................59

Page 8: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

7

TABLE OF CONTENTS – Continued

CHAPTER 5: FUTURE DIRECTION................................................................61

REFERENCES....................................................................................................84

APPENDIX A: INTERACTION OF H+ WITH THE EXTRACELLULAR AND

INTRACELLULAR ASPECTS OF hMATE1....................................................91

Page 9: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

8

LIST OF FIGURES

Figure 1: Chemical structure of selected organic cations (OCs)..........................64

Figure 2: Molecular mechanism of renal organic cation secretion ......................65

Figure 3: Predicted membrane topology of hOCT2.............................................66

Figure 4: Predicted membrane topology of rbMATE1.........................................67

Figure 5: hMATE1-mediated MPP transport: uptake vs. efflux...........................68

Figure 6: The culture dish insert...........................................................................69

Figure 7: Time course and kinetics of [3H]MPP uptake in CHO hMATE1 cells..70

Figure 8: IC50 of extracellular H+ as an inhibitor of hMATE1-mediated MPP

uptake....................................................................................................71

Figure 9: Time course of MPP loss from CHO hMATE1 cells............................72

Figure 10: Time course of NBD-TMA efflux in CHO hMATE1 cells.................73

Figure 11: Effect of extracellular H+ on hMATE1-mediated [3H]MPP efflux......74

Figure 12: Effect of increasing extracellular [H+] on the kinetics of

hMATE1-mediated [3H]MPP efflux...................................................75

Figure 13: Intracellular pH (pHi) in CHO hMATE1 cells....................................76

Figure 14: Effect of NH4Cl and MPP preload on hMATE1-mediated [3H]MPP

uptake..................................................................................................77

Figure 15: Effect of NH4Cl on hMATE1-mediated [3H]MPP efflux...................78

Figure 16: Kinetics of interaction between inward-facing hMATE1 and

intracellular H+...................................................................................79

Page 10: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

9

LIST OF FIGURES – Continued

Figure 17: Effect of increasing extracellular [MPP] on the kinetics of

hMATE1-mediated [3H]MPP efflux..................................................80

Figure 18: Effect of extracellular TEMA on hMATE1-mediated [3H]MPP

efflux..................................................................................................81

Page 11: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

10

LIST OF TABLES

Table 1: Selected organic cations of clinical or environmental significance…..82

Table 2: Effect of extracellular pH on the kinetics of hMATE1-mediated MPP

uptake...................................................................................................83

Page 12: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

11

LIST OF ACRONYMS

BBMV brush border membrane vesicle

CHO cells Chinese hamster ovary cells

CHO hMATE1 cells Chinese hamster ovary cells expressing hMATE1

MATE multidrug and toxin extrusion protein

MDR multidrug resistance protein

MPP 1-methyl-4-phenylpyridinium, a prototypic OC

NBD-TMA [2-(4-nitro-2,1,3-benzoxadiazol-7-yl)

aminoethyl]trimethylammonium, a fluorescent OC

NHE sodium-hydrogen exchanger

NMN N-methylnicotinamide

OC organic cation

OCT organic cation transporter

OCTN organic cation transporter novel

RPT renal proximal tubule

RPTEC renal proximal tubule epithelial cell

T2O tritiated water

TBuA tetrabutylammonium

TEA tetraethylammonium, a prototypic OC

TEMA triethylmethylammonium

TPeA tetrapentylammonium

WB Waymouth’s buffer

Page 13: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

12

ABSTRACT

Human multidrug and toxin extrusion 1 (hMATE1) is a major candidate for being

the molecular identity of organic cation/proton (OC/H+) exchange activity in the luminal

membrane of renal proximal tubules (RPT). Although physiological function of

hMATE1 supports luminal OC efflux, the kinetics of hMATE1-mediated OC transport

have typically been characterized through measurement of uptake i.e., the interaction

between outward-facing hMATE1 and OCs. To examine kinetics of hMATE1-mediated

transport in a more physiologically relevant direction i.e., an interaction between inward-

facing hMATE1 and cytoplasmic substrates, I measured the time course of hMATE1-

mediated efflux of the prototypic MATE1-substrate, [3H]1-methyl-4-phenylpyridinium

([3H]MPP), under a variety of conditions, including different values for intra- and

extracellular pH, from CHO cells that stably expressed hMATE1. I showed that an

IC50/K i for interaction between extracellular H+ and outward-facing hMATE1 determined

from conventional uptake experiments [12.9 ± 1.23 nM (pH 7.89); n = 9] and from the

efflux protocol [14.7 ± 3.45 nM (pH 7.83); n = 3] were not significantly different (P =

0.6). To test a hypothesis that H+ interacts symmetrically with each face of hMATE1,

kinetics of interaction between intracellular H+ and inward-facing hMATE1 were

determined using the efflux protocol. The IC50 for interaction with H+ was 11.5 nM (pH

7.91), consistent with symmetrical interactions of H+ with the inward-facing and

outward-facing aspects of hMATE1. The efflux protocols demonstrated in this study are

a potential means to examine kinetics at cytoplasmic face of hMATE1 and also a

practical tool to screen uptake of substrates at extracellular face of hMATE1.

Page 14: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

13

CHAPTER 1

INTRODUCTION

The human body synthesizes and consumes organic electrolytes on a daily basis.

Once distributed in the body at physiological pH, each of these organic electrolytes

(OEs), as indicated by the name, will typically behave as organic cations (OCs) or

organic anions (OAs). Although various endogenous OEs have significant roles in

homeostasis of the body, my interest is on how the body handles various exogenous i.e.,

xenobiotic, and toxic OEs. Within the scope of this dissertation, the focus is on renal

handling of OCs and, in particular, on an OC efflux transporter at the apical membrane of

the renal proximal tubule (RPT), namely, the Multidrug And Toxin Extrusion protein,

MATE1. Following is an overview of topics related to this issue:

1.1 Organic cations and the human body

Organic cations (OCs) are typically nitrogenous compounds (Fig. 1) that carry net

positive charge(s) at physiological pH, i.e., pH 7.35 to pH 7.45. The human body

synthesizes a wide array of physiologically important OCs, including choline, dopamine,

norepinephrine, and epinephrine. Nevertheless, it is likely that most OCs to which the

body is exposed are exogenous i.e., xenobiotic, and many of these can be toxic.

Importantly, the human body consumes toxic OCs on a daily basis, mostly in the form of

various plant alkaloids (71). This can be viewed from an evolutionary perspective as an

example of “chemical warfare” between plants and animals. In order to survive and to

be able to pass their genes to the next generation, plants synthesize various chemicals that

are toxic to animals, especially to those not participating in pollination. As an adaptation

Page 15: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

14

to the efforts of plants to dissuade the animals that eat them, only animals that are

equipped with a powerful system to handle botanical toxins will survive and be able to

produce offspring (1).

Besides plant products, OCs increasingly appear in modern daily life in the form

of drugs. In fact, approximately 40% of current prescribed medications are OCs (23)

(Table 1). Furthermore, there are increasing numbers of unwanted drug-drug interactions

(DDIs) with mild to fatal results in patients under treatment with multiple OC drugs (40).

Thus, it is crucial to understand how the body is cleared of toxic OCs. In this regard, the

two major organs responsible for the challenging task of OC secretion are the liver and

the kidney. For this dissertation, the focus is on handling of OCs by the kidney. The

interested reader is directed to recent reviews that describe OC transport by hepatocytes

(e.g., (24).

Before further discussion on renal handling of OCs, it is important to address a

definition of ‘OCs’ used in this study. It is very useful to classify OCs into two subtypes

based on structure and physicochemical properties (71). First, ‘type I’ OCs refer to

comparatively small (MW< 400 Da), and generally monovalent compounds, such as

tetraethylammonium (TEA) and 1-methyl-4-phenylpyridinium (MPP). Second, ‘type II’

OCs refer to larger, and frequently polyvalent, compounds that are also typically

comparatively hydrophobic. In this study, unless otherwise indicated, the focus is on a

renal handling of ‘type I’ OCs.

Page 16: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

15

1.2 Organic cations and the kidney

The role of the kidney as a site for OC secretion was first demonstrated in 1947.

Sperber (45) showed evidence in birds for a renal secretion of N-methylnicotinamide

(NMN), an organic cation by-product from metabolism of nicotinamide (vitamin B3). In

the same year, Rennick et al. (47) demonstrated in dogs that renal secretion has a role in

excretion of tetraethylammonium (TEA), a prototypic OC, by the kidney. After these

early reports of a role for the kidney in OC secretion, subsequent studies on renal OC

transport were conducted using a variety of experimental systems, including whole

animals, kidney slices, isolated perfused renal tubules, and renal membrane vesicles.

Results from the studies between the 1950s and the 1970s (reviewed in (45) advanced the

knowledge of renal OC handling toward an understanding of the tubular mechanism for

renal OC transport. The major findings included: (i) renal OC transport is mediated by

unique transporter protein(s) distinct from a previously described transport system for

para-aminohippurate (PAH), a prototypic organic anion (OA); and (ii) a major site for

renal OC secretion is the early portion of the renal proximal tubule (RPT).

1.3 A cellular model of renal organic cation secretion

In 1980-81, based on their work with renal cortical membrane vesicles, Holohan

and Ross published two consecutive articles that described a mechanism for a basolateral

(peritubular) OC uptake that is distinct from a mechanism for an apical (luminal) OC

efflux (18, 19). Furthermore, they suggested that the transporter(s) responsible for apical

OC efflux is functionally coupled to activity of the apical Na+/H+ exchanger (NHE) to

facilitate OC secretion. Three years later, Holohan and Ross (48) proposed a cellular

Page 17: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

16

model for OC secretion in the RPT (Fig. 2). In summary, renal OC secretion is a two-

step process. First, at the basolateral membrane of renal proximal tubule epithelial cells

(RPTECs), OC uptake from the blood is mediated by a single process that operates as (i)

facilitated diffusion, driven by an inside negative membrane potential, or as (ii) OC

exchange with another cytoplasmic OC. Second, at the apical membrane, OC efflux into

the tubular filtrate is mediated by a transport process that exchanges an OC for H+ that is

supplied continuously from the filtrate (and thus influenced by activity of NHEs). It is

important to acknowledge that this cellular model of renal OC secretion proposed by

Holohan and Ross is still valid and widely referred to, despite the fact that the body of

knowledge on OC transport has grown significantly as a result of advanced molecular

biology techniques during the past 20 years.

1.4 Study of organic cation transport in the kidney

Between the mid-1980s and the early-1990s, after Holohan and Ross proposed the

model for renal OC secretion, most studies in this area focused mainly on testing

hypotheses concerning the mechanism of OC/H+ exchange because of its accepted role as

the rate-limiting step in renal OC secretion. Following is a summary (see Pritchard and

Miller, 1993) of observations from selected studies using renal BBMV that are

particularly pertinent to this dissertation and its focus on apical OC efflux in RPTEC:

● Apical OC/H+ exchange is an obligatory exchange process i.e., substrate (OC or

H+) is required for the conformational change(s) of the protein responsible for this

process in both directions across the cell membrane.

Page 18: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

17

● A gradient of H+ drives OC transport in the opposite direction and is capable of

net OC movement against an electrochemical gradient. For example, in renal BBMV, an

outwardly-directed H+ gradient can support concentrative vesicular OC uptake.

● Results from uptake studies of monovalent OCs in BBMV suggested that the

typical stoichiometry of this exchange process is one-to-one. In other words, OC/H+

exchange is electroneutral.

In addition to studies on the mechanism of OC secretion, some workers focused

on substrate selectivity of both basolateral OC uptake and apical OC efflux transport

processes. In summary, for the basolateral membrane, an OC with higher

hydrophobicity, stronger ionic strength, or with amino group(s) on the benzene ring will

penetrate into the cells more effectively than one without these characteristics (61). For

the apical membrane, higher hydrophobicity and basicity are correlated with better

transport (61).

Up until the early-1990s, models for studying renal OC transport, both basolateral

uptake and apical efflux, were limited to preparations from the kidney as mentioned

previously, e.g., renal membrane vesicles and isolated proximal tubules. Although data

from study of native kidney tissues are invaluable, conclusions regarding a role for a

specific transport protein in renal OC secretion could not be readily drawn using such

physiologically complex systems. Yet it was crucial to identify the specific proteins

responsible for OC transport in the renal proximal tubule to form the basis of

understanding the molecular mechanisms for renal handling of OCs. Below I provide a

brief overview of the molecular characteristics of the basolateral entry step in renal OC

Page 19: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

18

secretion, followed by a more extensive discussion of the molecular characteristics of the

apical transport process, MATE1, which is the focus of this dissertation.

1.5 Cloning of the basolateral organic cation transporter

In 1994, Gründemann et al. (12) successfully cloned, from rat kidney, a protein

capable of transporting structurally diverse OCs, and named the protein: organic cation

transporter 1 (rOct1; Slc22a1). Importantly, rOct1 demonstrated kinetic and selectivity

profiles similar to those of basolateral OC transport characterized in studies of tubules in

intact rat kidney, and basolateral membrane vesicles from rat kidney. For example,

affinity of NMN and tetramethylammonium (TMA) for rOct1 expressed in Xenopus

oocytes (12) was similar to affinity of each OC from kinetic studies in renal proximal

tubule (62). It is obvious that cloning of OCTs provided an opportunity for the direct

characterization of specific transport proteins both functionally (kinetically) and

structurally. The landmark study by Gründemann et al. led to the subsequent cloning of

several other organic cation transporter homologs and orthologs, including rOct2;

Slc22a2 (41) and the human (10) and rabbit orthologs (57) of these proteins.

Importantly, it is clear from studies of these different OCT orthologs that there are

marked species differences regarding substrate selectivity, kinetics, and sites of

expression (76). For example,the interaction between hOCT1 and tetraalkylammonium

compounds, such as TEA, is significantly distinct from the interaction between rodent

and rabbit Oct1 orthologs and these compounds (76). Also, whereas both Oct1 and Oct2

are expressed in rat and mouse kidney (71), expression in human kidney is effectively

restricted to OCT2 (39). Thus, to clearly understand handling of OCs in human, it is

Page 20: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

19

important to study hOCTs directly, or to (at least) be aware of inter-species differences

when interpreting kinetic information gained in studies with orthologs of hOCTs.

1.6 hOCT2 as a major contributor to renal basolateral OC uptake

Human OCTs e.g., hOCT1 (SLC22A1) and hOCT2 (SLC22A2) were successfully

cloned in 1997. First, Koehler et al. reported a location of genes coding for hOCT1 and

hOCT2 on chromosome 6 (25), and later, both transporters were successfully cloned (10)

and characterized in various heterologous expression systems, including Xenopus oocytes

(10), HeLa cells (76), HEK-293 cells (16) , CHO cells (3), MDCK cells (63), and

proteoliposomes (21). As a result of the cloning of the OCTs, our understanding of

basolateral OC uptake increased significantly during the late-1990s to the mid-2000s.

Following is a list of selected information regarding expression profile, structure and

kinetics of OCTs:

● OCTs display comparatively high levels of expression in many epithelial

tissues, including renal proximal tubule and liver. In the human, OCT2 is the major OCT

expressed at the basolateral membrane of RPTECs, based on expression of both mRNA

and protein (39). Expression of OCT1 in human kidney is less than 1% of the level for

hOCT2 (72). OCT1 is, however, the major OCT expressed at the sinusoidal membrane

of the human liver (32). In rat, mouse, and rabbit, the kidney expresses both Oct1 and

Oct2 (71). Importantly, from a comparison of renal TEA clearance in wild-type mice

and in mice lacking both homologs of Oct, it is evident that renal secretion of TEA is

accounted for quantitatively by the combined activity of mOct1 and mOct2 (20).

Page 21: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

20

● When expressed in various heterologous expression systems, OCTs

demonstrate an electrogenic OC transport across the plasma membrane (8). Additionally,

OC uptake mediated by OCTs is driven by an inside-negative membrane potential (7).

Furthermore, OCTs can operate as an OC/OC exchanger (8), but an interaction with a

substrate is not required for a conformational change of OCTs. In other words, OCTs do

not operate as an obligatory exchanger (8). Importantly, changes in membrane potential,

but not in the transmembrane gradient of Na+ or H+, affects kinetics of OCT-mediated

transport.

● OCTs interact with a structurally diverse array of substrates and inhibitors, in

agreement with the hallmark of renal OC secretion as a polyspecific transport system

(61). Computationally derived pharmacophores for OCT1 (5) and OCT2 (54) identified

several key molecular determinants of effective substrate/inhibitor interaction with OCTs,

including the placement of hydrophobic mass relative to a basic nitrogen, and the

influence of specific molecular shape and size of a substrate.

● OCTs are members of the Major Facilitator Superfamily of the transport

proteins. They contain 550 to 560 amino acid residues with a membrane topology of

twelve transmembrane spanning domains (TMDs), which includes intracellular N- and C-

termini, and a long cytoplasmic loop between TMDs 6 and 7, and a very long

extracellular loop between TMDs 1 and 2 (12). Figure 3 illustrates proposed secondary

and tertiary structures of hOCT2 (44).

In summary, based on its expression and selectivity profiles (71), OCT2 is widely

regarded as the major contributor to OC uptake at the basolateral membrane of RPTEC in

Page 22: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

21

human kidneys. hOCT2-mediated OC uptake is driven by an inwardly-directed

electrochemical gradient for these cationic substrates, reflecting the inside-negative

membrane potential of RPTECs. Although this basolateral OC uptake is a passive

process, at steady state, OC uptake into the RPTEC, mediated by hOCT2, is capable of

accumulating an OC to a level higher (some 10- to 15-fold) than its concentration in the

peritubular capillary. As discussed in an upcoming section, this fact will prove to play a

critical role in development of an integrated model of renal OC secretion that involves

basolateral OC uptake and MATE-mediated OC efflux in RPTECs.

1.7 A molecular identity for the apical organic cation efflux transporter

The molecular identity for a transporter protein(s) responsible for apical OC

efflux proved to be more elusive than that for basolateral OC entry (the OCTs, described

previously). The physiological characteristics of apical OC transport were known from

studies performed in renal brush border membrane vesicles (BBMV), or in isolated

perfused proximal tubules prepared from the kidney. This “physiological fingerprint”

can be summarized as follows:

● Apical OC efflux is the active and a rate-limiting step for the net OC secretion

across the RPT epithelium (72), based on observations from isolated rabbit RPT that

showed that (i) the rate of basolateral OC uptake is similar along the length of the RPT,

whereas the rate of apical OC efflux decreases (51), and that (ii) BBMV from the outer

renal cortex have a higher rate for OC uptake than BBMV from the inner medulla have

(34).

Page 23: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

22

● Apical OC/H+ exchange operates as an obligatory exchanger with 1:1

stoichiometry for its substrates (OCs or H+) in each translocation cycle resulting in a net

electroneutral substrate exchange across the cell membrane (68).

● Effects of H+ on OC transport i.e., trans-stimulation (via an increase in the

turnover rate) and cis-inhibition (via a competitive inhibition) were demonstrated in renal

brush border membrane vesicles and supported the model of OC efflux as an OC/H+

exchange process (69).

● The apical exit step in renal OC secretion displays a multispecificity

comparable to that of the basolateral entry step for this process (60).

There were a few early candidates for the molecular identity of the apical OC/H+

exchanger, particularly organic cation transporter novel 1 (OCTN1; SLC22A4), OCTN2

(SLC22A5), and multidrug resistance transporter 1 (MDR1; ABCB1) i.e., P-glycoprotein

(P-gp). However, when expressed in various heterologous expression systems and tested

with common OCs, neither of these transport proteins displayed a comparable

kinetic/selectivity profile to that of the OC/H+ exchanger characterized in renal brush

border membrane vesicles.

OCTN1 was an early favorite in large part because of a report showing that it

supported TEA/H+ exchange when expressed in oocytes (73). Nevertheless, an

expression study in the human kidney showed no significant mRNA expression for

hOCTN1 (39). This observation attenuated the potential of hOCTN1 for being the apical

OC/H+ exchanger. A subsequent study in HEK-293 cells expressing hOCTN1 showed

that transport efficiencies (‘TEs’; i.e., Jmax/Kt) ((13) for TEA and other‘type I’ OCs are so

Page 24: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

23

small that it is unlikely that this process could play a significant role in the multispecific

OC secretion. Instead, it is more likely that OCTN1 is largely dedicated to the transport

of the antioxidant, ergothioneine (ET) (the TE for which is approximately 100 times

higher than that for TEA transport) (14). Interestingly, mutations in hOCTN1 gene are

found to associate with autoimmune diseases such as rheumatoid arthritis and Crohn’s

disease (26).

Another candidate for an apical OC efflux transporter was OCTN2, based on its

expression at the apical membrane of the RPTEC (39) and the observation that it can

mediate transport of prototypic OCs such as TEA. However, like its congener, hOCTN1,

hOCTN2-mediated OC transport has different kinetic/selectivity profiles from those

observed for OC/H+ exchange in renal BBMV. Interestingly, subsequent studies also

revealed that the transport activity of OCs by hOCTN2 probably has no physiological

significance (given the very low transport efficiencies for OC substrates). Instead,

hOCTN2 demonstrates high transport efficiency for carnitine that is required for normal

cellular lipid metabolism (55).

The final, ‘early’ candidate for apical OC export was MDR1. However, its

influence in renal OC secretion has long been problematic. Kinetic profiles for selected

prototypic OCs such as TEA for MDR1 are very different from those observed in renal

BBMV (72). Currently, there is a broad agreement that MDR1 is more likely to have a

role in an efflux of ‘type II’ OCs at the apical membrane of RPTEC (72). These

polyvalent and bulky OCs are proposed to passively diffuse across the basolateral

Page 25: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

24

membrane into the RPTECs. Subsequently, an apical MDR1 helps to accelerate an ATP-

dependent secretion of the ‘type II’ OCs.

