ArticlePDF Available

Abstract and Figures

In the mammalian kidneys, handling of various organic compounds is mediated by multispecific organic anion and cation transporters localized in the luminal and contraluminal cell membrane domains of specific nephron segments, largely in proximal tubules. These transporters are responsible for cellular uptake and/or elimination of endogenous and xenobiotic organic compounds, including various anionic and cationic drugs, thus contributing to their reabsorption and/or secretion along the nephron. Recent studies have indicated a pivotal role of these transporters in drug resistance, drug-drug interactions, and drug-induced nephrotoxicity, whereas the presence of disfunctional transporters due to truncated isoforms or point mutations can cause genetic diseases. In rat, mouse and rabbit nephrons, a number of these transporters exhibit sex differences in their protein and/or mRNA expression. In comparison with the expression in rodents and rabbits, in the human nephrons some transporters are absent, some exhibit different localization in the cell membrane domains, and none exhibit the sex-dependent expression. Species differences in some transporters have been further demonstrated concerning substrate selectivity, distribution in cells along the nephron, levels of mRNA and/or protein expression, sensitivity to inhibitors, and regulation. Overall these differences in the mammalian kidneys indicate that: a) data on the membrane transporters-related functions in one species can not simply be regarded as relevant for other species, and b) many physiological, pharmacological, and toxicological findings related to organic anion and cation transport and transporters in rodents and rabbits do not reflect the situation in humans.
Polar distribution of the common drug transporters in the rat PT cells. A. Definition of various nephron segments in relation to specific tissue zones. C, cortex; OS, outer stripe; IS, inner stripe; IM, inner medulla; PAP, papilla; UR, ureter; CG, cortical glomerulus; JMG, juxtamedullary glomerulus; PCT, proximal convoluted tubule; S3, PT straight segment; DT, distal tubule; CD, collecting duct; TALH, thick ascending limb of Henle; HL, Henle loop. B. Transmission electron micrograph of a PCT (cross section). BLM, basolateral (contraluminal) membrane; V, vacuole; BBM, brush-border (luminal, apical) membrane; M, mitochondria; N, nucleus). C. Membrane domain-specific distribution of various OA transporters (OA-Ts) in the PT cell. Slc (solute carriers) are secondary-or terciary-active transporters: sodium-dicarboxylate cotransporter NaDC3 and sodium-independent OA exchanger Oat1 and 3 are located in the BLM, whereas NaDC1, Oat2 and 5, and Oatp1 and 2 are located in the BBM. The driving force for all these transporters provides the ATP-driven (primary-active) Na/K-ATPase in the BLM, which generates ion gradients and the transmembrane (inside-negative) membrane potential. Abc (ATP-binding casette) are all ATP-driven (primary-active) transporters, located in either BLM (multidrug resistance associated proteins Mrp1, 3, 5, and 6) or BBM (Mrp2 and 4), which predominantly accept OC as substrates, and several multidrug resistance proteins from the Mdr1 (P-gp) subfamily in BBM, which predominantly accept OA, but also some OC as substrates. At the BBM, some OA from the glomerular filtrate (GF) can be reabsorbed, whereas at the BLM, most OA (drugs) are transported and accumulated in the cell, and then secreted across the BBM into the tubule luminal fluid. D. Membrane domain-specific distribution of various OC transporters (OC-Ts) in the PT cell. Slc: The OC transporters Oct1-3 in the BLM operate as facilitators that transport OC into the cell using the inside-negative membrane potential (generated by the Na/K-ATPase and ion gradients) as a driving force. Thus accumulated OC can exit the cell partially via the Abc transporters (Mrp1, 3, 5, 6) in the BLM, and predominantly via the BBM transporters, e.g., H + /OC exchangers Octn1 (organic cation novel membrane transporter) and MATE1 (multidrug and toxin extruder), Mdr1 (P-gp) proteins, and (less with) Mrp2 and 4. Some OC and the zwitterion L-carnitine can be reabsorbed from the glomerular filtrate by the action of Na + (or H + )-OC cotransporter Octn2. The non-reabsorbed and secreted OA or OC finish in urine. E-L, Representative pictures, obtained in immunocytochemical studies using specific antibodies (26–28, and our unpublished results), showing localization of a few OA and OC transporters in the PT BLM (E-I) and BBM (J-L). E, Na/K-ATPase; F, Oat1; G, Oat3; H, Oct1; I, Oct2; J, Oat2; K, Oat5; L, Mdr1.
… 
Content may be subject to copyright.
Are mice, rats, and rabbits good models for
physiological, pharmacological and toxicological
studies in humans?
Abstract
In the mammalian kidneys, handling of various organic compounds is
mediated by multispecific organic anion and cation transporters localized
in the luminal and contraluminal cell membrane domains of specific
nephron segments, largely in proximal tubules. These transporters are re-
sponsible for cellular uptake and/or elimination of endogenous and xeno-
biotic organic compounds, including various anionic and cationic drugs,
thus contributing to their reabsorption and/or secretion along the nephron.
Recent studies have indicated a pivotal role of these transporters in drug re-
sistance, drug-drug interactions, and drug-induced nephrotoxicity, whereas
the presence of disfunctional transporters due to truncated isoforms or point
mutations can cause genetic diseases. In rat, mouse and rabbit nephrons, a
number of these transporters exhibit sex differences in their protein and/or
mRNA expression. In comparison with the expression in rodents and rab-
bits, in the human nephrons some transporters are absent, some exhibit dif-
ferent localization in the cell membrane domains, and none exhibit the
sex-dependent expression. Species differences in some transporters have been
further demonstrated concerning substrate selectivity, distribution in cells
along the nephron, levels of mRNA and/or protein expression, sensitivity to
inhibitors, and regulation. Overall these differences in the mammalian kid-
neys indicate that: a) data on the membrane transporters-related functions
in one species can not simply be regarded as relevant for other species, and b)
many physiological, pharmacological, and toxicological findings related to
organic anion and cation transport and transporters in rodents and rabbits
do not reflect the situation in humans.
INTRODUCTION
Although the exact number of animals used annually for various ex-
perimental purposes worldwide is not known, it has been esti-
mated that between 40 million and 100 million of various animals are
used, most of them (~80%) being the purpose-bread rodents (mainly
mice and rats, but also hamsters, guinea pigs and gerbils) (http://en.wiki-
pedia.org/ wiki/Animal_testing). These animals are used for educa-
tional purposes, in basic research (genetics, physiology, biomedicine,
developmental biology, behavioral studies, basic pharmacology) and in
applied research (surgery, drug development, testing of various toxic
and cosmetic substances, military research). In pharmacological and
toxicological studies, especially when testing drugs, mice and rats are
the most common animals used. The 3R-alternatives (Reduction, Re-
IVAN SABOLI]
DAVORKA BRELJAK
MARIJA LJUBOJEVI]
HRVOJE BRZICA
Unit of Molecular Toxicology
Institute for Medical Research and
Occupational Health, Ksaverska cesta 2,
10001 Zagreb, Croatia
Correspondence:
Ivan Saboli}
Unit of Molecular Toxicology
Institute for Medical Research and
Occupational Health, Ksaverska cesta 2,
10001 Zagreb,Croatia
E-mail: sabolic@imi.hr
Abbreviations:
BBM – brush-border membrane
BLM – basolateral membrane
PT – proximal tubules
OA – organic anions
OC – organic cations
Key words: experimental animals,
gender differences, mammalian kidney,
nephrotoxicity, organic anions, organic
cations, proximal tubule
Received February 21, 2011.
PERIODICUM BIOLOGORUM UDC 57:61
VOL. 113, No 1, 7–16, 2011 CODEN PDBIAD
ISSN 0031-5362
Leading article
finement, Replacement), including non-animal alterna-
tives, have been widely accepted as the way to diminish
the use of experimental animals in research and testing,
but when studying interactions among cells, tissues and
organs, or when testing pharmacology and toxicology of
various substances, including drugs to be used in human
and veterinary medicine, in most cases there is no plausi-
ble substitute for the living animal. Moreover, the current
legislation requires that all new drugs, before being li-
cenced for human and animal use, have to be rigorously
tested in at least two mammalian species (rodents AND
non-rodents) for metabolism, pharmacokinetics, acute
and chronic toxicology in adult species, efficasy regard-
ing the expected actions, effects on reproduction, embry-
onic toxicity, and potential carcinogenicity. However, the
increasing evidence indicate that rodents and some other
common experimental animals, such as rabbits, may not
be good models for such studies relevant to humans due
to sex and species differences in various properties. Par-
ticularly relevant to this problem are the data on the role
and expression of various membrane transporters that
mediate transport of organic anions (OA) and cations
(OC) in the mammalian kidneys and other organs. In re-
cent years, a number of these transporters from different
protein families and species have been cloned and char-
acterized, and their localization, mainly in the liver and
kidneys, but also in the intestine, brain and placenta,
have been studied. Their kinetic and functional charac-
teristics, sex and species differences in their expression,
and their relevance for drug transport, drug-drug inter-
actions and drug toxicity, have been extensively studied
in normal and specific gene-inactivated (knock-out) ani-
mals, as well as in humans, and discussed in numerous
recent reviewes (1–22).
ORGANIC ANION AND CATION
TRANSPORTERS IN THE MAMMALIAN
KIDNEY
Humans and animals are constantly exposed to vari-
ous organic compounds that are either produced during
normal metabolism (endogenous substances) or enter
the body via food, air, or medication (exogenous com-
pounds, xenobiotics), and are potentially harmful to their
health. In the body, these compounds are either biotrans-
formed into more or less active metabolites, largely in
liver and kidneys, or remain unchanged. Dependent on
their physico-chemical characteristics, at the physiologi-
cal pH these organic compounds behave as OA (nega-
tively-charged) or OC (positively-charged), whereas so-
me compounds may be both (zwitter-ions). The unchan-
ged or biotransformed organic compounds are elimi-
nated from the body partially by the liver and largely by
the kidneys via secretory processes that are mediated by
various transporters localized in the cell membrane. The
representative groups of endogenous and xenobiotic OA
and OC, which are handled by the mammalian liver and
kidneys, are listed in Table 1. The cell membrane is not
freely permeable to these compounds; they are trans-
ported by various specialized proteins localized in the
membrane, collectively named as »drug transporters«.
Detailed lists of these compounds associated with rel-
evant transporters in the animal and human kidneys
have been reviewed elsewhere (1, 2, 4, 5, 8, 10, 11, 13, 16,
17, 22).
