Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Clinical Science (2008) 114, 567588 (Printed in Great Britain) doi:10.

1042/CS20070227

Hepatocellular transport in acquired


cholestasis: new insights into functional,
regulatory and therapeutic aspects

Marcelo G. ROMA, Fernando A. CROCENZI and Enrique A. SANCHEZ


POZZI
Instituto de Fisiologa Experimental (IFISE), Facultad de Ciencias Bioqumicas y Farmaceuticas (CONICETU.N.R.), Suipacha
570, S2002LRL, Rosario, Argentina

The recent overwhelming advances in molecular and cell biology have added enormously to our
understanding of the physiological processes involved in bile formation and, by extension, to
our comprehension of the consequences of their alteration in cholestatic hepatopathies. The
present review addresses in detail this new information by summarizing a number of recent
experimental findings on the structural, functional and regulatory aspects of hepatocellular
transporter function in acquired cholestasis. This comprises (i) a short overview of the
physiological mechanisms of bile secretion, including the nature of the transporters involved and
their role in bile formation; (ii) the changes induced by nuclear receptors and hepatocyte-enriched
transcription factors in the constitutive expression of hepatocellular transporters in cholestasis,
either explaining the primary biliary failure or resulting from a secondary adaptive response;
(iii) the post-transcriptional changes in transporter function and localization in cholestasis,
including a description of the subcellular structures putatively engaged in the endocytic
internalization of canalicular transporters and the involvement of signalling cascades in this effect;
and (iv) a discussion on how this new information has contributed to the understanding of the
mechanism by which anticholestatic agents exert their beneficial effects, or the manner in which
it has helped the design of new successful therapeutic approaches to cholestatic liver diseases.

INTRODUCTION
Cholestasis is a frequent, prominent and often severe
manifestation of many liver diseases. It can be defined as

an impairment of normal bile flow secondary to structural


or biochemical abnormalities of the liver and/or the biliary tree. These alterations may involve: (i) rapid changes
in transporter function, e.g. due to drug-induced direct

Key words: bile, cholestasis, hepatocellular transport, liver disease, nuclear receptor, therapeutic target.
Abbreviations: ABC transporter, ATP-binding-cassette transporter; AE2/Ae2, anion exchanger 2; AQP, aquaporin; ASBT/Asbt,
apical Na+ -dependent bile salt transporter; BSEP/Bsep, bile salt export pump; CAR, constitutively activated receptor; CYP/Cyp,
ctyochrome P450; E2 17G, oestradiol 17-d-glucuronide; EE, 17-ethynyloestradiol; ER, oestrogen receptor; ERK, extracellularsignal-regulated kinase; FXR, farnesoid X receptor; HNF, hepatocyte nuclear factor; IL, interleukin; JNK, c-Jun N-terminal kinase;
LPS, lipopolysaccharide; LRH1, liver receptor homologue 1; LXR, liver X receptor; MAPK, mitogen-activated protein
kinase; MRP/Mrp, multidrug resistance-associated protein; NHE, Na+ /H+ exchanger; NTCP/Ntcp, Na+ -taurocholate cotransporting polypeptide; OATP/Oatp, organic anion-transporting polypeptide; OST/Ost, organic solute transporter; PBC, primary
biliary cirrhosis; PFIC, progressive familial intrahepatic cholestasis; PI3K, phosphoinositide 3-kinase; PKC, protein kinase C;
PPAR, peroxisome-proliferator-activated receptor ; PSC, primary sclerosing cholangitis; PXR, pregnane X receptor; RAR,
retinoic acid receptor ; RXR, retinoid X receptor ; SHP, small heterodimer partner; Stat5, signal transducer and activator of
transcription 5; SULT/Sult, sulfotransferase; t-BOOH, t-butylhydroperoxide; TLC, taurolithocholate; TNF, tumour necrosis
factor ; UDC, ursodeoxycholate; TUDC, tauroUDC; UGT/Ugt, UDP-glucuronosyltransferase; VDR, vitamin D receptor.
Correspondence: Dr. Marcelo G. Roma (email mroma@fbioyf.unr.edu.ar).


C

The Authors Journal compilation


C

2008 Biochemical Society

567

568

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

inhibition of relevant transporters or changes in their


localization; and (ii) changes in transporter expression,
due to inhibition of carrier synthesis or exacerbated degradation. As a consequence of these primary cholestatic
alterations affecting primarily hepatocyte bile formation
(hepatocellular cholestasis), or in response to an intraor extra-hepatic blockage of the bile transit (obstructive
cholestasis), the liver develops a number of secondary
adaptive changes to minimize the detrimental effects of
toxic biliary compounds retained as a consequence of the
secretory failure. This adaptive response comprises,
among other modifications, changes in the expression
of both basolateral and canalicular transporters mediated
by the co-ordinated activation of a number of nuclear
hormone receptors. These regulators act as transcription
factors that directly interact with and control the expression of genomic DNA on binding a relevant ligand. These
ligands are, more commonly, retained bile salts or conjugated bilirubin, which function as sensors of secretory
failure.
The aim of the present review is to give an insight into
recent advances in the understanding of the mechanisms of canalicular transporter alterations in acquired
cholestasis, with particular emphasis in the structural and
functional alterations leading to transport dysfunction.
In addition, regulatory aspects involved in the adaptive
responses against the detrimental effects of the cholestatic
injury will be examined. Finally, we will discuss how
certain anticholestatic agents in current therapeutic use
may exert beneficial effects, and the mechanistic basis
of novel therapeutic strategies based on the information
obtained from these basic studies.

PHYSIOLOGICAL MECHANISMS OF BILE


FORMATION
Bile formation is an osmotic process driven by the
vectorial transport of solutes into bile. This induces
passive water movement from blood into bile in response
to osmotic gradients, via the water channels AQP
(aquaporin) 9 and AQP8 localized at the basolateral
and the apical membranes respectively [1,2]. For this to
occur, biliary solutes need to be actively concentrated and
retained into a confined space, i.e. the bile canaliculus.
Only bile salts, glutathione, either reduced (GSH) or
oxidized (GSSG) [3] and, perhaps, HCO3 [4] fulfil these
requirements. This primary canalicular secretion is modified further by cholangiocytes during its transit along
bile ducts. This process is a result of a balance between
hormone-dependent secretion of water and electrolytes, and the obligatory absorption of water, electrolytes
and organic solutes [5]. Figure 1 shows a schematic
representation of the main solutes and transporters
relevant to bile flow formation at both the hepatocellular
and the bile ductular levels.

C

The Authors Journal compilation


C

2008 Biochemical Society

Bile salts are the predominant organic solutes in


bile and the main determinants of bile flow. They are
mainly taken up by the NTCP/Ntcp (Na+ -taurocholate
co-transporting polypeptide; SLC10A1/Slc10a1), driven
by the Na+ gradient maintained by the Na+ /K+ -ATPase
pump [6]. A remaining fraction is taken up by a
Na+ -independent transport system mediated by OATP/
Oatp (organic anion-transporting polypeptide) family of
transporters, which are non-electrogenic in nature [7].
In addition to conjugated and unconjugated bile salts,
OATPs/Oatps accept other amphipathic compounds,
such as bilirubin glucuronides and, maybe, unconjugated bilirubin. Four OATPs have been cloned
and characterized from human liver, namely
OATP1A2 (SLCO1A2/SLC21A3; formerly OATPA), OATP1B1 (SLC21A6; formerly OATP-C or
LST-1), OATP1B3 (SLC21A8; formerly OATP-8) and
OATP2B1 (SLC21A9; formerly OATP-B). All but the
latter are involved in bile-salt uptake, with OATP1A2
being the most important one [8]. For unconjugated
bilirubin [9] and bilirubin monoglucuronides [10], both
OATP1B1 and OATP1B3 have been proposed to be
involved. Although OATP1B1 was thought to have a
far higher efficiency for unconjugated bilirubin than
OATP1B3 [10], this finding was not supported by others
[11]. Bilirubin diglucuronides appear to be transported
only by OATP1B1 [10]. There are three Oatps identified
in rats, namely Oatp1a1 (Slc21a1; formerly Oatp1),
Oatp1a4 (Slc21a5; formerly Oatp2) and Oatp1b2
(Slc21a10; formerly Oatp4 or Lst-1). Oatp1b2 is the
rodent orthologue of both OATP1B1 and OATP1B3
[12]. All of them are involved in the uptake of conjugated
and unconjugated bile salts [8,13]. Oatp1a1 and Oatp1a4
are involved in bilirubin monoglucuronide transport
[13,14].
After traversing the cell by Ficks diffusion bound to
high-affinity cytosolic proteins, monoanionic bile salts
are excreted in the canalicular pole by BSEP/Bsep (bile
salt export pump; ABCB11), an ABC transporter (ATPbinding-cassette transporter) [15]. In contrast, canalicular
efflux of divalent, bipolar sulfated or glucuronidated bile
salts is mediated by MRP2/Mrp2 (multidrug resistanceassociated protein 2; ABCC2/Abcc2). This carrier is also
engaged in the biliary excretion of bilirubin mono- and
di-glucuronides [16].
Another key determinant of bile flow is glutathione,
both in its reduced (GSH; approx. 80 %) and in its
oxidized (GSSG) forms [3]. The liver is the main site of
glutathione synthesis, and biliary glutathione comes from
this intracellular source. A high-affinity electrogenic
canalicular carrier for GSH has been functionally characterized, but not cloned [17]. This transporter actively
exports GSH into bile, although it can transfer GSSG
with lower affinity. On the contrary, GSSG is excreted with higher affinity by Mrp2, which can also transfer
GSH, but with low affinity [18].

Hepatocellular transport in acquired cholestasis

Figure 1 Schematic representation of the main transport proteins involved in bile flow generation in both hepatocytes
and cholangiocytes

ATP-dependent transporters are depicted as brown/orange circles. For the sake of simplicity, only rodent transporters are displayed, but human orthologues of all of
these transporters have been identified. See the text for details on both rodent and human transporters. Br, bilirubin; BS , bile salts; Glc, glucose; Glu, glutamate;
GSH, glutathione; OA , organic anions; VIP, vasoactive intestinal peptide.
Another putative candidate to partially determine
bile flow is HCO3 [4]; however, it remains questionable whether bile-to-plasma HCO3 gradients can be
maintained, due to the high paracellular permeability of
this anion [19]. Overall, HCO3 transport involves the
sequential participation of three different transporters,
namely Na+ /K+ -ATPase, NHE (Na+ /H+ exchanger),
localized at the basolateral domain, and AE2/Ae2 (anion
exchanger 2; SLC4A2/slc4a2), localized at the apical
domain. Na+ /K+ -ATPase activity maintains plasma-tocytosol Na+ gradients for basolateral efflux of H+ via
NHE. The extruded H+ neutralizes HCO3 in plasma,
enabling its passive diffusion as CO2 . After conversion
of CO2 back into HCO3 by intracellular carbonic
anhydrase, the anion is counter-transported with Cl at
the canalicular domain by AE2/Ae2.
Canalicular bile flow is modified further during its
transit along the bile ducts by both secretory and absorptive processes [20]. Ductular fluid secretion is driven

by the secretin-regulated cAMP-dependent output of an


HCO3 -rich fluid secreted via the Cl /HCO3 exchange
system AE2/Ae2. Exchange is driven by the out-toin Cl concentration gradient, which is maintained by
the Cl efflux across the apical membrane via the
secretin-activated CFTR (cystic fibrosis transmembrane
regulator). As with secretin, VIP (vasoactive intestinal
peptide) [21,22] and bombesin [23,24] stimulate ductular
HCO3 -rich fluid both in rodents and humans. The
effects of these peptides is cAMP-independent, at least in
rodents [22]. Blood-to-bile water movement at the ductular level is facilitated by constitutive AQP4 in the basolateral membrane, and secretin-stimulated AQP1 in the
apical membrane [1,2]. On the other hand, the absorption
of ductular water and electrolytes is driven by osmotic
gradients created by bile-to-plasma transport of electrolytes and organic solutes. They comprise glutamate,
glucose, transported by SGLT1 and GLUT1 at the apical
and basolateral domains respectively, and bile salts, taken

C

The Authors Journal compilation


C

2008 Biochemical Society

569

570

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

up by ASBT/Asbt (apical Na+ -dependent bile salt transporter; SLC10A2/slc10a2), followed by basolateral extrusion via the export pump MRP3/Mrp3 (ABCC3/Abcc3)
and the heterodimeric OSTOST/OstOst (where
OST/Ost is organic solute transporter) [5,25].