Because of these unsuccessful efforts to identify a transporter protein(s)

responsible for the OC/H+ exchange activity at the apical membrane of the RPTEC,

knowledge on the apical OC transport step lagged behind that for the basolateral element

in renal secretion of OCs.

1.8 The Multidrug And Toxin Extrusion (MATE) transporters

While students of renal OC transport were searching for the molecular identity of

the apical OC/H+ exchanger, a group of microbiologists also searched for an explanation

behind a resistance to cationic, quinolone-based antibiotics such as norfloxacin in Vibrio

parahaemolyticus. In 1998, Morita et al. demonstrated that a novel protein, NorM, is

responsible for norfloxacin resistance in V. parahaemolyticus, and suggested that NorM

represents a new group of energy-dependent, secondary active, multidrug efflux proteins

(36), that were later categorized as a new family of transporters named Multidrug And

Toxic compound Extruders, or MATEs (6). Mechanistically, NorM was proposed to

function as an Na+/OC exchanger (35). Since the description of NorM, more than 1,000

MATE family members in bacteria, plants, and fungi have been identified (28).

To introduce human MATE proteins (hMATEs) as the putative molecular identity

responsible for OC efflux at the apical membrane of RPTECs, I provide key information

in the following paragraphs on (i) discovery of hMATEs, (ii) functional characteristics of

hMATE1, (iii) structure of MATEs, and (iv) the kinetics and selectivity of hMATE1.

Page 26: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

25

1.9 Cloning of MATE proteins in humans and other mammals

In 2005, Otsuka et al. successfully cloned transporters now considered as the

major molecular candidates for being the long sought apical OC/H+ exchange activity of

RPTECs (42). Briefly, they used the DNA sequence encoding NorM, a prokaryotic

Na+/OC exchanger from V. parahaemolyticus, as a template, and then searched the

human genome for similar DNA sequences. They found two similar genes located on the

short arm of chromosome 17 and later named the product of each gene as a human

multidrug and toxin extrusion protein, i.e., hMATE1 (SLC47A1) and hMATE2

(SLC47A2).

Orthologs of hMATE1 were also successfully cloned in rabbit, rat and mouse.

MATE2 was found, in the human, to have two additional splice variances, hMATE2-K

and hMATE2-B. hMATE2-K is a functionally active product of SLC47A2, expression

of which is restricted to the kidney (in humans). On the other hand, functional roles for

both hMATE2 and hMATE2-B (so-called because expression appears to be restricted to

the brain) are still to be elucidated. A functional ortholog of hMATE2-K is also

expressed in rabbit kidney as rbMate2-K (77). In mouse, there is an expression of a

mMate1 homolog that is currently named mMate2. However, mMate2 is not an ortholog

of hMATE2 or of rbMate2 (74), and phylogenetic evidence suggests mMate2 should be

called mMate3 (74). Interestingly, rodent ‘MATE2’ is not expressed in the kidney and is

largely restricted to the testes (29).

Page 27: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

26

1.10 The physiological fingerprint of MATE-mediated transport

Several observations highlighted the potential role of hMATE proteins in renal

OC secretion. First, mRNA and protein for hMATE1 are highly expressed in the human

kidney (and human liver, the other major location for functional expression of OC/H+

exchange activity (38, 37). Next, from immunohistochemistry, hMATE1 is detected at

the apical region of the renal tubule including the proximal tubule (42). Importantly,

unlike other previous candidates for an apical OC/H+ exchanger, when expressed in a

heterologous expression system, hMATE1 revealed selectivity profiles (56) for various

OCs similar to the apical OC/H+ exchanger in renal BBMV and isolated renal proximal

tubules (71). Furthermore, significance of MATE1 on renal clearance of clinically

important substrates has been demonstrated in mice lacking mMate1, the only MATE

homolog expressed in mouse kidney (74). Renal clearance of the antidiabetic drug,

metformin (59) and the antibiotic, cephalexin (66), were significantly impaired in Mate1-

knockout mice.

As noted earlier, hMATE2-K is effectively restricted to the kidney (74) and has a

profile of substrate selectivity similar to that demonstrated for hMATE1 (74). However,

it is not known (i) if MATE1 and MATE2-K are coexpressed in the same RPTECs; or

(ii) if expression of either MATE homolog varies along the length of the RPT. It is

important to emphasize, therefore, that both hMATE1 and hMATE2-K are potential

contributors to the apical OC/H+ exchange activity, based on their expression profiles and

substrate interaction patterns, as discussed above. However, the aforementioned

evidence from studies of the importance of MATE1 in renal secretion of OCs in mouse

Page 28: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

27

kidney clearly implicates this process as a likely contributor of OC secretion in human

kidney. Consequently, my study focused on characterization of hMATE1-mediated OC

transport.

1.11 Structure of MATE transporters

Since its discovery in 2005, primary attention has been directed toward

understanding the function of mammalian MATE transporters. Comparatively little

attention has been given to understanding the structure of these proteins. Hydropathy

analyses suggest that MATE family members typically have a secondary structure with

12 TMDs. However, all the mammalian MATEs appear to have 13 TMDs, the result of

an extracellular C-terminal hydrophobic domain (78). Studies using epitope-tagging and

impermeant thiol-reactive probes have confirmed the presence of this 13th TMD in

human, rabbit and mouse MATE1 (78); and unpublished observations). But in order to

appreciate the molecular mechanism of MATE transporters, a detailed molecular

structure at the atomic level is required.

In 2010, He et al. (17) reported an x-ray crystal structure of the prototypic

bacterial MATE family member, NorM, with a resolution of 3.6 Å. The structure

confirmed the presence of 12 TMDs and provided the template for development of a

homology model of the 3D structure of mammalian MATE1 (Fig. 4) that serves as the

basis of ongoing studies of structure/function relationships for the mammalian MATEs.

Page 29: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

28

1.12 Kinetics and Selectivity of MATEs

Despite the acknowledged importance of MATE transporters in mediating renal

(and hepatic) OC secretion, comparatively little attention has been given to kinetics and

substrate selectivity of MATEs. Previous studies on MATEs have provided qualitative

information on their substrates; e.g., drug A interacts with hMATE1, whereas drug B

does not. Only a limited number of studies have been focused on the kinetic

characterization of MATE selectivity.

1.13 Kinetics and Selectivity of MATE-mediated transport: uptake vs. efflux

As noted above, there are comparatively few quantitative studies on the kinetics

of MATE-mediated transport. Importantly, what information is available is all directed

toward understanding the characteristics of MATE-mediated substrate ‘uptake.’ Under

physiological conditions at the apical membrane of the RPTEC, hMATE1 mediates OC

efflux i.e., an interaction between OCs and the cytoplasmic face of hMATE1, yet with

very few exceptions, what is known about hMATE1-mediated OC transport has been

obtained from uptake (influx) experiments, i.e., an interaction between the extracellular

face of hMATE1 and OC (Fig. 5). Results from hMATE1-mediated OC uptake have

been widely accepted to represent kinetics of hMATE1-mediated OC efflux based on an

assumption that interaction between each conformation of hMATE1 and substrates is

symmetrical (e.g., 31). Importantly, there is no experimental information on symmetry of

hMATE1-mediated transport.

There is, however, information on the symmetry of a poly-specific OC

transporter, namely, OCT2. Volk et al. (63) used the patch-clamp technique to examine

Page 30: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

29

symmetry of rat organic cation transporter 2 (rOct2, slc22a2) and showed that binding

affinity of TBuA (tetrabutylammonium) and corticosterone to each face of rOct2 is

significantly different. More interestingly, while TBuA has 4-fold higher affinity to

external over cytoplasmic surface of rOct2, corticosterone has binding preference to

cytoplasmic (20-fold) over external surface of the protein (64). This study suggests not

only that an interaction between a substrate to each conformation of a poly-specific

transporter can be significantly different, but also that differences in affinity for a given

substrate at the two sides can vary widely among different substrates. Therefore, it is

critical to examine symmetry of substrate interaction in a poly-specific transporters,

including hMATE1.

To characterize hMATE1-mediated transport in a physiologically relevant

direction i.e., hMATE1 as an OC efflux transporter, I developed a method to

quantitatively examine the interaction between inward-facing hMATE1 and its substrates,

including H+. The major hypothesis of this study was that H+, as a substrate for hMATE1,

interacts symmetrically with each conformation of hMATE1. To test this hypothesis,

hMATE1 was stably expressed in Chinese Hamster Ovary (CHO) cells, and kinetics of

interaction between H+ and the inward and outward-facing faces of hMATE1 and H+

were examined.

Page 31: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

30

The organization of upcoming Chapters

• Chapter 2 describes the general methods used throughout the studies that

comprise this dissertation.

• Chapter 3 presents the background, results and discussion of work described in

the research paper: Dangprapai Y, Wright SH. Interaction of H+ with the

extracellular and intracellular aspects of hMATE1. Am. J. Physiol. Renal Physiol,

2011 (in press).

• Chapter 4 describes the results of a set of currently unpublished experiments that

explore the utility of efflux measurements to assess the kinetics of hMATE1-

mediated substrate uptake.

• Chapter 5 provides an integrated view of this body of work and discusses the

future direction of MATE studies in the light of my research.

Page 32: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

31

CHAPTER 2

MATERIALS AND METHODS

2.1 Chemicals

[3H]1-methyl-4-phenylpyridinium ([3H]MPP; 80 Ci/mmol) and [2-(4-nitro-2,1,3-

benzoxadiazol-7-yl)aminoethyl]trimethylammonium [NBD-TMA (4)] were synthesized

by the Department of Chemistry and Biochemistry, University of Arizona. [14C]Mannitol

(58.8 mCi/mmol) was purchased from PerkinElmer. 2’,7’-bis-(2-carboxyethyl)-5-(and-

6)-carboxyfluorescein acetoxymethyl (BCECF-AM) and nigericin were obtained from

Invitrogen. Tetrapentylammonium (TPeA) bromide, saponin, and other chemicals

(unless otherwise noted) were purchased from Sigma.

2.2 Cloning of the hMATE1 gene

pcDNA5/FRT vector (Invitrogen) containing hMATE1 cDNA was provided by

Dr. Giacomini (UCSF). Briefly, cDNA of hMATE1 (GenBank accession no.

NM_018242) was amplified from a human kidney RNA sample using the reverse

transcription polymerase chain reaction (RT-PCR). Subsequently, the cDNA product

was cloned into the pcDNA5/FRT vector, and the sequence later confirmed with an

Applied Biosystems 3730xl DNA analyzer at the University of Arizona sequencing

facility.

2.3 Expression of hMATE1 in CHO Flp-In cells

Chinese Hamster Ovary cells containing a single FRT site (CHO Flp-In cells)

were purchased from Invitrogen. The cells were cultured in nutrient mixture F-12 Ham

Kaighn's Modification (Ham's F-12K) medium (Sigma) supplemented with 10% fetal

Page 33: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

32

bovine serum (HyClone) and 0.1 mg/ml Zeocin (Invitrogen). The cells were maintained

at 370C in an incubator (NuAire) supplied with 5% CO2. CHO Flp-In cells grown in T-

75 flasks (Cellstar) at ~ 80 – 90% confluency were used for electroporation.

pcDNA5/FRT/hMATE1 and pOG44 vectors containing Flp recombinase (Invitrogen)

were co-introduced by electroporation into a suspension of CHO Flp-In cells (~ 4 x 106

cells/ml in Ham’s F-12K medium). Subsequently, the electroporated cells were

maintained in serum-supplemented Ham’s F-12K medium containing 0.1 mg/ml of

Hygromycin B (Invitrogen).

2.4 Measurement of [3H]MPP uptake (for time course and kinetic studies) into CHO

hMATE1 cells

CHO hMATE1 cells were seeded in 24-well cell culture plate (Greiner Bio-one)

and allowed to reach ~100% confluency (typically 48 hours). The cells were rinsed twice

with WB pH 7.4 before being incubated in WB pH 8.5 containing [3H]MPP (~13 nM) at

room temperature (RT; 20 – 250C). For a time course study, [3H]MPP solution was

aspirated at various time points from each well and the cells were rinsed three times with

1 ml of ice-cold WB pH 8.5. For a kinetic study, the cells were incubated with WB pH

8.5 containing [3H]MPP with various concentrations of non-labeled MPP, and all

incubations were terminated at 5 minutes by removing the buffer and rinsing with ice-

cold WB as mentioned in the time course study. To release accumulated [3H]MPP, CHO

hMATE1 cells were lysed with 0.5 N NaOH containing 1% SDS and, subsequently,

neutralized with 1 N HCl. Cell lysate from each sample was aliquoted to a plastic vial

Page 34: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

33

and mixed with scintillation cocktail (MP Biomedicals), and radioactivity in the lysate

was determined by liquid scintillation spectroscopy (Beckman).

Two approaches were used to study the kinetics of cis-inhibition by hydrogen ions

(H+) of hMATE1-mediated MPP uptake . For the first protocol, CHO hMATE1 cells

were incubated for 5 minutes with ~13 nM of [3H]MPP in WB of various H+

concentrations. The second approach was based on measuring the kinetics of hMATE1-

mediated MPP uptake at two different values of extracellular pH, as will be described

later in this dissertation.

2.5 Measurement of MPP efflux from CHO hMATE1 cells

Two similar approaches were developed to characterize hMATE1-mediated MPP

efflux. Both involved a two-step process that started with loading cells with [3H]MPP,

followed by assessment of [3H]MPP efflux from the preloaded cells. The first approach

was applied to CHO hMATE1 cells grown to complete confluence in 48-well cell culture

plates, whereas the second one was performed using a laminar flow perfusion chamber

created by assembly of a dish insert (Warner Instruments, Fig. 6) into a 35-mm cell

culture dish (Falcon) containing CHO hMATE1 cells.

For efflux studies using 48-well plates, cells were first incubated with WB pH 7.4

for 30 minutes. After aspirating this medium, the cells were loaded with labeled substrate

by adding WB pH 8.5 containing ~13 nM of [3H]MPP, followed by a 20 minute

incubation. After the buffer was aspirated, the cells were rinsed quickly (within 1 – 2

seconds) with WB pH 8.5 at room temperature. Efflux of the labeled substrate from

CHO hMATE1 cells was initiated by adding WB of various composition, according to

Page 35: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

34

the purpose of each specific experiment. The time course of efflux under these test

conditions was determined by aspirating the efflux medium at desired time intervals and

then determining the amount of labeled substrate remaining in the cells (as described

earlier).

For experiments that used the perfusion chamber approach, CHO hMATE1 cells

were grown in a 35-mm cell culture dish. When reaching confluence, cell medium was

discarded and the cells were incubated with WB pH 7.4 for 30 minutes at room

temperature. After the WB was completely aspirated, a dish insert (Fig. 6) was assembled

on top of the layer of CHO hMATE1 cells in the cell culture dish. The flat surfaces of

the dish insert were covered with a thin layer of grease to create a sealed chamber

containing CHO hMATE1 cells (Fig. 6). Spring clips were used to firmly stabilize the

chamber. The exposed cells in the chamber were loaded with labeled substrate by

introducing WB pH 8.5 containing ~39 nM of [3H]MPP to the chamber via polyethylene

(PE) tubing. Additionally, this loading buffer also contained ~10 µM of [14C]Mannitol,

which was used to correct for extracellular volume during subsequent calculations. At

the end of the loading step (~20 minutes), the radioactive buffer was cleared from the

chamber by purging it with air (which cleared the label-containing medium via the

second PE tubing line; Fig. 6).

To initiate MPP efflux, WB was introduced into the chamber at a constant rate of

5 ml/minutes by a syringe pump (Harvard apparatus). The flow characteristics of the

chamber resulted in a laminar flow of medium through the chamber. The perfusate was

simultaneously collected into a sample vial from the distal end of the chamber at a rate of

Page 36: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

35

6 seconds per sample. At the end of efflux duration, amount of radioactivity from each

radionuclide in each perfusate sample was determined, as was the amount of each label

remaining in the cells in the chamber, using a dual-label counting protocol.

2.6 Manipulation of intracellular pH

For measurement of intracellular pH (pHi), CHO hMATE1 cells were grown to

approximately 50% confluence in 35-mm cell culture dishes. The low level of cell

confluency facilitated monitoring pHi of a selected single cells during the study. To

measure pHi, the cells were first rinsed twice with WB pH 7.4 at room temperature, and

then incubated in the dark at room temperature in WB containing 5 µM of the pH-

sensitive fluorescent probe BCECF-AM (membrane permeable). Subsequently, the cells

were rinsed at least five times with WB pH 7.4 to remove excess probe. After the last

rinse, the cells were incubated in WB pH 7.4 for at least 10 minutes to allow the complete

enzymatic cleavage of intracellular BCECF-AM (less fluorescent) to membrane

impermeable BCECF (strongly fluorescent).

To visualize the cells, the 35-mm cell culture dish was securely placed on the

stage of a fluorescence microscope (Nikon TS100) equipped with PE tubing that was

connected to a peristaltic pump to provide a suction of test buffer during the

measurement. BCECF was excited alternately at 460 nm and 488 nm, with an emission

wavelength of 530 nm. Under the microscope, a visual field was selected that contained

cells with a homogeneous fluorescence signal. From this field, the pHi of at least 20 cells

was monitored. The fluorescence signal during each measurement was recorded using an

InCyt Im2 imaging system (Intracellular Imaging, Inc.). A value of pHi for each

Page 37: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

36

fluorescence ratio (488/460 nm) was calculated from a calibration curve performed at the

end of each experiment. A calibration curve was determined by exposing cells to various

standard pH buffers mimicking fluid composition of the cytoplasm. Nigericin (10 µM,

freshly prepared) was added into each standard pH buffer to facilitate an equilibration of

pHi to pH of the buffer. Note that only cells that gave a stable signal throughout a course

of the measurement (10 – 15 minutes) were included in a final analysis for calculation of

the pHi.

2.7 Measurement of CHO cell volume

CHO hMATE1 cells were grown to complete confluence in a 48, 24, or 12-well

cell culture plate. For a time course study, the cells were incubated with WB pH 7.4

containing tritiated water (T2O) and [14C]Mannitol (approximately 60 mM and 4 µM,

respectively). The incubation was terminated after various durations by aspirating the

radioactive buffer (without any rinsing); steady state for T2O accumulation was reached

within 40 minutes. Amount of total T2O and [14C]Mannitol in the samples was

determined as described previously using a dual-labeled counting protocol. The

contribution of extracellular T2O to the total amount of T2O in each sample was

calculated from the amount of [14C]Mannitol, which represented extracellular volume

(separate experiments showed that mannitol was restricted to the extracellular space).

Note that due to the highly volatile nature of T2O, all incubations in this study were

performed with complete coverage of the cell culture wells using a plastic paraffin film

(Parafilm).

Page 38: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

37

2.8 Statistics

Data were analyzed using Prism (GraphPad Software). Statistical tools such as

paired t-test or unpaired t-test were applied according to the nature of each data set.

Page 39: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

38

CHAPTER 3

SYMMETRY OF H + INTERACTION WITH hMATE1 1

3.1 Background

The designation ‘organic cations’ (OCs) refers to a large group of organic

compounds that carry net positive charge(s) at physiological pH. The majority of OCs

are xenobiotic i.e., of exogenous origin, and many are toxic. On a regular basis,

individuals consume potentially toxic OCs, typically as plant products and/or prescribed

medicines. Currently, approximately 40 percent of commonly prescribed drugs can be

classified as OCs (23), including metformin (anti-diabetic), cimetidine (anti-peptic ulcer),

and clonidine (anti-hypertensive and analgesic). In the human body, the renal proximal

tubule (RPT) is the major site for OC secretion (71). In RPT epithelial cells (RPTECs),

OC secretion is a two-step process, with OC uptake across the peritubular (basolateral)

membrane by a process of facilitated diffusion, and OC efflux at the luminal (apical)

membrane, mainly by an OC/H+ exchange process (24).

There is broad agreement that the basolateral entry step in OC secretion by the

human RPT is dominated by human organic cation transporter 2 (hOCT2, SLC22A2),

which has been well characterized both structurally and functionally (71). There is also

growing consensus that the exit step, also the rate-limiting step (71), for OC secretion

involves the combined activity of two members of the Multidrug And Toxin Extrusion

(MATE) family, MATE1 (SLC47A1) and MATE2-K (SLC47A2) (24, 42). Although in

1 Content of this chapter was accepted for publication (9) as ‘Dangprapai Y, Wright SH. Interaction of H+ with the extracellular and intracellular aspects of hMATE1. Am. J. Physiol. Renal Physiol, 2011 May 25, in press.’ (Appendix A) during preparation of this dissertation.

Page 40: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

39

their proposed physiological role MATEs mediate the efflux of OCs, the characterization

of MATE-mediated OC transport has, with few exceptions, relied on measurement of

uptake i.e., influx, with the tacit assumption that MATEs interact symmetrically with

their substrates, including H+ (31). Although this is a reasonable assumption with respect

to the catalytic and energetic mechanism of transport, i.e., mediated exchange of H+ for

OC (42, 58), it may well not be the case with respect to the kinetics and selectivity of

substrate interaction with these transporters. In that regard, it is significant that rat Oct2

(rOct2, slc22a2) has been shown to have markedly asymmetrical interactions with (at

least) selected OCs. For example, the affinity of tetrabutylammonium (TBuA) to

outward-facing rOct2 is four-fold higher than its affinity to inward-facing rOct2, whereas

the affinity of corticosterone to outward-facing rOct2 is twenty-fold lower than its

affinity to inward-facing rOct2 (64).