In the mammalian kidneys, transport of OA and OC
is maintained by numerous, largely multispecific trans-
porters that are localized in the apical (luminal) and/or
basolateral membrane domains in the epithelial cells
along the nephron. To drive vectorial transport of their
substrates in direction of secretion or reabsorption, most
OA transporters operate as bidirectional anion exchan-
gers, and use transmembrane ion gradients (secondary-
or tertiary-active transporters) generated by the activity
of primary-active Na/K-ATPase and/or secondary-active
ion exchangers (for example, Na+/H+antiporter), whe-
reas most OC transporters operate as the bidirectional fa-
cilitators, while some OA and OC transporters use ATP
(primary-active transporters). The characteristics and the
nomenclature of all these transporters have been recently
collected and published; most of them belong to the large
family of solute carriers (Slc in animals/SLC in hu-
mans), whereas the ATP-driven transporters are mem-
8Period biol, Vol 113, No 1, 2011.
I. Saboli} et al. Sex and species differences in renal drug transporters
TABLE 1
Representative endogenous and exogenous (xenobiotic)
OA and OC that are handled by various membrane
transporters in the mammalian kidneys.
Organic anions
Endogenous: cyclic nucleotides, dicarboxylates, urate, folate,
some prostaglandins, neurotransmitter metabolites, steroid
hormones conjugated with sulfate, cysteine, glycine, and glu-
curonide.
Exogenous (xenobiotics): Medicaments (antibiotics, anti-vi-
ral drugs, nonsteroid anti-inflammatory drugs, diuretics, an-
giotensin-converting enzyme inhibitors, angiotensin II an-
tagonists, anti-neoplastics, anti-epileptics, histamine-H2-re-
ceptor antagonists); Conjugates (steroid hormones conju-
gated with sulfate, cysteine, glycine, and glucuronide); Diag-
nostic and experimental drugs (p-aminohippuric acid (PAH));
Food constituents (flavonoids); Environmental toxins (my-
cotoxins, herbicides, pesticides, some toxic metals).
Organic cations
Endogenous: choline, corticosterone, progesterone, endoge-
nous cardiac glycosides, bioactive monoamines (dopamine,
histamine, epinephrin, norepinephrin, serotonin), some pro-
staglandins.
Exogenous (xenobiotics): Medicaments ((ant)agonists of va-
rious receptors, ion channel blockers, transporter blockers,
psychoactive drugs, some antiviral drugs, antidiabetics, ane-
stetics, antimalarics, muscle relaxants, cardiac glycosides);
Toxins and experimental drugs (tetraethylammonium (TEA),
tetramethylammonium (TMA), nicotine).
Zwitter ion – L-carnitine
Detailed lists of these compounds associated with relevant
transporters in the animal and human kidneys have been re-
viewed elsewhere (1, 2, 4, 5, 8, 10, 11, 13, 16, 17, 22).
Period biol, Vol 113, No 1, 2011. 9
Sex and species differences in renal drug transporters I. Saboli} et al.
Figure 1. Polar distribution of the common drug transporters in the rat PT cells. A. Definition of various nephron segments in relation to specific tissue
zones. C, cortex; OS, outer stripe; IS, inner stripe; IM, inner medulla; PAP, papilla; UR, ureter; CG, cortical glomerulus; JMG, juxtamedullary
glomerulus; PCT, proximal convoluted tubule; S3, PT straight segment; DT, distal tubule; CD, collecting duct; TALH, thick ascending limb of
Henle; HL, Henle loop. B. Transmission electron micrograph of a PCT (cross section). BLM, basolateral (contraluminal) membrane; V, vacuole;
BBM, brush-border (luminal, apical) membrane; M, mitochondria; N, nucleus). C. Membrane domain-specific distribution of various OA trans-
porters (OA-Ts) in the PT cell. Slc (solute carriers) are secondary- or terciary-active transporters: sodium-dicarboxylate cotransporter NaDC3 and so-
dium-independent OA exchanger Oat1 and 3 are located in the BLM, whereas NaDC1, Oat2 and 5, and Oatp1 and 2 are located in the BBM. The
driving force for all these transporters provides the ATP-driven (primary-active) Na/K-ATPase in the BLM, which generates ion gradients and the
transmembrane (inside-negative) membrane potential. Abc (ATP-binding casette) are all ATP-driven (primary-active) transporters, located in ei-
ther BLM (multidrug resistance associated proteins Mrp1, 3, 5, and 6) or BBM (Mrp2 and 4), which predominantly accept OC as substrates, and sev-
eral multidrug resistance proteins from the Mdr1 (P-gp) subfamily in BBM, which predominantly accept OA, but also some OC as substrates. At the
BBM, some OA from the glomerular filtrate (GF) can be reabsorbed, whereas at the BLM, most OA (drugs) are transported and accumulated in the
cell, and then secreted across the BBM into the tubule luminal fluid. D. Membrane domain-specific distribution of various OC transporters (OC-Ts)
in the PT cell. Slc: The OC transporters Oct1-3 in the BLM operate as facilitators that transport OC into the cell using the inside-negative membrane
potential (generated by the Na/K-ATPase and ion gradients) as a driving force. Thus accumulated OC can exit the cell partially via the Abc trans-
porters (Mrp1, 3, 5, 6) in the BLM, and predominantly via the BBM transporters, e.g., H+/OC exchangers Octn1 (organic cation novel membrane
transporter) and MATE1 (multidrug and toxin extruder), Mdr1 (P-gp) proteins, and (less with) Mrp2 and 4. Some OC and the zwitterion
L-carnitine can be reabsorbed from the glomerular filtrate by the action of Na+(or H+)-OC cotransporter Octn2. The non-reabsorbed and secreted
OA or OC finish in urine. E-L, Representative pictures, obtained in immunocytochemical studies using specific antibodies (26–28, and our unpub-
lished results), showing localization of a few OA and OC transporters in the PT BLM (E-I) and BBM (J-L). E, Na/K-ATPase; F, Oat1; G, Oat3; H,
Oct1; I, Oct2; J, Oat2; K, Oat5; L, Mdr1.
bers of the ATP-binding casette (Abc in animals/ABC in
humans) family of transporters (23, 24). The number of
newly discovered, cloned, and characterized transporters
from both families increases every year. In the nephron,
these transporters mediate: a) transport (net secretion) of
various endogenous organic compounds that are gener-
ated in normal metabolism in the kidneys and other or-
gans, b) transport (net secretion) of exogenous (xenobio-
tic) organic compounds that enter the body for iatrogen
(medicaments/drugs) or alimentary (food constituents)
reasons, or as the enviromental toxins, c) drug-drug in-
teractions, d) drug resistance, e) development of drug-
-induced nephrotoxicity, and f) specific genetic diseases,
caused by malfunctional transporters due to truncated
isoforms or point mutations. The convoluted (S1/S2)
and straight (S3) proximal tubule (PT) segments are the
principal nephron parts in which handling of OA and
OC takes place. In the PT epithelial cells, the relevant
transporters are differently distributed in the luminal
(brush-border, BBM) and contraluminal (basolateral,
BLM) membrane domains. A polar distribution of some,
well characterized transporters of OA and OC in the rat
PT cells is illustrated in Fig. 1C (OA transporters) and
Fig. 1D (OC transporters).
SEX AND SPECIES DIFFERENCES
IN THE EXPRESSION OF RENAL
ORGANIC ANION TRANSPORTERS
Majority of the cloned and well-characterized OA
transporters belong to the subfamily Slc22/SLC22 (Oat1/
OAT1, Oat2/OAT2, Oat3/OAT3, OAT4, Oat5/OAT5,
etc...), which operate as anion exchangers (23), whereas
the ATP-driven efflux pumps Mrp1, 3, 5, and 6, and
Mrp2 and 4 belong to the subfamily Abcc/ABCC (24).
Recent studies have shown that in the rodent and rabbit
kidneys, some members of both transporter families ex-
hibit species and/or sex differences in the expression of
mRNA and/or protein, and in their localization along
the nephron, and that these differences influence secre-
tory functions of the organ. These differences are related
to the sex hormone-regulated expression of specific trans-
porters in one of the membrane domains of (largely) PT
cells. From the available data for a limited number of re-
nal OA transporters in rats, mice, rabbits, and humans,
collected and shown in Table 2, one can conclude the fol-
lowing: (a) Oat1/OAT1 is always expressed in the PT
BLM, but the male (M)-dominant sex differences in its
expression are present in rats and mice, but not in rabbits
and humans, (b) Oat2/OAT2 in rats and mice is locali-
zed to the PT BBM, where it exhibits the female (F)-do-
minant sex differences; its localization in rabbits is not
known (at the level of mRNA, M = F), whereas in hu-
mans, this transporter is localized to the PT BLM and
exhibits no sex differences, (c) Oat3/OAT3 in rodents
and humans is localized to the PT BLM (in rabbits, the
PT membrane domain has not been defined), but the ex-
pression is M-dominant in rats, F-dominant in mice, and
sex-independent in rabbits and humans, (d) OAT4 is the
human-specific transporter (not detected in rodents and
rabbits) in the PT BBM, similarly expressed in M and F,
(e) Oat5 is the rodent-specific transporter (not present in
humans) in the PT BBM (and in intracellular organelles
in mice), with F-dominant expression in both rats and
10 Period biol, Vol 113, No 1, 2011.
I. Saboli} et al. Sex and species differences in renal drug transporters
TABLE 2
Sex and species differences in the expression of several OA transporters in the mammalian kidneys.
Species and sex differences, localization in the nephron segment, membrane domain
Family Transporter Rats Mice Rabbits Humansa
Slc/SLC Oat1/OAT1 M>F M>F M=F M=F
PT, BLM PT, BLM PT,? PT, BLM
Oat2/OAT2 M<F M<F M=F
bM=F
PT, BBM PT, BBM ? PT, BLM
Oat3/OAT3 M>F M<F
bM=F M=F
PT, BLM PT, BLM PT,? PT, BLM
OAT4 ND ND ND M = F
PT, BBM
Oat5 M<F M<F ? ND
PT, BBM PT, BBM, IO ? ?
Abc/ABC Mrp4 M>F M<F ? ?