ALTERATIONS IN CANALICULAR
TRANSPORTERS IN ACQUIRED CHOLESTASIS
Changes in hepatobiliary transporter expression/activity
in acquired cholestasis have been reported to occur at the
transcriptional, post-transcriptional or post-translational
level. These pathomechanisms result in changes in
transporter activity in days, hours or minutes/seconds
respectively.

Changes in transporter expression in


cholestasis
Decreased expression of hepatobiliary transport systems
at the protein level can be the primary cause of the cholestatic injury or, alternatively, the secondary consequence of adaptive changes in transporter expression. In
the former case, a clear distinction between the mechanisms that actually cause the secretory failure from the
cascade of changes resulting from this initial cholestatic
insult is difficult to establish. These secondary adaptive
changes can be dissected better in experimental models of
obstructive cholestasis (bile duct ligation). In this case, the
primary cholestatic insult occurs at an extrahepatic level,
so that all of the hepatic changes can be regarded as adaptive. It should be stressed, however, that these animal
models cannot be extrapolated unequivocally to human
cholestatic conditions, due to differences in transcriptional regulation, transporter specificity and the length
of the cholestatic insult (acute in laboratory animal
compared with chronic in humans).

Adaptive changes in the expression of transporters and


metabolic systems in cholestasis
In several animal models of experimental cholestasis,
and in several human cholestatic liver diseases as well,
there is a common pattern of adaptive responses involving
changes in transporter expression. This response protects
the hepatocyte from the retention of toxic biliary
compounds, particularly bile salts and bilirubin [26]. The
adaptive changes comprise down-regulation of basolateral uptake systems, accompanied by the up-regulation
of basolateral extrusion systems (Figure 2). This greatly
helps to maintain intracellular concentrations of internalized metabolites, or de-novo-synthesized ones, at a safe
low level, despite ongoing secretory failure. This adaptive
transporter regulation is assisted further by repression
of the bile-salt-synthesizing enzymes Cyp7a1 and
Cyp8b1 (where Cyp is cytochrome P450), and the up
C

The Authors Journal compilation


C

2008 Biochemical Society

regulation of phase I and phase II bile-acid-metabolizing


enzymes. Phase I bile-acid-hydroxylating enzymes
comprise Cyp3a11 (the rodent homologue of human
CYP3A4) and Cyp2b10 (the rodent homologue of
human CYP2B6). Phase II-conjugating enzymes are
Sult2a1 (where Sult is sulfotransferase) and Ugt2b4
(where Ugt is UDP-glucuronosyltransferase). Bile acid
hydroxylation (mainly at 1, 2, 4, 6, 6, 22 or 23
positions) results in an increased intracellular pool of
less toxic polyhydroxylated bile salts. Most of these
transformations facilitate the excretion of bile acids
into the bile (sulfation) or into the blood for subsequent urinary elimination (hydroxylation, sulfation and
glucuronidation) [27].
Ntcp [2831], Oatp1a1 [28,31,32] and Oatp1b2 [33]
protein levels are decreased in several animal models
of experimental cholestasis at a transcriptional level
[34]. This helps to maintain at a minimum the uptake
of potentially toxic compounds arriving to the liver,
such as the bile salts reabsorbed by the intestine and
bilirubin. On the other hand, Oatp1a4 is up-regulated in
bile-duct-ligated mice [33]. Organic substrate transport
carried out by Oatp1a4 is potentially bidirectional [35]
and, therefore, can reverse its transport direction in
conditions of bilirubin conjugate/bile acid overload, thus
acting as an alternative export system in cholestasis.
In addition to impairing uptake, the cholestatic
insult enhances routes for the basolateral extrusion of
organic anions. The basolateral membrane of rodent
cholestatic hepatocytes overexpresses several ATPdependent organic anion efflux pumps belonging to
the Mrp family, such as Mrp1 (ABCC1/Abcc1) [34,36,37],
Mrp3 (ABCC3/Abcc3) (in rat [28,3843], but not in
mouse [44]), Mrp4 (ABCC4/Abcc4) [34,44,45] and Mrp5
(ABCC5/Abcc5) [34,39]. In experimental obstructive
cholestasis in rodents, all of these increased expressions
occur at a protein level and are, at least in part,
transcriptional in nature [34]. These ATP-dependent
carriers extrude divalent bile acids (e.g. bile acid sulfoconjugates) and monovalent bile acids (glyco- and
tauro-conjugates) in rodents [13]. In humans, specificity
and transport mechanisms may vary. For example,
MRP3 transports only bile acid glycoconjugates with
low affinity [46,47], and MRP4 co-transports bile acids
with GSH [48]. In addition, the recently identified
OSTOST/OstOst, a heterodimeric transporter
that exports bile salts and their conjugates back to
plasma, is also up-regulated at the transcriptional level in
both the bile-duct-ligated mice and in cholestatic patients
with PBC (primary biliary cirrhosis) [49]. The relative
importance of each of these basolateral extrusion carriers
to transport biliary substrates, and their role in protecting
the hepatocyte against a cholestatic insult, may be
differential. Experiments in Mrp4/ mice with bile duct
ligation revealed that hepatic Mrp4 plays an important
role in the adaptive response to obstructive cholestatic

Hepatocellular transport in acquired cholestasis

Figure 2 Adaptive changes in obstructive cholestasis in the expression of transporters and enzymes involved in bile salt
synthesis and bile salt/bilirubin conjugation in the liver, kidney and intestines

Note that, for simplicity, only changes in rodent gene products are displayed. Most of the adaptive changes at the hepatocellular level have been confirmed
in patients with cholestasis. On the contrary, most of the adaptive response in ductular epithelia, kidney and intestines described in rodents remains to be confirmed in
humans. For details, see the text. Br, unconjugated bilirubin; Br-G, bilirubin glucuronides; CFTR, cystic fibrosis transmembrane regulator; OA , organic anions; BS ,
bile salts.
liver injury by preferentially improving the extrusion of
bile salts into the blood. Indeed, more extensive damage
and lower serum bile salt levels occurred in Mrp4/
mice compared with the wild-type, despite both Mrp3
and OstOst being up-regulated [44]. Conversely,
bile-duct-ligated Mrp3/ mice had no differences
compared with the wild-type in both serum bile salt
levels and liver histology [43,50]; Mrp3/ mice had
lower serum levels of bilirubin, suggesting a preferential
role for Mrp3 in the basolateral extrusion of bilirubin
glucuronides.

The bile salt conjugates and the bilirubin glucuronides


that are not taken up by the hepatocyte, or that are
effluxed into the systemic circulation after uptake, can
be eliminated in urine. This alternative excretory route is
enhanced under cholestatic conditions [12]. The increased
efficiency of the kidney to detoxify these compounds
occurs by (i) increased passive glomerular filtration,
(ii) repressed transcription, leading to diminished protein
levels of Asbt [51], a transporter involved in tubular
re-absorption of bile salts, and (iii) the co-ordinated overexpression at the protein level of basolateral Oat1 [52]

C

The Authors Journal compilation


C

2008 Biochemical Society

571

572

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

and of the apical transporter Mrp2 [38,51]; Mrp2 mRNA


levels did not increase, suggesting post-transcriptional
mechanisms [51]. Similarly, renal Mrp4 which, unlike
liver, has an apical localization in kidney, is up-regulated
transcriptionally in mouse kidney [53,54], although it is
down-regulated at the post-transcriptional level in the
rat [45].
The final hepatic fate of the cholephilic compounds
under physiological conditions is canalicular transfer,
which represents the rate-limiting step in the overall hepatic handling. In obstructive cholestasis, secondary inhibition of the expression of canalicular transporters occurs.
These changes can be regarded, however, as beneficial,
since this may help to protect bile ducts from mechanical
damage by an increase in biliary pressure [53,55] or from
cholangiocyte necro-apoptosis induced by toxic biliary
solutes [56]. Both Mrp2 protein expression and mRNA
levels were considerably impaired in obstructive cholestasis in rats [5759] and humans [60,61]. Bsep expression
is less altered by far [59,62]. An adaptive response against
this is the enhancement in the bile salt cholehepatic
shunt pathway. This mechanism removes bile salts
from the biliary space and maintains in operation
the hepato-cholangiocyte flux of bile salts, despite
ongoing bile duct obstruction. A key ductular bile salt
transporter involved in this response is Asbt. Although
the expression of this transporter is maintained per
cholangiocyte, the number of intrahepatic bile ducts
markedly increases in bile-duct-ligated rats (approx. 10fold after 1 week) [25]. Improvement in Asbt-mediated
bile acid transport through the cholangiocyte apical
membrane in bile-duct-ligated animals is assisted by a
concomitant overexpression of the basolateral bile acid
transporters Mrp3 [40] and OstOst [49]; Ost
Ost induction was only confirmed at the mRNA level.
Multiplication of bile ducts also induces a proportional
increase in the biliary tree death space. This helps to
substitute the intestines as the major reservoir of the
bile salt pool when bile salt enterohepatic recirculation is
impaired [63]. Down-regulation of intestinal Asbt protein
levels also occurs. This decreases the amount of bile salts
returning to the liver, thus reducing further the liver
bile salt load. Adaptive down-regulation of Asbt was
observed in bile-duct-ligated rats, the mechanism being
post-transcriptional in nature [64].
Commonly, the acquired changes in transporter expression in human cholestatic liver disease are consistent
with those occurring in experimental animal models of
cholestasis, although the mechanisms accounting for
these expression changes may vary. Generally, the contribution of transcriptional mechanisms is more limited,
or absent, in humans compared with rodents, with posttranscriptional/post-translational mechanisms being
more relevant [13,6567].
As in rodents, the down-regulation of basolateral uptake transport systems is apparent in chronic cholestatic