To study MATE-mediated OC transport in its physiologically relevant direction

i.e., hMATE1 as an OC efflux transporter, we quantified the MATE-mediated efflux of

the model OC, MPP. In this study, we show that kinetics of interaction between outward-

facing hMATE1 and H+ measured using an efflux protocol are not significantly different

from those obtained using conventional uptake experiments. Furthermore, for the first

time, we report kinetics of interaction between inward-facing hMATE1 and H+. These

results support the conclusion that, despite the likelihood of structurally distinct binding

regions exposed to the cytoplasmic vs. extracellular faces of hMATE1, the kinetics of H+

interaction with each face of the transporter are similar.

Page 41: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

40

3.2 Cis-inhibition by extracellular H + of hMATE1-mediated OC uptake

We first characterized hMATE1-mediated transport of the test substrate, MPP, in

CHO cells that stably expressed the transporter. Uptake of [3H]MPP was linear for

approximately ten minutes (Fig. 7A) and five minute uptakes were selected for use in

subsequent kinetic studies as it covered the initial rate of MPP uptake in CHO hMATE1

cells. Figure 7B shows that unlabeled MPP blocked the uptake of labeled MPP with a

profile that was adequately described by the Michaelis-Menten equation for the

competitive interaction of labeled and unlabeled substrate (30):

max

t

J [MPP*]J* D[MPP*]

K [MPP*] [MPP]= +

+ +

where J* is the rate of [3H]MPP transport from a concentration equal to [MPP*]; Jmax is

the maximum rate of mediated MPP transport; Kt is the MPP concentration that resulted

in half-maximal transport (Michaelis constant); [MPP] is the concentration of unlabeled

MPP in the uptake reaction, and D is a constant that describes the non-saturable (first-

order) component of total substrate uptake (over the range of substrate concentrations

tested) that presumably reflected the combined influence of diffusive flux, nonspecific

binding, and/or incomplete rinsing of the cell layer. In four separate experiments, the

Jmax was 2.1 ± 0.18 pmol min-1 cm-2 and the apparent Kt (at pH 8.5) was 4.0 ± 0.29 µM

(Table 2).

The interaction between outward-facing hMATE1 and H+ was initially

characterized using two experimental approaches that each involved measurement of

uptake. First, the IC50 for H+ inhibition of hMATE1-mediated MPP uptake was

Page 42: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

41

determined (Fig. 8). The decrease in MPP uptake resulting from an increase in

extracellular [H+] ([H+]o) was adequately described by the following relationship (11):

app

o

J [MPP*]J D[MPP*]

IC [H ]50

+= +

+

where J is the rate of [3H]MPP uptake; Japp is the product of the maximum rate of

[3H]MPP uptake (Jmax) and the ratio of the Ki of H+ and Kt for MPP transport; and IC50 is

the concentration of [H+]o that reduced mediated (i.e., blockable) [3H]MPP transport by

50%. The IC50 for H+-inhibited hMATE1-mediated MPP uptake was 12.4 ± 2.21 nM (pH

7.91, n = 5). Although this is the first kinetic assessment of the inhibitory effect of

extracellular H+ on hMATE1-mediated transport, several previous studies showed that

transport was reduced as extracellular pH was decreased below a value of 8.5 (42, 56),

(58). The most detailed of these (56) when replotted as the effect of [H+]o on MATE1-

mediated transport of [14C] tetraethylammonium (TEA, a prototypic OC) suggested an

IC50 of about 15 nM (pH ~7.8), similar to the value noted here.

It is also generally observed that hMATE1-mediated OC transport decreases as

pH values increase beyond 8.5 (42, 56) suggesting that H+ interaction with hMATE1 is

unlikely to be limited to a simple competitive interaction and probably includes indirect

effects of reduced [H+]o on the structure of hMATE1 and/or other proteins. Therefore,

we decided to use another approach to determine an ‘apparent Ki’ for hydrogen ion (KHi-

app), namely, the influence of extracellular [H+] on the kinetics of MPP transport.

Kinetics of hMATE1-mediated [3H]MPP uptake at pH 7.5 ([H+] = 31.6 nM) and pH 8.5

([H+] = 3.2 nM) are presented in Table 2. The increase in [H+]o resulted in a 3-fold

Page 43: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

42

increase in the apparent Kt for MPP transport. This effect was anticipated given the

previous observation that extracellular H+ acts as a competitive inhibitor of OC/H+

exchange in isolated renal brush border membrane vesicles (BBMV) (69). However, in

our current experiments on [3H]MPP uptake in intact cells, it was also evident that the

increase in extracellular [H+] resulted in a modest (1.8-fold) but significant (P < 0.01)

increase in the Jmax for MPP transport, which is not consistent with a purely competitive

interaction of H+ with MPP at the external face of hMATE1. This result was, in fact,

seen in all four experiments that measured the kinetics of MPP transport at two different

pHs (Table 2). Nevertheless, although these data suggest that extracellular hydrogen ion

interacts with hMATE1 at several sites, resulting in a ‘mixed-type’ inhibitory profile, we

suggest that the predominant influence of H+ on apparent Kt reflected competition

between H+ and MPP for a common site or (more likely) a set of mutually exclusive sites

within a common binding surface.

The simplifying assumption that H+ acts predominantly as a competitive inhibitor

of hMATE1-mediated MPP transport permitted calculation of KHi-app using the

relationships (11):

max

t app

J [MPP]J

K [MPP]−

=+

, where ot app t H

i app

[H ]K K

K1 +

+

−−

= +

For these relationships, J and Jmax are as previously described; Kt is the Michaelis

constant for hMATE1-mediated MPP transport; and Kt-app is the apparent Kt for MPP

transport (reflecting the competitive influence of [H+]o). From the experiments shown in

Table 2, the KHi-app for the competitive interaction of H+ with extracellular aspect of

Page 44: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

43

hMATE1 was 13.5 ± 0.84 nM (pH 7.87) which was not different from the apparent IC50

for H+ inhibition of hMATE1-mediated MPP transport, noted above (P = 0.73).

3.3 Characterization of hMATE1-mediated OC efflux

The intent of this study was development of a quantitative view of the kinetics of

hMATE1-mediated transport when operating in its ‘physiological’ direction, i.e.,

exporting OCs. To this end, we used two similar approaches to quantify hMATE1-

mediated efflux from cells stably expressing this protein. The first involved

measurement of the amount of substrate retained in cells over time after an initiation of

efflux. Figure 9A shows the 10-minute time course of [3H]MPP efflux from CHO

hMATE1 cells obtained using this method. As expected, there was a time-dependent

decrease in cell [3H]MPP and, as supported by observations described below, the loss of

[3H]MPP from the cells was effectively restricted to efflux via hMATE1. Because the

concentration of [3H]MPP in the cells at time zero was likely to be well below the Kt at

the cytoplasmic face of the transporter, the kinetics of efflux could have been expected to

be first-order2. This was not, however, the case, as shown by the non-linearity of the

semi-log presentation of these data (Fig. 9A). The curvilinearity of the plot suggested

that MPP efflux involved at least two compartments, arranged in series (e.g., an

2The volume of CHO cells, determined using tritiated water corrected for extracellular space with [14C]Mannitol, was approximately 0.3 µl/cm2 of CHO cell (confluent layer; data not shown), similar to previous reports on the volume of CHO cells (49). If accumulated [3H]MPP was distributed uniformly in this volume (which, as discussed in the text, is an oversimplification), the calculated ‘bulk’ concentration of [3H]MPP in CHO hMATE1 cells in this study was as high as ~0.4 µM at initiation of efflux. This level of accumulated MPP is about 35 times lower than the apparent Kt for interaction between MPP and external face of hMATE1 at pH 7.5 (12.3 ± 1.08 µM; n = 4). Although an apparent Kt for MPP interaction with cytoplasmic face of hMATE1 has not been characterized, we suggest that the [MPP]cell was sufficiently low to anticipate first-order behavior for hMATE1-mediated MPP efflux.

Page 45: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

44

endosomal compartment and the cytoplasm) with the extracellular compartment.

Furthermore, the evident curvilinearity necessitates that passage from the first

(‘endosomal’) into the second (cytoplasm), is rate-limiting. This is not an unexpected

result in light of evidence that organic cations can, in fact, be sequestered within

intracellular compartments (33), including uptake of TEA into endosomal vesicles

isolated from rat renal cortex (46).

To assess, at least qualitatively, if such sequestration could account for the non-

first order efflux profile shown in Figure 9A, we examined the time course of efflux of a

fluorescent substrate for OC transporters, NBD-TMA (λex = 458 nm, λem = 530 nm; 4).

NBD-TMA blocked hMATE1-mediated MPP transport in CHO cells with an IC50 of 14.8

µM (data not shown), and though NBD-TMA may well display a quantitatively distinct

distribution profile in CHO cells than the structurally distinct MPP, it provided a useful

probe for visualizing the complexity of substrate distribution in these cells. CHO

hMATE1 cells were incubated in WB pH 8.5 containing 0.5 mM NBD-TMA for 15

minutes in an incubator with 5% CO2 at 37oC. The NBD-TMA loaded cells were then

transferred to the stage of an epifluorescence microscope (Olympus IX71). Intracellular

accumulation of fluorescent signal was only observed in cells that expressed hMATE1

(data not shown). After brief rinsing, NBD-TMA efflux was initiated by adding buffers

without substrate. Figure 10 provides a sequence of images showing the time course of

NBD-TMA efflux from CHO hMATE1 cells under several conditions.

Before the efflux was initiated, i.e., at t = 0, fluorescence within CHO hMATE1

cells could be categorized into two distinct patterns: a comparatively diffuse,

Page 46: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

45

homogeneous signal and a punctate signal. The diffuse signal was observed throughout

the cells and probably represented free NBD-TMA in the cytoplasm. In contrast, the

punctate signal was scattered, more intense, and probably represented NBD-TMA

sequestered within cytoplasmic organelles. Our finding was similar to a result from rat

choroid plexus cells in primary culture showing that distribution of accumulated

quinacrine (another fluorescent OC) includes both a diffuse and a punctate fluorescent

signal (33). Furthermore, as noted earlier, endosomes isolated from the rat renal cortex

accumulate TEA via TEA/H+ exchange (68). Based on these results, we suggest that the

punctate distribution of NBD-TMA in CHO cells most likely represented a similar

sequestration in cytoplasmic endosomes.

But is efflux of sequestered OC from this intracellular compartment slow

compared to efflux from the cytoplasm across the plasma membrane? When efflux of

NBD-TMA was performed at extracellular pH 7.4, the diffuse signal rapidly diminished

and almost disappeared after 10 minutes, whereas the punctate signal was still largely

retained (Fig. 10, panel a). In contrast, when 0.1 mM TPeA, a non-transported inhibitor

of OC/H+ exchange in renal BBMV (67), was present in the extracellular medium during

the efflux period, both patterns of NBD-TMA signal were effectively retained for at least

10 minutes (Fig. 10, panel b). A brief exposure to 0.01% saponin can selectively

permeabilize cholesterol-rich membranes, such as the plasma membrane (52), and when

NBD-TMA loaded cells were exposed to saponin (co-exposed with 0.1 mM TPeA), the

diffuse signal cleared as early as 15 seconds while the punctate fluorescent signal still

remained after 10 minutes of efflux (Fig. 10, panel c).

Page 47: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

46

These results support the contention that at the start of the efflux period, [3H]MPP

was distributed in at least two compartments: a ‘fast’ cytoplasmic compartment that

should readily access hMATE1 at the plasma membrane; and a ‘slow’ compartment

consisting of [3H]MPP sequestered in various cytoplasmic organelles. The solid line fit

to the data presented in Figure 3A assumed a two-phase model for the exponential loss of

[3H]MPP from CHO hMATE1 cells: a large compartment (77% of accumulated

[3H]MPP]) that turned over with a half-time of 1.6 minutes; and a smaller compartment

(23% of accumulated [3H]MPP) that turned over with a half-time of over ten minutes.

For this study, we focused on the early time course of [3H]MPP efflux that we suggest

was dominated by the fast, cytoplasmic compartment. We found that [3H]MPP in the

slow, sequestered compartment could be treated as a constant, permitting the data to be

analyzed using a one-compartment model that decayed to a plateau, using the

relationship: [3H]MPPt = ([3H]MPPt0 – C) e(-kt) + C, where [3H]MPPt is total labeled

MPP in the cells at any time t after initiation of efflux (t0); C is sequestered [3H]MPP; and

k is the first-order rate constant for hMATE1-mediated [3H]MPP efflux from the

cytoplasmic compartment. Correcting total [3H]MPP in the cells for the sequestered

compartment resulted in an efflux profile that was described by the relationship:

[3H]MPPt = ([3H]MPPt0) e(-kt)

The adequacy of this model to describe the efflux of [3H]MPP from CHO hMATE1 cells

is evident in the linearity of the semi log plot of these data presented in Figure 9B, and

the similarity in the rate constant of efflux (k) calculated after subtraction of sequestered

[3H]MPP (0.37 min-1) to the rate constant of efflux from the fast compartment of the two

Page 48: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

47

compartment model (0.43 min-1; Fig. 9A). For the rest of this study, each efflux data set

was analyzed by a single-phase decay equation after subtraction of the estimated

sequestered pool of [3H]MPP, as shown above.

The analysis of [3H]MPP efflux from pre-loaded CHO hMATE1 cells also

permitted a direct test of the hypothesis that an inwardly-directed H+ gradient should

increase the rate of hMATE1-mediated MPP efflux. These experiments used the second

of the two efflux protocols described in the methods, the flow chamber that permitted

continuous monitoring of [3H]MPP that left the cells into the passing stream of buffer.

Within the limit of resolution of the method, the efflux of MPP from the cytoplasmic

compartment of CHO hMATE1 cells (i.e., total efflux corrected for the ‘slow’

compartment) was adequately described by first-order kinetics (Fig. 11). In these

experiments, the half-life (t0.5) of hMATE1-mediated MPP efflux in buffer of the

extracellular pH 8.5 was 9.8 ± 1.43 min (n = 4), and decreasing the external pH to 7.4

(i.e., increasing [H+]o from 3.2 to 39.8 nM) resulted in a 7-fold decrease (P = 0.0004) in

the t0.5, down to 1.4 ± 0.08 min (n = 5). These results confirmed previous observations

(57) that inwardly-directed H+ gradients trans-stimulate MATE1-mediated OC efflux

and, of particular importance to the present context, provided a validation for our

approach to measuring hMATE1-mediated OC efflux.

Quantification of MPP efflux also provided an alternative means for assessing the

kinetics of H+ interaction with the external face of hMATE1. Figure 12A shows the

effect of systematically increasing extracellular [H+] on the rate of MPP efflux. As

expected, hMATE1-mediated MPP efflux was stimulated by increasing the concentration

Page 49: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

48

of extracellular H+, i.e., the exchangeable substrate. Furthermore, the rate constant for

MPP efflux supported by each extracellular [H+], when plotted as a function of [H+] (Fig.

12B), was adequately described by the Michaelis-Menten equation; the half-maximal

stimulation of MPP efflux occurred at an extracellular pH of 7.83 ([H+] = 14.7 ± 3.45 nM;

n = 3), and this result was not significantly different from either the IC50 (P = 0.79) or the

K i (P = 0.63) for extracellular H+ inhibition of hMATE1-mediated MPP uptake. Note

that hMATE1-mediated MPP efflux was also performed at pHo 6.5 and 6.0, but the rate

constants for efflux at these values of pHo (0.37 ± 0.013 and 0.36 ± 0.017 minute-1,

respectively) did not differ from that measured at pHo 7.0 (0.37 ± 0.02 minute-1) and, so

(for clarity) were not shown in Figure 12A.

3.4 Interaction of intracellular H + with the cytoplasmic face of hMATE1

To examine kinetics of interaction between H+ and the inward-facing aspect of

hMATE1, MPP efflux was determined as a function of intracellular H+ concentration.

From multiple measurements at room temperature in WB pH 7.4, CHO hMATE1 cells

demonstrated a baseline pHi of 7.57 ± 0.03 ([H+] i = 27.1 ± 1.79 nM; n = 21). This

baseline value varied by less than 0.2 pH units during a 15-minute exposure to WB of

different pH. Figure 13A shows a representative result of the change in pHi resulting

from an acute shift from an extracellular pH of 7.4 to one of 8.5; baseline pHi increased

by only 0.1 pH units (from pH 7.53 to pH 7.63). Manipulation of pHi was achieved

through acute exposure of cells to buffers containing increasing concentrations of NH4Cl.

Figure 13B shows the effect on the pHi of CHO hMATE1 cells of an acute exposure to

20 mM NH4Cl. Cytoplasmic pH shifted within 8 seconds from a steady-state value of 7.7

Page 50: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

49

to 8.3, which was maintained up to 3 minutes. Acute exposure of these cells to 2.5, 5, 10,

or 20 mM NH4Cl effectively established cytoplasmic pH values of pH 7.8 ([H+] i = 15.8 ±

0.44 nM; n = 3), pH 7.9 (12.5 ± 1.42 nM; n = 6), pH 8.1 (8.7 ± 0.76 nM; n = 3), or pH 8.2

(5.8 ± 0.35 nM, n = 4), respectively.

We also considered it necessary to determine if extracellular NH4+ had a direct

effect on the transport process. The presence of 20 mM NH4Cl in an uptake buffer (pH

8.5) decreased the 5-minute uptake of [3H]MPP by about 70% (Fig. 14). An inhibition of

uptake was expected because of the associated decrease in exchangeable H+ in the

cytoplasm noted above. However, it could also have included a direct inhibitory effect of

NH4+ on the extracellular face of hMATE1. We reasoned that, if the observed decrease

in MPP uptake was due simply to a decrease in intracellular [H+], then providing an

alternative exchangeable substrate at the cytoplasmic face of hMATE1 should rescue the

inhibitory effect of extracellular NH4+ on MPP uptake. To test this hypothesis, CHO

hMATE1 cells were pre-loaded with a high concentration of non-labeled MPP (5 mM

MPP for 10 minutes) prior to measuring [3H]MPP uptake in WB containing 20 mM

NH4Cl. Results in Figure 8 demonstrated that [3H]MPP uptake inhibited by extracellular

NH4Cl was quantitatively rescued in the cells pre-loaded with non-labeled MPP,

suggesting that extracellular NH4+ did not directly inhibit hMATE1-mediated MPP

uptake and that observed effects of NH4Cl on transport reflected changes in the

cytoplasmic [H+].

The rate of MPP efflux should increase with decreasing intracellular H+

concentration because of the associated decrease in competition between the two

Page 51: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

50

substrates at the cytoplasmic face of hMATE1. To test this, cells were pre-loaded with

[3H]MPP and efflux was initiated at extracellular pH 8.5 with no NH4Cl in the

extracellular medium. After one minute, buffer was switched to WB pH 7.4 (to stimulate

efflux; see Fig. 11) containing different concentrations of NH4Cl to manipulate

intracellular pH. Efflux was then continuously monitored for another 2 minutes. As

shown in Figure 15, decreasing the pH from 8.5 to 7.4 (i.e., increasing the concentration

of extracellular H+) produced the anticipated increase (approximately 7 fold) in rate of

[3H]MPP efflux. In this condition, with no extracellular NH4Cl, the pHi of CHO cells

was 7.57 ([H+] i = 27.1 nM). As expected, alkalinization of the cytoplasm by addition of

10 mM NH4Cl (pHi = 8.1, [H+] i = 8.7 nM) or 20 mM NH4Cl (pHi = 8.2, [H+] i = 5.8 nM)

did progressively increase the rate of [3H]MPP efflux.

To determine the kinetics of interaction between inward-facing hMATE1 and

intracellular H+, hMATE1-mediated MPP efflux was performed at an extracellular pH of

7.4 with no NH4Cl, and with 2.5, 5, 10, or 20 mM NH4Cl. As shown in Figure 16, the

efflux rate constant (k) increased in a concentration-dependent manner as the cells were

exposed to increasing concentrations of NH4Cl, i.e., as intracellular [H+] was

systematically decreased from 27.1 nM (pH 7.57) to 5.8 nM (pH 8.2), see inset to Fig. 16.

The IC50 for cytoplasmic H+ on hMATE1-mediated MPP efflux was at pH 7.94 ([H+] i =

11.5 nM). This value did not differ substantially from the range of values (12 nM to 15

nM) for the apparent Ki/IC50 of H+ interaction with the extracellular face of hMATE1

described earlier. Acknowledging that the pH range upon which this estimate of the

cytoplasmic IC50 was based was, of necessity, narrower than that used to study the

Page 52: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

51

interaction of H+ with the extracellular aspect of hMATE1, we suggest that these data

support the conclusion that, within the limits of resolution of currently available methods,

the intra- and extracellular faces of hMATE1 display kinetically symmetrical interactions

with H+.

3.5 Interaction between H+ and hMATE1: relevance to renal OC secretion

It is relevant to place the kinetic profile of H+ interaction with hMATE1 into the

context of OC secretion in intact RPT. In the ‘early’ proximal tubule, pH of the

glomerular filtrate is moderately alkaline, ~7.1 (15), reflecting a plasma pH of ~7.4. The

current results suggest that, at this pH, the extracellular face of hMATE1 is ~ 85%

occupied by H+ in the filtrate. These protons not only facilitate an exchange for

cytoplasmic OCs, but would also limit interaction between luminal OCs and outward-

facing hMATE1, i.e., luminal H+ should reduce ‘re-uptake’ of secreted OCs via

hMATE1. Furthermore, as a result of H+ secretion, and bicarbonate and water

reabsorption, along the length of the proximal tubule, the concentration of luminal H+

increases. In the late proximal tubule, pH of the filtrate can be as acidic as pH 6.8 to pH

6.7 (2), representing a concentration of H+ that should occupy at least 92% of available

hMATE1 transporters. This increase in luminal H+ would continuously reduce a ‘re-

uptake’ of luminal OCs, the concentration of which can also be expected to increase

along the length of the RPT (due to the combined influence of MATE secretory activity

and water reabsorption).