PT, BBM PT, BBM
Species-specific sex differences in the expression of OA transporters were observed at the level of protein and/or mRNA. For Oat1, Oat2,
Oat3, and Oat5 expression in experimental animals, sex differences are determined by either stimulatory effects of androgens or inhibi-
tory effects of estrogens and progesterone, or both, and are absent in prepubertal animals. Most data have been collected from various
publications (15, 26–29). aOur unpublished data. bData from (29, 30), and our unpublished data. M, males; F, females; PT, proximal tu-
bules; BBM, brush-border (luminal) membrane; BLM, basolateral (contraluminal) membrane; IO, intracellular organelles; ND, not de-
tected in the species; ?, unknown.
mice, and (f) the expression of Mrp4 in the PT BBM is
M-dominant in rats and F-dominant in mice; the data
for rabbits and humans are not available. A number of
other renal OATs in adult rats, mice, and in few other
species, also exhibit sex differences in their expression at
the level of protein and/or mRNA, whereas in prepube-
rtal animals, the expression of thus far tested OA trans-
porters was low and sex-independent (15, 25).
As has been tested in rats, mice and rabbits, the urine
excretion of relevant OA correlates well with the renal
expression of Oats. Thus, sex differences in the renal ex-
pression of Oat1, Oat2, and Oat3 in adult rats clearly cor-
relate with similar differences in the urine excretion of
their substrates (Table 3), whereas in the adult rabbits, in
which the expression of these Oats is similar in both
sexes, the excretion of relevant OA is also similar (30).
Furthermore, the prepubertal M and F rats excrete
OA with similar and much lower rate than the adults,
which is in good correlation with the sex-independent
and much lower expression of renal Oats (for references
see (15)). In addition, in mice with the knocked-out Oat1
and Oat3 genes, the urine excretion of p-aminohippurate
(PAH) and a few other OA is strongly impaired (29, 31,
32). Since the localization and the level of protein expres-
sion in the cell membrane is one of the major determi-
nants (next to the substrate specificity and kinetic charac-
teristics) of the transporter function, the observations in
humans that: a) none of the indicated transporters shows
sex differences in the expression, b) OAT2 is localized in
the membrane domain (BLM) which is opposite from
that in rodents (BBM), and c) OAT4 is present only in
humans, whereas Oat5 is rodent-specific, indicate that
the renal handling of OA with the all possible conse-
quences, such as interactions and nephrotoxicity of an-
ionic drugs, may be in humans different from that in ro-
dents and rabbits. Moreover, different sex-related expres-
sion of Oat3 in rats, mice, and rabbits, and of Mrp4 in rats
and mice, indicates that the overall handling and secre-
tion of many OA may be different among these species.
The respective transport studies in humans are scarce;
only a few of them have shown that the renal clearance of
some drugs and/or their metabolites may be sex-related,
but these differences may rather reflect the sex-depend-
ent metabolism of anionic (and other) drugs catalized by
drug-metabolizing enzymes, not the transporters-medi-
ated active secretion in the renal tubules (3, 15).
SEX AND SPECIES DIFFERENCES
IN THE EXPRESSION OF RENAL
ORGANIC CATION TRANSPORTERS
A number of OC transporters from different protein
families and species have been cloned and characterized,
and their functional roles have been studied mainly in
the liver and kidneys (8). The most important OC trans-
porters are grupped into the families Slc22/SLC22 (Oct1/
OCT1, Oct2/OCT2, Oct3/OCT3, Octn1/OCTN1, Octn2/
OCTN2), Slc47/SLC47 (MATE1, MATE2, MATE2-
-K), and Abcb/ABCB (Mdr1/MDR1 (P-glycoprotein, P-gp)).
Oct1-3/OCT1-3 represent polyspecific bidirectional trans-
porters that mediate electrogenic, sodium- and pH-inde-
pendent intracellular uptake of OC via facilitated diffu-
sion. In the rat PT, these transporter are predominantly
localized to the BLM, where they mediate the first step of
the renal OC excretion. The second, exit step across the
BBM is largely mediated by the electroneutral H+-OC
exchangers MATE1 (in rodents, rabbits and humans),
MATE2 (in mice, not known for rats, not present in rab-
bits and humans), and MATE2-K (in rabbits and hu-
mans, not present in rats and mice), and by the ATP-
-driven efflux pump Mdr1/MDR1. Besides in expression
and localization, OC transporters in various species dif-
fer in substrate specificity, inhibitor sensitivity, transport
mechanism, and regulation (8, 33).
The renal OC transporters have been extensively stu-
died recent years, but most of this research showed their
expression at the level of mRNA, whereas only a few
transporters were described also at the protein level. As
listed in Table 4, the mRNA expression of various renal
OC transporters in different species exhibits different
pattern of sex dependency. Thus, in the rat, mouse, rabbit
and human kidneys: (a) mRNA expression of various
OC transporters is species-dependent, but sex differences
are present only in some cases, (b) expression of both
mRNA and protein have been thus far studied and corre-
lated only in a few cases, (c) in the Oct/OCT subfamily,
rats and mice express the Oct1 mRNA in similar abun-
dance in M and F, whereas the protein is localized to the
proximal tubule BLM with (largely) M-dominant ex-
pression. However, rabbits do not express the Oct1 mRNA
and protein at all, whereas in humans, OCT1 is localized
to the apical membrane of proximal and distal tubules
with similar expression in M and F. (d) The expression of
renal Oct2/OCT2 in different species has been studied in
more detail at both mRNA and protein level. The expres-
sion of Oct2 mRNA in rats, mice and rabbits exhibits the
M-dominant pattern, but at the protein level this ba-
solateral transporter in PT is clearly stronger in M than F
rats and mice, but sex-independent in rabbits and hu-
mans. (e) In the Octn/OCTN subfamily, the mRNA ex-
pression of the indicated transporters is either sex-inde-
pendent or still controversial, whereas the Octn2 protein
in rats and mice is localized to the PT BBM. (f) In the
Period biol, Vol 113, No 1, 2011. 11
Sex and species differences in renal drug transporters I. Saboli} et al.
TABLE 3
In rats, sex differences in the expression of some renal
OA transporters correlate well with the excretion of rele-
vant OA in urine.
Oat Protein
Expression
Organic anion Urine excretion
Oat1 M > F PAH, Furosemide M > F
Oat2 M < F PFOA M < F
Oat3 M > F PAH, Taurocholate M > F
Data have been collected from the available literature and
from own publications (for references see (15)).M, males; F,
females; PAH, p-aminohippurate; PFOA, perfluorooctanoic
acid.
12 Period biol, Vol 113, No 1, 2011.
I. Saboli} et al. Sex and species differences in renal drug transporters
TABLE 4
Sex and species differences in the renal expression of OC transporters at the level of mRNA and/or protein.
OC Transporter Species mRNA M vs. F Protein (Nephron segment,
Membrane domain)
M vs. F
Oct1/OCT1 Rat M £F PT, BLM M ³F
Mouse M = F PT, BLM M > F
Rabbit ND ND ND
Human +/? PT & DT, AM M = F
Oct2/OCT2 Rat M > F PT, BLM M > F
Mouse M > F PT, BLM M > F
Rabbit M > F PT,? M = F
Human +/? PT, BLM M = F
Oct3/OCT3 Rat M = F ?
Mouse M = F ?
Rabbit M = F ?
Human +/? ?
Octn1/OCTN1 Rat M = F ?
Mouse M = F ?
Rabbit +/? ?
Human +/? ?
Octn2/OCTN2 Rat M ³F PT, BBM
Mouse M = F PT, BBM
Rabbit +/? ?
Human +/? ?
MATE1 Rat M > F PT, BBM M > F
Mouse M > F Various segments, AM ?
Rabbit M = F ?
Human +/? PT, BBM ?
MATE2 Rat +/? ?
Mouse M = F
Rabbit ND ND
Human ND ND
MATE2-K Rat ND ND
Mouse ND ND
Rabbit M = F ?
Human +/? PT, BBM ?
Mdr1a Rat M > F ?
Mouse M £F?
Mdr1b Rat +/? ?
Mouse M < F ?
Mdr2 Rat +/? ?
Mouse M = F ?
MDR1 Human +/? ?
Data have been collected from the previously reviewed literature (15), from other studies (34, 36–49), and from own unpublished stud-
ies. The mRNA expression was determined in the kidney cortex or whole kidney tissue, whereas protein expression was determined by
immunocytochemistry in tissue cryosections and/or by Western blotting in cell membranes isolated from various kidney zones. F, fe-
males; M, males; ND, not detected; +/?, mRNA detected, but sex-dependency unknown; ?, unknown data; PT, proximal tubules;
BLM, basolateral membrane; DT, distal tubules; AM, apical membrane.
MATE subfamily, MATE1 in the rat kidney is localized
to the PT BBM and M-dominant in both mRNA and
protein expression. Mice exhibit higher expression of
mRNA in M, but the protein was detected in the apical
membrane of various tubule segments with unknown
levels of expresssion in M and F. MATE1 is present also
in the rabbit and human kidneys, but in these species the
levels of MATE1 mRNA and protein expression in M
and F has been poorly investigated. However, clear spe-
cies differences exist in the expression of MATE2, which
is present in mice (not clear in rats), but not in rabbits
and humans, and MATE2-K, which is present in rabbits
and humans, but not in rodents. (g) In the Mdr/MDR
subfamily, the Mdr1a mRNA expression in the rat kid-
neys isM>F,whereas in mice, the mRNA expression of
this transporters may be just opposite, e.g., M < F. Whe-
re tested, sex hormones responsible for the observed sex
differences in the expression of mRNA and/or protein of
the specific OC transporters have been defined, whereas
in prepubertal animals, the expression of both parame-
ters is low and sex-independent (reviewed in (15)).
Several in vivo studies in variously-treated rats and
mice, and/or in vitro studies in tissue slices or isolated re-
nal BLM vesicles from the same animals, have correlated
the protein expression of some OC transporters and the
rates of OC secretion (34, reviewed in (15)). The data
have shown that: (a) M rats and mice exhibit higher rate
of OC tetraethylammonium (TEA) clearance than the F
animals, (b) in rodents, tissue slices from the M kidneys
exhibit higher accumulation of TEA than the slices from
the F kidneys, (c) in BLM vesicles isolated from the rat
kidneys, TEA uptake in the vesicles from M kidneys is
greater than in the vesicles from F kidneys, (d) gonade-
ctomy of M rodents downregulates, the treatment of
these animals with testosterone strongly upregulates, whe-
reas the treatment with estradiol slightly downregulates
the renal expression of Oct2 protein and the accumula-
tion of TEA in kidney tissue slices. These data thus indi-
cate the major role of androgens in regulating the Oct2-
-mediated OC secretion in rodents. An important role of
this transporter for OC secretion in the mouse kidneys
can be further demonstrated in the animals defficient
(knock-out) in Oct1 and Oct2, in which the renal secre-
Period biol, Vol 113, No 1, 2011. 13
Sex and species differences in renal drug transporters I. Saboli} et al.