C

The Authors Journal compilation


C

2008 Biochemical Society

diseases in humans. NTCP and OATP1B1 protein levels


decrease in inflammation-induced icteric cholestasis (e.g.
cholestatic alcoholic hepatitis) [65], late-stage PBC [66],
PFIC2 and PFIC3 (type 2 and 3 progressive familial
intrahepatic cholestasis respectively) [68], PSC (primary
sclerosing cholangitis) [67] and extrahepatic biliary atresia
[69]. OATP1B3 is also down-regulated in PFIC1 (type 1
PFIC) and PFIC2 at the protein level [68]. Similarly,
adaptive increments in protein levels of the hepatocellular basolateral export pumps MRP1 [70,71], MRP3
[66,70,71], MRP4 [71,72] and MRP5 [71] have been reported in late-stage PBC. Similarly, MRP3 [73] and MRP4
[68] protein levels were increased in PFIC3. Finally, increases in MRP3 have been reported in both biliary atresia
[73] and biliary obstruction, secondary to carcinomas
of the biliary tract or pancreas [61]. The mRNA levels of
the basolateral extrusion transporter OSTOST are
also augmented in the liver of patients with advanced
PBC [49]. Extensive up-regulation at the mRNA level of
OSTOST, but not of MRP3 and MRP4, may help
to explain why the induction of OSTs is more profound
than that of MRP3 and MRP4 in PBC [49]. OATP1B3,
which remains highly expressed in liver under cholestatic
conditions, behaves as a GSH-dependent symporter, and
has recently been proposed to be an additional route for
the basolateral extrusion of cholephilic organic anions,
including bile salts, in human cholestatic diseases [74].
At the canalicular pole, and similar to the findings
in bile-duct-ligated rats, MRP2 expression is decreased
at the protein level in cholestatic liver diseases, although
the changes are only observed in advanced stages of the
disease and are less dramatic in intensity. For example,
MRP2 protein expression only decreased in some, but
not all, patients with stage IV PBC [75] and remained unaffected in stages IIII PBC [66,75]. Similarly, MRP2 and
BSEP mRNA levels fell only in livers from poorly drained
patients with obstructive extrahepatic cholestasis, but
not in the well-drained ones [61]. Finally, BSEP protein
expression was relatively well preserved in patients with
extrahepatic biliary atresia, even in the untreated ones
[76].
In human cells, other than hepatocytes, the existence of
an adaptive response against cholestasis remains largely
speculative, although some indications are emerging.
Human cholangiocytes actively proliferate in response
to many cholestatic conditions, including obstructive
cholestasis and PBC [77]. MRP3 protein levels were
frequently increased in proliferative cholangiocytes from
livers of patients with various forms of cholestasis, as
revealed by immunohistochemical studies [70,73]. In
the intestine, adaptive down-regulation of ASBT in
patients with obstructive cholestasis has been reported
[78]. Finally, although all of the transporters involved in
the renal response to cholestasis have been identified in
human kidney, the occurrence of these adaptive changes
in patients with cholestasis remains to be confirmed.

Hepatocellular transport in acquired cholestasis

Role of nuclear receptors and hepatocyte-enriched


transcription factors in adaptive response in cholestasis
Adaptive changes in expression of hepatocellular transporters are mediated mainly by the co-ordinated modulation of a number of transcription factors, including the
nuclear orphan receptors and liver-enriched HNFs (hepatocyte nuclear factors). Most of the information on these
mechanisms was obtained from rodent models of cholestasis, by using either specific ligands to activate nuclear
receptors or knockout animals with selective deficiency in
the synthesis of transcription factors. However, these regulations remain largely unconfirmed in human cholestatic
disease. It should be borne in mind that changes involving
these regulators are transcriptional in nature and, as stated
above, transcriptional mechanisms may be less relevant in
humans compared with rodents [13,6567].
The nuclear receptors thought to be involved in feedback or feedforward regulation of carrier genes in
cholestasis comprise, among others, FXR (farnesoid X
receptor; NR1H4), PXR (pregnane X receptor; NR1I2),
CAR (constitutively activated receptor; NR1I3), VDR
(vitamin D receptor; NR1I1), LXR (liver X receptor;
NR1H3), RAR (retinoic acid receptor ), LRH1 (liver
receptor homologue 1; NR5A2), PPAR (peroxisomeproliferator-activated receptor ; NR1C1) and SHP
(small heterodimer partner; NR0B2). All except the two
latter heterodimerize with RXR (retinoid X receptor
; NR2B1), enabling high-affinity DNA binding and
further activation of gene transcription [12]. The ligands
involved in this adaptive response are biliary constituents
retained by secretory failure, such as bilirubin [79] and
hydrophobic hepatotoxic bile salts (e.g. chenodeoxycholate, deoxycholate and lithocholate) [80,81].
In rodents, FXR-mediated induction of the nuclear
repressor SHP inhibits Ntcp expression in experimental
obstructive cholestasis [82,83], which surpasses the
RXR/RAR-mediated activation of the Ntcp promoter
[83]. The SHP effect may be, at least in part, mediated
by a multistep, regulatory cascade involving repression
of the hepatocyte-enriched transcription factor HNF4,
which suppresses its activating effect on HNF1 [also
known as TCF1 (transcription factor 1)], a potent Ntcp
activator [84]. The human SHP promoter region also
contains FXR-response elements [85]. The involvement
of SHP was, however, challenged by studies showing
that Ntcp is down-regulated in bile-duct-ligated SHP/
mice, suggesting that SHP is not essential for Ntcp
repression [86]. As bile salts can repress HNF4
(NR2A1) transcription genes, a direct bile salt inhibition
of this transcription factor can help to explain this finding
[87]. In humans, the relevance of this latter transcriptional
cascade is doubtful, as neither HNF4, HNF1 nor
RXR/RAR binds the Ntcp promoter, suggesting
species differences in NTCP/Ntcp gene regulation
[84].

A similar FXR-mediated repression of the SHP/


HNF4/HNF1 cascade has been proposed to be a
primary mediator of the transcriptional impairment of
Oatp1a1 and Oatp1b2 in rodent models of cholestasis
[13,87]. Interestingly, OATP1B1, the gene of the human
orthologue of Oatp1b2, contains an HNF1-binding
site in its promoter region [88], suggesting that this latter
mechanism may be operative in patients with cholestasis
[89]. In addition, PXR and CAR up-regulate the Na+ independent bile-acid uptake transporter Oatp1a4 [33].
CAR is one of the nuclear receptors involved in the upregulation of Mrp3 and Mrp4 in rodents, thus enhancing
basolateral export pathways in these animal models [90
92]. Mrp3, but not Mrp4, overexpression is assisted by
PXR [90,93] and LRH1 [94]. In this latter case, another
factor mediating bile-acid-dependent induction would be
required, as bile acids are not LRH1 ligands. In contrast,
induction of Mrp3 and Mrp4 are FXR-independent
[53,95,96]. Furthermore, FXR appears to inhibit Mrp4
in rodent cholestatic liver, but CAR overrides this inhibitory effect [97]. In humans, the mechanisms of MRP3
induction may be different to rodents, as a CAR
cis-binding element could not be identified in the human
MRP3 promoter [98]. PXR-mediated up-regulation of
human MRP3 is an alternative possibility, as ligand-activated PXR has also been shown to induce MRP3/Mrp3
expression in human hepatoma cell lines [93]. LRH1induced MRP3 expression is also possible, as the MRP3
promoter contains a binding element for LRH1 [99].
Finally, MRP3 overexpression may also involve a
decrease in the expression of RXR/RAR in human
cholestasis. This nuclear receptor down-regulates MRP3
expression by abrogating the activity of the transcription
factor Sp1 on the MRP3 gene promoter [98].
FXR activates the basolateral bile-salt-exporting carrier OSTOST/OstOst, both in healthy rodents
[96] and in cholestatic rodents and humans [49]. Similarly, FXR transactivates OATP1B3, but not other OATP
isoforms [100]. Differential activation of OATP1B3 may
be related to its bile-salt-exporting function at the
basolateral domain [74].
In the canalicular pole, bile-salt-activated induction
of FXR is responsible for the maintenance of Bsep
expression under these cholestatic conditions by upregulating the otherwise decreased Bsep expression. This
regulatory role for FXR has been only confirmed in a
rodent model of obstructive cholestasis [53]; however,
the BSEP (ABCB11) gene also contains an IR1 (inverted
repeat 1) element in its promoter regions that directly
binds to the FXR/RXR heterodimer upon activation
by bile acids, resulting in enhanced transactivation of the
BSEP gene [101103].
Unlike Bsep, Mrp2 expression is extensively repressed
in cholestatic liver [57]. Obstructive cholestasis is
accompanied by systemic release of pro-inflammatory
cytokines [104]. The decrease in Mrp2 content in this

C

The Authors Journal compilation


C

2008 Biochemical Society

573

574

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

condition appears to be due to a cytokine [IL (interleukin)


1]-dependent impairment in RAR expression
[105,106]. Cytokines also induce re-localization of the
nuclear receptor partner RXR from the nucleus to
cytosol, a finding shown in LPS (lipopolysaccharide)induced cholestasis [107]. If the latter event also occurs in
obstructive cholestasis, this may contribute further to the
reduced occurrence of RAR/RXR heterodimerization
and that of other nuclear receptors that transactivate
MRP2/Mrp2, such as FXR, PXR and CAR [108].
Nuclear receptors also mediate adaptive changes
in enzymes involved in bile salt synthesis and bile
salt/bilirubin detoxification [27,109,110]. Most of these
regulations have been observed in rodent models of
cholestasis, and the involvement of these regulatory
mechanisms in patients with cholestasis has been
inferred, but not determined, from studies in patients
without cholestasis receiving specific nuclear receptor
ligands for therapy or from studies in human cells lines.
FXR (via the SHP-mediated repression of LHR1 [85])
and PXR [80,111] inhibit de novo bile salt synthesis
by repressing the transcription of the genes encoding
Cyp7a1, the rate-limiting enzyme in the classic bile salt
biosynthetic pathway. In primary human hepatocytes,
activation of PXR by rifampicin promotes PXR interaction with HNF4 and further CYP7A1 inhibition
[112]. Cyp8b1, which controls the dihydroxy-totrihydroxy bile acid synthesis ratio, was also repressed,
thus regulating the bile salt hydrophobicity index. A
similar down-regulation of both Cyp7a1 and Cyp8b1
occurs after PPAR activation in rodents [113,114].
PXR and CAR enhance the expression of Cyp3a-mediated bile salt hydroxylation in rodents [80,92,111,115],
and this also appears to apply to humans. Indeed, a
response element in the human CYP3A promoter has
been identified for PXR [116] and CAR [117], and ligands
of both nuclear receptors stimulate CYP3A expression in
human hepatocytes [118]. Interestingly, administration
of the PXR ligand rifampicin stimulates CYP3A4
expression in otherwise healthy patients with cholesterol
gallstones [119]. HNF4 appears to be critically involved
in PXR- and CAR-mediated transcriptional activation
of CYP3A in humans. A specific cis-acting element in the
CYP3A4 gene enhancer has been identified that confers
HNF4 binding, thus permitting PXR- and CARmediated gene activation [120]. Finally, a role for LXR
in CYP3A4 induction has been proposed recently [121].
Rodent Ugt1a1 (UDP-glucuronosyltransferase 1A1)mediated bilirubin glucuronoconjugation is also activated
by PXR and CAR [90,92]. The PXR activator rifampicin
[122] and the CAR activator phenobarbital [123] both
induced human UGT1A1 protein expression.
CAR [90], and to a lesser extent FXR [124], PXR
[125,126], VDR [127] and LXR [128], activate SULT2A1/
Sult2a1. This enzyme enhances bile acid sulfation and
further renal excretion of the sulfated bile salts. Finally,

C

The Authors Journal compilation


C

2008 Biochemical Society

both FXR [129] and PPAR [130] increase bile acid


glucuronidation by transactivating human UGT2B4.

Changes in transporter expression as a primary cause of


cholestasis
Changes in transporter expression in acquired hepatocellular cholestasis may represent the primary causal
factor of cholestatic dysfunction. Although adaptive
changes similar to those occurring in obstructive
cholestasis can also take place to protect the hepatocyte
against secondary manifestations of the cholestatic insult,
these adaptive changes may further aggravate the primary
secretory failure by reversing hepatocellular polarity.
Many acquired non-obstructive cholestatic disorders
involve primary changes in hepatocellular transporter
expression (hepatocellular cholestasis). They can be
divided in two large subgroups, namely (i) inflammationinduced cholestasis, which have cytokines as the common
deleterious effector, and (ii) pure (bland) cholestasis,
which is caused by drugs or hormones. It should,
however, be borne in mind that cholestatic diseases with
features of both obstructive and hepatocellular cholestasis
also exist. For example, PBC [75] and PSC [67], two
prototypical chronic cholestatic liver disease in humans,
develop bile duct obstruction by cholangiodestruction,
but inflammation-induced release of cytokines is also critically involved in the progression of the disease [131,132].