The interaction of H+ with hMATE1 also underscores its role in providing the

‘driving force’ for OC secretion in RPT. As a secondary active H+ exchanger, MATE1

Page 53: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

52

can mediate export of OCs against their electrochemical gradient driven by the coupled

influx of H+ moving down its electrochemical gradient. The active OC transport via the

OC/H+ exchanger is frequently described as being driven by an inward H+ gradient and,

indeed, is typically assessed in this way in studies using isolated renal BBMV – including

our own studies (68, 69). It is, however, evident from the previous discussion that this

can give a false impression of the energetic basis of OC export mediated by MATEs in

RPTEC. In the early RPT, which appears to be the primary site of OC secretion (51), the

inwardly-directed chemical gradient for H+ is very modest, about 1.25 fold, reflecting a

luminal pH of about 7.1 vs. a cytoplasmic pH of about 7.2 (75). Nevertheless, the tubule

is still poised to support a robust active transepithelial OC secretion because uptake of

OC from the blood into the RPTEC across the basolateral membrane is mediated by the

electrogenic activity of OCT2. Consequently, the inside negative membrane potential of

these cells, -60 to -70 mV, can be expected to support concentrative (yet passive)

accumulation of OCs to levels 10-15 times that in the blood. Importantly, the luminal

OC export step, mediated by MATE1 (and/or MATE2-K) involves obligatory,

electroneutral exchange (71). Thus, even in the absence of an inwardly-directed H+

chemical gradient, exchange of a proton in the filtrate for OC in the cytoplasm can permit

development of a luminal OC concentration that approaches the elevated level (compared

to the blood) of OC in the cytoplasm, with the result being active transepithelial

secretion. The MATE1-mediated step is still the ‘active’ element in this process because,

even in the absence of a chemical gradient for H+, protons are still far from

electrochemical equilibrium and kept that way through the activity of Na+/H+ exchange.

Page 54: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

53

The increasing acidity of the tubular filtrate along the length of the RPT further enhances

the energetic effectiveness of MATE-mediated OC secretion into the tubular filtrate.

It is also relevant to discuss the potential impact on renal OC secretion of the

comparatively high affinity of the cytoplasmic face of hMATE1 for H+. With an

apparent Ki of 11.5 nM (pH 7.94), the cytoplasmic pH in RPTEC of pH 7.2 ([H+] = 63.1

nM) (75) suggests that the cytoplasmic face of MATE1 should be ~85% occupied by

hydrogen ion. Based on selectivity studies in our lab and others (42, 58), the affinity for

OCs of the outward-facing aspect of hMATE1 is typically 10 to 1000-times lower than

that for H+, and at least in general terms this is likely to be the case for the inwardly-

directed face of MATE1, as well. This would seem to ‘set the stage’ for rather brisk

competition between accumulated OCs and near-saturating levels of cytoplasmic H+. But

that scenario assumes that the ‘bulk’ pH of the cytoplasm (which is what the BCECF

measurements represent) is an accurate indicator of the hydrogen ion concentration

within the unstirred ‘microdomain’ at the cytoplasmic face of the cell membrane. While

this is arguably the case in the current experiments with CHO cells, in which cytoplasmic

pH values were 7.5 and higher [well below the ‘set-point’ for NHEs (43)], the same may

not be the case in salt transporting epithelia like the renal proximal tubule. Indeed, it is

worth noting that there is experimental evidence that the activity of Na+/H+ exchange

creates a markedly acidic microenvironment at the extracellular face of the luminal

membrane of enterocytes (2). We suggest, therefore, that the activity of the major

Na+/H+ exchangers (NHEs) in the luminal membrane of RPTEC i.e., NHE3 and NHE8,

could create local alkaline conditions near the cytoplasmic face of the membrane, thereby

Page 55: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

54

facilitating the effective interaction of accumulated OCs with the inward face of MATE1.

Interestingly, results from a mathematical model of the renal proximal tubule brush

border membrane indicated that Na+/H+ exchange activity is (in principle) capable of

establishing a gradient of luminal H+ along the length of each microvillus, with the

cytoplasmic tip of the microvillus as alkaline as pH 8.0 (27). Another consequence of the

differential impact of NHE activity, i.e., local acidification of the extracellular face of the

membrane vs. local alkalinization of the cytoplasmic face of the membrane, would be an

effective increase in the inwardly-directed electrochemical gradient for H+ that would

enhance the secretory ‘power’ of MATE1-mediated OC export. Further study regarding

the expression profile of hMATE1 at the luminal membrane of the hRPTEC and the

influence of NHEs in hMATE1-mediated OC efflux are necessary for a better

understanding of this physiologically, pharmacologically, and toxicologically important

process.

Although our current results suggest that hMATE1 is symmetrical with respect to

its kinetic interactions with intra- and extracellular H+, it is premature to assume that

organic substrates will display a similar symmetry. Given the intrinsic structural

asymmetry of transport proteins, in conjunction with the ‘alternating access’ model of

transporter function, it is unlikely that binding surfaces of the ‘translocation pathway’

will be identical when accessed from the cytoplasmic vs. extracellular faces of the

membrane. Whereas protons titrate single carboxyl residues, the transient, weak

interactions of an organic substrate reflect interactions with multiple residues (65) and

even modest differences in the 3D structure of binding surfaces are likely to influence

Page 56: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

55

binding kinetics. As noted previously, the affinities of the inner vs. outer faces of rOct2

for corticosterone and TBuA differ markedly. Importantly, the differences are in the

opposite direction; whereas the inward face has a higher (20x) affinity for corticosterone,

the outward face has a higher affinity (4x) for TBuA (64). We suggest that a similar

degree of kinetic asymmetry can be expected for other multispecific transporters,

including MATE1. In this regard, it is also appropriate to note that the kinetic profiles

reported here reflect the interaction of a single substrate with MATE1, i.e., MPP.

Although MPP is a widely used to characterize organic cation transporters, both OCTs

(10) and MATEs (42), structurally distinct substrates for the multiselective MATE

transporters could display a different kinetic interaction with H+.

In summary, the interaction between H+, both intracellular and extracellular, and

the MATE1-mediated transport of MPP was examined, and the results suggested that the

kinetics of interaction between inward-facing hMATE1 and intracellular H+ are not

significantly different from the kinetics of the interaction between outward-facing

hMATE1 and extracellular H+. Nevertheless, despite this apparent symmetry of H+

interaction with hMATE1, it can be anticipated that at least some OCs will display

asymmetrical interactions with intra- vs. extracellular faces of hMATE1. The approach

for assessing the kinetics of OC efflux from intact cultured cells introduced in the present

study offers a potential means to address this issue.

Page 57: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

56

CHAPTER 4

AN APPLICATION OF THE EFFLUX PROTOCOL TO THE STUDY OF

hMATE1-MEDIATED OC INFLUX

4.1 Background

Since the successful cloning of mammalian MATEs in 2005, a number of OCs,

including prescribed drugs such as metformin (56), have been demonstrated to be

substrates for hMATE1. The typical approach to determine if a test compound is a

substrate for a defined transport protein, including MATEs, is direct measurement of

uptake of that compound into cells that heterologously express the protein, such as CHO

hMATE1 cells. An advantage of such uptake experiments is that they are comparatively

simple to do and straightforward to interpret. However, a major limitation of this

approach is the very limited access to radiolabeled compounds; few are commercially

available and they are very expensive to have synthesized. Measurement of unlabeled

compounds using mass spectrophotometry (MS) is an alternative approach, but also

expensive and time consuming. Consequently, the interaction of most compounds with

MATEs has been limited to measurement of their inhibitory effect on MATE-mediated

transport of a radiolabeled prototypic OC such as TEA, or MPP. For example, an

inhibition of [3H]MPP uptake into CHO hMATE1 cells by drug Y is generally viewed as

being sufficient to infer that drug Y is (probably) a substrate for hMATE1. A major

disadvantage of the inhibitory approach in uptake experiments is that both substrates and

non-transported inhibitors for hMATE1 can inhibit uptake of a radiolabeled OC into

CHO hMATE1 cells. An example of a non-transported inhibitor for apical OC/H+

Page 58: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

57

exchange in renal BBMV is tetrapentylammonium (TPeA) (70), which was also shown in

my studies (Chapter 3) to block hMATE1 transport activity (Fig. 10).

hMATE1 is an obligatory exchanger, and the trans-stimulation effect of

hMATE1-mediated efflux was clearly demonstrated in Chapter 3. Thus, this efflux

protocol, when compared to using radiolabeled compounds or MS, has the potential to be

a more practical (simple, straightforward, and economical) approach to distinguish

substrates from non-transported inhibitors for MATEs-mediated transport. For example,

at low extracellular [H+], drug Y will not trans-stimulate [3H]MPP efflux from CHO

hMATE1 cells if drug Y is a non-transported inhibitor. The experiments in this chapter

provide a ‘proof-of-concept’ for the hypothesis that known substrates for hMATE1, MPP

and triethylmethylammonium (TEMA), will trans-stimulate hMATE1-mediated

[3H]MPP efflux in a manner consistent with their directly determined characteristics of

hMATE1-mediated uptake.

To test the hypothesis, [3H]MPP efflux from CHO hMATE1 cells was measured

using the flow chamber protocol, as described in Chapter 3. The main difference was

that, in this chapter, the focus was on the effect of extracellular MPP or TEMA, not H+,

on hMATE1-mediated [3H]MPP efflux. As shown below, both MPP and TEMA trans-

stimulated [3H]MPP efflux in a dose-dependent manner, and the trans-effect reached a

maximal limit at saturating concentrations of these substrates.

4.2 Trans-stimulation of hMATE1-mediated OC efflux by MPP

CHO hMATE1 cells grown to confluence in a 35-mm cell culture dish were pre-

loaded with [3H]MPP using the protocol described in Chapter 3. A dish insert was placed

Page 59: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

58

inside the cell culture dish to create a closed chamber containing the cells. To examine

kinetics of interaction between outward-facing hMATE1 and extracellular MPP,

[3H]MPP efflux from CHO hMATE1 cells was initiated by introducing into the cell

chamber a continuous flow of efflux buffer (pH 8.5) containing various concentrations of

MPP.

Figure 17 shows that extracellular MPP trans-stimulated hMATE1-mediated

[3H]MPP efflux in a dose-dependent manner, and this stimulatory effect was not different

when extracellular [MPP] was increased from 100 µM to 200 µM. Based on the Kt value

of 5 µM for MPP with outward-facing hMATE1 (Fig. 7B), extracellular [MPP] of 100

µM should occupy ~ 95% of available, outwardly-directed hMATE1, and increasing

[MPP] to 200 µM would increase this to ~98%. In other words, MPP at 100 µM

concentration effectively saturated hMATE1 and increasing [MPP] to 200 µM would not

be expected to, and did not, provide a further stimulatory effect. Interestingly, the rate of

MPP efflux achieved by a saturating concentration of extracellular [MPP] was less than

that associated with a near saturating level of H+ (Fig. 17, pHo = 7.4). This implies that

the turnover number for hMATE1 occupied by H+ is greater than when occupied by

MPP.

To calculate the K50 for the stimulatory effect of MPP on hMATE1-mediated

[3H]MPP efflux, efflux rate constants (k) for [3H]MPP efflux from CHO hMATE1 cells

at each extracellular [MPP] were plotted as a function of extracellular [MPP] (Fig. 17,

inset). Analysis with the Michaelis-Menten equation revealed a K50 value for

extracellular MPP of 1.7 µM. This K50 value was similar to the Kt (4.00 ± 0.29 µM ) for

Page 60: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

59

interaction between MPP and the external surface of hMATE1 determined from direct

measurement of uptake (Table 2). This observation emphasizes the potential of efflux as

a means for examining kinetics of OC interaction with the external surface of hMATE1.

4.3 Trans-stimulation of hMATE1-mediated OC efflux by TEMA

Triethylmethylammonium (TEMA) is a quaternary ammonium OC. The

molecular structure of TEMA is similar to that of TEA, the only difference being that an

ethyl group is replaced by a methyl group. Because it is comparatively easy to synthesize

3H-labeled TEMA, it has proven to be a useful test compound to characterize OC

transporters (Wright SH, unpublished observations). Furthermore, because the structure

of TEMA is very different from that of the planar, heterocyclic MPP, it provides a

structurally distinct test substrate for studying multispecific OC transporters. Not

surprisingly, the kinetics of TEMA (IC50 = 51.6 ± 4.57 µM ) are similar to those for TEA

(IC50 = 40.6 ± 4.57 µM) when presented as an inhibitor for hMATE1-mediated [3H]MPP

uptake (Astorga B., unpublished results).

The influence of extracellular TEMA on [3H]MPP efflux from CHO hMATE1

cells was studied using the chamber protocol as described earlier. Although I only

performed a single experiment, I consider the result worth reporting for two reasons.

First, as expected, TEMA trans-stimulated hMATE1-mediated [3H]MPP efflux in a

concentration-dependent manner, thereby extending the ‘proof-of-concept’ evident in the

more extensive studies with MPP reported above. Second, the maximum rate of TEMA-

stimulated MPP efflux was comparable to that supported by a saturating concentration of

MPP, suggesting that the turnover number of a TEMA-loaded transporters was (at least)

Page 61: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

60

comparable to that of an MPP-loaded transporter. To provide kinetics of interaction

between TEMA and outward-facing hMATE1, more experiments are needed.

Nevertheless, I suggest that these limited observations support the view that measurement

of hMATE1-mediated efflux can extend information gained by determining the inhibitory

effect of a test compound on hMATE1 transport activity by assessing the ability of that

compound to stimulate hMATE1-mediated efflux.

Page 62: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

61

CHAPTER 5

FUTURE DIRECTION

Apical OC/H+ exchange is the active and a rate-limiting step for renal secretion of

a structurally diverse array of xenobiotic and toxic OCs. Based on currently available

information, the major candidate for the molecular identity of apical OC/H+ exchange

activity in RPTECs is hMATE1 (along with its functionally similar congener, hMATE2-

K). Indeed, evidence in mice lacking Mate1 suggests that Mate1 is necessary for renal

secretion of the cationic drugs, metformin (anti-diabetic) and cephalexin (antibiotic).

Although study on MATEs has steadily increased since 2005 after the successful cloning

of hMATE1, data on kinetics of MATE-mediated transport are limited. Most reports

have focused on qualitative aspects of OC transport by MATEs, such as whether or not a

particular class of drug is a substrate for MATEs. To examine the molecular mechanism

of MATE-mediated transport, not only qualitative data on the process, but also

quantitative (kinetic) information is required.

In a native renal proximal tubule, hMATE1 functions as an OC efflux transporter

at the apical membrane. Significantly, even the limited information available for the

kinetic profiles of MATEs has been derived from uptake experiments, rather than from

more physiologically relevant efflux experiments. In a heterologous expression system,

data from an uptake study reflect an interaction between a substrate and the extracellular

surface of a transporter. On the other hand, data from an efflux experiment represent an

interaction between a substrate and cytoplasmic face of a transporter protein. The

emphasis on measurement of ‘uptake’ reflects the fact that such experiments are easier to

Page 63: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

62

perform than are efflux studies. Although it is clear that characterization of hMATE1-

mediated transport in an efflux setting, compared to an uptake experiment, is a better

model to study hMATE1-mediated transport, prior to the current study, no kinetic profile

of hMATE1-mediated efflux has been reported.

In this study, for the first time, kinetics of interaction between H+ and the

cytoplasmic face of hMATE1 were examined and compared to H+ interaction at the

extracellular face. The results suggested a quantitatively symmetrical interaction of

hMATE1 with H+. However, it would be premature to assume that each face of

hMATE1 also interacts symmetrically with OCs, because of their structural complexity

compared to H+. Thus, I think it is critical that the kinetics of interaction between

cytoplasmic OC and hMATE1 be determined. Theoretically, the kinetics of OC efflux

(reflecting interaction with the intracellular face of hMATE1) can be calculated from a

time course of hMATE1-mediated OC efflux that begins with a near-saturating

intracellular OC concentration. The subsequent decline in the rate of efflux (as substrate

concentration falls) can be analyzed using the integrated Michaelis-Menten equation.

This approach was used successfully to determine the kinetics of fluorescein efflux from

pre-loaded renal proximal tubules (53). I have made preliminary attempts to conduct

such experiments with CHO hMATE1 cells. However, my attempt to saturate the

cytoplasmic face of hMATE1 with MPP has been challenging. Although these

observations could be interpreted as suggesting that the affinity of the cytoplasmic face of

hMATE1 for MPP is lower than that of the extracellular face, much more work needs to

be done to support such a conclusion. An optimized efflux protocol that assesses the first

Page 64: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

63

seconds of efflux more efficiently, or a protocol that uses OCs other than MPP, have the

potential to more fully explore this topic. Alternatively, a study focusing on the

cytoplasmic face of a transporter protein, including MATEs, could perhaps be performed

using isolated secretory vesicles, which expose the intracellular face of a heterologously

expressed membrane transporter. Yeast have been used to generate such preparations

(50). Another approach could involve purified transport protein in reconstituted in

artificial proteoliposomes (22).

The current work also provides the way for addressing another critical issue,

namely, the extent to which inhibitors of MATE-mediated activity are also transported

substrates. Novel labeled, e.g., radioactive or fluorescent, substrates are expensive, and

the efflux method can be developed to provide high-throughput screening of the extent to

which unlabeled substrates stimulate efflux of inexpensive test probes such as [3H]MPP.

Finally, the need to understand the molecular basis of selectivity underscores the need to

address more completely the structure of MATE proteins. The results of empirical

measures of selectivity for test substrates should be compared to developing models of

the structure of MATE proteins to provide predictive models of substrate/inhibitor

interaction, with both faces, of these critically important elements of the renal pathway

for drug secretion.

Page 65: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

64

Figure 1. Chemical structure of selected organic cations (OCs). This example

emphasizes that OCs are a group of structurally diverse chemicals. Thus, it is necessary

for organic cation transporters including MATEs to display a functional ‘multispecificity’

for OCs.

N+

CH3 CH3

CH3

CH3

N+ CH3

CH3

N

N

NHNH

N

CH3

S

NH

NH2

NH

NH

NH

NH2

Tetraethylammonium (TEA)

1-methyl-4-phenylpyridinium (MPP)

1-cyano-2-methyl-3-[2-[(5-methyl-1H-imidazol-4-yl)methylsulfanyl]ethyl]guanidine

(cimetidine)

1-(diaminomethylidene)guanidine (metformin)

Page 66: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

65

Figure 2. Molecular mechanism of renal organic cation secretion. A model for

organic cation secretion at the renal proximal tubule epithelium, as discussed in the text,

was modified from the original model proposed by Holohan and Ross (48) with

additional detail regarding molecular identity of transporter proteins from currently

available information on renal OC secretion. (OC+ = organic cation, NKA = sodium-

potassium ATPase, NHE = sodium-hydrogen exchanger)

Peritubular

capillary

Renal

proximal tubule

Page 67: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

66

Figure 3. Predicted membrane topology of hOCT2. Secondary (A) and tertiary (B)

structure of hOCT2 was proposed based on an atomic structure of a related bacterial

glycerol-3-phosphate transporter (GlpT). (This figure was modified from Figure 1 of

reference (44).

A B

Page 68: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

67

Figure 4. Predicted membrane topology of rbMATE1. A homology model of

rbMATE1 (Chang and Wright, unpublished observation), without the 13th transmembrane

domain, was constructed from the atomic structure of bacterial NorM (17).

Page 69: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

68

Figure 5. hMATE1-mediated MPP transport: uptake vs. efflux. A: hMATE1-

mediated MPP efflux from renal proximal tubular epithelial cells (RPTEC) is an

interaction between the cytoplasmic face of hMATE1 and MPP. B: hMATE1-mediated

MPP uptake in CHO hMATE1 cells is an interaction between extracellular face of

hMATE1 and MPP.

A OC efflux

RPTEC

B OC uptake

CHO cell

Page 70: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

69

Figure 6. The culture dish insert. A top-view diagram of the closed bath chamber

insert (RC-37FC) with perfusion ports for 35-mm cell culture dish was modified from a

picture shown in a product brochure of Warner Instruments.

Page 71: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

70

Figure 7. Time course and kinetics of [3H]MPP uptake in CHO hMATE1 cells. A:

[3H]MPP accumulated linearly in CHO hMATE1 cells up to approximately 10 minutes

(solid circles). [3H]MPP uptake was inhibited by co-incubating the cells with 1 mM

nonlabeled MPP (open circles). Buffer pH was 8.5. Each point is the mean (±SEM) of

uptake measured in two wells of a 24-well plate. B: Uptake (5 min; pH 8.5) of [3H]MPP

transport measured in the presence of increasing concentrations of unlabeled MPP (from

which the kinetics of MPP transport were determined). Each point is the mean (±SEM)

of uptakes measured in three wells of a 24-well plate.

0 5 10 15 20 25 300

30

60

90

1 µCi/ml [3H]MPP

1 µCi/ml [3H]MPPwith 1 mM MPP

CHO hMATE1 cells

A

Time (minutes)

[3H

]MP

P (

fmol

cm

-2)

0

5

10

15

20

1 10 100 1000

Jmax = 2.15 pmol min-1 cm-2

0

Kt = 4.89 µM

B

[MPP] (µM)

[3 H]M

PP

(fm

ol m

in-1

cm-2

)

Page 72: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

71

Figure 8. IC50 of extracellular H+ as an inhibitor of hMATE1-mediated MPP

uptake. Five-minute uptakes of 27 nM [3H]MPP into CHO hMATE1 cells were plotted

as a function of extracellular [H+]. Each point is the mean (±SEM) of uptakes determined

in five separate experiments (n = 5).