TABLE 5
Species differences in the rates of OA transport, affinity for OC substrates, inhibitory constant of OC transport, and inhibition
of an OC transport with other OC in various experimental models.
Parameter Experimental model Species differences
Substrate specificity:
Quinidine transport Oct1/OCT1-transfected XO rat –, human +
Na+-L-carnitine cotransport Octn1/OCTN1-transfected cells rat –, mouse +, human +
Relative rate of transport:
TEA uptake Octn2/OCTN2-transfected cells rat > mouse > human
Na+-L-carnitine cotransport Octn2/OCTN2-transfected cells rat < mouse < human
Kmvalue for:
Choline Oct2/OCT2-transfected XO rat > human
TEA (Octn1-mediated) Renal cortical BBMV rat > rabbit
Ki value for:
Inhibition of MPP uptake by n-TAA Oct1/OCT1-transfected XO rat, mouse, rabbit < human
Inhibition of TEA transport by various OC Oct1/OCT1-transfected cells rat < human
Oct2/OCT2-transfected cells rat > human
IC50 value for:
Inhibition of TEA uptake by TBA, TPA, cimetidine guanidine
and famotidine
Oct2/OCT2-transfected cells rabbit < human
Inhibition of TEA uptake by tyramine, carbachol and choline Oct2/OCT2-transfected cells rabbit > human
Inhibition of OC transport with other OC:
Inhibition of L-carnitine transport by TEA and choline Octn2/OCTN2-transfected cells rat > human
Inhibition of TEA transport by cimetidine and rhodamine 123 MATE1-transfected cells mouse > human
Inhibition of TEA transport by quinidine, verapamil, nicotine,
corticosterone, testosterone and quercetin
MATE1-transfected cells mouse < human
Data have been collected from the literature (7, 42, 50–62). XO, Xenopus oocytes; Transfected cells, various kinds of cultured cells
transfected with the indicated OC transporters; BBMV, isolated brush-border membrane vesicles; (-) Absence or (+) presence of trans-
port. Km, Michaelis constant. MPP, 1-methyl-4-phenylpyridinium; n-TAA, n-tetraalkylammonium compounds; TEA, tetraethyl-
ammonium; TBA, tetrabutylammonium.
tion of OA is nearly abolished (35). However, the studies
in isolated PT from the M and F rabbit kidneys have
shown similar uptake of TEA in both sexes, which com-
pares to similar and sex-independent expression of Oct2
protein in their kidneys (30).
Species differences in some OC transporters have also
been demonstrated concerning substrate selectivity and
transport rates, sensitivity to inhibitors, and regulation. A
comparison of kinetic characteristics of various OC trans-
porters from different species have been tested largely
following their expression in Xenopus oocytes and cul-
tured cells. The data collected from the current literature
clearly indicate species differences in many characteris-
tics among the comparable OC transporters in rats, mice,
rabbits and humans. As listed in Table 5, the related OC
transporters in rodents, rabbits and humans differ in sub-
strate specificity, relative transport rate with specific sub-
strates, affinity (Km) for specific substrates, and inhibi-
tion of the transporter activity with various substrates (Ki
and IC50 values, inhibition of the OC transport with
other OCs). Overall, these data indicate that the OC
transporters from each subfamily exhibit species differ-
ences in a variety of kinetic characteristics that may result
in different, species-specific functional performance in
the mammalian kidneys and other organs.
Various aspects of long-term (developmental, hormo-
nal and nutritional regulation, regulation under patolo-
gical conditions) and short-term regulation of OC trans-
port and expression and/or activity of various OC trans-
porters in the mammalian kidneys in vivo and in various
experimental models in vitro, have been recently revie-
wed (33). A set of information that point to species differ-
ences in short-term regulation of the activity of some of
these transporters has been collected from the literature
and summarized in Table 6. Most of these information
have been generated in the cell lines transfected with the
defined animal or human OC transporters, and in some
cases the regulation of their activity (transporter-medi-
ated uptake of an OC) in the cells could be correlated
with the relevant transport in PT segments isolated from
the same species. Thus (Table 6), (a) in the cells trans-
fected with the rat Oct1, activation of protein kinases A
(PKA) and C (PKC) resulted in upregulation of the OC
transport, whereas in the same cell line transfected with
the human OCT1, activation of these enzymes caused
downregulation of the OC transport (in one study the ef-
fect was not observed), (b) in the cells transfected with
the rabbit Oct2, activation of PKA activity caused upre-
gulation of the OC uptake, and the same effect was pres-
ent in the isolated rabbit PT segments, whereas (c) in the
cells transfected with the human OCT2, and in isolated
human PT segments, the activation of both kinases down-
regulated the OC transport. These experiments in vitro
indicate that the short-term regulation of OC transporter
activity in the mammalian kidneys may be species-spe-
cific also in vivo.
CONCLUSION
In rodents, various renal transporters of OA and OC
exhibit sex differences in their protein (and mRNA) ex-
pression and functional characteristics. This phenome-
non may be relevant during life in these animals in con-
ditions that are associated with significant changes in
blood levels of sex hormones (female hormonal cycle/
oestrus in rodents, pregnancy, ageing). When compared
in rats, mice, rabbits and humans, some renal OA and
OC transporters also exhibit species differences in their
presence, expression level, sex-dependence, membrane
domain-specific localization in the cells, various kinetic
characteristics, and regulation. Humans exhibit no sex
differences in the expression of thus far tested OA and
OC transporters, and other characteristics related to the-
14 Period biol, Vol 113, No 1, 2011.
I. Saboli} et al. Sex and species differences in renal drug transporters
TABLE 6
Species differences in short term regulation of the mammalian OC transporters.
Species & Experimental model Transport Effector Effect on transport
Rat
Oct1-transfected HEK-293 cells ASP+uptake PKA activation Increase
PKC activation Increase
Rabbit
Oct2-transfected CHO-K1 cells TEA uptake PKA activation Increase
Isolated PT segments TEA uptake PKA activation Increase
Human
OCT1-transfected HEK-293 cells Amiloride & PKA activation Decrease
ASP+uptake PKC activation Decrease/NE
OCT2-transfected HEK-293 cells Amiloride & PKA activation Decrease
ASP+uptake PKC activation Decrease/NE
Isolated PT segments ASP+uptake PKC activation Decrease
Data have been collected from the literature (33, 63–69). ASP+, fluorescent cationic dye 4-(4-dimethylamino)styryl-N-methylpy-
ridinium; TEA, tetraetylammonium; PT, proximal tubules; PKA, protein kinase A; PKC, protein kinase C; NE, no effect.
se transporters are in many respects different from those
in rodents and rabbits. Although in humans sex differ-
ences in pharmacokinetics have been identified for some
drugs, they are small, and their clinical relevance is mi-
nor (70). Therefore, (a) data on OA and OC transport
and transporters in one species can not simply be re-
garded as relevant for other species, and (b) physiologi-
cal, pharmacological and toxicological data in rats, mice,
and rabbits, that are related to the functions of these
transporters in the kidneys, may not be relevant to the sit-
uation in humans.
Acknowledgement: This work was supported by grant
No. 022-0222148-2146 (Mammalian Renal Transporters;
Gender Differences and Effects of Toxic Metals) from the
Ministry of Science, Education and Sports, Republic of
Croatia (I.S.).
REFERENCES
1. ANZAI N, KANAI Y, ENDOU H 2006 Organic anion transporter
family: Current knowledge. J Pharmacol Sci 100: 411–426
2. EL-SHEIKHAAK,MASEREEUW R, RUSSELFGM2008
Mechanisms of renal anionic drug transport. Eur J Pharmacol 585:
245–255
3. FRANCONI F, BRUNELLESCI S, STEARDO L, CUOMO V
2007 Gender differences in drug responses. Pharmacol Res 55: 81–95
4. GIACOMINI K M, HUANG S M, TWEEDIE D J, BENET L Z,
BROUWER K L, CHU X, DAHLIN A, EVERS R, FISCHER V,
HILLGREN K M, HOFFMASTER K A, ISHIKAWA T, KEP-
PLER D, KIM R B, LEE C A, NIEMI M, POLLI J W, SUGIYAMA
Y, SWAAN P W, WARE J A, WRIGHT S H, YEE S W, ZAMEK-
-GLISZCZYNSKI M J, ZHANG L 2010 Membrane transporters
and drug development. Nat Rev Drug Discov 9: 215–236
5. HO R H, KIM R B 2005 Transporters and drug therapy: Implica-
tions for drug disposition and disease. Clin Pharmacol Ther 78:
260–277
6. KOEPSELL H 2004 Polyspecific organic cation transporters: their
functions and interactions with drugs. Trends Pharmacol Sci 25:
375–381
7. KOEPSELL H, GORBOULEV V, ARNDT P 1999 Molecular phar-
macology of organic cation transporters in kidney. J Membrane Biol
167: 103–117
8. KOEPSELL H, SCHMITT B M, GORBOULEV V 2003 Organic
cation transporters. Rev Physiol Biochem Pharmacol 150: 1–35
9. LAUNAY-VACHER V, IZZEDINE H, KARIE S, HULOT JS,
BAUMELOU A, DERAY G 2006 Renal tubular drug transporters.
Nephron Physiol 103: 97–p106
10. LEE W, KIM R B 2004 Transporters and renal drug elimination.
Ann Rev Pharmacol Toxicol 44: 137–166
11. LI M, ANDERSON G D, WANG J 2006 Drug-drug interactions
involving membrane transporters in the human kidney. Expert Opin
Drug Metab Toxicol 2: 505–532
12. MORRIS M E, LEE H J, PREDKO L M 2003 Gender differences in
the membrane transport of endogenous and exogenous compounds.
Pharmacol Rev 55: 229–240
13. RIZWAN A N, BURCKHARDT G 2007 Organic anion transport-
ers of the SLC22 family: biopharmaceutical, physiological, and pa-
thological roles. Pharmaceut Res 24: 450–470
14. ROBERTSON E E, RANKIN G O 2006 Human renal organic an-
ion transporters: Characteristics and contributions to drug and drug
metabolite excretion. Pharmacol Ther 109: 399–412
15. SABOLIC I, ASIF A R, BUDACH W E, WANKE C, BAHN A,
BURCKHARDT G 2007 Gender differences in kidney function.