Inflammation-induced cholestasis
Pro-inflammatory cytokines produced as a consequence
of either infectious or non-infectious diseases are potent
inducers of intrahepatic cholestasis by acting as potent inhibitors of transporter expression [133]. Among the multiple pathologies in which inflammation-induced cholestasis is involved, sepsis-induced cholestasis is the prototypical one [134,135]. The cholestatic complication of
this disease has been associated with the action of the bacterial wall component LPS, and the consequent hepatic
release of the inflammatory cytokines IL1, TNF (tumour necrosis factor ) and IL6 [136]. Therefore LPS administration to rodents has been widely used as an experimental model to characterize sepsis-induced cholestasis.
This model was also helpful in understanding the cholestatic mechanisms of cytokines. This information is relevant to human cholestatic diseases other than sepsis, as cytokines are critical mediators of alcoholic, viral, autoimmune and drug-induced cholestatic hepatitis [133].
Protein expression of both Bsep and Mrp2 are
impaired in experimental sepsis in rodents [37,137,138],
although the alteration of Bsep appears to be less pronounced [62]. In rats, Mrp2, but not Bsep, expression
is modulated transcriptionally [37,137], whereas, in
humans, down-regulation of both BSEP and MRP2 is
post-transcriptional in nature, indicating marked species
differences [137].

Hepatocellular transport in acquired cholestasis

The decrease in canalicular transporter expression


in LPS-induced cholestasis in rats is accompanied by
transcriptional down-regulation of Ntcp [139,140] and
several Oatps [136,139,140]. Up-regulation of Mrp1
and Mrp3 was also reported [37,140], although the upregulation of Mrp3 was not observed by others [136,141].
At least part of these alterations occur in human liver
slices exposed to LPS, in particular those involving
NTCP transcriptional down-regulation [137]. This
event correlated inversely with TNF and IL1 levels
[137].
Down-regulation of Mrp2, Ntcp and Oatps in
rodents may be due, in part, to repressed transcriptional
expression of both RXR/RAR and HNF1 [142],
which are transactivators of these transporters. LPSinduced cytokine release may play a key role [141,142].
Indeed, depletion of Kuppfer cells, the main hepatic
source of cytokines, counteracted the decrease induced
by LPS in the binding activity of the transcription
factors RXR/RAR and HNF1, and the associated
decrease in Ntcp expression in rats [143]. Cytokines
released after LPS administration to rodents also reduce
the binding activity of RXR/RAR by inducing a
shift in RXR from the nucleus into the cytosol, which
induces the removal of the heterodimeric partner from
the nucleus [107]. Phosphorylation of RXR by JNK
(c-Jun N-terminal kinase) after LPS-induced stimulation
of Kupffer cells [144] has been proposed to trigger
this re-localization [107]. JNK activation has been also
proposed to be a key determinant in the decreased
binding activity of HNF1. This effect appears to be
secondary to a decrease in HNF4 promoter activity
and protein expression, with JNK-mediated HNF4
phosphorylation having a critical role [145].
Selective inhibition of different cytokines in LPSinduced cholestasis in vivo revealed that IL1 is the
major cytokine involved in these effects [146], whereas
IL6 only plays a crucial role during the later stages of
endotoxaemia [147]. However, mechanisms independent
of cytokines, and not involving the repression of RXR/
RAR and HNF1, may also occur [146]. For example,
LPS-induced PXR activation has been shown to
contribute to Mrp2 down-regulation in mice [148].
Alterations in hepatobiliary transport systems
have been reported in inflammation-induced acute
icteric cholestasis caused by cholestatic alcoholic hepatitis, cholestatic autoimmune hepatitis and drug-induced
hepatitis [65]. A decreased expression of basolateral
uptake transporters (NTCP and OATP2) and canalicular
export pumps (BSEP and MRP2) was reported. This
was accompanied by low mRNA levels of NTCP and
OATP2 and, to a lesser extent, of BSEP. In contrast,
MRP2 down-regulation appears to occur by post-transcriptional mechanisms. Finally, MRP3 expression is
maintained, which may protect hepatocytes from further
accumulation of potentially toxic biliary constituents.

Pure (bland) cholestasis


Causes of pure (bland) cholestasis include hormones
and hormone derivatives, such as oestrogens, C17 -substituted testosterone and anabolic steroids, as well as a large
number of cholestatic drugs [149]. Among the several
cholestatic pathologies belonging to the latter group,
oestrogen-induced cholestasis in rats and its clinical
correlate, obstetric cholestasis, were studied with more
detail in terms of hepatocellular transporter expression
and these will be focused upon below.
Oestrogens have been proposed to be a key causal
factor in intrahepatic cholestasis in susceptible women
during pregnancy [150], or following administration of
oral contraceptives and postmenopausal replacement
therapy [151]. Owing to this relevant clinical correlate,
cholestasis induced by oestrogens in rodents is a common
experimental model of hepatocellular cholestasis. The
cholestatic injury is produced either over the course
of a few days by administration of EE (17-ethynyloestradiol), a model oestrogen, or within minutes by
E2 17G (oestradiol 17-d-glucuronide), an oestrogen
metabolite.
In the EE-induced cholestatic model, Bsep expression
is impaired slightly [62,152], whereas Mrp2 expression is
decreased to a higher degree at the post-transcriptional
level [57]. EE also induces transcriptional down-regulation of Ntcp [32,152,153] and Oatps [32,153,154],
together with the up-regulation of Mrp3 [42,58,154]. The
nuclear receptor ER (oestrogen receptor ) has been
recently proposed to mediate, at least in part, EE-induced
cholestasis and the changes in transporter expression
induced by the cholestatic agent [155]. Indeed, ER /
mice lack the EE-induced repression of the mRNA
expression of Ntcp, Oatp1a1, Oatp1a4, Bsep and bileacid-biosynthesis enzymes (Cyp7a1, Cyp7b1 and
Cyp8b1) [155]. On the contrary, other nuclear receptors,
such us ER, FXR, SHP, PXR and CAR, appear not to be
involved [155]. However, a role for FXR or SHP in other
species cannot be ruled out. Unlike this study in mice, EE
down-regulates FXR and up-regulates SHP, via ER, in
rats [156,157]. In addition, an oestrogen-induced decrease
in nuclear binding activity of Ntcp transactivators, such as
HNF1, C/EBP (CAAT/enhancer binding protein) and
PXR, has also been proposed to play a role in the downregulation of Ntcp [153]. Finally, an indirect mechanism
by which oestrogens influence the secretion of pituitary
hormones which, in turn, modify liver gene expression,
has been proposed recently [157]. In summary, EE
induces changes in bile acid metabolism and transport
through: (i) an interaction with nuclear receptors,
such as ER, FXR and SHP, (ii) a indirect interaction
with pituitary (or pituitary-dependent) hormones, and
(iii) as yet unidentified nuclear-receptor- and pituitary-independent mechanisms.
Although oestrogens are thought to be key causal
factors in obstetric cholestasis [150], the extent to which

C

The Authors Journal compilation


C

2008 Biochemical Society

575

576

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

experimental EE-induced cholestasis in rats resembles


obstetric cholestasis during pregnancy is unknown.
Differences can be anticipated, however, since women
who develop obstetric cholestasis appear to have a
genetically linked susceptibility to the disease, and are
exposed to high levels of other hormones apart from
oestrogens (e.g., progesterone, prolactin and placental
lactogens) [13,150]. Owing to this complexity, animal
models of pregnancy-induced cholestasis are currently
unavailable, but the pattern of expression of transporters
in normal pregnant rats may provide some clues. As in
EE-induced cholestasis, transcriptional down-regulation
of Ntcp was apparent in pregnant rats, a finding
attributed to the down-regulation of HNF1 and
decreased promoter binding of RXR/RAR [158]. This
phenomenon was, however, not observed by others [159].
The limited, if any, decrease in Ntcp expression might
be attributed to the simultaneous stimulation of Ntcp
transcription by prolactin, an effect involving the binding
of Stat5 (signal transducer and activator of transcription 5)
to the Ntcp promoter [13,13,160]. Unlike in EE-induced
cholestasis, in which all Oatps were moderately downregulated, Oatp1a1 remains unchanged in pregnant rats,
whereas Oatp1a4 is only marginally decreased at both
the protein and mRNA levels [159]. Although a counterstimulatory effect of prolactin was not confirmed
for Oatp1a1 and Oatp1a4, the hormone up-regulates
Oatp1b2 expression via the Stat5 signal transduction
pathway [161]. The expression of basolateral export
pumps in pregnant rats also differs from that in
EE-induced cholestasis. Expression of both Mrp1
and Mrp3 is decreased at a post-transcriptional and
transcriptional level respectively [162]. Mrp6 mRNA
levels are also decreased [162]. Finally, at the canalicular
pole, Mrp2 expression was extensively reduced at a
post-transcriptional level, whereas Bsep expression is
not impaired. Bsep expression may be sustained by the
selective counter-stimulatory effect of prolactin on Bsep
expression [159]. Although moderate when compared
with the changes occurring in a pure oestrogen
model of cholestasis, the alterations in transporter
expression observed in pregnant rats suggest that
pregnancy represents an oestrogen-linked cholestaticprone condition that, when overlapping with subclinical
genetically linked alterations in bile formation in
susceptible women, may lead to overt cholestasis.

Manipulation of the expression of hepatocellular transporters


as a therapeutic strategy in cholestasis
The identification of a critical role for nuclear receptors
as mediators of the adaptive changes in hepatocellular
transporter expression in cholestasis has raised expectation about the employment of specific optimized ligands
of these nuclear receptors for the treatment of cholestatic
disorders [163165]. These new therapeutic approaches
are aimed not only at restoring the normal plasma-to
C

The Authors Journal compilation


C

2008 Biochemical Society

bile excretory route by stimulating defective transporter


expression, but also at enabling alternative elimination
routes. This knowledge has also provided new rationales
for the understanding of the beneficial effects of wellestablished anticholestatic agents, which have been used
in an empirical manner in the past.
Among the anticholestatic agents, UDC (ursodeoxycholate) is the prototypical one. Feeding UDC to
normal rodents stimulated hepatocellular expression of
both Mrp2 and Bsep by transcriptional and post-transcriptional mechanisms respectively [166]. Expression at
both mRNA and protein levels of both of the basolateral
efflux pumps Mrp3 [167] and Mrp4 [96] was increased,
whereas that of Oatp1a1 was decreased (Figure 3). On the
other hand, expression of the uptake transporter Ntcp
[166] and the basolateral export transporter OstOst
[96] remained unaffected. Feeding UDC also stimulates
the protein expression of Mrp2 [167] and Mrp4 [96]
efflux pumps in mice kidney, and that of Mrp2 [167],
Mrp3 [167] and Mrp4 [96] in mouse intestines. Intestinal
OstOst mRNA was also elevated [96]. Finally, the
induction of bile-salt-hydroxylating enzymes [95] and
repression of bile-salt-synthesizing enzymes [96] also
occurs after UDC administration. Taken together, this
response enhances the overall capacity of the body to
detoxify toxic biliary constituents.
The effects of UDC in rodents were only partially
reproduced in humans. When administered to otherwise
healthy patients with cholesterol gallstones, UDC stimulated post-transcriptionally the expression of BSEP
and MRP4, but not MRP2, MRP3, OATP1B1 or
bile-salt-metabolizing enzymes in liver [168]. However,
the situation in patients with cholestasis may be far
more complex. For example, a positive correlation was
observed between the hepatic enrichment of UDC in
liver of UDC-treated patients with PBC and the hepatic
levels of OATP1B1 and MRP2, whereas BSEP levels had
no correlation [66]. This suggests that, unlike healthy
subjects, patients with PBC lack UDC-stimulatory
mechanisms of BSEP overexpression. Furthermore,
UDC would improve MRP2 or OATP1B1 levels only
when their expression is impaired by the disease.
Some of the effects induced by UDC in rodents appear
to involve the activation of nuclear factors. With the exception of the induction of Bsep by UDC, which appears
to be FXR-dependent in mice [167], the remaining responses to UDC are FXR-independent and may involve,
in part, other nuclear factors, probably PXR [95]. As most
of these effects occur spontaneously in the cholestatic
liver, UDC administration to bile-duct-ligated mice only
have a limited protective response [13]. More specific and
effective therapies using newly synthesized ligands that
bind FXR with higher affinity, e.g. GW4064 and 6-ethyl
chenodeoxycholic acid, are being actively tested; they
might counteract the reduced expression and activity of
FXR in cholestasis, particularly in cholestasis induced by