10 - 210 - 10

100

200

300

400

100 101 102 1031

IC50H+ = 12.4 ± 2.21 nM(pH 7.91)

0

[H+] (nM)

[3 H]M

PP

(%

of

pH 7

.5)

Page 73: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

72

Figure 9. Time course of MPP loss from CHO hMATE1 cells. A: Following a 20-

minute preload of 13.5 nM [3H]MPP, the amount of labeled MPP remaining in the cells

was measured as a function of time after efflux was initiated in an MPP-free buffer of pH

7.4 ([H+]o = 39.8 nM). Each point is the mean [3H]MPP content (±1/2 the range) of cells

in two wells of a 48-well plate in a single representative experiment. The semi-log plot

reveals that there were multiple compartments involved in [3H]MPP efflux from CHO

hMATE1 cells as discussed in the text. The solid line represents two-phase exponential

decline in cell [3H]MPP (kinetic parameters listed in the figure). B: Time course of the

first-order (single phase) decline in cell [3H]MPP, reflecting correction for sequestered

[3H]MPP (see text for discussion). The dotted lines in both graphs represent the 95%

confidence interval of the fitted values (solid line).

0 5 101

10

100

kfast = 0.37 ± 0.01 min-1

t0.5, fast = 1.88 min

B

Efflux duration (minutes)

Cel

l [3 H

]MP

P (

% o

f t

= 0

)

0 5 1010

100

kfast = 0.43 ± 0.12 min-1

t0.5, fast = 1.62 min

kslow = 0.06 ± 0.09 min-1

t0.5, slow = 10.7 min

A

Efflux duration (minutes)

Cel

l [3H

]MP

P (

% o

f t

= 0

)

Page 74: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

73

Figure 10. Time course of NBD-TMA efflux in CHO hMATE1 cells. CHO hMATE1

cells were pre-loaded with 0.5 mM NBD-TMA at 370C for 15 minutes. Following

aspiration of the medium the cells were transferred to a fluorescent microscope, rinsed

with WB pH 8.5 at which point an image was collected representing ‘t = 0’. Subsequent

images were collected at the indicated time points following introduction of test media.

For each condition, the cells were illuminated only during collection of the image. The

10-minute images were from different fields of cells (to avoid the influence of

photobleaching).

a. pH 7.4

b. pH 7.4 + 0.1 mM TPeA

c. pH 8.5 + 0.1 mM TPeA + 0.01% saponin

t = 0 1 min. 10 min.

t = 0

t = 0 1 min.

1 min.

10 min.

10 min.

0.5 min.

0.5 min.

0.25 min.

Page 75: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

74

Figure 11. Effect of extracellular H+ on hMATE1-mediated [3H]MPP efflux. Efflux

was performed at extracellular pH 8.5 ([H+]out = 3.2 nM) and at pH 7.4 ([H+]out = 39.8

nM) using the efflux chamber protocol (see text).

0.0 0.5 1.0 1.5 2.010

100

pH 8.5

pH 7.4

Efflux duration (minutes)

Cel

l [3H

]MP

P (

% o

f t =

0)

Page 76: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

75

Figure 12. Effect of increasing extracellular [H+] on the kinetics of hMATE1-

mediated [3H]MPP efflux. A: Time course of [3H]MPP efflux from the ‘fast’

cytoplasmic compartment (as defined in the text) was determined in buffers of decreasing

external pH. The value of the slow compartment used for each condition was determined

from the efflux profile at pHo 7.0. Each point is the mean of results obtained in three

separate experiments. B: The rate constants for [3H]MPP efflux from the fast

compartment (the slopes of the lines fit to the data in Fig. 12A) plotted as a function of

extracellular [H+] (line fit using Michaelis-Menten equation).

0 25 50 75 1000.00

0.15

0.30

0.45

B

K50 = 14.67 ± 3.45 nM(pH 7.83)

[H+] (nM)

Effl

ux

rate

con

stan

t (k

; m

in-1

)

0 1 2 3 4 510

100

A

pH 8.0

pH 7.7

pH 8.2

pH 7.0pH 7.4

pH 8.5

Efflux duration (minutes)

Cel

l [3H

]MP

P (

% o

f t =

0)

Page 77: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

76

Figure 13. Intracellular pH (pH i) in CHO hMATE1 cells. A: A representative

experiment showing the modest change in pHi associated with acute change of pHo; in

this case from pHo of 7.4 to pHo of 8.5 shifted pHi from 7.53 to 7.63. B: Upon acute

exposure to 20 mM NH4Cl the pHi of CHO hMATE1 cells increased rapidly (within 8

seconds) from its baseline pHi value (pHi of 7.57, i.e., [H+] i = 27.1 ± 1.79 nM; n = 21) to

a new value (pHi of 8.2, i.e., [H+] i = 5.8 ± 0.35 nM; n = 4) that was relatively stable for at

least three minutes. Data shown are from a single representative experiment. [Each point

of the fluorescence ratio in both A. and B. was an average value measured from twenty

eight CHO hMATE1 cells.]

0 3 6 9

1.1

1.2

1.3

pHo 7.4 pHo 8.5

pHi 7.53

pHi 7.63

A

Time (minutes)

Flu

ores

cenc

e ra

tio (

488

/460

nm

)

7.0

7.2

7.4

7.6

7.8

Intr

acel

lula

r p

H (

pH

i)

0 3 6 9 12

1.2

1.3

1.4

1.5

20 mMNH4Clcontrol control

pHi 7.7

pHi 8.3B

Time (minutes)

Flu

ores

cenc

e ra

tio (

488/

460

nm)

7.5

8.0

8.5

Intr

acel

lula

r p

H (

pHi)

Page 78: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

77

Figure 14. Effect of NH4Cl and MPP preload on hMATE1-mediated [3H]MPP

uptake. The time course of hMATE1-mediated uptake of [3H]MPP (13.5 nM) was

measured under the conditions indicated in the figure. In every case, pHo was 8.5. Each

point is the mean (±SEM) of uptake values (measured in triplicate) from three separate

experiments (n = 3).

0 1 2 3 4 50

40

80

120

160 no preload, no NH4Cl (control)5 mM MPP preload, no NH4Clno preload, 20 mM NH4Cl during uptake5 mM MPP preload and20 mM NH4Cl during uptake

Duration (minutes)

[3 H]M

PP

(%

of

upta

ke a

t 5

min

.)

Page 79: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

78

Figure 15. Effect of NH4Cl on hMATE1-mediated [3H]MPP efflux. Cells were

preloaded with [3H]MPP (36.8 nM) for 15 min. Efflux into a buffer of pH 8.5 was

measured for one minute, at which point the cells were exposed to a buffer of pH 7.4

containing the indicated concentrations of NH4Cl (to decrease [H+] i). Efflux into these

new solutions was then determined for two minutes. Single representative examples are

shown for each condition.

0 1 2 3

10

100

pH 8.5

[NH4Cl](mM)

20

pH 7.4

10

0

no NH4Cl

Efflux duration (minutes)

Cel

l [3 H

]MP

P (

% o

f am

ount

at

t =

0)

Page 80: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

79

Figure 16. Kinetics of interaction between inward-facing hMATE1 and

intracellular H +. The time course of hMATE1-mediated [3H]MPP efflux was measured

for each of the indicated values of intracellular pH. Each point is the mean of values

determined in 3-6 separate experiments. The inset shows the relationship between

cytoplasmic [H+] and the calculated rate constants (mean ±SEM) for hMATE1-mediated

[3H]MPP efflux.

0.0 0.5 1.0 1.5 2.0

10

100

7.67.8

7.9

8.2

8.1

pHi

Duration (minutes)

Cel

l [3H

]MP

P (

% o

f t =

0)

0 10 20 300.0

0.5

1.0

1.5

2.0

2.5

IC50H+ = 11.47 nM (pH 7.94)

Intracellular [H+] (nM)

Eff

lux

rate

con

stan

t (m

in-1

)

Page 81: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

80

Figure 17. Effect of increasing extracellular [MPP] on the kinetics of hMATE1-

mediated [3H]MPP efflux. Time course of [3H]MPP efflux from the ‘fast’ cytoplasmic

compartment (as defined in the text) was determined in buffers (pH 8.5) of increasing

extracellular concentration of unlabeled MPP. The value of the slow compartment used

for each condition was determined from the efflux profile at pHo 7.4. Each point is the

mean of results obtained in one or two separate experiments. The rate constants for

[3H]MPP efflux from the fast compartment plotted as a function of extracellular [MPP]

is shown in the inset. The line was created from the Michaelis-Menten equation.

0.0 0.5 1.0 1.5 2.0

32

40

50

63

79

100

00.1110

100200

0(pH 7.4)

[MPP](mM)

Efflux duration (minutes)

Cel

l [3 H

]MP

P (

% o

f t =

0)

0 50 100 150 2000.0

0.1

0.2

0.3

[MPP] (µµµµM)

K (

min

-1)

K50 = 1.7 µM

[MPP] (µM)

Page 82: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

81

Figure 18. Effect of extracellular TEMA on hMATE1-mediated [3H]MPP efflux.

Efflux was performed using chamber protocol at extracellular pH 8.5 ([H+]out = 3.2 nM)

with various concentration of TEMA, and at extracellular pH 7.4 ([H+]out = 39.8 nM) as a

positive control.

0.0 0.5 1.0 1.5 2.0

32

40

50

63

79

100

0

0(pH 7.4)

0.05

0.11

100

[TEMA](mM)

Efflux duration (minutes)

Cel

l [3 H

]MP

P (

% o

f t =

0)

10

Page 83: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

82

Antidiabetic – metformin

Antiarrhythmic – procainamide, quinidine

Antibiotic – levofloxacin

Antiviral – acyclovir, ganciclovir, tenofovir

Antiparasitic – quinine, pyrimethamine

Chemotherapeutic – platinum-containing agents such as cisplatin, oxaliplatin

Recreational – nicotine

Misc. – cimetidine (anti-peptic ulcer), clonidine (anti-hypertensive and analgesic)

(Endogenous – choline, dopamine)

Table 1. Selected organic cations of clinical or environmental significance.

Page 84: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

83

Table 2. Effect of extracellular pH on the kinetics of hMATE1-mediated MPP

uptake. Five-minute uptakes of [3H]MPP (~13 nM) were determined as a function of

increasing concentration of unlabeled MPP at two values for external pH (pHo), pH 7.5 ±

0.1 (n = 4) and pH 8.5 ± 0.02 (n = 4). Both Jmax and Kt of MPP uptake in CHO hMATE1

cells from these two pH values were significantly different (* p = 0.01).

pHo

7.5 ± 0.1

8.5 ± 0.02 4.0 ± 0.29

12.3 ± 1.08* 3.7 ± 0.36*

2.1 ± 0.18

Kt

(µM)

Jmax

(pmol min-1 cm-2)

Page 85: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

84

REFERENCES

1. Ames BN, Profet M, Gold LS. Nature’s chemicals and synthetic chemicals: comparative toxicology. Proc. Natl. Acad. Sci. U.S.A 87: 7782-7786, 1990.

2. Anderson CMH, Thwaites DT. Hijacking solute carriers for proton-coupled drug transport. Physiology (Bethesda) 25: 364-377, 2010.

3. Barendt WM, Wright SH . The human organic cation transporter (hOCT2) recognizes the degree of substrate ionization. J. Biol. Chem 277: 22491-22496, 2002.

4. Bednarczyk D, Mash EA, Aavula BR, Wright SH. NBD-TMA: a novel fluorescent substrate of the peritubular organic cation transporter of renal proximal tubules. Pflugers Arch 440: 184-192, 2000.

5. Bednarczyk D, Ekins S, Wikel JH, Wright SH. Influence of molecular structure on substrate binding to the human organic cation transporter, hOCT1. Mol. Pharmacol 63: 489-498, 2003.

6. Brown MH, Paulsen IT, Skurray RA. The multidrug efflux protein NorM is a prototype of a new family of transporters. Mol. Microbiol 31: 394-395, 1999.

7. Budiman T, Bamberg E, Koepsell H, Nagel G. Mechanism of electrogenic cation transport by the cloned organic cation transporter 2 from rat. J. Biol. Chem 275: 29413-29420, 2000.

8. Busch AE, Quester S, Ulzheimer JC, Waldegger S, Gorboulev V, Arndt P, Lang F, Koepsell H. Electrogenic properties and substrate specificity of the polyspecific rat cation transporter rOCT1. J. Biol. Chem 271: 32599-32604, 1996.

9. Dangprapai Y, Wright SH. Interaction of H+ with the extracellular and intracellular aspects of hMATE1. Am J Physiol Renal Physiol (May 25, 2011). doi: 10.1152/ajprenal.00075.2011.

10. Gorboulev V, Ulzheimer JC, Akhoundova A, Ulzheimer-Teuber I, Karbach U, Quester S, Baumann C, Lang F, Busch AE, Koepsell H. Cloning and characterization of two human polyspecific organic cation transporters. DNA Cell Biol 16: 871-881, 1997.

11. Groves CE, Evans KK, Dantzler WH, Wright SH. Peritubular organic cation transport in isolated rabbit proximal tubules. Am. J. Physiol 266: F450-458, 1994.

12. Gründemann D, Gorboulev V, Gambaryan S, Veyhl M, Koepsell H. Drug excretion mediated by a new prototype of polyspecific transporter. Nature 372: 549-552, 1994.

Page 86: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

85

13. Gründemann D, Liebich G, Kiefer N, Köster S, Schömig E. Selective substrates for non-neuronal monoamine transporters. Mol. Pharmacol 56: 1-10, 1999.

14. Gründemann D, Harlfinger S, Golz S, Geerts A, Lazar A, Berkels R, Jung N, Rubbert A, Schömig E. Discovery of the ergothioneine transporter. Proc. Natl. Acad. Sci. U.S.A 102: 5256-5261, 2005.

15. Hamm LL, Nakhoul N . Renal Acidification. In: Brenner and Rector’s The Kidney. Saunders, 2007, p. 248 - 267.

16. Hayer-Zillgen M, Brüss M, Bönisch H. Expression and pharmacological profile of the human organic cation transporters hOCT1, hOCT2 and hOCT3. Br. J. Pharmacol 136: 829-836, 2002.

17. He X, Szewczyk P, Karyakin A, Evin M, Hong W-X, Zhang Q, Chang G. Structure of a cation-bound multidrug and toxic compound extrusion transporter. Nature 467: 991-994, 2010.

18. Holohan PD, Ross CR. Mechanisms of organic cation transport in kidney plasma membrane vesicles: 1. Countertransport studies. J. Pharmacol. Exp. Ther 215: 191-197, 1980.

19. Holohan PD, Ross CR. Mechanisms of organic cation transport in kidney plasma membrane vesicles: 2. delta pH studies. J. Pharmacol. Exp. Ther 216: 294-298, 1981.

20. Jonker JW, Wagenaar E, Van Eijl S, Schinkel AH. Deficiency in the organic cation transporters 1 and 2 (Oct1/Oct2 [Slc22a1/Slc22a2]) in mice abolishes renal secretion of organic cations. Mol. Cell. Biol 23: 7902-7908, 2003.

21. Keller T, Elfeber M, Gorboulev V, Reiländer H, Koepsell H. Purification and functional reconstitution of the rat organic cation transporter OCT1. Biochemistry 44: 12253-12263, 2005.

22. Keller T, Schwarz D, Bernhard F, Dötsch V, Hunte C, Gorboulev V, Koepsell H. Cell free expression and functional reconstitution of eukaryotic drug transporters. Biochemistry 47: 4552-4564, 2008.

23. Kim MK, Shim C-K . The transport of organic cations in the small intestine: current knowledge and emerging concepts. Arch. Pharm. Res 29: 605-616, 2006.

24. Klaassen CD, Aleksunes LM. Xenobiotic, bile acid, and cholesterol transporters: function and regulation. Pharmacol. Rev 62: 1-96, 2010.

Page 87: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

86

25. Koehler MR, Wissinger B, Gorboulev V, Koepsell H, Schmid M. The two human organic cation transporter genes SLC22A1 and SLC22A2 are located on chromosome 6q26. Cytogenet. Cell Genet 79: 198-200, 1997.

26. Koepsell H, Lips K, Volk C. Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications. Pharm. Res 24: 1227-1251, 2007.

27. Krahn TA, Weinstein AM . Acid/base transport in a model of the proximal tubule brush border: impact of carbonic anhydrase. Am. J. Physiol 270: F344-355, 1996.

28. Kuroda T, Tsuchiya T. Multidrug efflux transporters in the MATE family. Biochim. Biophys. Acta 1794: 763-768, 2009.

29. Lickteig AJ, Cheng X, Augustine LM, Klaassen CD, Cherrington NJ. Tissue distribution, ontogeny and induction of the transporters Multidrug and toxin extrusion (MATE) 1 and MATE2 mRNA expression levels in mice. Life Sci 83: 59-64, 2008.

30. Malo C, Berteloot A. Analysis of kinetic data in transport studies: new insights from kinetic studies of Na+-D-glucose cotransport in human intestinal brush-border membrane vesicles using a fast sampling, rapid filtration apparatus. J. Membr. Biol 122: 127-141, 1991.

31. Matsushima S, Maeda K, Inoue K, Ohta K-ya, Yuasa H, Kondo T, Nakayama H, Horita S, Kusuhara H, Sugiyama Y. The inhibition of human multidrug and toxin extrusion 1 is involved in the drug-drug interaction caused by cimetidine. Drug Metab. Dispos 37: 555-559, 2009.

32. Meyer-Wentrup F, Karbach U, Gorboulev V, Arndt P, Koepsell H. Membrane localization of the electrogenic cation transporter rOCT1 in rat liver. Biochem. Biophys. Res. Commun 248: 673-678, 1998.

33. Miller DS, Villalobos AR, Pritchard JB . Organic cation transport in rat choroid plexus cells studied by fluorescence microscopy. Am. J. Physiol 276: C955-968, 1999.

34. Montrose-Rafizadeh C, Roch-Ramel F, Schäli C. Axial heterogeneity of organic cation transport along the rabbit renal proximal tubule: studies with brush-border membrane vesicles. Biochim. Biophys. Acta 904: 175-177, 1987.

35. Morita Y, Kataoka A, Shiota S, Mizushima T, Tsuchiya T. NorM of vibrio parahaemolyticus is an Na+-driven multidrug efflux pump. J. Bacteriol 182: 6694-6697, 2000.

Page 88: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

87

36. Morita Y, Kodama K, Shiota S, Mine T, Kataoka A, Mizushima T, Tsuchiya T. NorM, a putative multidrug efflux protein, of Vibrio parahaemolyticus and its homolog in Escherichia coli. Antimicrob. Agents Chemother 42: 1778-1782, 1998.

37. Moseley RH, Jarose SM, Permoad P. Organic cation transport by rat liver plasma membrane vesicles: studies with tetraethylammonium. Am. J. Physiol 263: G775-785, 1992.

38. Moseley RH, Morrissette J, Johnson TR. Transport of N1-methylnicotinamide by organic cation-proton exchange in rat liver membrane vesicles. Am. J. Physiol 259: G973-982, 1990.

39. Motohashi H, Sakurai Y, Saito H, Masuda S, Urakami Y, Goto M, Fukatsu A, Ogawa O, Inui K-ichi. Gene expression levels and immunolocalization of organic ion transporters in the human kidney. J. Am. Soc. Nephrol 13: 866-874, 2002.

40. Nies AT, Koepsell H, Damme K, Schwab M. Organic cation transporters (OCTs, MATEs), in vitro and in vivo evidence for the importance in drug therapy. Handb Exp Pharmacol 201: 105-167, 2011.

41. Okuda M, Saito H, Urakami Y, Takano M, Inui K . cDNA cloning and functional expression of a novel rat kidney organic cation transporter, OCT2. Biochem. Biophys. Res. Commun 224: 500-507, 1996.

42. Otsuka M, Matsumoto T, Morimoto R, Arioka S, Omote H, Moriyama Y . A human transporter protein that mediates the final excretion step for toxic organic cations. Proc. Natl. Acad. Sci. U.S.A 102: 17923-17928, 2005.

43. Ozkan P, Mutharasan R. A rapid method for measuring intracellular pH using BCECF-AM. Biochim. Biophys. Acta 1572: 143-148, 2002.

44. Pelis RM, Dangprapai Y, Wunz TM, Wright SH. Inorganic mercury interacts with cysteine residues (C451 and C474) of hOCT2 to reduce its transport activity. Am. J. Physiol. Renal Physiol 292: F1583-1591, 2007.

45. Pritchard JB, Miller DS . Mechanisms mediating renal secretion of organic anions and cations. Physiol. Rev 73: 765-796, 1993.

46. Pritchard JB, Sykes DB, Walden R, Miller DS. ATP-dependent transport of tetraethylammonium by endosomes isolated from rat renal cortex. Am. J. Physiol 266: F966-976, 1994.

47. Rennick BR, Moe GK. Absorption and renal excretion of the tetraethylammonium ion. J. Pharmacol. Exp. Ther 91: 210-217, 1947.

Page 89: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

88

48. Ross CR, Holohan PD. Transport of organic anions and cations in isolated renal plasma membranes. Annu. Rev. Pharmacol. Toxicol 23: 65-85, 1983.