Pfluegers Arch Eur J Physiol 455: 397–429
16. SEKINE T, CHA S H, ENDOU H 2000 The multispecific organic
anion transporter (OAT) family. Pfluegers Arch Eur J Physiol 440:
337–350
17. SEKINE T, MIYAZAKIH, ENDOU H 2006 Molecular physiology
or renal organic anion transporters. Am J Physiol Renal Physiol 290:
F251–F261
18. SRIMAROENG C, PERRY L J, PRITCHARD J B 2008 Physiology,
structure, and regulation of the cloned organic anion transporters.
Xenobiotica 38: 889–935
19. SWEET D H 2005 Organic anion transporter (Slc22a) family mem-
bers as mediators of toxicity. Toxicol Appl Pharmacol 204: 198–215
20. TERLOW S A, MASEREEUW R, RUSSELFGM2003 Modu-
latory effects of hormones, drugs, and toxic events on renal organic
anion transport. Biochem Pharmacol 65: 1393–1405
21. VANAUBELRAMH,MASEREEUW R, RUSSEL F G M 2000
Molecular pharmacology of renal organic anion transporters. Am J
Physiol Renal Physiol 279: F216–F232
22. WRIGHT S H 2005 Role of organic cation transporters in the renal
handling of therapeutic agents and xenobiotics. Toxicol Appl Phar-
macol 204: 309–319
23. HEDIGER M A (Guest Editor) 2004 Special issue: The ABCs of
solute carriers: physiological, pathological, and therapeutic implica-
tions of human membrane transport proteins. Pflügers Arch Eur J
Physiol 447 (5)
24. STIEGER B, HIGGINS C F (Guest Editors) 2007 Special issue:
Twenty years of ABC transporters. Pflügers Arch Eur J Physiol 453 (5)
25. BRZICA H, BRELJAK D, KRICK W, LOVRIC M, BURCK-
HARDT, G, BURCKHARDT B C, SABOLIC I 2009 The liver and
kidney expression of sulfate anion transporter sat-1 in rats exhibits
male-dominant gender differences. Pflügers Arch Eur J Physiol 457:
1381–1392
26. BRELJAK D, LJUBOJEVIC M, BALEN D, ZLENDER V, BRZI-
CA H, MICEK V, KUSAN M, ANZAI N, SABOLIC I 2010 Renal
expression of organic anion transporter Oat5 in rats and mice exhib-
its the female-dominant sex differences. Histol Histopathol 25: 1385–1402
27. LJUBOJEVIC M, BALEN D, BRELJAK D, KUSAN M, ANZAI
N, BAHN A, BURCKHARDT G, SABOLIC I 2007 Renal expres-
sion of organic anion transporter OAT2 in rats and mice is regulated
by sex hormones. Am J Physiol Renal Physiol 292: F361–F372
28. LJUBOJEVIC M, HERAK-KRAMBERGER C M, HAGOS Y,
BAHN A, ENDOU H, BURCKHARDT G, SABOLIC I 2004 Rat
renal cortical OAT1 and OAT3 exhibit gender differences deter-
mined by both androgen stimulation and estrogen inhibition. Am J
Physiol Renal Physiol 287: F124–F138
29. VAN WERT A L, BAILEY R M, SWEET D H 2007 Organic anion
transporter 3 (Oat3/Slc22a8) knockout mice exhibit altered clear-
ance and distribution of penicillin G. Am J Physiol Renal Physiol 293:
F1332–F1341
30. GROVES C E, SUHRE W B, CHERRINGTON N J, WRIGHT S
H 2006 Sex differences in the mRNA, protein, and functional ex-
pression of organic anion transporter (Oat) 1, OAT3, and organic
cation transporter (OCT) 2 in rabbit renal proximal tubules. J Phar-
macol Exp Therap 316: 743–752
31. ERALY S A, VALLON V, VAUGHAN D A, GANGOITI J A,
RICHTER K, NAGLE M, MONTE J C, RIEG T, TRUONG D M,
LONG J M, BARSHOP B A, KALER G, NIGAM S K 2006 De-
creased renal organic anion secretion and plasma accumulation of
endogenous organic anions in OAT1 knock-out mice. J Biol Chem
281: 5072–5083
32. SWEET D H, MILLER D S, PRITCHARD J B, FUJIWARA Y,
BEIER D R, NIGAM S K 2002 Impaired organic anion transport in
kidney and choroid plexus of organic anion transporter 3 (Oat3
(Slc22a8)) knockout mice. J Biol Chem 277: 26934–26943
33. CIARIMBOLI G, SCHLATTER E 2005 Regulation of organic cat-
ion transport. Pflugers Arch Eur J Physiol 449: 423–441
34. MEETAM P, SRIMAROENG C, SOODVILAI S, CHATSUD-
THIPONG V 2009 Regulatory role of testosterone in organic cation
transport: in vivo and in vitro studies. Biol Pharm Bull 32: 982–987
35. JONKER J W, WAGENAAR E, VAN EIJL S, SCHINKEL A H
2003 Defficiency 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
36. CUI Y J, CHENG X, WEAVER Y M, KLAASSEN C D 2009 Tis-
sue distribution, gender-divergent expression, ontogeny, and chemi-
cal induction of multidrug resistance transporter genes (Mdr1a,
Mdr1b,Mdr2) in Mice. Drug Metab Dispos 37: 203–210
37. KARBACH U, KRICKE J, MEYER-WENTRUP F, GORBOU-
LEV V, VOLK C, LOFFING-CUENI D, KAISSLING B, BACH-
Period biol, Vol 113, No 1, 2011. 15
Sex and species differences in renal drug transporters I. Saboli} et al.
MANN S, KOEPSELL H 2000 Localization of organic cation
transporters OCT1 and OCT2 in rat kidney. Am J Physiol Renal
Physiol 279: F679–F687
38. LICKTEIG A J, CHENG X, AUGUSTINE L M, KLAASSEN C
D, CHERRINGTON N J 2008 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
39. LU H, KLAASSEN C D 2008 Gender differences in mRNA expres-
sion of ATP-binding cassette efflux and bile acid transporters in kid-
ney, liver, and intestine of 5/6 nephrectomized rats. Drug Metab
Dispos 36: 16–23
40. MASUDA S, TERADA T, YONEZAWA A, TANIHARA Y, KI-
SHIMOTO K, KATSURA T, OGAWA O, INUI K 2006 Identifica-
tion and functional characterization of a new human kidney–spe-
cific H+/organic cation antiporter, kidney-specific multidrug and
toxin extrusion 2. J Am Soc Nephrol 17: 2127–2135
41. NISHIHARA K, MASUDA S, JI L, KATSURA T, INUI K 2007
Pharmacokinetic significance of luminal multidrug and toxin extru-
sion 1 in chronic renal failure rats. Biochem Pharmacol 73:1482–1490
42. OTSUKA M, MATSUMOTO T, MORIMOTO R, ARIOKA S,
OMOTE H, MORIYAMA Y 2005 A human transporter protein that
mediates the final excretion step for toxic organic cations. PNAS 102:
17923–17928
43. SLITT AL, CHERRINGTON NJ, HARTLEY DP, LEAZER TM,
KLAASSEN C D 2002 Tissue distribution and renal developmental
changes in rat organic cation transporter mRNA levels. Drug Metab
Disp 30: 212–219
44. TERADA T, MASUDA S, ASAKA J, TSUDA M, KATSURA T,
INUI K 2006 Molecular cloning, functional characterization and tis-
sue distribution of rat H+/organic cation antiporter MATE1. Pharm
Res 23: 1696–1701
45. TZVETKOV M V, VORMFELDE S V, BALEN D, MEINEKE I,
SCHMIDT T, SEHRT D, SABOLIC I, KOEPSELL H, BROCK-
MOLLER J 2009 The effects of genetic polymorphisms in the or-
ganic cation transporters OCT1, OCT2, and OCT3 on the renal
clearance of metformin. Clin Pharmacol Therap 86: 299–306
46. URAKAMI Y, OKUDA M, SAITO H, INUI K 2000 Hormonal reg-
ulation of organic cation transporter OCT2 expression in rat kidney.
FEBS Lett 473: 173–176
47. WRIGHT S H, EVANS K K, ZHANG X, CHERRINGTON N J,
SITAR D S, DANTZLERW H 2004 Functional map of TEA trans-
port activity in isolated rabbit renal proximal tubules. Am J Physiol
Renal Physiol 287: F442–F451
48. ZHANG X, CHERRINGTON N J, WRIGHT S H 2007 Molecu-
lar identification and functional characterization of rabbit MATE1
and MATE2-K. Am J Physiol Renal Physiol 293: F360–F370
49. ZHANG X, EVANS K K, WRIGHT S H 2002 Molecular cloning
of rabbit organic cation transporter rbOCT2 and functional compar-
isons with rbOCT1. Am J Physiol Renal Physiol 283: F124–F133
50. DRESSER M J, GRAY A T, GIACOMINI K M 2000 Kinetic and se-
lectivity differences between rodent, rabbit, and human organic cat-
ion transporters (OCT1). J Pharmacol Exp Ther 292: 1146–1152
51. HIASA M, MATSUMO T, KOMATSU T, MORIYAMA Y 2006
Wide variety of locations for rodent MATE1, a transporter protein
that mediates the final excretion step for toxic organic cations. Am J
Physiol Cell Physiol 291: C678–C686
52. NAGEL G, VOLK C, FRIEDRICH T, ULZHEIMER JC, BAM-
BERG E, KOEPSELL H 1997 A reevaluation of substrate specificity
of the rat cation transporter rOCT1. J Biol Chem 272: 31953–31956
53. SCHALI C, SCHILD L, OVERNEY J, ROCH-RAMEL F 1983
Secretion of tetraethylammonium by proximal tubules of rabbit kid-
neys. Am J Physiol Renal Physiol 245: F238–F246
54. SUHRE W M, EKINS S, CHANG C, SWAAN P W, WRIGHT S
H 2005 Molecular determinants of substrate/inhibitor binding to the
human and rabbit renal organic cation transporters hOCT2 and
rbOCT2. Mol Pharmacol 67: 1067–1077
55. SWEET D H, BUSH K T, NIGAM S K 2001 The organic anion
transporter family: from physiology to ontogeny and the clinic. Am J
Physiol Renal Physiol 281: F197–F205
56. SWEET D H, MILLER D S, PRITCHARD J B 2001 Ventricular
choline transport. A role for organic cation transporter 2 expressed in
choroid plexus. J Biol Chem 276: 41611–41619
57. TAKANO M, INUI K, OKANO T, SAITO H, HORI R 1984 Car-
rier-mediated transport systems of tetraethylammonium in rat renal
brush-border and basolateral membrane vesicles. Biochim Biophys
Acta 773: 113–24
58. TAMAI I, OHASHI R, NEZU J, SAI Y, KOBAYASHI D, OKU A,
SHIMANE M, TSUJI A 2000 Molecular and functional character-
ization of organic cation/carnitine transporter family in mice. J Biol
Chem 275: 40064–40072.