Hepatocellular transport in acquired cholestasis

Figure 3 Beneficial effects of the anticholestatic therapeutic agent UDC on the expression of transporters and enzymes
involved in bile salt synthesis, and bile salt and bilirubin metabolism in the liver, kidney and intestines in normal rodents

At the hepatocellular level, UDC stimulates the expression of the canalicular transporters Bsep and Mrp2 and the basolateral efflux pumps Mrp3 and Mrp4. Expression
of Ntcp, Oatp1 and OstOst remain unaffected. The increased expression of Bsep has been attributed to UDC-induced activation of FXR, with the remaining
responses presumably involving other nuclear factors, such as PXR. UDC also induces beneficial changes in bile salt metabolism, particularly the repression of bile
acid synthesis from cholesterol and the stimulation of phase I bile acid metabolism, leading to more hydroxylated, less toxic, bile salts, which can be more easily
sulfated/glucuronidated and exported further via urine. This latter process is also stimulated by UDC by up-regulating both Mrp2 and Mrp4 efflux pumps in the kidney.
Finally, UDC up-regulates Mrp2, Mrp3 and Mrp4 efflux pumps in the intestine. Overexpression of the latter two efflux pumps contributes to preserve enterohepatic
bile salt recirculation. Note that the Figure shows the effects of UDC on healthy rodents. The extent to which these mechanisms are in operation in patients with
cholestasis is uncertain. See the text for further details.
pro-inflammatory cytokines (e.g. endotoxaemia) [169],
pregnancy [170] or oestrogens [156,157]. For example,
the FXR agonist 6-ethyl chenodeoxycholic acid has
beneficial effects in oestrogen-induced cholestasis [154],
a pathology that occurs with a decrease in FXR mRNA
levels [157]. This FXR ligand increases the expression of
Mrp2 and Bsep in EE-induced cholestasis and reduces
the transcriptional expression of Cyp7a1, Cyp8b1 and

Ntcp [154]. Although FXR-mediated stimulation of Bsep


expression can be beneficial in hepatocellular cholestasis
with impaired Bsep transport, it can be detrimental in obstructive cholestasis, where pressure in the obstructed bile
ducts can build up to hazardous levels. This explains why
FXR/ mice are more resilient to obstructive cholestasis
[97] and that UDC induces the FXR-mediated harmful
effects in bile-duct-ligated animals [55]. As most of the

C

The Authors Journal compilation


C

2008 Biochemical Society

577

578

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

relevant acquired cholestasis in humans, such as PBC


and PSC, develops an obstructive/cholangiodestructive
component, this factor may narrow the therapy window
for treatments with FXR ligands [171].
Apart from improving the expression of key transporters in certain cholestatic diseases, in others UDC
largely enhances the activity of transporters without
affecting expression. For example, EE impairs Mrp2
expression and the biliary excretion of Mrp2 substrates,
but UDC co-administration largely restored this alteration without counteracting the Mrp2 down-regulation
[172]. The same holds true for the anticholestatic
hepatoprotector silymarin [173]. This might involve a
direct positive interaction of the anticholestatic agents
either with a putative regulatory site of the transporter
or with its lipid microenvironment.
The capability of UDC to induce beneficial changes in
bile acid metabolism in rodents, particularly repression
of Cyp7a1 and stimulation of Cyp3a11 (the rodent
homologue of human CYP3A4), has proved to be
limited in humans, but can be complemented with
simultaneous administration of PXR agonists, such as
rifampicin [119]. Rifampicin, which has also been used
for a long time in the treatment of cholestatic diseases,
preferentially stimulated beneficial changes in bile acid
metabolism [119]. This drug also enhances bilirubin-conjugating capability (via UGT1A1), bilirubin excretion (via
MRP2) and bile acid/bilirubin glucuronide retrograde
exportation (via MRP3) [119]. It should be highlighted
again that these stimulatory effects have been tested in
patients without cholestasis, but these mechanism may be
different in patients with cholestasis. If confirmed under
cholestatic conditions, similar complementary effects
of UDC with CAR agonists, such as phenobarbital or
6,7-dimethylesculetin (the main active component of a
herbal concoction used in traditional YinChin medicine
to treat neonatal jaundice) are to be expected, as CAR
co-ordinately regulates a similar group of detoxifying
genes to those induced by PXR (see above). In line with
this, FXR, PXR and CAR agonists protected against
hepatic bile acid toxicity in a complementary manner in
rats [92,115]. This preliminary encouraging information
supports the implementation of clinical trials using combinations of specific ligands targeting multiple but
complementary nuclear receptors.

Post-translational alterations of
hepatobiliary transporters
Bile flow can be impaired by certain cholestatic agents
within hours or even minutes, a time period incompatible
with changes in transporter expression at the protein
level. These acute cholestatic changes account, for
example, for the rapid cholestatic effects induced by drugs
in patients [174]. Moreover, certain chronic cholestatic
conditions do not involve changes in transporter expressions of a suitable magnitude to explain the extent of bile

C

The Authors Journal compilation


C

2008 Biochemical Society

flow impairment, and anticholestatic agents may have beneficial effects without improving transporter expression.
In these cases, alterations in transporter intrinsic activity
or dynamic localization may play a crucial role [175].

Transporter inhibition by cholestatic drugs


A number of cholestatic agents cis-inhibit ATP-dependent Bsep-mediated bile salt transport in isolated rat
liver canalicular membrane vesicles, mostly in a competitive manner [174]. These agents include cyclosporin
A [176], glibenclamide [176], rifamycin [176], rifampicin [176], bosentan [177,178] and thiazolidinedione derivatives [179,180]. This effect was confirmed for human
BSEP with some of these compounds [178,181].
Flutamide was also shown to cis-inhibit BSEP [182].
On the other hand, some cholestatic compounds may
induce trans-inhibition of BSEP/Bsep. For example,
E2 17G has been suggested to trans-inhibit Bsep in rat
liver, as it requires an intact Mrp2-mediated E2 17Gtransporting activity to exert its cholestatic effect
[176,183]. This has been confirmed further in Xenopus
laevis oocytes co-expressing rat Bsep, the inhibition
mechanism being non-competitive in nature [183].
However, the relevance of this mechanism in the intact
liver is doubtful, as experimental manoeuvres that
increase the biliary E2 17G concentration attenuate
E2 17G-induced cholestasis [184,185], and others that
dilute the cholestatic compound in bile are not protective
at all [186]. The dependency of E2 17G-induced
cholestasis on intact Mrp2 activity, which indeed occurs
in vivo [187], can be explained alternatively by the Mrp2
inhibitory modulation of Bsep transport activity via, for
example, proteinprotein interactions.
PM4-S
(5-pregnan-3-ol-20-one
sulfate),
a
progesterone metabolite that accumulates in the serum of
women with pregnancy-induced cholestasis, has also
been shown to trans-inhibit rat Bsep in X. laevis oocytes
co-expressing rat Bsep in a non-competitive manner
[183]. This suggests that this metabolite impairs bile
flow in the isolated perfused rat liver, while being
excreted at concentrations higher than the K i for Bsep
trans-inhibition [183].

Changes in transporter localization


Impairment of transporter activity in cholestasis can also
occur by endocytic internalization of relevant canalicular
transporters, which re-localize into intracellular vesicular
structures, presumably the subapical endosomal compartment. If maintained with time in chronic cholestatic
conditions, this may lead to delivery of the protein to the
lysosomal compartment, followed by degradation [188].
This may contribute to the decreased post-transcriptional
expression of canalicular transporters shown to occur in
different cholestatic diseases.
Endocytic internalization of the canalicular transporters Mrp2 and Bsep occurs in several models of

Hepatocellular transport in acquired cholestasis

experimental cholestasis, including bile duct ligation


[188], hyperosmotic perfusion [189,190] and oxidative
challenge [191193]. Administration of cholestatic
compounds, such as LPS [190], E2 17G [194,195], TLC
(taurolithocholate) [196,197] and cyclosporine A [198],
also induces this effect. In addition, Mrp2 internalization
has been reported to occur in post-cold ischaemic
rat liver, a phenomenon dependent on Kupffer cell
activation, with thromboxane A2 being the most likely
mediator [199]. Together with endocytic internalization,
abnormal expression of this canalicular transporter in
the apical end of the lateral membrane was also shown to
occur under sustained E2 17G-induced cholestasis. This
was attributed to mis-sorting of endocytosed transportercontaining vesicles localized at the pericanalicular area
to the closer lateral membrane domain [200].
Changes in localization of canalicular export pumps
have been also shown to occur in human cholestatic
hepatopathies, including PBC [201], obstructive
extrahepatic cholestasis [61,202], PSC [202], autoimmune
hepatitis [202] and acute intrahepatic cholestasis induced
by drugs, such as antibiotics, tiopronin, chlorpromazine,
NSAIDs (non-steroidal anti-inflammatory drugs)
and antidepressants [202204]. In the latter case,
redistribution of Mrp2 occurs towards the basolateral
membrane rather than into intracellular vesicles [204].
The mechanism by which endocytosis of canalicular
transporters occurs remains largely unknown, although
it is known that the process is microtubule-independent
[205]. Alterations of actin cytoskeletal integrity onset
transporter endocytosis [206], although retrieval of
canalicular transporters can also occur with intact
actin organization [195,197]. Proteins involved in the
cross-linking of actin filaments with plasma membrane
proteins, such as the ERM (ezrin/radixin/moesin) family
of proteins, may be altered in these cases, without any
apparent impairment of actin organization [207,208].
Interestingly, a deficient localization of radixin at the
canalicular membrane occurs in patients with PBC, which
is associated with retrieval of MRP2, MRP1, MDR3
and BSEP, together with co-localization of these carriers
with radixin in intracellular vesicles [201]. This finding
was confirmed for MRP2 in other human cholestatic
diseases, such as obstructive jaundice, PSC, autoimmune
hepatitis and drug-induced liver injury [202].
Accumulating evidence indicates that changes in canalicular transporter localization occurring in cholestasis
depend upon activation of critical signalling pathways
(Figure 4). One of the pathways involves classical Ca2+ dependent PKC (protein kinase C) isoforms (mainly
PKC in hepatocytes). Selective activation of Ca2+ dependent PKCs induces cholestasis and retrieval of Bsep
from the canalicular membrane and cholestasis in the
isolated perfused rat liver [209]. PKC activation also
induces redistribution of Mrp2 from the canalicular to
the basolateral membrane in HepG2 cells [210]. A critical

participation of Ca2+ -dependent PKCs in the retrieval of


Bsep and the associated bile salt secretory failure in tBOOH (t-butylhydroperoxide)-induced oxidative stress
has been reported recently by our group [193]. However,
the kind of canalicular protein that is internalized under
oxidative stress conditions and the signalling molecule(s)
involved appear to depend on the pro-oxidant agent employed. For example, the oxidizing compound ethacrynic
acid, which does not translocate classical but rather
novel PKC isoforms, internalizes Mrp2 selectively
without affecting Bsep [192]. The novel PKC isoform
PKC is also activated in TLC-induced cholestasis and
has been suggested to be involved in TLC-induced
cholestatic effect [211]. This phenomenon occurs in a
PI3K (phosphoinositide 3-kinase)-dependent manner, as
products of PI3K are potent activators of PKC [212].