49. Rotoli BM, Bussolati O, Dall’Asta V, Gazzola GC. Membrane potential and amino acid transport in a mutant Chinese hamster ovary cell line. J. Cell. Physiol 146: 417-424, 1991.

50. Ruetz S, Gros P. Functional expression of P-glycoproteins in secretory vesicles. J. Biol. Chem 269: 12277-12284, 1994.

51. Schäli C, Schild L, Overney J, Roch-Ramel F. Secretion of tetraethylammonium by proximal tubules of rabbit kidneys. Am. J. Physiol 245: F238-246, 1983.

52. Schulz I. Permeabilizing cells: some methods and applications for the study of intracellular processes. Meth. Enzymol 192: 280-300, 1990.

53. Shuprisha A, Wright SH, Dantzler WH. Method for measuring luminal efflux of fluorescent organic compounds in isolated, perfused renal tubules. Am. J. Physiol. Renal Physiol 279: F960-964, 2000.

54. Suhre WM, Ekins S, Chang C, Swaan PW, Wright SH. Molecular determinants of substrate/inhibitor binding to the human and rabbit renal organic cation transporters hOCT2 and rbOCT2. Mol. Pharmacol 67: 1067-1077, 2005.

55. Tamai I, Ohashi R, Nezu J, Yabuuchi H, Oku A, Shimane M, Sai Y, Tsuji A. Molecular and functional identification of sodium ion-dependent, high affinity human carnitine transporter OCTN2. J. Biol. Chem 273: 20378-20382, 1998.

56. Tanihara Y, Masuda S, Sato T, Katsura T, Ogawa O, Inui K-I . Substrate specificity of MATE1 and MATE2-K, human multidrug and toxin extrusions/H+-organic cation antiporters. Biochem. Pharmacol 74: 359-371, 2007.

57. Terashita S, Dresser MJ, Zhang L, Gray AT, Yost SC, Giacomini KM . Molecular cloning and functional expression of a rabbit renal organic cation transporter. Biochim. Biophys. Acta 1369: 1-6, 1998.

58. Tsuda M, Terada T, Asaka J-ichi, Ueba M, Katsura T, Inui K-ichi . Oppositely directed H+ gradient functions as a driving force of rat H+/organic cation antiporter MATE1. Am. J. Physiol. Renal Physiol 292: F593-598, 2007.

59. Tsuda M, Terada T, Mizuno T, Katsura T, Shimakura J, Inui K-ichi . Targeted disruption of the multidrug and toxin extrusion 1 (mate1) gene in mice reduces renal secretion of metformin. Mol. Pharmacol 75: 1280-1286, 2009.

Page 90: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

89

60. Ullrich KJ, Rumrich G . Luminal transport system for choline+ in relation to the other organic cation transport systems in the rat proximal tubule. Kinetics, specificity: alkyl/arylamines, alkylamines with OH, O, SH, NH2, ROCO, RSCO and H2PO4

-groups, methylaminostyryl, rhodamine, acridine, phenanthrene and cyanine compounds. Pflugers Arch 432: 471-485, 1996.

61. Ullrich KJ . Renal transporters for organic anions and organic cations. Structural requirements for substrates. J. Membr. Biol 158: 95-107, 1997.

62. Ullrich KJ, Papavassiliou F, David C, Rumrich G, Fritzsch G. Contraluminal transport of organic cations in the proximal tubule of the rat kidney. Pflugers Arch. 419: 84-92, 1991.

63. Urakami Y, Okuda M, Masuda S, Saito H, Inui KI. Functional characteristics and membrane localization of rat multispecific organic cation transporters, OCT1 and OCT2, mediating tubular secretion of cationic drugs. J. Pharmacol. Exp. Ther 287: 800-805, 1998.

64. Volk C, Gorboulev V, Budiman T, Nagel G, Koepsell H. Different affinities of inhibitors to the outwardly and inwardly directed substrate binding site of organic cation transporter 2. Mol. Pharmacol 64: 1037-1047, 2003.

65. Watanabe A, Choe S, Chaptal V, Rosenberg JM, Wright EM, Grabe M, Abramson J. The mechanism of sodium and substrate release from the binding pocket of vSGLT. Nature 468: 988-991, 2010.

66. Watanabe S, Tsuda M, Terada T, Katsura T, Inui K-I. Reduced renal clearance of a zwitterionic substrate cephalexin in MATE1-deficient mice. J. Pharmacol. Exp. Ther 334: 651-656, 2010.

67. Wright SH, Wunz TM, Wunz TP . Structure and interaction of inhibitors with the TEA/H+ exchanger of rabbit renal brush border membranes. Pflugers Arch 429: 313-324, 1995.

68. Wright SH, Wunz TM . Transport of tetraethylammonium by rabbit renal brush-border and basolateral membrane vesicles. Am. J. Physiol 253: F1040-1050, 1987.

69. Wright SH, Wunz TM . Mechanism of cis- and trans-substrate interactions at the tetraethylammonium/H+ exchanger of rabbit renal brush-border membrane vesicles. J. Biol. Chem 263: 19494-19497, 1988.

70. Wright SH, Wunz TM . Influence of substrate structure on turnover of the organic cation/H+ exchanger of the renal luminal membrane. Pflugers Arch 436: 469-477, 1998.

Page 91: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

90

71. Wright SH, Dantzler WH . Molecular and cellular physiology of renal organic cation and anion transport. Physiol. Rev 84: 987-1049, 2004.

72. Wright SH . Role of organic cation transporters in the renal handling of therapeutic agents and xenobiotics. Toxicol. Appl. Pharmacol 204: 309-319, 2005.

73. Yabuuchi H, Tamai I, Nezu J, Sakamoto K, Oku A, Shimane M, Sai Y, Tsuji A. Novel membrane transporter OCTN1 mediates multispecific, bidirectional, and pH-dependent transport of organic cations. J. Pharmacol. Exp. Ther 289: 768-773, 1999.

74. Yonezawa A, Inui K-I. Importance of the Multidrug and Toxin Extrusion MATE/SLC47A Family to Pharmacokinetics, Pharmacodynamics/Toxicodynamics and Pharmacogenomics. Br J Pharmacol (April 1, 2011). doi: 10.1111/j.1476-5381.2011.01394.x.

75. Yoshitomi K, Frömter E . Cell pH of rat renal proximal tubule in vivo and the conductive nature of peritubular HCO3

- (OH-) exit. Pflugers Arch 402: 300-305, 1984.

76. Zhang L, Schaner ME, Giacomini KM. Functional characterization of an organic cation transporter (hOCT1) in a transiently transfected human cell line (HeLa). J. Pharmacol. Exp. Ther 286: 354-361, 1998.

77. Zhang X, Cherrington NJ, Wright SH. Molecular identification and functional characterization of rabbit MATE1 and MATE2-K. Am. J. Physiol. Renal Physiol 293: F360-370, 2007.

78. Zhang X, Wright SH. MATE1 has an external COOH terminus, consistent with a 13-helix topology. Am. J. Physiol. Renal Physiol 297: F263-271, 2009.

Page 92: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

91

APPENDIX A

INTERACTION OF H+ WITH THE EXTRACELLULAR AND

INTRACELLULAR ASPECTS OF hMATE1

Dangprapai Y, Wright SH. Interaction of H+ with the extracellular and intracellular

aspects of hMATE1. Am J Physiol Renal Physiol (May 25, 2011). doi:

10.1152/ajprenal.00075.2011

Page 93: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

92

Page 94: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

93

Interaction of H+ with the extracellular and intracellular aspects of hMATE1

Authors: Yodying Dangprapai and Stephen H. Wright Affiliation: Department of Physiology, University of Arizona, Tucson, AZ 85724 Running head: Symmetry of H+ interaction with hMATE1 Corresponding author: Stephen H. Wright University of Arizona College of Medicine Department of Physiology Address: P.O. Box 245051 Tucson, AZ 85724 Phone: (520) 626-4253 Fax: (520) 626-2382 E-Mail: [email protected]

Page 95: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

94

Abstract

Interaction of H+ with the extracellular and intracellular aspects of hMATE1– Human

multidrug and toxin extrusion 1 (hMATE1, SLC47A1) is a major candidate for being the

molecular identity of organic cation/proton (OC/H+) exchange activity in the luminal

membrane of renal proximal tubules (RPT). Although physiological function of

hMATE1 supports luminal OC efflux, the kinetics of hMATE1-mediated OC transport

have typically been characterized through measurement of uptake i.e., the interaction

between outward-facing hMATE1 and OCs. To examine kinetics of hMATE1-mediated

transport in a more physiologically relevant direction i.e., an interaction between inward-

facing hMATE1 and cytoplasmic substrates, we measured the time course of hMATE1-

mediated efflux of the prototypic MATE1-substrate, [3H]1-methyl-4-phenylpyridinium

([3H]MPP), under a variety of intra- and extracellular pH conditions, from CHO cells that

stably expressed the transporter. In this study, we showed that an IC50/K i for interaction

between extracellular H+ and outward-facing hMATE1 determined from conventional

uptake experiments [12.9 ± 1.23 nM (pH 7.89); n = 9] and from the efflux protocol [14.7

± 3.45 nM (pH 7.83); n = 3] were not significantly different (P = 0.6). Furthermore,

kinetics of interaction between intracellular H+ and inward-facing hMATE1 determined

using the efflux protocol revealed an IC50 for H+ of 11.5 nM (pH 7.91), consistent with

symmetrical interactions of H+ with the inward-facing and outward-facing aspects of

hMATE1.

Key Words: organic cation, transport, secretion, renal proximal tubule, pH

Page 96: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

95

The designation ‘organic cations’ (OCs) refers to a large group of organic

compounds that carry net positive charge(s) at physiological pH. The majority of OCs

are xenobiotic i.e., of exogenous origin, and many are toxic. On a regular basis,

individuals consume potentially toxic OCs, typically as plant products and/or prescribed

medicines. Currently, approximately 40 percent of commonly prescribed drugs can be

classified as OCs (7), including metformin (anti-diabetic), cimetidine (anti-peptic ulcer),

and clonidine (anti-hypertensive and analgesic). In the human body, the renal proximal

tubule (RPT) is the major site for OC secretion (24). In RPT epithelial cells (RPTECs),

OC secretion is a two-step process, with OC uptake across the peritubular (basolateral)

membrane by a process of facilitated diffusion, and OC efflux at the luminal (apical)

membrane, mainly by an OC/H+ exchange process (8).

There is broad agreement that the basolateral entry step in OC secretion by the

human RPT is dominated by human organic cation transporter 2 (hOCT2, SLC22A2),

which has been well characterized both structurally and functionally (24). There is also

growing consensus that the exit step, also the rate-limiting step (24), for OC secretion

involves the combined activity of two members of the Multidrug And Toxin Extrusion

(MATE) family, MATE1 (SLC47A1) and MATE2-K (SLC47A2) (8, 12). Although in

their proposed physiological role MATEs mediate the efflux of OCs, the characterization

of MATE-mediated OC transport has, with few exceptions, relied on measurement of

uptake i.e., influx, with the tacit assumption that MATEs interact symmetrically with

their substrates, including H+ (10). While this is a reasonable assumption with respect to

the catalytic and energetic mechanism of transport, i.e., mediated exchange of H+ for OC

Page 97: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

96

(12, 19), it may well not be the case with respect to the kinetics and selectivity of

substrate interaction with these transporters. In that regard, it is significant that rat Oct2

(rOct2, slc22a2) has been shown to have markedly asymmetrical interactions with (at

least) selected OCs. For example, the affinity of tetrabutylammonium (TBuA) to

outward-facing rOct2 is four-fold higher than its affinity to inward-facing rOct2, whereas

the affinity of corticosterone to outward-facing rOct2 is twenty-fold lower than its

affinity to inward-facing rOct2 (20).

To study MATE-mediated OC transport in its physiologically relevant direction

i.e., hMATE1 as an OC efflux transporter, we quantified the MATE-mediated efflux of

the model OC, MPP. In this study, we show that kinetics of interaction between outward-

facing hMATE1 and H+ measured using an efflux protocol are not significantly different

from those obtained using conventional uptake experiments. Furthermore, for the first

time, we report kinetics of interaction between inward-facing hMATE1 and H+. These

results support the conclusion that, despite the likelihood of structurally distinct binding

regions exposed to the cytoplasmic vs. extracellular faces of hMATE1, the kinetics of H+

interaction with each face of the transporter are similar.

Materials and Methods

Chemicals. [3H]1-methyl-4-phenylpyridinium ([3H]MPP; 80 Ci/mmol) and [2-(4-nitro-

2,1,3-benzoxadiazol-7-yl)aminoethyl]trimethylammonium [NBD-TMA (2)] were

synthesized by the Department of Chemistry and Biochemistry, University of Arizona.

[14C]Mannitol (58.8 mCi/mmol) was purchased from PerkinElmer. 2’,7’-bis-(2-

Page 98: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

97

carboxyethyl)-5-(and-6)-carboxyfluorescein acetoxymethyl (BCECF-AM) and nigericin

were obtained from Invitrogen. Tetrapentylammonium (TPeA) bromide, saponin, and

other chemicals (unless otherwise noted) were purchased from Sigma.

Cell culture. CHO Flp-In cells (Invitrogen) that stably expressed hMATE1 (CHO

hMATE1 cells), created in our lab as previously described (26), were grown in Ham’s

F12 Kaighn’s modification medium (Sigma) supplemented with 10% fetal bovine serum

(HyClone) and 0.1 mg/ml of Hygromycin B (Invitrogen). These cells were maintained in

an incubator (NuAire) at 37oC supplied with 5% CO2. Subculture of the cells was

performed every 3 to 4 days. For transport studies, the CHO hMATE1 cells were seeded

in multi-well cell culture plates (Cellstar) or in 35-mm cell culture dishes (Falcon).

hMATE1-mediated MPP uptake. CHO hMATE1 cells were grown to confluence in 12-

well or 24-well cell culture plates. Before each uptake, cells were incubated in

Waymouth buffer (WB; in mM, NaCl 135, Hepes 13, CaCl2-2H2O 2.5, MgCl2-6H2O 1.2,

MgSO4-7H2O 0.8, and D-glucose 28) pH 7.4 at room temperature (between 20oC to

25oC) for 30 minutes. After removal of WB pH 7.4, MPP uptake was initiated by adding

WB pH 8.5 containing [3H]MPP (approximately 13 nM). For a time course study,

[3H]MPP uptake was terminated at specific times by aspirating and rinsing each well with

ice-cold (4oC) WB pH 8.5. Cells were lysed by adding 0.5 N NaOH/1% SDS, and

neutralized with 1 N HCl. After being mixed with scintillation cocktail (MP

Biomedicals), lysate from each sample was transferred to a liquid scintillation counter

(Beckman LS6000IC) to determine the amount of [3H]MPP.

Page 99: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

98

hMATE1-mediated MPP efflux. Two (similar) protocols were used. The first used

CHO hMATE1 cells grown in 48-well cell culture plates. Cells grown to confluence

were incubated in WB pH 7.4 for 30 minutes at room temperature. Then, WB pH 7.4

was replaced by WB pH 8.5 containing [3H]MPP (~13 nM) for 20 minutes to allow

accumulation of labeled MPP into the cells. After the solution containing [3H]MPP was

removed, the cells were briefly rinsed with WB pH 8.5 at room temperature. Efflux of

the labeled MPP was initiated by adding various buffers, according to the purpose of each

experiment. The efflux was terminated by removing the buffer at a specific time, and the

remaining intracellular [3H]MPP was determined by lysing the cells with 0.5 N NaOH

containing 1% SDS. Radioactivity in each cell lysate was determined as described

above. The amount of [3H]MPP remaining in the cells at each time point was expressed

as a percentage of the amount of accumulated [3H]MPP before efflux was initiated.

The second efflux protocol measured continuous efflux from CHO hMATE1 cells

grown in a 35-mm cell culture dish. After the cells were incubated in WB pH 7.4 at room

temperature for 30 minutes, a dish insert (Warner Instruments, RC-37FC; Supplemental

Fig. S1) was assembled within the 35-mm dish, creating a closed chamber with a growth

surface area of 1.85 cm2 and a volume of 185 µl. WB pH 8.5 containing [3H]MPP and

[14C]mannitol (approximately 39 nM and 10 µM, respectively) was introduced into the

chamber via polyethylene (PE) tubing and [3H]MPP was allowed to accumulate for 20

minutes within the CHO hMATE1 cells (the mannitol served as a marker for extracellular

volume). Subsequently, the buffer containing labeled solutes was removed from the

chamber through PE tubing at the distal end of the chamber and hMATE1-mediated

Page 100: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

99

[3H]MPP efflux was initiated by introducing laminar flow of various buffers through the

chamber using a syringe pump (Harvard Apparatus) at a constant rate (5 ml/minute). A

fraction collector was used to continuously collect buffer at the distal end of the chamber;

fractions were typically collected every six seconds (usually for 2 to 2.5 minutes). The

amounts of [3H]MPP and [14C]mannitol were measured in each sample as described

earlier using a double-label counting protocol. A representative result of [3H]MPP efflux

measured using the chamber protocol is shown in Supplemental Figure S2.

Intracellular pH measurement. CHO hMATE1 cells were incubated in WB containing

5 µM BCECF-AM at room temperature for 15 minutes. Using excitation wavelengths of

488 nm and 460 nm, the fluorescence signal from intracellular BCECF was measured at

an emission wavelength of 535 nm. The ratio of fluorescence from the two excitation

wavelengths was recorded over time using an InCyt Im2 imaging system (Intracellular

Imaging, Inc.). Intracellular pH (pHi) of CHO hMATE1 cells was calculated from a

calibration curve performed after each measurement in standard pH buffers containing 10

µM nigericin (see Supplemental Fig. S3 for a representative example).

Statistics. Data were analyzed using Prism (GraphPad Software). Statistical tools such

as paired t-test or unpaired t-test were applied according to the nature of each data set.

Results and Discussion

Cis-inhibition by extracellular H+ of hMATE1-mediated OC uptake. We first

characterized hMATE1-mediated transport of the test substrate, MPP, in CHO cells that

stably expressed the transporter. Uptake of [3H]MPP was linear for approximately ten

Page 101: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

100

minutes (Fig. 1A) and five minute uptakes were selected for use in subsequent kinetic

studies as it covered the initial rate of MPP uptake in CHO hMATE1 cells. Figure 1B

shows that unlabeled MPP blocked the uptake of labeled MPP with a profile that was

adequately described by the Michaelis-Menten equation for the competitive interaction of

labeled and unlabeled substrate (Malo and Berteloot, 1991):

max

t

J [MPP*]J* D[MPP*]

K [MPP*] [MPP]= +

+ +

where J* is the rate of [3H]MPP transport from a concentration equal to [MPP*]; Jmax is

the maximum rate of mediated MPP transport; Kt is the MPP concentration that resulted

in half-maximal transport (Michaelis constant); [MPP] is the concentration of unlabeled

MPP in the uptake reaction, and D is a constant that describes the non-saturable (first-

order) component of total substrate uptake (over the range of substrate concentrations

tested) that presumably reflected the combined influence of diffusive flux, nonspecific

binding, and/or incomplete rinsing of the cell layer. In four separate experiments, the

Jmax was 2.1 ± 0.18 pmol min-1 cm-2 and the apparent Kt (at pH 8.5) was 4.0 ± 0.29 µM

(Table 1).

The interaction between outward-facing hMATE1 and H+ was initially

characterized using two experimental approaches that each involved measurement of

uptake. First, the IC50 for H+ inhibition of hMATE1-mediated MPP uptake was

determined (Fig. 2). The decrease in MPP uptake resulting from an increase in

extracellular [H+] ([H+]o) was adequately described by the following relationship (4):

app

o

J [MPP*]J D[MPP*]

IC [H ]50

+= ++

Page 102: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

101

where J is the rate of [3H]MPP uptake; Japp is the product of the maximum rate of

[3H]MPP uptake (Jmax) and the ratio of the Ki of H+ and Kt for MPP transport; and IC50 is

the concentration of [H+]o that reduced mediated (i.e., blockable) [3H]MPP transport by

50%. The IC50 for H+-inhibited hMATE1-mediated MPP uptake was 12.4 ± 2.21 nM (pH

7.91, n = 5). Although this is the first kinetic assessment of the inhibitory effect of

extracellular H+ on hMATE1-mediated transport, several previous studies showed that

transport was reduced as extracellular pH was decreased below a value of 8.5 (12, 18,

19). The most detailed of these (18), when replotted as the effect of [H+]o on MATE1-

mediated transport of [14C] tetraethylammonium (TEA, a prototypic OC) suggested an

IC50 of about 15 nM (pH ~7.8), similar to the value noted here.

It is also generally observed that hMATE1-mediated OC transport decreases as

pH values increase beyond 8.5 (12, 18), suggesting that H+ interaction with hMATE1 is

unlikely to be limited to a simple competitive interaction and probably includes indirect

effects of reduced [H+]o on the structure of hMATE1 and/or other proteins. Therefore,

we decided to use another approach to determine an ‘apparent Ki’ for hydrogen ion (KHi-

app), namely, the influence of extracellular [H+] on the kinetics of MPP transport.

Kinetics of hMATE1-mediated [3H]MPP uptake at pH 7.5 ([H+] = 31.6 nM) and pH 8.5

([H+] = 3.2 nM) are presented in Table 1. The increase in [H+]o resulted in a 3-fold

increase in the apparent Kt for MPP transport. This effect was anticipated given the

previous observation that extracellular H+ acts as a competitive inhibitor of OC/H+

exchange in isolated renal brush border membrane vesicles (BBMV) (23). However, in

our current experiments on [3H]MPP uptake in intact cells, it was also evident that the

Page 103: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

102

increase in extracellular [H+] resulted in a modest (1.8-fold) but significant (P < 0.01)

increase in the Jmax for MPP transport, which is not consistent with a purely competitive

interaction of H+ with MPP at the external face of hMATE1. This result was, in fact,

seen in all four experiments that measured the kinetics of MPP transport at two different

pHs (Table 1). Nevertheless, although these data suggest that extracellular hydrogen ion

interacts with hMATE1 at several sites, resulting in a ‘mixed-type’ inhibitory profile, we

suggest that the predominant influence of H+ on apparent Kt reflected competition

between H+ and MPP for a common site or (more likely) a set of mutually exclusive sites

within a common binding surface.