59. WRIGHT S H, WUNZ T M 1987 Transport of tetraethylammo-
nium by rabbit renal brush-border and basolateral membrane vesi-
cles. Am J Physiol Renal Physiol 253: F1040–F1050
60. WU X, GEORGE R L, HUANG W, WANG H, CONWAY S J,
LEIBACH F H, GANAPATHY V 2000 Structural and functional
characteristics and tissue distribution pattern of rat OCTN1, an or-
ganic cation transporter, cloned from placenta. Biochim Biophys Acta
1466: 315–327
61. WU X, HUANG W, PRASAD P D, SETH P, RAJAN D P, LEI-
BACH F H, CHEN J, CONWAY S J, GANAPATHY V 1999 Func-
tional characteristics and tissue distribution pattern of organic cation
transporter 2 (OCTN2), an organic cation/carnitine transporter. J
Pharmacol Exp Ther 290: 1482–1492
62. YABUUCHI H, TAMAI I, NEZU J, SAKAMOTO K, OKU A,
SHIMANE M, SAI Y, TSUJI A 1999 Novel membrane transporter
OCTN1 mediates multispecific, bidirectional, and pH-dependent
transport of organic cations. J Pharmacol Exp Ther 289: 768–773
63. BIERMANN J, LANG D, GORBOULEV G, KOEPSELL H,
SINDIC A, SCHRÖTER R, ZVIRBLIENE A, PAVENSTÄDT H,
SCHLATTER E, CIARIMBOLI G 2006 Characterization of regu-
latory mechanisms and states of human organic cation transporter 2.
Am J Physiol Cell Physiol 290: C1521–C1531
64. ÇETINKAYA I, CIARIMBOLI G, YALÇINKAYA G, MEHRENS
T, VELIC A, HIRSCH J R, GORBOULEV V, KOEPSELL H,
SCHLATTER E 2003 Regulation of human organic cation trans-
porter hOCT2 by PKA, PI3K, and calmodulin-dependent kinases.
Am J Physiol Renal Physiol 284: F293–F302
65. CIARIMBOLI G, STRUWE K, ARNDT P, GORBOULEV V, KOE-
PSELL H, SCHLATTER E, HIRSCH J R 2004 Regulation of the
human organic cation transporter hOCT1. J Cell Physiol 201: 420–428
66. HOHAGE H, MORTH D M, QUERL I U, GREVEN J 1994 Reg-
ulation by PKC of the contraluminal transport system for organic
cations in rabbit kidney S2 proximal tubules. J Pharmacol Exp Ther
268: 897–901
67. MEHRENS T, LELLECK S, ÇETINKAYA I, KNOLLMANN M,
HOHAGE H, GORBOULEV V, BOKNIK P, KOEPSELL H,
SCHLATTER E 2000 The affinity of the organic cation transporter
rOCT1 is increased by protein kinase C-dependent phosphory-
lation. J Am Soc Nephrol 11: 1216–1224
68. PIETIG G, MEHRENS T, HIRSCH J R, ÇETINKAYA I, PIE-
CHOTA H, SCHLATTER E 2001 Properties and regulation of or-
ganic cation transport in freshly isolated human proximal tubules. J
Biol Chem 276: 33741–33746
69. SOODVILAI S, CHATSUDTHIPONG A, CHATSUDTHIPONG
V 2007 Role of MAPK and PKA in regulation of rbOCT2-mediated
renal organic cation transport. Am J Physiol Renal Physiol 293: F21–F27
70. MEIBOHM B, BEIERLE I, DERENDORF H 2002 How impor-
tant are gender differences in pharmacokinetics? Clin Pharmacokinet
41: 329–342
16 Period biol, Vol 113, No 1, 2011.
I. Saboli} et al. Sex and species differences in renal drug transporters
... There are several works demonstrating that sex can influence OCT1 expression at mRNA and protein levels. In mice and rats, renal OCT1 protein expression was higher in male than in female animals (Sabolic et al., 2011). However, renal OCT1 mRNA expression was higher in female than in male rats (Sabolic et al., 2011). ...
... In mice and rats, renal OCT1 protein expression was higher in male than in female animals (Sabolic et al., 2011). However, renal OCT1 mRNA expression was higher in female than in male rats (Sabolic et al., 2011). Therefore, regulation of OCT1 mRNA and protein expression can be divergent, at least in rodents. ...
Article
Full-text available
The organic cation transporter 1 (OCT1) belongs together with OCT2 and OCT3 to the solute carrier family 22 (SLC22). OCTs are involved in the movement of organic cations through the plasma membrane. In humans, OCT1 is mainly expressed in the sinusoidal membrane of hepatocytes, while in rodents, OCT1 is strongly represented also in the basolateral membrane of renal proximal tubule cells. Considering that organic cations of endogenous origin are important neurotransmitters and that those of exogenous origin are important drugs, these transporters have significant physiological and pharmacological implications. Because of the high expression of OCTs in excretory organs, their activity has the potential to significantly impact not only local but also systemic concentration of their substrates. Even though many aspects governing OCT function, interaction with substrates, and pharmacological role have been extensively investigated, less is known about regulation of OCTs. Possible mechanisms of regulation include genetic and epigenetic modifications, rapid regulation processes induced by kinases, regulation caused by protein–protein interaction, and long-term regulation induced by specific metabolic and pathological situations. In this mini-review, the known regulatory processes of OCT1 expression and function obtained from in vitro and in vivo studies are summarized. Further research should be addressed to integrate this knowledge to known aspects of OCT1 physiology and pharmacology.
... Point (2) is the extrapolation of the transporter activity by simple allometric scaling. It is well known that kidney transporters may have pronounced species-specific expression and that some transporters are sex specific in rats, but not in other species, including man (Sabolic et al. 2011). For other perfluoroalkyl substances, like perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid (PFOA), sex-specific renal clearance has been reported for rats (Harada et al. 2005), involving OAT2 and OAT3. ...
... For other perfluoroalkyl substances, like perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid (PFOA), sex-specific renal clearance has been reported for rats (Harada et al. 2005), involving OAT2 and OAT3. These anion transporters are not sex specific in humans (Sabolic et al. 2011). Hence, sex-specific differences in renal clearance do not directly translate from rats to humans. ...
... Therefore, 3R-alternatives (Reduction, Refinement, Replacement), including non-animal alternatives, have been widely accepted as a way to diminish the use of experimental animals in research and testing. On the contrary, when studying interactions among cells, tissues and organs, or when testing the pharmacology and toxicology of various substances, including drugs to be used in human and veterinary medicine, in most cases there is no plausible substitute for the living animal [451][452][453][454]. Moreover, the current legislation requires that all new drugs, before being licenced for human and animal use, have to be rigorously tested in at least two mammalian species (rodents and non-rodents) for metabolism, pharmacokinetics, acute and chronic toxicology in adult species, efficacy regarding the expected actions, effects on reproduction, embryonic toxicity, and potential carcinogenicity. ...
... Moreover, the current legislation requires that all new drugs, before being licenced for human and animal use, have to be rigorously tested in at least two mammalian species (rodents and non-rodents) for metabolism, pharmacokinetics, acute and chronic toxicology in adult species, efficacy regarding the expected actions, effects on reproduction, embryonic toxicity, and potential carcinogenicity. However, increasing evidence indicates that rodents and other common experimental animals, such as rabbits, may not be good models for studies relevant to humans due to sex and species differences in various properties [451,[454][455][456]. Therefore, one should bear in mind when choosing the appropriate species for toxicological testing that the results are comparable to humans. ...
Article
The comet assay has become one of the methods of choice for the evaluation and measurement of DNA damage. It is sensitive, quick to perform and relatively affordable for the evaluation of DNA damage and repair at the level of individual cells. The comet assay can be applied to virtually any cell type derived from different organs and tissues. Even though the comet assay is predominantly used on human cells, the application of the assay for the evaluation of DNA damage in yeast, plant and animal cells is also quite high, especially in terms of biomonitoring. The present extensive overview on the usage of the comet assay in animal models will cover both terrestrial and water environments. The first part of the review was focused on studies describing the comet assay applied in invertebrates. The second part of the review, (Part 2) will discuss the application of the comet assay in vertebrates covering cyclostomata, fishes, amphibians, reptiles, birds and mammals, in addition to chordates that are regarded as a transitional form towards vertebrates. Besides numerous vertebrate species, the assay is also performed on a range of cells, which includes blood, liver, kidney, brain, gill, bone marrow and sperm cells. These cells are readily used for the evaluation of a wide spectrum of genotoxic agents both in vitro and in vivo. Moreover, the use of vertebrate models and their role in environmental biomonitoring will also be discussed as well as the comparison of the use of the comet assay in vertebrate and human models in line with ethical principles. Although the comet assay in vertebrates is most commonly used in laboratory animals such as mice, rats and lately zebrafish, this paper will only briefly review its use regarding laboratory animal models and rather give special emphasis to the increasing usage of the assay in domestic and wildlife animals as well as in various ecotoxicological studies.
... The use of animal models, however, has become the subject of increasing criticism and ethically a strong need exists for the development of novel in vitro screening models to effectively reduce the number of animals used in research (Baumans, 2004;Liedtke et al., 2013;Soldatow et al., 2013;Freires et al., 2017). Expression of liverspecific enzymes relevant to drug metabolism and toxicity differ between animals and humans, influencing pre-clinical outcomes (Martigoni et al., 2006;Sabolic et al., 2011;Dalgaard, 2015). Furthermore, in vivo animal studies are costly and time-consuming (Soldatow et al., 2013;Sabolic et al., 2011). ...
... Expression of liverspecific enzymes relevant to drug metabolism and toxicity differ between animals and humans, influencing pre-clinical outcomes (Martigoni et al., 2006;Sabolic et al., 2011;Dalgaard, 2015). Furthermore, in vivo animal studies are costly and time-consuming (Soldatow et al., 2013;Sabolic et al., 2011). ...