Experimental therapeutic approaches


based upon the modulation of
post-translational alterations of
transporters in cholestasis
Direct transporter inhibition by cholestatic drugs is, in
most cases, reversed by withdrawing the drug, due to
the reversible nature of the phenomenon [213]. Similarly,
mechanisms involving changes in transporter localization
can be spontaneously reversed if the cholestatic insult is
transient. For example, canalicular transporters that are
internalized by a rapid bolus administration of E2 17G
are spontaneously re-targeted to the canalicular membrane by a microtubule-dependent mechanism within
2 h [214]. Some experimental therapeutic approaches
have been designed to prevent transporter internalization
and/or accelerate this re-insertion in order to avoid
irreversible consequences of sustained internalization, as
outlined below.

cAMP
cAMP is a second messenger that stimulates hepatocellular exocytic pathways [215] and partially prevents
the impairment of bile flow and internalization of ABC
transporters in E2 17G- and TLC-induced cholestasis.
Reduction in bile flow and activity of Bsep [195] and
Mrp2 [194] in the acute phase of E2 17G-induced cholestasis is partially prevented by the membrane-permeant
cAMP analogue dibutyryl cAMP. More significantly,
dibutyryl cAMP shortened the spontaneous recovery to
normal bile flow, Mrp2 function and Mrp2 localization
[194]. A similar acceleration of the re-insertion of endocytosed transporters has also been described by us for
Bsep in TLC-induced cholestasis [197]. In the IRHC
(isolated rat hepatocyte couplet) model, a preventive
effect of dibutyryl cAMP was observed on both E2 17G[195,216] and TLC- [197,216] induced Bsep relocation.
In this case, however, prevention by dibutyryl cAMP
was complete. This protective effect was significantly

C

The Authors Journal compilation


C

2008 Biochemical Society

579

580

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

Figure 4 Post-translational alterations in hepatobiliary transporters in cholestasis

Cholestatic drugs may cis- or trans -inhibit BSEP/Bsep or induce endocytic retrieval of canalicular transporters. Cyclosporin A, glibenclamide, rifamycin, rifampicin,
bosentan, thiazolidinedione derivatives and flutamide all cis-inhibit either or both Bsep and BSEP, whereas PM4-S and E2 17G trans-inhibit the transporter in a
non-competitive manner. Alternatively, E2 17G inhibition can be explained by Mrp2 inhibitory modulation of Bsep transport activity via proteinprotein interaction, while
E2 17G is being transported by Mrp2. A number of cholestatic manoeuvres in rats also lead to endocytic internalization of canalicular transporters into the subapical
compartment (SAC); this may lead to delivery to the lysosomal compartment, followed by degradation. Canalicular transporter retrieval has also been confirmed in
several human cholestatic diseases (see text for details). Several signalling pathways have been proposed to mediate this retrieval in experimental models of cholestasis.
Activation of PKC, when activated by ethacrynic acid or TLC, induces transporter internalization in rats. In addition, activation of PKC in t -BOOH-induced oxidative
stress in rats leads to transporter retrieval as well. Experimental therapeutic manoeuvres may prevent the retrieval of the transporters or accelerate exocytic re-insertion.
Increases in intracellular cAMP levels induced by administration of the permeant cAMP analogue dibutyryl cAMP or by the phosphodiesterase inhibitor silibinin prevents
internalization and accelerates re-insertion, via cytosolic Ca2+ elevation, in rat cholestatic hepatocytes. On the other hand, TUDC prevents transporter endocytosis,
probably via ERK- and p38 MAPK-dependent pathways.
blocked by the Ca2+ chelator BAPTA/AM [1,2-bis-(oaminophenoxy)ethane-N,N,N ,N -tetra-acetic acid tetrakis(acetoxymethyl ester)], but not by the PKA (protein
kinase A) inhibitor KT5720, suggesting the involvement
of Ca2+ or Ca2+ -dependent signalling pathways in the
protective effect of dibutyryl cAMP. A similar protective
effect in terms of the signalling modulators involved was
afforded by silibinin, the active component of the hepatoprotector silymarin. This was most probably due to the
ability of silibinin to inhibit cAMP phosphodiesterase,
thus increasing endogenous cAMP levels [216].

TUDC (tauroUDC)
TUDC, the naturally occuring taurine conjugate of
UDC, stimulates exocytosis in the cholestatic liver [217]
and counteracts endocytic internalization of both Bsep
[218] and Mrp2 [196] in TLC-induced cholestasis. The
Ca2+ -sensitive PKC isoform PKC has been proposed
to mediate the anticholestatic effects of TUDC against
TLC-induced cholestasis [196]. This is in apparent
contradiction with more recent findings that PKC
is cholestatic rather than hepatoprotective [209]. In
previous studies, pan-specific PKC inhibitors were used,
and the biological response evoked by the interplay
between different PKC isoforms may be different from
that evoked by just one of them. Furthermore, TUDC

C

The Authors Journal compilation


C

2008 Biochemical Society

activates the members of the MAPK (mitogen-activated


protein kinase) family ERK (extracellular-signalregulated kinase) [219] and p38 MAPK [220], and the
cholestatic effect of PKC may be overridden by the
choleretic effects of these signal transduction pathways.

CONCLUDING REMARKS AND FUTURE


DIRECTIONS
The recent enormous progress in molecular biology has
prompted the identification and characterization of several novel ATP-dependent and ATP-independent hepatocellular transporters, including many critically involved
in bile flow generation. This new information has been
quickly transferred to the liver physiopathology area,
providing an extensive understanding of the alterations
accounting for cholestatic diseases at the molecular level.
In acquired hepatocellular cholestasis, these alterations
may involve early changes in the dynamic localization
of canalicular transporters, followed by long-term transcriptional or post-transcriptional factors affecting
transporter synthesis. As a consequence of these primary
alterations, an adaptive response occurs to minimize the
detrimental effects of biliary compounds retained due
to the secretory failure. This includes decreased uptake,

Hepatocellular transport in acquired cholestasis

increased biotransformation to less toxic metabolites,


decreased expression of canalicular transporters and
enhanced expression of alternative basolateral pumps,
which export these metabolites into the blood for further
renal excretion. This adaptive response is promoted by
the co-ordinated expression of a complex network of
nuclear receptors and hepatocyte-enriched transcription
factors, whose activity is modulated by biliary metabolites retained by the cholestatic failure. These adaptive
changes have been largely characterized in animal models
of experimental cholestasis. The occurrence of a similar
protective response in human cholestatic hepatopathies is
becoming increasingly apparent, but species differences
in the mechanisms of action (e.g. post-transcriptional in
humans compared with transcriptional in rodents)
appears to exist and will require characterization in each
case.
This adaptive spontaneous mechanism is, however,
not always sufficient to prevent hepatocellular damage
and therapeutic intervention is often required. A number
of newly designed therapeutic strategies are currently
being explored to enhance adaptability by targeted
activation of nuclear receptors with high-affinity specific
ligands. Furthermore, the recognition of the involvement
of these transcription factors in the anticholestatic
effects of remedies classically used in the treatment of
cholestatic disease (e.g. UDC and rifampicin) has added
a rationale to their usage and extended their applications.
The development of new tailor made strategies for
each hepatopathy by targeting specific nuclear receptors
according to the particular deficiency is a key future challenge for both basic and clinical hepatologists. This goal
will require characterization of the impairments in transporter and/or nuclear receptor expression in each human
cholestatic disease. The finding of more selective potent
nuclear receptor ligands for an appropriate gene control
will be also an area of active research in a near future.
Finally, a number of compounds have been tested
in animal models of cholestasis to prevent/revert the
endocytic internalization of canalicular transporters
that occurs shortly after cholestasis. These experimental
therapeutic strategies are based either on the wellrecognized capability of the therapeutic agents to favour
exocytic carrier re-insertion (e.g. dibutyryl cAMP and
TUDC) or to counteract potentially cholestatic signalling
pathways (e.g. PKC or PI3K inhibitors). Taking into
account that post-transcriptional changes involving
transporter retrieval are increasingly recognized in
cholestatic disease in humans and that they may account
for post-transcriptional changes in this hepatopathy,
this strategy may effectively complement the effect of
inducers of carrier expression by co-operatively assuring
proper localization and constitutive expression. Whether
such a concerted beneficial action will occur effectively
in patients with cholestatic hepatopathies remains to be
ascertained, and this will represent a challenge for clinical

researchers in the way to explore better therapeutic


alternatives in cholestatic liver disease.

ACKNOWLEDGMENTS
This work was supported by CONICET (Consejo
Nacional de Investigaciones Cientficas y Tecnicas; PIP

6442), and ANPCyT (Agencia Nacional de Promocion

Cientfica y Tecnologica; PICT 05-26115), Argentina.

REFERENCES
1 Masyuk, A. I. and LaRusso, N. F. (2006) Aquaporins in
the hepatobiliary system. Hepatology 43 (Suppl. 1),
S75S81
2 Marinelli, R. A., Gradilone, S. A., Carreras, F. I.,
Calamita, G. and Lehmann, G. L. (2004) Liver
aquaporins: significance in canalicular and ductal bile
formation. Ann. Hepatol. 3, 130136
3 Ballatori, N. and Truong, A. T. (1992) Glutathione as a
primary osmotic driving force in hepatic bile formation.
Am. J. Physiol. 263, G617G624
4 Hardison, W. G. and Wood, C. A. (1978) Importance of
bicarbonate in bile salt independent fraction of bile flow.
Am. J. Physiol. 235, E158E164
5 Marinelli, R. A. and LaRusso, N. F. (1996) Solute and
water transport pathways in cholangiocytes.
Semin. Liver Dis. 16, 221229
6 Bohan, A. and Boyer, J. L. (2002) Mechanisms of hepatic
transport of drugs: implications for cholestatic drug
reactions. Semin. Liver Dis. 22, 123136
7 Kullak-Ublick, G. A., Stieger, B., Hagenbuch, B. and
Meier, P. J. (2000) Hepatic transport of bile salts. Semin.
Liver Dis. 20, 273292
8 Pauli-Magnus, C., Stieger, B., Meier, Y., Kullak-Ublick,
G. A. and Meier, P. J. (2005) Enterohepatic transport of
bile salts and genetics of cholestasis. J. Hepatol. 43,
342357
9 Briz, O., Serrano, M. A., MacIas, R. I.,
Gonzalez-Gallego, J. and Marin, J. J. (2003) Role of
organic anion-transporting polypeptides, OATP-A,
OATP-C and OATP-8, in the human placenta-maternal
liver tandem excretory pathway for foetal bilirubin.
Biochem. J. 371, 897905
10 Cui, Y., Konig, J., Leier, I., Buchholz, U. and Keppler, D.
(2001) Hepatic uptake of bilirubin and its conjugates by
the human organic anion transporter SLC21A6.
J. Biol. Chem. 276, 96269630
11 Wang, P., Kim, R. B., Chowdhury, J. R. and Wolkoff,
A. W. (2003) The human organic anion transport protein
SLC21A6 is not sufficient for bilirubin transport.
J. Biol. Chem. 278, 2069520699
12 Trauner, M. and Boyer, J. L. (2003) Bile salt transporters:
molecular characterization, function, and regulation.
Physiol. Rev. 83, 633671
13 Geier, A., Wagner, M., Dietrich, C. G. and Trauner, M.
(2007) Principles of hepatic organic anion transporter
regulation during cholestasis, inflammation and liver
regeneration. Biochim. Biophys. Acta 1773, 283308
14 Kamisako, T., Kobayashi, Y., Takeuchi, K. et al. (2000)
Recent advances in bilirubin metabolism research: the
molecular mechanism of hepatocyte bilirubin transport
and its clinical relevance. J. Gastroenterol. 35, 659664
15 Suchy, F. J. and Ananthanarayanan, M. (2006) Bile salt
excretory pump: biology and pathobiology. J. Pediatr.
Gastroenterol. Nutr. 43, (Suppl. 1), S10S16