The simplifying assumption that H+ acts predominantly as a competitive inhibitor

of hMATE1-mediated MPP transport permitted calculation of KHi-app using the

relationships (4):

max

t app

J [MPP]J

K [MPP]−

=+

, where ot app t H

i app

[H ]K K

K1 +

+

−−

= +

For these relationships, J and Jmax are as previously described; Kt is the Michaelis

constant for hMATE1-mediated MPP transport; and Kt-app is the apparent Kt for MPP

transport (reflecting the competitive influence of [H+]o). From the experiments shown in

Table 1, the KHi-app for the competitive interaction of H+ with extracellular aspect of

hMATE1 was 13.5 ± 0.84 nM (pH 7.87) which was not different from the apparent IC50

for H+ inhibition of hMATE1-mediated MPP transport, noted above (P = 0.73).

Page 104: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

103

Characterization of hMATE1-mediated OC efflux. The intent of this study was

development of a quantitative view of the kinetics of hMATE1-mediated transport when

operating in its ‘physiological’ direction, i.e., exporting OCs. To this end, we used two

similar approaches to quantify hMATE1-mediated efflux from cells stably expressing this

protein. The first involved measurement of the amount of substrate retained in cells over

time after an initiation of efflux. Figure 3A shows the 10-minute time course of [3H]MPP

efflux from CHO hMATE1 cells obtained using this method. As expected, there was a

time-dependent decrease in cell [3H]MPP and, as supported by observations described

below, the loss of [3H]MPP from the cells was effectively restricted to efflux via

hMATE1. Because the concentration of [3H]MPP in the cells at time zero was likely to

be well below the Kt at the cytoplasmic face of the transporter, the kinetics of efflux

could have been expected to be first-order1. This was not, however, the case, as shown

by the non-linearity of the semi-log presentation of these data (Fig. 3A). The

curvilinearity of the plot suggested that MPP efflux involved at least two compartments,

arranged in series (e.g., an endosomal compartment and the cytoplasm) with the

extracellular compartment. Furthermore, the evident curvilinearity necessitates that

passage from the first (‘endosomal’) into the second (cytoplasm), is rate-limiting. This is

not an unexpected result in light of evidence that organic cations can, in fact, be

sequestered within intracellular compartments (11), including uptake of TEA into

endosomal vesicles isolated from rat renal cortex (14).

To assess, at least qualitatively, if such sequestration could account for the non-

first order efflux profile shown in Figure 3A, we examined the time course of efflux of a

Page 105: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

104

fluorescent substrate for OC transporters, NBD-TMA (λex = 458 nm, λem = 530 nm) (2).

NBD-TMA blocked hMATE1-mediated MPP transport in CHO cells with an IC50 of 14.8

µM (data not shown), and though NBD-TMA may well display a quantitatively distinct

distribution profile in CHO cells than the structurally distinct MPP, it provided a useful

probe for visualizing the complexity of substrate distribution in these cells. CHO

hMATE1 cells were incubated in WB pH 8.5 containing 0.5 mM NBD-TMA for 15

minutes in an incubator with 5% CO2 at 37oC. The NBD-TMA loaded cells were then

transferred to the stage of an epifluorescence microscope (Olympus IX71). Intracellular

accumulation of fluorescent signal was only observed in cells that expressed hMATE1

(data not shown). After brief rinsing, NBD-TMA efflux was initiated by adding buffers

without substrate. Figure 4 provides a sequence of images showing the time course of

NBD-TMA efflux from CHO hMATE1 cells under several conditions.

Before the efflux was initiated, i.e., at t = 0, fluorescence within CHO hMATE1

cells could be categorized into two distinct patterns: a comparatively diffuse,

homogenous signal and a punctate signal. The diffuse signal was observed throughout

the cells and probably represented free NBD-TMA in the cytoplasm. In contrast, the

punctate signal was scattered, more intense, and probably represented NBD-TMA

sequestered within cytoplasmic organelles. Our finding was similar to a result from

primary rat choroid plexus cells showing that distribution of accumulated quinacrine

(another fluorescent OC) includes both a diffuse and a punctate fluorescent signal (11).

Furthermore, as noted earlier, endosomes isolated from the rat renal cortex accumulate

TEA via TEA/H+ exchange (14). Based on these results, we suggest that the punctate

Page 106: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

105

distribution of NBD-TMA in CHO cells most likely represented a similar sequestration in

cytoplasmic endosomes.

But is efflux of sequestered OC from this intracellular compartment slow

compared to efflux from the cytoplasm across the plasma membrane? When efflux of

NBD-TMA was performed at extracellular pH 7.4, the diffuse signal rapidly diminished

and almost disappeared after 10 minutes, whereas the punctate signal was still largely

retained (Figure 4, panel a). In contrast, when 0.1 mM TPeA, a non-transported inhibitor

of OC/H+ exchange in renal BBMV (22), was present in the extracellular medium during

the efflux period, both patterns of NBD-TMA signal were effectively retained for at least

10 minutes (Figure 4, panel b). A brief exposure to 0.01% saponin can selectively

permeabilize cholesterol-rich membranes, such as the plasma membrane (17), and when

NBD-TMA loaded cells were exposed to saponin (co-exposed with 0.1 mM TPeA), the

diffuse signal cleared as early as 15 seconds while the punctate fluorescent signal still

remained after 10 minutes of efflux (Fig. 4, panel c).

These results support the contention that at the start of the efflux period, [3H]MPP

was distributed in at least two compartments: a ‘fast’ cytoplasmic compartment that

should readily access hMATE1 at the plasma membrane; and a ‘slow’ compartment

consisting of [3H]MPP sequestered in various cytoplasmic organelles. The solid line fit

to the data presented in Figure 3A assumed a two-phase model for the exponential loss of

[3H]MPP from CHO hMATE1 cells: a large compartment (77% of accumulated [3H]

MPP]) that turned over with a half-time of 1.6 minutes; and a smaller compartment (23%

of accumulated [3H]MPP) that turned over with a half-time of over ten minutes. For this

Page 107: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

106

study, we focused on the early time course of [3H]MPP efflux that we suggest was

dominated by the fast, cytoplasmic compartment. We found that [3H]MPP in the slow,

sequestered compartment could be treated as a constant, permitting the data to be

analyzed using a one-compartment model that decayed to a plateau, using the

relationship: [3H]MPPt = ([3H]MPPt0 – C) e(-kt) + C, where [3H]MPPt is total labeled

MPP in the cells at any time t after initiation of efflux (t0); C is sequestered [3H]MPP; and

k is the first-order rate constant for hMATE1-mediated [3H]MPP efflux from the

cytoplasmic compartment. Correcting total [3H]MPP in the cells for the sequestered

compartment resulted in an efflux profile that was described by the relationship:

[3H]MPPt = ([3H]MPPt0) e(-kt)

The adequacy of this model to describe the efflux of [3H]MPP from CHO hMATE1 cells

is evident in the linearity of the semi log plot of these data presented in Figure 3B, and

the similarity in the rate constant of efflux (k) calculated after subtraction of sequestered

[3H]MPP (0.37 min-1) to the rate constant of efflux from the fast compartment of the two

compartment model (0.43 min-1; Fig. 3A). For the rest of this study, each efflux data set

was analyzed by a single-phase decay equation after subtraction of the estimated

sequestered pool of [3H]MPP, as shown above.

The analysis of [3H]MPP efflux from pre-loaded CHO hMATE1 cells also

permitted a direct test of the hypothesis that an inwardly-directed H+ gradient should

increase the rate of hMATE1-mediated MPP efflux. These experiments used the second

of the two efflux protocols described in the methods, the flow chamber that permitted

continuous monitoring of [3H]MPP that left the cells into the passing stream of buffer.

Page 108: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

107

Within the limit of resolution of the method, the efflux of MPP from the cytoplasmic

compartment of CHO hMATE1 cells (i.e., total efflux corrected for the ‘slow’

compartment) was adequately described by first-order kinetics (Fig. 5). In these

experiments, the half-life (t0.5) of hMATE1-mediated MPP efflux in buffer of the

extracellular pH 8.5 was 9.8 ± 1.43 min (n = 4), and decreasing the external pH to 7.4

(i.e., increasing [H+]o from 3.2 to 39.8 nM) resulted in a 7-fold decrease (P = 0.0004) in

the t0.5, down to 1.4 ± 0.08 min (n = 5). These results confirmed previous observations

(12, 19) that inwardly-directed H+ gradients trans-stimulate MATE1-mediated OC efflux

and, of particular importance to the present context, provided a validation for our

approach to measuring hMATE1-mediated OC efflux.

Quantification of MPP efflux also provided an alternative means for assessing the

kinetics of H+ interaction with the external face of hMATE1. Figure 6A shows the effect

of systematically increasing extracellular [H+] on the rate of MPP efflux. As expected,

hMATE1-mediated MPP efflux was stimulated by increasing the concentration of

extracellular H+, i.e., the exchangeable substrate. Furthermore, the rate constant for MPP

efflux supported by each extracellular [H+], when plotted as a function of [H+] (Fig. 6B),

was adequately described by the Michaelis-Menten equation; the half-maximal

stimulation of MPP efflux occurred at an extracellular pH of 7.83 ([H+] = 14.7 ± 3.45 nM;

n = 3), and this result was not significantly different from either the IC50 (P = 0.79) or the

K i (P = 0.63) for extracellular H+ inhibition of hMATE1-mediated MPP uptake. Note

that hMATE1-mediated MPP efflux was also performed at pHo 6.5 and 6.0, but the rate

constants for efflux at these values of pHo (0.37 ± 0.013 and 0.36 ± 0.017 minute-1,

Page 109: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

108

respectively) did not differ from that measured at pHo 7.0 (0.37 ± 0.02 minute-1) and, so

(for clarity) were not shown in Figure 6A.

Interaction of intracellular H+ with the cytoplasmic face of hMATE1. To examine

kinetics of interaction between H+ and the inward-facing aspect of hMATE1, MPP efflux

was determined as a function of intracellular H+ concentration. From multiple

measurements at room temperature in WB pH 7.4, CHO hMATE1 cells demonstrated a

baseline pHi of 7.57 ± 0.03 ([H+] i = 27.1 ± 1.79 nM; n = 21). This baseline value varied

by less than 0.2 pH units during a 15-minute exposure to WB of different pH. Figure 7A

shows a representative result of the change in pHi resulting from an acute shift from an

extracellular pH of 7.4 to one of 8.5; baseline pHi increased by only 0.1 pH units (from

pH 7.53 to pH 7.63). Manipulation of pHi was achieved through acute exposure of cells

to buffers containing increasing concentrations of NH4Cl. Figure 7B shows the effect on

the pHi of CHO hMATE1 cells of an acute exposure to 20 mM NH4Cl. Cytoplasmic pH

shifted within 8 seconds from a steady-state value of 7.7 to 8.3, which was maintained up

to 3 minutes. Acute exposure of these cells to 2.5, 5, 10, or 20 mM NH4Cl effectively

established cytoplasmic pH values of pH 7.8 ([H+] i = 15.8 ± 0.44 nM; n = 3), pH 7.9

(12.5 ± 1.42 nM; n = 6), pH 8.1 (8.7 ± 0.76 nM; n = 3), or pH 8.2 (5.8 ± 0.35 nM, n = 4),

respectively.

We also considered it necessary to determine if extracellular NH4+ had a direct

effect on the transport process. The presence of 20 mM NH4Cl in an uptake buffer (pH

8.5) decreased the 5-minute uptake of [3H]MPP by about 70% (Fig. 8). An inhibition of

Page 110: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

109

uptake was expected because of the associated decrease in exchangeable H+ in the

cytoplasm noted above. However, it could also have included a direct inhibitory effect of

NH4+ on the extracellular face of hMATE1. We reasoned that, if the observed decrease

in MPP uptake was due simply to a decrease in intracellular [H+], then providing an

alternative exchangeable substrate at the cytoplasmic face of hMATE1 should rescue the

inhibitory effect of extracellular NH4+ on MPP uptake. To test this hypothesis, CHO

hMATE1 cells were pre-loaded with a high concentration of non-labeled MPP (5 mM

MPP for 10 minutes) prior to measuring [3H]MPP uptake in WB containing 20 mM

NH4Cl. Results in Figure 8 demonstrated that [3H]MPP uptake inhibited by extracellular

NH4Cl was quantitatively rescued in the cells pre-loaded with non-labeled MPP,

suggesting that extracellular NH4+ did not directly inhibit hMATE1-mediated MPP

uptake and that observed effects of NH4Cl on transport reflected changes in the

cytoplasmic [H+].

The rate of MPP efflux should increase with decreasing intracellular H+

concentration because of the associated decrease in competition between the two

substrates at the cytoplasmic face of hMATE1. To test this, cells were pre-loaded with

[3H]MPP and efflux was initiated at extracellular pH 8.5 with no NH4Cl in the

extracellular medium. After one minute, buffer was switched to WB pH 7.4 (to stimulate

efflux; see Fig. 5) containing different concentrations of NH4Cl to manipulate

intracellular pH. Efflux was then continuously monitored for another 2 minutes. As

shown in Figure 9, decreasing the pH from 8.5 to 7.4 (i.e., increasing the concentration of

extracellular H+) produced the anticipated increase (approximately 7 fold) in rate of

Page 111: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

110

[3H]MPP efflux. In this condition, with no extracellular NH4Cl, the pHi of CHO cells

was 7.57 ([H+] i = 27.1 nM). As expected, alkalinization of the cytoplasm by addition of

10 mM NH4Cl (pHi = 8.1, [H+] i = 8.7 nM) or 20 mM NH4Cl (pHi = 8.2, [H+] i = 5.8 nM)

did progressively increase the rate of [3H]MPP efflux.

To determine the kinetics of interaction between inward-facing hMATE1 and

intracellular H+, hMATE1-mediated MPP efflux was performed at an extracellular pH of

7.4 with no NH4Cl, and with 2.5, 5, 10, or 20 mM NH4Cl. As shown in Figure 10, the

efflux rate constant (k) increased in a concentration-dependent manner as the cells were

exposed to increasing concentrations of NH4Cl, i.e., as intracellular [H+] was

systematically decreased from 27.1 nM (pH 7.57) to 5.8 nM (pH 8.2), see inset to Fig. 10.

The IC50 for cytoplasmic H+ on hMATE1-mediated MPP efflux was at pH 7.94 ([H+] i =

11.5 nM). This value did not differ substantially from the range of values (12 nM to 15

nM) for the apparent Ki/IC50 of H+ interaction with the extracellular face of hMATE1

described earlier. Acknowledging that the pH range upon which this estimate of the

cytoplasmic IC50 was based was, of necessity, narrower than that used to study the

interaction of H+ with the extracellular aspect of hMATE1, we suggest that these data

support the conclusion that, within the limits of resolution of currently available methods,

the intra- and extracellular faces of hMATE1 display kinetically symmetrical interactions

with H+.

It is relevant to place the kinetic profile of H+ interaction with hMATE1 into the

context of OC secretion in intact RPT. In the ‘early’ proximal tubule, pH of the

glomerular filtrate is moderately alkaline, ~7.1 (6) reflecting a plasma pH of ~7.4. The

Page 112: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

111

current results suggest that, at this pH, the extracellular face of hMATE1 is ~ 85%

occupied by H+ in the filtrate. These protons not only facilitate an exchange for

cytoplasmic OCs, but would also limit interaction between luminal OCs and outward-

facing hMATE1, i.e., luminal H+ should reduce ‘re-uptake’ of secreted OCs via

hMATE1. Furthermore, as a result of H+ secretion, and bicarbonate and water

reabsorption, along the length of the proximal tubule, the concentration of luminal H+

increases. In the late proximal tubule, pH of the filtrate can be as acidic as pH 6.8 to pH

6.7 (1), representing a concentration of H+ that should occupy at least 92% of available

hMATE1 transporters. This increase in luminal H+ would continuously reduce a ‘re-

uptake’ of luminal OCs, the concentration of which can also be expected to increase

along the length of the RPT (due to the combined influence of MATE secretory activity

and water reabsorption).

The interaction of H+ with hMATE1 also underscores its role in providing the

‘driving force’ for OC secretion in RPT. As a secondary active H+ exchanger, MATE1

can mediate export of OCs against their electrochemical gradient driven by the coupled

influx of H+ moving down its electrochemical gradient. The active OC transport via the

OC/H+ exchanger is frequently described as being driven by an inward H+ gradient and,

indeed, is typically assessed in this way in studies using isolated renal BBMV – including

our own studies (23). It is, however, evident from the previous discussion that this can

give a false impression of the energetic basis of OC export mediated by MATEs in

RPTEC. In the early RPT, which appears to be the primary site of OC secretion (16), the

inwardly-directed chemical gradient for H+ is very modest, about 1.25 fold, reflecting a

Page 113: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

112

luminal pH of about 7.1 vs. a cytoplasmic pH of about 7.2 (25). Nevertheless, the tubule

is still poised to support a robust active transepithelial OC secretion because uptake of

OC from the blood into the RPTEC across the basolateral membrane is mediated by the

electrogenic activity of OCT2. Consequently, the inside negative membrane potential of

these cells, -60 to -70 mV (25), can be expected to support concentrative (yet passive)

accumulation of OCs to levels 10-15 times that in the blood. Importantly, the luminal

OC export step, mediated by MATE1 (and/or MATE2-K) involves obligatory,

electroneutral exchange (24). Thus, even in the absence of an inwardly-directed H+

chemical gradient, exchange of a proton in the filtrate for OC in the cytoplasm can permit

development of a luminal OC concentration that approaches the elevated level (compared

to the blood) of OC in the cytoplasm, with the result being active transepithelial

secretion. The MATE1-mediated step is still the ‘active’ element in this process because,

even in the absence of a chemical gradient for H+, protons are still far from

electrochemical equilibrium and kept that way through the activity of Na+/H+ exchange.

The increasing acidity of the tubular filtrate along the length of the RPT further enhances

the energetic effectiveness of MATE-mediated OC secretion into the tubular filtrate.

It is also relevant to discuss the potential impact on renal OC secretion of the

comparatively high affinity of the cytoplasmic face of hMATE1 for H+. With an

apparent Ki of 11.5 nM (pH 7.94), the cytoplasmic pH in RPTEC of pH 7.2 ([H+] = 63.1

nM) (25) suggests that the cytoplasmic face of MATE1 should be ~85% occupied by

hydrogen ion. Based on selectivity studies in our lab and others (12, 19), the affinity for

OCs of the outward-facing aspect of hMATE1 is typically 10 to 1000-times lower than

Page 114: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

113

that for H+, and at least in general terms this is likely to be the case for the inwardly-

directed face of MATE1, as well. This would seem to ‘set the stage’ for rather brisk

competition between accumulated OCs and near-saturating levels of cytoplasmic H+. But

that scenario assumes that the ‘bulk’ pH of the cytoplasm (which is what the BCECF

measurements represent) is an accurate indicator of the hydrogen ion concentration

within the unstirred ‘microdomain’ at the cytoplasmic face of the cell membrane. While

this is arguably the case in the current experiments with CHO cells, in which cytoplasmic

pH values were 7.5 and higher [well below the ‘set-point’ for NHEs (13)], the same may

not be the case in salt transporting epithelia like the renal proximal tubule. Indeed, it is

worth noting that there is experimental evidence that the activity of Na+/H+ exchange

creates a markedly acidic microenvironment at the extracellular face of the luminal

membrane of enterocytes (1). We suggest, therefore, that the activity of the major

Na+/H+ exchangers (NHEs) in the luminal membrane of RPTEC i.e., NHE3 and NHE8,

could create local alkaline conditions near the cytoplasmic face of the membrane, thereby

facilitating the effective interaction of accumulated OCs with the inward face of MATE1.

Interestingly, results from a mathematical model of the renal proximal tubule brush

border membrane indicated that Na+/H+ exchange activity is (in principle) capable of

establishing a gradient of luminal H+ along the length of each microvillus, with the

cytoplasmic tip of the microvillus as alkaline as pH 8.0 (9). Another consequence of the

differential impact of NHE activity, i.e., local acidification of the extracellular face of the

membrane vs. local alkalinization of the cytoplasmic face of the membrane, would be an

effective increase in the inwardly-directed electrochemical gradient for H+ that would

Page 115: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

114

enhance the secretory ‘power’ of MATE1-mediated OC export. Further study regarding

the expression profile of hMATE1 at the luminal membrane of the hRPTEC and the

influence of NHEs in hMATE1-mediated OC efflux are necessary for a better

understanding of this physiologically, pharmacologically, and toxicologically important

process.