Article
Abstract Ethnopharmacology relevance Traditional herbal medicines are utilized by 27 million South Africans. Xysmalobium undulatum (Uzara) is one of the most widely used traditional medicinal plants in Southern Africa. A false belief in the safety of herbal medicine may result in liver injury. Herb-induced liver injury (HILI) range from asymptomatic elevation of liver enzymes, to cirrhosis and in certain instances even acute liver failure. Various in vitro and in vivo models are available for the pre-clinical assessment of drug and herbal hepatotoxicity. However, more reliable and readily available in vitro models are needed, which are capable of bridging the gap between existing models and real human exposure. Three-dimensional (3D) spheroid cultures offer higher physiological relevance, overcoming many of the shortcomings of traditional two-dimensional cell cultures. Aims of this study This study investigated the hepatotoxic and anti-prolific effects of the crude X. undulatum aqueous extract during a sub-chronic study (21 days), in both a 3D HepG2/C3A spheroid model and the Sprague Dawley rat model. Methods HepG2/C3A spheroids were treated with a known hepatotoxin, valproic acid, and crude X. undulatum aqueous extract for 21 days with continuous evaluation of cell viability and proliferation. This was done by evaluating cell spheroid growth, intracellular adenosyl triphosphate (ATP) levels and extracellular adenylate kinase (AK). Sprague Dawley rats were treated with the same compounds over 21 days, with evaluation of in vivo toxicity effects on serum chemistry. Results The results from the in vitro study clearly indicated hepatotoxic effects and possible liver damage following treatment with valproic acid, with associated growth inhibition, loss of cell viability and increased cytotoxicity as indicated by reduced intracellular ATP levels and increased AK levels. These results were supported by the increased in vivo levels of AST, ALT and LDH following treatment of the Sprague Dawley rats with valproic acid, indicative of hepatic cellular damage that may result in hepatotoxicity. The in vitro 3D spheroid model was also able to predict the potential concentration dependant hepatotoxicity of the crude X undulatum aqueous extract. Similarly, the results obtained from the in vivo Sprague Dawley model indicated moderate hepatotoxic potential. Conclusion The data from both the 3D spheroid model and the Sprague Dawley model were able to indicate the potential concentration dependant hepatotoxicity of the crude X undulatum aqueous extract. The results obtained from this study also confirmed the ability of the 3D spheroid model to effectively and reliably predict the long-term outcomes of possible hepatotoxicity.
... In the in vitro transport study using HEK293 and CHO cell lines with different chain lengths of PFCAs, the compounds with carbon counts 7-9 have strong affinity to Oat1 and Oat3 (Weaver et al. 2010). Also in this report, the authors suggested that Oatp1a1 could Sabolic et al. (2011) compared the expression of renal tubular transporters in rodents, rabbits and humans, more Oat1 and Oat3 were expressed in male rats than in female rats. However, in humans, there were no gender differences in the expression of these transporters. ...
Article
Full-text available
Perfluoroalkyl and polyfluoroalkyl substances (PFASs) have been used in various industrial applications for many years. Long-chain PFASs are ubiquitous in wildlife and are reported to have a long elimination half-life in biological systems. Moreover, significant gender difference exists in the elimination of PFASs, where less is eliminated in male than in female. Recently, PFASs manufacturers and agencies have tried to replace the use of long-chain PFASs with short-chain PFASs, since they are expected to exhibit less bioaccumulation potential. Nevertheless, the potential risk and the pharmacokinetic (PK) characteristics of the short-chain PFASs still remain unknown. This study aims to fill the knowledge gap on short-chain PFASs, perfluoropentanoic acid (PFPeA), in terms of its PK properties using non-linear mixed-effect modeling and to explore gender differences in rats. Animal studies were carried out following oral or intravenous administration of PFPeA in male and female rats at a dose of 0.5–10 mg/kg. Plasma, urine, feces and nine tissues were collected and analyzed using ultra-performance liquid chromatography–tandem mass spectrometry. PK findings revealed that the clearance and the inter-compartmental clearance were 1.75 and 3.12 times higher in female rats than in male rats, respectively. According to the result, PFPeA is eliminated more rapidly in female rats than in male rats. Also, the tissue distribution study confirmed that distribution characteristics exhibited gender difference. This study provides scientific evidence for conducting further investigation into short-chain PFASs, biomonitoring plans and decision making regarding human health risk assessment.
... Experimental animal models are known to be useful and effective in this process. [6,7,8]. In addition, there are several articles that use in vitro models to investigate the medicinal properties of essential oils [9,10,11]. ...
Article
The objective of this study was to assess the species composition, abundance and habitat association of non‐volant small mammals from grassland, Acacia woodland, farmland, riverine forest, bushland and wooded grassland habitats in Gibe Sheleko National Park, southwestern Ethiopia. Data were collected using 49 Sherman live traps in 70 × 70 m sized girds and visual observations from December 2018 to August 2020. A total of 937 small mammals belonging to 10 species were captured and directly identified. The identified small mammal species and their relative abundance include Arvicanthis niloticus (7.04%), Crocidura olivieri (1.92%), Grammomys dolichurus (3.84%), Lemniscomys striatus (10.25%), Mastomys awashensis (24.55%), Mastomys natalensis (33.83%), Mus tenellus (0.85%), Myomys fumatus (6.30%), Rattus rattus (4.70%) and Stenocephalemys albipes (6.72%). Hystrix cristata, Xerus rutilus and Tachyoryctes splendens were documented through observation. The highest number of species (6) was registered at farmland and wooded grassland followed by bushland and riverine forest (5) and Acacia woodland and grassland (4). Lemniscomys striatus, M. awashensis and M. natalensis were common in all habitats. More, 216 (23.05%) individuals were recorded in farmland, while the least, 111 (11.85%) were recorded in Acacia woodland. The study area has various topographical land settings additional study is needed using different traps. Cette étude avait pour objectif d'évaluer la composition des espèces, l'abondance et l'association des habitats des petits mammifères non‐volants des prairies, des forêts d'acacias, des terres agricoles, des forêts riveraines, des broussailles et des prairies boisées du parc national de Gibe Sheleko, dans le sud‐ouest de l'Ethiopie. Les données ont été collectées à l'aide de 49 pièges vivants Sherman dans des gaines de 70 × 70 m et d'observations visuelles de décembre 2018 à août 2020. Un total de 937 petits mammifères appartenant à 10 espèces ont été capturés et ont été directement identifiés. Les espèces de petits mammifères identifiées et leur abondance relative sont les suivantes : Arvicanthisniloticus (7.04%), Crociduraolivieri (1.92%), Grammomysdolichurus (3.84%), Lemniscomysstriatus (10.25%), Mastomysawashensis (24.55%), Mastomysnatalensis (33.83%), Mus tenellus (0.85%), Myomysfumatus (6.30%), Rattusrattus (4.70%) et Stenocephalemysalbipes (6.72%). Hystrixcristata, Xerusrutilus et Tachyoryctessplendens ont été documentés par observation. Le nombre le plus élevé d'espèces (6) a été enregistré dans les terres agricoles et les prairies boisées, suivi par les zones de brousse et les forêts riveraines (5) et les zones boisées et les prairies d'acacia (4). Lemniscomysstriatus, M. awashensis et M. natalensis étaient des espèces communes dans tous les habitats. Plus de 216 (23.05%) individus ont été enregistrés dans les terres agricoles, tandis que le nombre le plus faible, 111 (11.85%) ont été enregistrés dans les forêts d'acacias. La zone d'étude présente différents contextes topographiques ; une étude supplémentaire est nécessaire en utilisant différents pièges.
Article
K-oximes were developed as modern drug candidates acting as AChE reactivators. In this study, it has been investigated which interspecies and intergender differences changes could be observed in Wistar rats and Swiss mice, both genders, after the treatment with increasing doses of selected acetylcholinesterase reactivators - asoxime, obidoxime, K027, K048, and K075. After the 24 h, a number of died animals was counted and the median lethal dose (LD50) for each oxime was calculated. By using the intramuscular route of administration, asoxime and K027 had the least toxicity in female rats (640.21 mg/kg and 686.08 mg/kg), and in female mice (565.75 mg/kg and 565.74 mg/kg), respectively. Moreover, asoxime and K027 showed 3, 4 or 8 times less acute toxicity in comparison to K048, obidoxime and K075, respectively. Beyond, K075 had the greatest toxicity in male rats (81.53 mg/kg), and in male mice (57.34 mg/kg), respectively. Our results can help to predict likely adverse toxic effects, target organ systems and possible outcome in the event of massive human overexposure, and in establishing risk categories or in dose selection for the initial repeated dose toxicity tests to be conducted for each oxime.
Article
Full-text available
Organic anion transporters play important roles in the elimination of a variety of endogenous substances, xenobiotics and their metabolites from the body. During the last decade, molecular cloning has identified several families of multispecific organic anion transporters mediating the renal and hepatic elimination of organic anions and, most recently, the OAT (organic anion transporter) family, the founding member of which (OAT1) is the basolateral p-aminohippurate (PAH) transporter in the renal proximal tubule. So far, four isoforms have been identified. OATs are membrane proteins with 12 putative membrane-spanning domains and function as sodium-independent exchangers or facilitators. OATs show weak structural similarity to organic cation transporters (OCTs) and OCTN/carnitine transporters. OATs are multispecific organic anion transporters, the substrates of which include both endogenous (e.g. cyclic nucleotides, prostaglandins, urate, dicarboxylates) and exogenous anions (various anionic drugs and environmental substances). All members of the OAT family are expressed in the kidney, while some are also expressed in the liver, brain and placenta. The OAT family represents the renal secretory pathway for organic anions and is also involved in the distribution of organic anions in the body.
Article
Full-text available
The organic anion transporter 5 (Oat5, Slc22a19) was previously localized to the brush-border of proximal tubule (PT) S3 segment in rat and mouse kidneys. Here we report on sex hormone-regulated expression of Oat5 in rat kidneys, after reinvestigating: a) expression of its mRNA by end-point and real time RT-PCR in the tissue, b) abundance of its protein by Western blotting (WB) in isolated membranes, and c) immunolocalization in tissue cryosections. In untreated male (M) and female (F) adult rats, the expression of Oat5 mRNA was predominant in the outer stripe (OS), exhibiting sex differences (M<F), upregulated by castration, and unaffected by ovariectomy. In castrated M, testosterone treatment strongly downregulated, whereas estradiol and progesterone treatment weakly upregulated its expression. By WB, a single protein band of ~72 kDa in variously-treated animals exhibited a density pattern comparable to that of mRNA. By immunostaining, Oat5 protein was localized to the brush-border of S1/S2 in the cortex (CO) (weakly) and in S3 of the OS and medullary rays (strongly) with the F-dominant intensity. In variously-treated rats, the immunostaining pattern matched that of mRNA and WB data. In prepubertal rats, the renal expression of Oat5 mRNA and protein was weak and sex-independent. In adult mice, the sex-dependent pattern of renal Oat5 protein expression was comparable to that in rats. Therefore, the renal expression of Oat5 in rats (and mice) exhibits zonal (CO<OS) and sex differences (M<F), which appear after puberty, largely due to androgen-driven downregulation of its mRNA and protein expression.