C

The Authors Journal compilation


C

2008 Biochemical Society

581

582

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

16 Akita, H., Suzuki, H., Ito, K. et al. (2001)


Characterization of bile acid transport mediated by
multidrug resistance associated protein 2 and bile
salt export pump. Biochim. Biophys. Acta 1511,
716
17 Ballatori, N. and Dutczak, W. J. (1994) Identification and
characterization of high and low affinity transport
systems for reduced glutathione in liver cell canalicular
membranes. J. Biol. Chem. 269, 1973119737
18 Yang, B. and Hill, C. E. (2001) Nifedipine modulation of
biliary GSH and GSSG/conjugate efflux in normal and
regenerating rat liver. Am. J. Physiol. Gastrointest.
Liver Physiol. 281, G85G94
19 Krenhuber, H. G., Felberbauer, F. X. and Graf, J. (1997) Is
intracellular pH and/or intracellular bicarbonate a
determinant of bile salt independent canalicular bile
formation? The subject revisited. Yale J. Biol. Med. 70,
459469
20 Bogert, P. T. and LaRusso, N. F. (2007) Cholangiocyte
biology. Curr. Opin. Gastroenterol. 23, 299305
21 Nyberg, B., Sonnenfeld, T. and Einarsson, K. (1991)
Vasoactive intestinal peptide and secretin: effects of
combined and separate intravenous infusions on bile
secretion in man. Scand. J. Gastroenterol. 26,
109118
22 Cho, W. K. and Boyer, J. L. (1999) Vasoactive intestinal
polypeptide is a potent regulator of bile secretion from rat
cholangiocytes. Gastroenterology 117, 420428
23 Kortz, W. J., Nashold, J. R., Delong, E. and Meyers, W. C.
(1986) Effects of bombesin on fasting bile formation.
Ann. Surg. 203, 17
24 Cho, W. K., Mennone, A., Rydberg, S. A. and Boyer, J. L.
(1997) Bombesin stimulates bicarbonate secretion from
rat cholangiocytes: implications for neural regulation of
bile secretion. Gastroenterology 113, 311321
25 Xia, X., Francis, H., Glaser, S., Alpini, G. and Lesage, G.
(2006) Bile acid interactions with cholangiocytes.
World J. Gastroenterol. 12, 35533563
26 Lee, J. and Boyer, J. L. (2000) Molecular alterations in
hepatocyte transport mechanisms in acquired cholestatic
liver disorders. Semin. Liver Dis. 20, 373384
27 Trottier, J., Milkiewicz, P., Kaeding, J., Verreault, M. and
Barbier, O. (2006) Coordinate regulation of hepatic bile
acid oxidation and conjugation by nuclear receptors.
Mol. Pharm. 3, 212222
28 Ogawa, K., Suzuki, H., Hirohashi, T. et al. (2000)
Characterization of inducible nature of MRP3 in rat liver.
Am. J. Physiol. Gastrointest. Liver Physiol. 278,
G438G446
29 Gartung, C., Ananthanarayanan, M., Rahman, M. A. et al.
(1996) Down-regulation of expression and function of the
rat liver Na+ /bile acid cotransporter in extrahepatic
cholestasis. Gastroenterology 110, 199209
30 Moseley, R. H., Wang, W., Takeda, H. et al. (1996) Effect
of endotoxin on bile acid transport in rat liver: a potential
model for sepsis-associated cholestasis. Am. J. Physiol.
271, G137G146
31 Dumont, M., Jacquemin, E., DHont, C. et al. (1997)
Expression of the liver Na+ -independent organic anion
transporting polypeptide (oatp-1) in rats with bile duct
ligation. J. Hepatol. 27, 10511056
32 Simon, F. R., Fortune, J., Iwahashi, M., Gartung, C.,
Wolkoff, A. W. and Sutherland, E. (1996) Ethinylestradiol
cholestasis involves alterations in expression of liver
sinusoidal transporters. Am. J. Physiol. 271,
G1043G1052
33 Stedman, C. A., Liddle, C., Coulter, S. A. et al. (2005)
Nuclear receptors constitutive androstane receptor and
pregnane X receptor ameliorate cholestatic liver injury.
Proc. Natl. Acad. Sci. U.S.A. 102, 20632068


C

The Authors Journal compilation


C

2008 Biochemical Society

34 Slitt, A. L., Allen, K., Morrone, J. et al. (2007) Regulation


of transporter expression in mouse liver, kidney, and
intestine during extrahepatic cholestasis. Biochim.
Biophys. Acta 1768, 637647
35 Li, L., Meier, P. J. and Ballatori, N. (2000) Oatp2
mediates bidirectional organic solute transport: a role
for intracellular glutathione. Mol. Pharmacol. 58,
335340
36 Pei, Q. L., Kobayashi, Y., Tanaka, Y. et al. (2002)
Increased expression of multidrug resistance-associated
protein 1 (mrp1) in hepatocyte basolateral membrane and
renal tubular epithelia after bile duct ligation in rats.
Hepatol. Res. 22, 5864
37 Vos, T. A., Hooiveld, G. J. E. J., Koning, H. et al. (1998)
Up-regulation of the multidrug resistance genes, Mrp1
and Mdr1b, and down-regulation of the organic anion
transporter, Mrp2, and the bile salt transporter, Spgp, in
endotoxemic rat liver. Hepatology 28, 16371644
38 Tanaka, Y., Kobayashi, Y., Gabazza, E. C. et al. (2002)
Increased renal expression of bilirubin glucuronide
transporters in a rat model of obstructive jaundice.
Am. J. Physiol. Gastrointest. Liver Physiol. 282,
G656G662
39 Donner, M. G., Warskulat, U., Saha, N. and Haussinger,
D. (2004) Enhanced expression of basolateral multidrug
resistance protein isoforms Mrp3 and Mrp5 in rat liver by
LPS. Biol. Chem. 385, 331339
40 Soroka, C. J., Lee, J. M., Azzaroli, F. and Boyer, J. L.
(2001) Cellular localization and up-regulation of
multidrug resistance-associated protein 3 in hepatocytes
and cholangiocytes during obstructive cholestasis in rat
liver. Hepatology 33, 783791
41 Donner, M. G. and Keppler, D. (2001) Up-regulation of
basolateral multidrug resistance protein 3 (Mrp3) in
cholestatic rat liver. Hepatology 34, 351359
42 Ruiz, M. L., Villanueva, S. S., Luquita, M. G., Vore, M.,
Mottino, A. D. and Catania, V. A. (2006) Ethynylestradiol
increases expression and activity of rat liver Mrp3.
Drug Metab. Dispos. 34, 10301034
43 Belinsky, M. G., Dawson, P. A., Shchaveleva, I. et al.
(2005) Analysis of the in vivo functions of Mrp3.
Mol. Pharmacol. 68, 160168
44 Mennone, A., Soroka, C. J., Cai, S. Y. et al. (2006)
Mrp4/ mice have an impaired cytoprotective response
in obstructive cholestasis. Hepatology 43, 10131021
45 Denk, G. U., Soroka, C. J., Takeyama, Y., Chen, W. S.,
Schuetz, J. D. and Boyer, J. L. (2004) Multidrug
resistance-associated protein 4 is up-regulated in liver but
down-regulated in kidney in obstructive cholestasis in the
rat. J. Hepatol. 40, 585591
46 Zeng, H., Liu, G., Rea, P. A. and Kruh, G. D. (2000)
Transport of amphipathic anions by human multidrug
resistance protein 3. Cancer Res. 60, 47794784
47 Akita, H., Suzuki, H., Hirohashi, T., Takikawa, H. and
Sugiyama, Y. (2002) Transport activity of human MRP3
expressed in Sf9 cells: comparative studies with rat
MRP3. Pharm. Res. 19, 3441
48 Rius, M., Nies, A. T., Hummel-Eisenbeiss, J.,
Jedlitschky, G. and Keppler, D. (2003) Cotransport of
reduced glutathione with bile salts by MRP4 (ABCC4)
localized to the basolateral hepatocyte membrane.
Hepatology 38, 374384
49 Boyer, J. L., Trauner, M., Mennone, A. et al. (2006)
Upregulation of a basolateral FXR-dependent bile acid
efflux transporter OST-OST in cholestasis in humans
and rodents. Am. J. Physiol. Gastrointest. Liver Physiol.
290, G1124-G1130
50 Zelcer, N., van de, W. K., de, W. R. et al. (2006) Mice
lacking Mrp3 (Abcc3) have normal bile salt transport, but
altered hepatic transport of endogenous glucuronides.
J. Hepatol. 44, 768775