Although our current results suggest that hMATE1 is symmetrical with respect to

its kinetic interactions with intra- and extracellular H+, it is premature to assume that

organic substrates will display a similar symmetry. Given the intrinsic structural

asymmetry of transport proteins, in conjunction with the ‘alternating access’ model of

transporter function, it is unlikely that binding surfaces of the ‘translocation pathway’

will be identical when accessed from the cytoplasmic vs. extracellular faces of the

membrane. Whereas protons titrate single carboxyl residues, the transient, weak

interactions of an organic substrate reflect interactions with multiple residues (5, 21) and

even modest differences in the 3D structure of binding surfaces are likely to influence

binding kinetics. As noted previously, the affinities of the inner vs. outer faces of rOct2

for corticosterone and TBuA differ markedly. Importantly, the differences are in the

opposite direction; whereas the inward face has a higher (20x) affinity for corticosterone,

the outward face has a higher affinity (4x) for TBuA (20). We suggest that a similar

degree of kinetic asymmetry can be expected for other multispecific transporters,

including MATE1. In this regard, it is also appropriate to note that the kinetic profiles

reported here reflect the interaction of a single substrate with MATE1, i.e., MPP.

Although MPP is a widely used to characterize organic cation transporters, both OCTs

Page 116: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

115

(3) and MATEs (12), structurally distinct substrates for the multiselective MATE

transporters could display a different kinetic interaction with H+.

In summary, the interaction between H+, both intracellular and extracellular, and

the MATE1-mediated transport of MPP was examined, and the results suggested that the

kinetics of interaction between inward-facing hMATE1 and intracellular H+ are not

significantly different from the kinetics of the interaction between outward-facing

hMATE1 and extracellular H+. Nevertheless, despite this apparent symmetry of H+

interaction with hMATE1, it can be anticipated that at least some OCs will display

asymmetrical interactions with intra- vs. extracellular faces of hMATE1. The approach

for assessing the kinetics of OC efflux from intact cultured cells introduced in the present

study offers a potential means to address this issue.

Page 117: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

116

Footnote

1The volume of CHO cells, determined using tritiated water corrected for extracellular

space with [14C]mannitol, was approximately 0.3 µl/cm2 of CHO cell (confluent layer;

data not shown), similar to previous reports on the volume of CHO cells (15). If

accumulated [3H]MPP was distributed uniformly in this volume (which, as discussed in

the text, is an oversimplification), the calculated ‘bulk’ concentration of [3H]MPP in

CHO hMATE1 cells in this study was as high as ~0.4 µM at initiation of efflux. This

level of accumulated MPP is about 35 times lower than the apparent Kt for interaction

between MPP and external face of hMATE1 at pH 7.5 (12.3 ± 1.08 µM; n = 4). Although

an apparent Kt for MPP interaction with cytoplasmic face of hMATE1 has not been

characterized, we suggest that the [MPP]cell was sufficiently low to anticipate first-order

behavior for hMATE1-mediated MPP efflux.

Page 118: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

117

Acknowledgments

The authors would like to thank Dr. Amritlal Mandal for his assistance on intracellular

pH measurement. The work presented here was supported, in part, by NIH grants

DK58251, DK080801, and ES06694. During the course of this study, funding for Y.

Dangprapai was partially supported by the Department of Physiology, Faculty of

Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.

Page 119: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

118

References

1. Anderson CMH, Thwaites DT. Hijacking solute carriers for proton-coupled drug transport. Physiology (Bethesda) 25: 364-377, 2010.

2. Bednarczyk D, Mash EA, Aavula BR, Wright SH. NBD-TMA: a novel fluorescent substrate of the peritubular organic cation transporter of renal proximal tubules. Pflugers Arch 440: 184-192, 2000.

3. Gorboulev V, Ulzheimer JC, Akhoundova A, Ulzheimer-Teuber I, Karbach U, Quester S, Baumann C, Lang F, Busch AE, Koepsell H. Cloning and characterization of two human polyspecific organic cation transporters. DNA Cell Biol 16: 871-881, 1997.

4. Groves CE, Evans KK, Dantzler WH, Wright SH. Peritubular organic cation transport in isolated rabbit proximal tubules. Am. J. Physiol 266: F450-458, 1994.

5. Guan L, Kaback HR. Lessons from lactose permease. Annu Rev Biophys Biomol Struct 35: 67-91, 2006.

6. Hamm LL, Nakhoul N. Renal Acidification. In: Brenner and Rector’s The Kidney, edited by Brenner BM. Saunders, 2007, p. 248 - 267.

7. Kim MK, Shim C-K. The transport of organic cations in the small intestine: current knowledge and emerging concepts. Arch. Pharm. Res 29: 605-616, 2006.

8. Klaassen CD, Aleksunes LM. Xenobiotic, bile acid, and cholesterol transporters: function and regulation. Pharmacol. Rev 62: 1-96, 2010.

9. Krahn TA, Weinstein AM. Acid/base transport in a model of the proximal tubule brush border: impact of carbonic anhydrase. Am. J. Physiol 270: F344-355, 1996.

10. Matsushima S, Maeda K, Inoue K, Ohta K-ya, Yuasa H, Kondo T, Nakayama H, Horita S, Kusuhara H, Sugiyama Y. The inhibition of human multidrug and toxin extrusion 1 is involved in the drug-drug interaction caused by cimetidine. Drug Metab. Dispos 37: 555-559, 2009.

11. Miller DS, Villalobos AR, Pritchard JB. Organic cation transport in rat choroid plexus cells studied by fluorescence microscopy. Am. J. Physiol 276: C955-968, 1999.

12. Otsuka M, Matsumoto T, Morimoto R, Arioka S, Omote H, Moriyama Y. A human transporter protein that mediates the final excretion step for toxic organic cations. Proc. Natl. Acad. Sci. U.S.A 102: 17923-17928, 2005.

13. Ozkan P, Mutharasan R. A rapid method for measuring intracellular pH using BCECF-AM. Biochim. Biophys. Acta 1572: 143-148, 2002.

Page 120: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

119

14. Pritchard JB, Sykes DB, Walden R, Miller DS. ATP-dependent transport of tetraethylammonium by endosomes isolated from rat renal cortex. Am. J. Physiol 266: F966-976, 1994.

15. Rotoli BM, Bussolati O, Dall’Asta V, Gazzola GC. Membrane potential and amino acid transport in a mutant Chinese hamster ovary cell line. J. Cell. Physiol 146: 417-424, 1991.

16. Schäli C, Schild L, Overney J, Roch-Ramel F. Secretion of tetraethylammonium by proximal tubules of rabbit kidneys. Am. J. Physiol 245: F238-246, 1983.

17. Schulz I. Permeabilizing cells: some methods and applications for the study of intracellular processes. Meth. Enzymol 192: 280-300, 1990.

18. Tanihara Y, Masuda S, Sato T, Katsura T, Ogawa O, Inui K-I. Substrate specificity of MATE1 and MATE2-K, human multidrug and toxin extrusions/H+-organic cation antiporters. Biochem. Pharmacol 74: 359-371, 2007.

19. Tsuda M, Terada T, Asaka J-ichi, Ueba M, Katsura T, Inui K-ichi. Oppositely directed H+ gradient functions as a driving force of rat H+/organic cation antiporter MATE1. Am. J. Physiol. Renal Physiol 292: F593-598, 2007.

20. Volk C, Gorboulev V, Budiman T, Nagel G, Koepsell H. Different affinities of inhibitors to the outwardly and inwardly directed substrate binding site of organic cation transporter 2. Mol. Pharmacol 64: 1037-1047, 2003.

21. Watanabe A, Choe S, Chaptal V, Rosenberg JM, Wright EM, Grabe M, Abramson J. The mechanism of sodium and substrate release from the binding pocket of vSGLT. Nature 468: 988-991, 2010.

22. Wright SH, Wunz TM, Wunz TP. Structure and interaction of inhibitors with the TEA/H+ exchanger of rabbit renal brush border membranes. Pflugers Arch 429: 313-324, 1995.

23. Wright SH, Wunz TM. Mechanism of cis- and trans-substrate interactions at the tetraethylammonium/H+ exchanger of rabbit renal brush-border membrane vesicles. J. Biol. Chem 263: 19494-19497, 1988.

24. Wright SH, Dantzler WH. Molecular and cellular physiology of renal organic cation and anion transport. Physiol. Rev 84: 987-1049, 2004.

25. Yoshitomi K, Frömter E. Cell pH of rat renal proximal tubule in vivo and the conductive nature of peritubular HCO3- (OH-) exit. Pflugers Arch 402: 300-305, 1984.

Page 121: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

120

26. Zhang X, Cherrington NJ, Wright SH. Molecular identification and functional characterization of rabbit MATE1 and MATE2-K. Am. J. Physiol. Renal Physiol 293: F360-370, 2007.

Page 122: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

121

Figure legends

Figure 1. Time course and kinetics of [3H]MPP uptake in CHO hMATE1 cells. A:

[3H]MPP accumulated linearly in CHO hMATE1 cells up to approximately 10 minutes

(solid circles). [3H]MPP uptake was inhibited by co-incubating the cells with 1 mM non-

labeled MPP (open circles). Buffer pH was 8.5. Each point is the mean (±SEM) of

uptake measured in two wells of a 24-well plate. B: Uptake (5 min; pH 8.5) of [3H]MPP

transport measured in the presence of increasing concentrations of unlabeled MPP (from

which the kinetics of MPP transport were determined). Data shown are from a single

representative experiment. Each point is the mean (±SEM) of uptakes measured in three

wells of a 24-well plate.

Figure 2. IC50 of extracellular H+ as an inhibitor of hMATE1-mediated MPP

uptake. Five-minute uptakes of 13 nM [3H]MPP into CHO hMATE1 cells were plotted

as a function of extracellular [H+]. Each point is the mean (±SEM) of uptakes determined

in five separate experiments (n = 5).

Figure 3. Time course of MPP loss from CHO hMATE1 cells. A: Following a 20-

minute preload of 13.5 nM [3H]MPP, the amount of labeled MPP remaining in the cells

was measured as a function of time after efflux was initiated in an MPP-free buffer of pH

7.4 ([H+]o = 39.8 nM). Each point is the mean [3H]MPP content (±SEM) of cells in two

wells of a 48-well plate in a single representative experiment. The semi-log plot reveals

that there were multiple compartments involved in [3H]MPP efflux from CHO hMATE1

Page 123: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

122

cells as discussed in the text. The solid line represents two-phase exponential decline in

cell [3H]MPP (kinetic parameters listed in the figure). B: Time course of the first-order

(single phase) decline in cell [3H]MPP, reflecting correction for sequestered [3H]MPP

(see text for discussion). The dotted lines in both graphs represent the 95% confidence

interval of the fitted values (solid line).

Figure 4. Time course of NBD-TMA efflux in CHO hMATE1 cells. CHO hMATE1

cells were pre-loaded with 0.5 mM NBD-TMA at 370C for 15 minutes. Following

aspiration of the medium the cells were transferred to a fluorescent microscope, rinsed

with WB pH 8.5 at which point an image was collected representing ‘t = 0.’ Subsequent

images were collected at the indicated time points following introduction of test media.

For each condition, the cells were illuminated only during collection of the image. The

10-minute images were from different fields of cells (to avoid the influence of

photobleaching).

Figure 5. Effect of extracellular H+ on hMATE1-mediated [3H]MPP efflux. Efflux

was performed at extracellular pH 8.5 ([H+]out = 3.2 nM) and at pH 7.4 ([H+]out = 39.8

nM) using the efflux chamber protocol (see text).

Figure 6. Effect of increasing extracellular [H+] on the kinetics of hMATE1-

mediated [3H]MPP efflux. A: Time course of [3H]MPP efflux from the ‘fast’

cytoplasmic compartment (as defined in the text) was determined in buffers of decreasing

Page 124: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

123

external pH. The value of the slow compartment used for each condition was determined

from the efflux profile at pHo 7.0. Each point is the mean of results obtained in three

separate experiments. B: The rate constants for [3H]MPP efflux from the fast

compartment (the slopes of the lines fit to the data in Fig. 6A) plotted as a function of

extracellular [H+] (line fit using Michaelis-Menten equation).

Figure 7. Intracellular pH (pH i) in CHO hMATE1 cells. A: A representative

experiment showing the modest change in pHi associated with acute change of pHo; in

this case a shift from pHo of 7.4 to pHo of 8.5 increased pHi from 7.53 to 7.63. B: Upon

acute exposure to 20 mM NH4Cl the pHi of CHO hMATE1 cells increased rapidly

(within 8 seconds) from a baseline pHi value (pHi of 7.57, i.e., [H+] i = 27.1 ± 1.79 nM; n

= 21) to a new value (pHi of 8.2, i.e., [H+] i = 5.8 ± 0.35 nM; n = 4) that was relatively

stable for at least three minutes. Data shown are from a single representative experiment.

(Each point of the fluorescence ratio in both A. and B. was an average value measured

from twenty eight CHO hMATE1 cells.)

Figure 8. Effect of NH4Cl and MPP preload on hMATE1-mediated [3H]MPP

uptake. The time course of hMATE1-mediated uptake of [3H]MPP (13.5 nM) was

measured under the conditions indicated in the figure. In every case, pHo was 8.5. Each

point is the mean (±SEM) of uptake values (measured in triplicate) from three separate

experiments (n = 3).

Page 125: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

124

Figure 9. Effect of NH4Cl on hMATE1-mediated [3H]MPP efflux. Cells were

preloaded with [3H]MPP (36.8 nM) for 15 min. Efflux into a buffer of pH 8.5 was

measured for one minute, at which point the cells were exposed to a buffer of pH 7.4

containing the indicated concentrations of NH4Cl (to decrease [H+] i). Efflux into these

new solutions was then determined for two minutes. Single representative examples are

shown for each condition.

Figure 10. Kinetics of interaction between inward-facing hMATE1 and

intracellular H +. The time course of hMATE1-mediated [3H]MPP efflux was measured

for each of the indicated values of intracellular pH. Each point is the mean of values

determined in 3-6 separate experiments. The inset shows the relationship between

cytoplasmic [H+] and the calculated rate constants (mean ±SEM) for hMATE1-mediated

[3H]MPP efflux.

Page 126: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

125

Table

Table 1. Effect of extracellular pH on the kinetics of

hMATE1-mediated MPP uptake Five-minute uptakes of [3H]MPP (~13 nM) were determined

as a function of increasing concentration of unlabeled MPP at two values for external pH (pHo), pH 7.5 ± 0.1 (n = 4) and pH 8.5 ± 0.02 (n = 4). Both Jmax and Kt of MPP uptake in CHO hMATE1 cells from these two pH values were significantly different (* p = 0.01).

pHo

K t

(µM)

Jmax

(pmol min-1 cm-2)

7.5 ± 0.1

8.5 ± 0.02 4.0 ± 0.29

12.3 ± 1.08* 3.7 ± 0.36*

2.1 ± 0.18

Page 127: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

126

Figure 1

0 5 10 15 20 25 300

30

60

90

1 µCi/ml [3H]MPP

1 µCi/ml [3H]MPPwith 1 mM MPP

CHO hMATE1 cells

A

Time (minutes)

[3 H]M

PP

(fm

ol c

m-2

)

0

5

10

15

20

1 10 100 1000

Jmax = 2.1 ± 0.18 pmol min-1 cm-2

0

Kt = 4.0 ± 0.29 µM

B

[MPP] (µM)[3 H

]MP

P (

fmol

min

-1cm

-2)

Page 128: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

127

Figure 2

10 - 210 - 10

100

200

300

400

100 101 102 1031

IC50H+ = 12.4 ± 2.21 nM(pH 7.91)

0

[H+] (nM)

[3H

]MP

P (

% o

f pH

7.5

)

Page 129: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

128

Figure 3

0 5 101

10

100

kfast = 0.37 ± 0.01 min-1

t0.5, fast = 1.88 min

B

Efflux duration (minutes)C

ell [

3 H]M

PP

(%

of

t =

0)

0 5 1010

100

kfast = 0.43 ± 0.12 min-1

t0.5, fast = 1.62 min

kslow = 0.06 ± 0.09 min-1

t0.5, slow = 10.7 min

A

Efflux duration (minutes)

Cel

l [3H

]MP

P (

% o

f t

= 0

)

Page 130: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

129

Figure 4

a. pH 7.4

b. pH 7.4 + 0.1 mM TPeA

c. pH 8.5 + 0.1 mM TPeA + 0.01% saponin

t = 0 1 min. 10 min.

t = 0

t = 0 1 min.

1 min.

10 min.

10 min.

0.5 min.

0.5 min.

0.25 min.

Page 131: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

130

Figure 5

0.0 0.5 1.0 1.5 2.010

100

pH 8.5

pH 7.4

Efflux duration (minutes)

Cel

l [3H

]MP

P (

% o

f t

= 0

)

Page 132: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

131

Figure 6

0 25 50 75 1000.00

0.15

0.30

0.45

B

K50 = 14.67 ± 3.45 nM(pH 7.83)

[H+] (nM)E

fflux

rat

e co

nst

ant (

k; m

in-1

)

0 1 2 3 4 510

100

A

pH 8.0

pH 7.7

pH 8.2

pH 7.0pH 7.4

pH 8.5

Efflux duration (minutes)

Cel

l [3H

]MP

P (

% o

f t =

0)

Page 133: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

132

Figure 7

0 3 6 9

1.1

1.2

1.3

pHo 7.4 pHo 8.5

pHi 7.53

pHi 7.63A

Time (minutes)

Flu

ores

cen

ce r

atio

(4

88/4

60

nm)

7.0

7.2

7.4

7.6

7.8

Intr

acel

lula

r p

H (

pHi)

0 3 6 9 12

1.2

1.3

1.4

1.5

20 mMNH4Clcontrol control

pHi 7.7

pHi 8.3B

Time (minutes)

Flu

ores

cenc

e ra

tio (

488/

460

nm)

7.5

8.0

8.5

Intr

acel

lula

r p

H (

pH

i)

Page 134: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

133

Figure 8

0 1 2 3 4 50

40

80

120

160 no preload, no NH4Cl (control)5 mM MPP preload, no NH4Clno preload, 20 mM NH4Cl during uptake5 mM MPP preload and20 mM NH4Cl during uptake

Duration (minutes)

[3 H]M

PP

(%

of

upta

ke a

t 5

min

.)

Page 135: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

134

Figure 9

0 1 2 3

10

100

pH 8.5

[NH4Cl](mM)

20

pH 7.4

10

0

no NH4Cl

Efflux duration (minutes)

Cel

l [3 H

]MP

P (

% o

f am

ount

at

t =

0)

Page 136: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

135

Figure 10

0.0 0.5 1.0 1.5 2.0

10

100

7.67.8

7.9

8.2

8.1

pHi

Duration (minutes)

Cel

l [3H

]MP

P (

% o

f t =

0)

0 10 20 300.0

0.5

1.0

1.5

2.0

2.5

IC50H+ = 11.47 nM (pH 7.94)

Intracellular [H+] (nM)

Eff

lux

rate

con

stan

t (m

in-1

)

Page 137: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

136

Supplemental Figure Legends

Figure S1. The culture dish insert. A top-view diagram of the closed bath chamber

insert (RC-37FC) with perfusion ports for 35-mm cell culture dish was modified from a

picture shown in a product brochure of Warner Instruments.

Figure S2. Efflux of [ 3H]MPP from CHO hMATE1 cells (determined in three separate

experiments using the chamber protocol described in the text). After preloading the cells

with [3H]MPP (and [14C]mannitol) at pH 8.5, the chamber was perfused with WB pH 7.4.

Total [3H]MPP and [14C]mannitol was determined in perfusate samples collected every 6

seconds. A: The amount of [3H]MPP in each perfusate sample that effluxed from the

cells (solid circles) was calculated by subtracting the amount of residual [3H]MPP from

the pre-loading solution that was in the extracellular volume (calculated from the level of

[14C]mannitol in each sample) from total [3H]MPP in each sample (open circles). B:

Amount of [3H]MPP remaining in the cells at each time point during the efflux period

(solid circles) was calculated from the total amount of [3H]MPP lost from the cells, plus

the amount of [3H]MPP left in the cells at the end of the efflux period. Here, the cellular

MPP is expressed as a percentage of total [3H]MPP (i.e., cellular plus extracellular)

present at the start of the efflux period, to provide insight into (i). the relative amounts of

the compound in each compartment; and (ii). the precision of the measurement. Only the

corrected amount of cellular [3H]MPP was used for further analysis. [Dashed lines

represented a 95% confidence interval resulted from analyzing the data with a single-

phase decay (Prism).]

Page 138: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

137

Figure S3. Intracellular pH (pH i) in CHO hMATE1 cells. CHO hMATE1 cells were

pre-loaded with BCECF-AM, and fluorescence of intracellular BCECF was measured at

room temperature as described in the Methods section. A representative result shown in

this figure demonstrated the effect of extracellular NH4Cl on pHi. The inset shows the

calibration of intracellular pH using 10 µM nigericin and standardized pH buffers

(performed after each experiment).

Page 139: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

138

Figure S1

Page 140: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

139

Figure S2

0.0 0.5 1.0 1.5 2.0 2.50

20

40

60

100

total [3H]MPPcorrected [3H]MPP

A

Efflux duration (minutes)

[3 H]M

PP

(%

eff

lux

at t

= 0

.1 m

in)

0.0 0.5 1.0 1.5 2.0 2.510

100

total

corrected

B

Efflux duration (minutes)

[3 H]M

PP

(%

am

ount

at

t =

0)

Page 141: HUMAN MULTIDRUG AND TOXIN EXTRUSION PROTEIN 1 ...Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation

140

Figure S3

0 10 20 30 40 50 60 700.0

0.5

0.8

1.0

1.2

1.4

1.65 mMNH4Cl

10 mMNH4Cl

WBpHo7.4

WBpHo7.4

WBpHo7.4

10µµµµM NigericinpH 4.3 5.0 6.0 7.47.0 8.2 9.0

Time (minutes)

Ave

rage

rat

io (

488

nm/4

60 n

m)

00

0.75 1.00 1.25 1.50

4.0

6.0

8.0

10.0

R2 = 0.98

Ratio 488 nm/460 nm

Intr

acel

lula

r pH