Article
Full-text available
Membrane transporters can be major determinants of the pharmacokinetic, safety and efficacy profiles of drugs. This presents several key questions for drug development, including which transporters are clinically important in drug absorption and disposition, and which in vitro methods are suitable for studying drug interactions with these transporters. In addition, what criteria should trigger follow-up clinical studies, and which clinical studies should be conducted if needed. In this article, we provide the recommendations of the International Transporter Consortium on these issues, and present decision trees that are intended to help guide clinical studies on the currently recognized most important drug transporter interactions. The recommendations are generally intended to support clinical development and filing of a new drug application. Overall, it is advised that the timing of transporter investigations should be driven by efficacy, safety and clinical trial enrolment questions (for example, exclusion and inclusion criteria), as well as a need for further understanding of the absorption, distribution, metabolism and excretion properties of the drug molecule, and information required for drug labelling.
Article
Full-text available
Organic cation transporters (OCTs) can mediate metformin transmembrane transport. We explored metformin pharmacokinetics in relation to genetic variations in OCT1, OCT2, OCT3, OCTN1, and MATE1 in 103 healthy male Caucasians. Renal clearance varied 3.8-fold and was significantly dependent on creatinine clearance (r(2) = 0.42, P < 0.0001), age (r(2) = 0.09, P = 0.002), and OCT1 polymorphisms. Carriers of zero, one, and two low-activity OCT1 alleles (Arg61Cys, Gly401Ser, 420del, or Gly465Arg) had mean renal clearances of 30.6, 33.1, and 37.1 l/h, respectively (P = 0.04, after adjustment for creatinine clearance and age). Immunohistochemical staining of human kidneys demonstrated OCT1 expression on the apical side of proximal and distal tubules. Increased renal clearance, in parallel with the known decreased hepatic uptake, may contribute to reduced metformin efficacy in low-activity genotypes. Renal OCT1 expression may be important not only in relation to metformin but with respect to other drugs as well.
Article
Full-text available
The renal proximal tubule (RPT) plays a crucial role in organic cation (OC) secretion and has a major impact on pharmacokinetics of OC drugs. Secretory transport is vectorial. Thus, it involves transporters located at both basolateral and apical membranes. Although sex hormones have been shown to regulate OC transport, there is little data on the effect of testosterone on OC secretion in a whole animal. Therefore, we determined the clearance of tetraethylammonium (TEA), a model OC substrate, in intact and castrated male mice. Castration significantly decreased renal TEA secretion by 30%, and testosterone supplementation returned TEA secretion to control levels in castrated mice. The mechanism of this effect was further examined in isolated mouse renal proximal tubules (mRPT). TEA uptake in isolated mRPT from castrated mice was reduced by 36%. This effect was reversed in tubules from castrated mice supplemented with testosterone. Kinetic analysis of [(3)H]-TEA uptake in isolated mRPT showed a decreased V(max) with no change in K(m), implying that the decrease in transport rate was caused by lowering in the number of transporters in castrated mice rather than a change in transporter affinity. Quantitative real time polymerase chain reaction (real time PCR) revealed that organic cation transporter (OCT)2 is the major TEA transporter in male mice. Moreover, OCT2 mRNA level was significantly reduced after castration. Castrated mice also showed a modest increase in organic cation/carnitine transporter 1 (OCTN1) mRNA level, indicating that testosterone may also regulate apical OCTN1 expression. These data suggest that testosterone regulates transepithelial transport of OC through modulation of OCT2 expression in male mice.
Article
Full-text available
The sulfate anion transporter (sat-1, Slc26a1) has been cloned from rat liver, functionally characterized, and localized to the sinusoidal membrane in hepatocytes and basolateral membrane (BLM) in proximal tubules (PT). Here, we confirm previously described localization of sat-1 protein in rat liver and kidneys and report on gender differences (GD) in its expression by immunochemical, transport, and excretion studies in rats. The approximately 85-kDa sat-1 protein was localized to the sinusoidal membrane in hepatocytes and BLM in renal cortical PT, with the male-dominant expression. However, the real-time reverse-transcription polymerase chain reaction data indicated no GD at the level of sat-1 mRNA. In agreement with the protein data, isolated membranes from both organs exhibited the male-dominant exchange of radiolabeled sulfate for oxalate, whereas higher oxalate in plasma and 24-h urine indicated higher oxalate production and excretion in male rats. Furthermore, the expression of liver, but not renal, sat-1 protein was: unaffected by castration, upregulated by ovariectomy, and downregulated by estrogen or progesterone treatment in males. Therefore, GD (males > females) in the expression of sat-1 protein in rat liver (and, possibly, kidneys) are caused by the female sex-hormone-driven inhibition at the posttranscriptional level. The male-dominant abundance of sat-1 protein in liver may conform to elevated uptake of sulfate and extrusion of oxalate, causing higher plasma oxalate in males. Oxalate is then excreted by the kidneys via the basolateral sat-1 (males > females) and the apical CFEX (Slc26a6; GD unknown) in PT and eliminated in the urine (males > females), where it may contribute to the male-prevailing development of oxalate urolithiasis.
Article
Full-text available
1. The transport of negatively charged drugs, xenobiotics, and metabolites by epithelial tissues, particularly the kidney, plays critical roles in controlling their distribution, concentration, and retention in the body. Thus, organic anion transporters (OATs) impact both their therapeutic efficacy and potential toxicity. 2. This review summarizes current knowledge of the properties and functional roles of the cloned OATs, the relationships between transporter structure and function, and those factors that determine the efficacy of transport. Such factors include plasma protein binding of substrates, genetic polymorphisms among the transporters, and regulation of transporter expression. 3. Clearly, much progress has been made in the decade since the first OAT was cloned. However, unresolved questions remain. Several of these issues--drug-drug interactions, functional characterization of newly cloned OATs, tissue differences in expression and function, and details of the nature and consequences of transporter regulation at genomic and intracellular sites--are discussed in the concluding Perspectives section.
Article
Transport of [3H]tetraethylammonium, an organic cation, has been studied in brush-border and basolateral membrane vesicles isolated from rat kidney cortex. Some characteristics of carrier-mediated transport for tetraethylammonium were demonstrated in brush-border and basolateral membrane vesicles; the uptake was saturable, was stimulated by the countertransport effect, and showed discontinuity in an Arrhenius plot. In brush-border membrane vesicles, the presence of an H+ gradient ([H+]i > [H+]o) induced a marked stimulation of tetraethylammonium uptake against its concentration gradient (overshoot phenomenon), and this concentrative uptake was completely inhibited by HgCl2. In contrast, the uptake of tetraethylammonium by basolateral membrane vesicles was unaffected by an H+ gradient. Tetraethylammonium uptake by basolateral membrane vesicles was significantly stimulated by a valinomycin-induced inside-negative membrane potential, while no effect of membrane potential was observed in brush-border membrane vesicles. These results suggest that tetraethylammonium transport across brush-border membranes is driven by an H+ gradient via an electroneutral H+-tetraethylammonium antiport system, and that tetraethylammonium is transported across basolateral membranes via a carrier-mediated system and this process is stimulated by an inside-negative membrane potential.
Article
Multidrug resistance (Mdr) transporters are ATP-binding cassette transporters that efflux amphipathic cations from cells and protect tissues from xenobiotics. Unfortunately, Mdr transporters also efflux anticancer drugs from some tumor cells, resulting in multidrug resistance. There are two groups of Mdrs in mice: group I includes Mdr1a and Mdr1b that transport xenobiotics, whereas group II is Mdr2, a flipase that facilitates phospholipid excretion into bile. Little is known about the regulation of Mdr genes in vivo. The purpose of this study was to determine tissue distribution, gender differences, ontogeny, and chemical induction of Mdrs in mice. The mRNA of Mdr1a is highest in gastrointestinal tract, Mdr1b in ovary and placenta, and Mdr2 in liver. Both Mdr1a and Mdr1b in kidney show female-predominant expression patterns due to repression by androgens. The ontogeny of mouse Mdr1a in duodenum and brain as well as Mdr1b in brain, kidney, and liver all share a similar developmental pattern: low expression at birth, followed by a gradual increase to mature levels at approximately 30 days of age. In contrast, Mdr2 mRNA in liver is markedly up-regulated at birth, which returns to low levels by 5 days of age and then gradually increases to mature levels. None of the Mdrs in liver are readily inducible by any class of microsomal enzyme inducers. In conclusion, the three Mdr transporters in mice are expressed in a tissue-specific and age-dependent pattern, there are gender differences in expression, and Mdr transporters are inducible by only a few microsomal enzyme inducers.
Article
Brush-border and basolateral membrane vesicles (BBMV and BLMV, respectively) from rabbit renal cortex were used to study transport of the organic cation, tetraethylammonium (TEA). Outwardly directed proton gradients stimulated uptake of TEA into BBMV and supported concentrative accumulation. Furthermore, an inwardly directed H+ gradient accelerated TEA efflux from BBMV. These data suggest that TEA transport in BBMV involved exchange with H+. The Jmax and Kt for TEA transport into BBMV under pH equilibrium conditions (pH 7.5) were 2.1 nmol.mg-1.min-1 and 0.15 mM, respectively. Under pH gradient conditions (6.0in:7.5out), Jmax increased by 270% with no effect on Kt. Uptake of TEA into BBMV was stimulated by an inside-positive electrical potential difference (PD), although exchange of TEA for H+ appeared to be one for one. In BLMV, H+ gradients had little effect on TEA uptake and were incapable of supporting concentrative transport. The Jmax and Kt for TEA transport in BLMV were 0.33 nmol.mg-1.min-1 and 0.37 mM, respectively. Inside-negative PDs stimulated this uptake, suggesting that it involved an electrically conductive pathway. TEA transport in BBMV and BLMV was inhibited by amiloride and cimetidine, although p-aminohippuric acid was without effect. Thus, secretion of TEA involves carrier-mediated transport steps at both the luminal and peritubular membranes, although an active step is not evident in isolated BLMV.