Hepatocellular transport in acquired cholestasis

51 Lee, J., Azzaroli, F., Wang, L. et al. (2001) Adaptive


regulation of bile salt transporters in kidney and liver in
obstructive cholestasis in the rat. Gastroenterology 121,
14731484
52 Brandoni, A., Villar, S. R., Picena, J. C., Anzai, N.,
Endou, H. and Torres, A. M. (2006) Expression of rat
renal cortical OAT1 and OAT3 in response to acute
biliary obstruction. Hepatology 43, 10921100
53 Wagner, M., Fickert, P., Zollner, G. et al. (2003) Role of
farnesoid X receptor in determining hepatic ABC
transporter expression and liver injury in bile duct-ligated
mice. Gastroenterology 125, 825838
54 Marschall, H. U., Wagner, M., Bodin, K. et al. (2006)
Fxr/ mice adapt to biliary obstruction by enhanced
phase I detoxification and renal elimination of bile acids.
J. Lipid Res. 47, 582592
55 Fickert, P., Zollner, G., Fuchsbichler, A. et al. (2002)
Ursodeoxycholic acid aggravates bile infarcts in bile
duct-ligated and Mdr2 knockout mice via disruption of
cholangioles. Gastroenterology 123, 12381251
56 Lamireau, T., Zoltowska, M., Levy, E. et al. (2003) Effects
of bile acids on biliary epithelial cells: proliferation,
cytotoxicity, and cytokine secretion. Life Sci. 72,
14011411
57 Trauner, M., Arrese, M., Soroka, C. J. et al. (1997) The rat
canalicular conjugate export pump (Mrp2) is
down-regulated in intrahepatic and obstructive
cholestasis. Gastroenterology 113, 255264
58 Kamisako, T. and Ogawa, H. (2005) Alteration of the
expression of adenosine triphosphate-binding cassette
transporters associated with bile acid and cholesterol
transport in the rat liver and intestine during cholestasis.
J. Gastroenterol. Hepatol. 20, 14291434
59 Kanno, K., Tazuma, S., Niida, S. and Chayama, K. (2003)
Unique reciprocal changes of hepatocellular membrane
transporter expression and fluidity in rats with selective
biliary obstruction. Hepatol. Res. 26, 157163
60 Yamada, T., Arai, T., Nagino, M. et al. (2005) Impaired
expression of hepatic multidrug resistance protein 2 is
associated with posthepatectomy hyperbilirubinemia in
patients with biliary cancer. Langenbecks Arch. Surg. 390,
421429
61 Shoda, J., Kano, M., Oda, K. et al. (2001) The expression
levels of plasma membrane transporters in the cholestatic
liver of patients undergoing biliary drainage and their
association with the impairment of biliary secretory
function. Am. J. Gastroenterol. 96, 33683378
62 Lee, J. M., Trauner, M., Soroka, C. J., Stieger, B., Meier,
P. J. and Boyer, J. L. (2000) Expression of the bile salt
export pump is maintained after chronic cholestasis in the
rat. Gastroenterology 118, 163172
63 Desmet, V. J. (1992) Modulation of the liver in cholestasis.
J. Gastroenterol. Hepatol. 7, 313323
64 Sauer, P., Stiehl, A., Fitscher, B. A. et al. (2000)
Downregulation of ileal bile acid absorption in
bile-duct-ligated rats. J. Hepatol. 33, 28
65 Zollner, G., Fickert, P., Zenz, R. et al. (2001)
Hepatobiliary transporter expression in percutaneous
liver biopsies of patients with cholestatic liver diseases.
Hepatology 33, 633646
66 Zollner, G., Fickert, P., Silbert, D. et al. (2003) Adaptive
changes in hepatobiliary transporter expression in
primary biliary cirrhosis. J. Hepatol. 38, 717727
67 Oswald, M., Kullak-Ublick, G. A., Paumgartner, G. and
Beuers, U. (2001) Expression of hepatic transporters
OATP-C and MRP2 in primary sclerosing cholangitis.
Liver 21, 247253
68 Keitel, V., Burdelski, M., Warskulat, U. et al. (2005)
Expression and localization of hepatobiliary transport
proteins in progressive familial intrahepatic cholestasis.
Hepatology 41, 11601172

69 Shneider, B. L., Fox, V. L., Schwarz, K. B. et al. (1997)


Hepatic basolateral sodium-dependent-bile acid
transporter expression in two unusual cases of
hypercholanemia and in extrahepatic biliary atresia.
Hepatology 25, 11761183
70 Ros, J. E., Libbrecht, L., Geuken, M., Jansen, P. L. and
Roskams, T. A. (2003) High expression of MDR1, MRP1,
and MRP3 in the hepatic progenitor cell compartment
and hepatocytes in severe human liver disease. J. Pathol.
200, 553560
71 Barnes, S. N., Aleksunes, L. M., Augustine, L. et al. (2007)
Induction of hepatobiliary efflux transporters in
acetaminophen-induced acute liver failure cases.
Drug Metab. Dispos. 35, 19631969
72 Zollner, G., Wagner, M., Fickert, P. et al. (2007)
Expression of bile acid synthesis and detoxification
enzymes and the alternative bile acid efflux pump MRP4
in patients with primary biliary cirrhosis. Liver Int. 27,
920929
73 Scheffer, G. L., Kool, M., de Haas, M. et al. (2002) Tissue
distribution and induction of human multidrug resistant
protein 3. Lab. Invest. 82, 193201
74 Briz, O., Romero, M. R., Martinez-Becerra, P. et al. (2006)
OATP8/1B3-mediated cotransport of bile acids and
glutathione: an export pathway for organic anions from
hepatocytes? J. Biol. Chem. 281, 3032630335
75 Kullak-Ublick, G. A., Baretton, G. B., Oswald, M.,
Renner, E. L., Paumgartner, G. and Beuers, U. (2002)
Expression of the hepatocyte canalicular multidrug
resistance protein (MRP2) in primary biliary cirrhosis.
Hepatol. Res. 23, 7882
76 Kogan, D., Ananthanarayanan, M., Emre, S., Suchy, F. J.
and Shneider, B. L. (1999) The bile salt export pump
(BSEP/SPGP) is not down-regulated in human cholestasis
associated with extrahepatic biliary atresia (EHBA).
Hepatology 30, 468A
77 Strazzabosco, M., Fabris, L. and Spirli, C. (2005)
Pathophysiology of cholangiopathies.
J. Clin. Gastroenterol. 39, S90-S102
78 Hruz, P., Zimmermann, C., Gutmann, H. et al. (2006)
Adaptive regulation of the ileal apical sodium dependent
bile acid transporter (ASBT) in patients with obstructive
cholestasis. Gut 55, 395402
79 Huang, W., Zhang, J., Chua, S. S. et al. (2003) Induction of
bilirubin clearance by the constitutive androstane receptor
(CAR). Proc. Natl. Acad. Sci. U.S.A. 100, 41564161
80 Staudinger, J. L., Goodwin, B., Jones, S. A. et al. (2001)
The nuclear receptor PXR is a lithocholic acid sensor
that protects against liver toxicity. Proc. Natl. Acad.
Sci. U.S.A. 98, 33693374
81 Parks, D. J., Blanchard, S. G., Bledsoe, R. K. et al. (1999)
Bile acids: natural ligands for an orphan nuclear receptor.
Science 284, 13651368
82 Zollner, G., Wagner, M., Fickert, P. et al. (2005) Role of
nuclear receptors and hepatocyte-enriched transcription
factors for ntcp repression in biliary obstruction in mouse
liver. Am. J. Physiol. Gastrointest. Liver Physiol. 289,
G798G805
83 Denson, L. A., Sturm, E., Echevarria, W. et al. (2001) The
orphan nuclear receptor, Shp, mediates bile acid-induced
inhibition of the rat bile acid transporter, Ntcp.
Gastroenterology 121, 140147
84 Jung, D., Hagenbuch, B., Fried, M., Meier, P. J. and
Kullak-Ublick, G. A. (2004) Role of liver-enriched
transcription factors and nuclear receptors in regulating
the human, mouse, and rat NTCP gene. Am. J. Physiol.
Gastrointest. Liver Physiol. 286, G752G761
85 Goodwin, B., Jones, S. A., Price, R. R. et al. (2000) A
regulatory cascade of the nuclear receptors FXR, SHP-1,
and LRH-1 represses bile acid biosynthesis. Mol. Cell 6,
517526


C

The Authors Journal compilation


C

2008 Biochemical Society

583

584

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

86 Wang, L., Lee, Y. K., Bundman, D. et al. (2002)


Redundant pathways for negative feedback regulation of
bile acid production. Dev. Cell 2, 721731
87 Zollner, G., Marschall, H. U., Wagner, M. and Trauner, M.
(2006) Role of nuclear receptors in the adaptive response
to bile acids and cholestasis: pathogenetic and therapeutic
considerations. Mol. Pharm. 3, 231251
88 Jung, D., Hagenbuch, B., Gresh, L., Pontoglio, M.,
Meier, P. J. and Kullak-Ublick, G. A. (2001)
Characterization of the human OATP-C (SLC21A6) gene
promoter and regulation of liver-specific OATP genes by
hepatocyte nuclear factor 1 alpha. J. Biol. Chem. 276,
3720637214
89 Jung, D. and Kullak-Ublick, G. A. (2003) Hepatocyte
nuclear factor 1: a key mediator of the effect of bile
acids on gene expression. Hepatology 37, 622631
90 Wagner, M., Halilbasic, E., Marschall, H. U. et al. (2005)
CAR and PXR agonists stimulate hepatic bile acid and
bilirubin detoxification and elimination pathways in mice.
Hepatology 42, 420430
91 Cherrington, N. J., Hartley, D. P., Li, N., Johnson, D. R.
and Klaassen, C. D. (2002) Organ distribution of
multidrug resistance proteins 1, 2, and 3 (Mrp1, 2, and 3)
mRNA and hepatic induction of Mrp3 by constitutive
androstane receptor activators in rats. J. Pharmacol.
Exp. Ther. 300, 97104
92 Zhang, J., Huang, W., Qatanani, M., Evans, R. M. and
Moore, D. D. (2004) The constitutive androstane receptor
and pregnane X receptor function coordinately to prevent
bile acid-induced hepatotoxicity. J. Biol. Chem. 279,
4951749522
93 Teng, S., Jekerle, V. and Piquette-Miller, M. (2003)
Induction of ABCC3 (MRP3) by pregnane X receptor
activators. Drug Metab. Dispos. 31, 12961299
94 Bohan, A., Chen, W. S., Denson, L. A., Held, M. A. and
Boyer, J. L. (2003) Tumor necrosis factor -dependent
up-regulation of Lrh-1 and Mrp3(Abcc3) reduces liver
injury in obstructive cholestasis. J. Biol. Chem. 278,
3668836698
95 Schuetz, E. G., Strom, S., Yasuda, K. et al. (2001)
Disrupted bile acid homeostasis reveals an unexpected
interaction among nuclear hormone receptors,
transporters, and cytochrome P450. J. Biol. Chem. 276,
3941139418
96 Zollner, G., Wagner, M., Moustafa, T. et al. (2006)
Coordinated induction of bile acid detoxification and
alternative elimination in mice: role of FXR-regulated
organic solute transporter-/ in the adaptive response to
bile acids. Am. J. Physiol. Gastrointest. Liver Physiol.
290, G923-G932
97 Stedman, C., Liddle, C., Coulter, S. et al. (2006) Benefit
of farnesoid X receptor inhibition in obstructive
cholestasis. Proc. Natl. Acad. Sci. U.S.A. 103,
1132311328
98 Chen, W., Cai, S. Y., Xu, S., Denson, L. A., Soroka, C. J.
and Boyer, J. L. (2007) Nuclear receptors RXR:RAR
are repressors for human MRP3 expression.
Am. J. Physiol. Gastrointest. Liver Physiol. 292,
G1221-G1227
99 Tanaka, T., Uchiumi, T., Hinoshita, E. et al. (1999) The
human multidrug resistance protein 2 gene: functional
characterization of the 5 -flanking region and expression
in hepatic cells. Hepatology 30, 15071512
100 Jung, D., Podvinec, M., Meyer, U. A. et al. (2002)
Human organic anion transporting polypeptide 8
promoter is transactivated by the farnesoid X receptor/
bile acid receptor. Gastroenterology 122, 19541966
101 Sinal, C. J., Tohkin, M., Miyata, M., Ward, J. M.,
Lambert, G. and Gonzalez, F. J. (2000) Targeted
disruption of the nuclear receptor FXR/BAR impairs bile
acid and lipid homeostasis. Cell 102, 731744


C

The Authors Journal compilation


C

2008 Biochemical Society

102 Ananthanarayanan, M., Balasubramanian, N.,


Makishima, M., Mangelsdorf, D. J. and Suchy, F. J. (2001)
Human bile salt export pump promoter is transactivated
by the farnesoid X receptor/bile acid receptor.
J. Biol. Chem. 276, 2885728865
103 Plass, J. R., Mol, O., Heegsma, J. et al. (2002) Farnesoid X
receptor and bile salts are involved in transcriptional
regulation of the gene encoding the human bile salt export
pump. Hepatology 35, 589596
104 Liu, T. Z., Lee, K. T., Chern, C. L., Cheng, J. T., Stern, A.
and Tsai, L. Y. (2001) Free radical-triggered hepatic injury
of experimental obstructive jaundice of rats involves
overproduction of proinflammatory cytokines and
enhanced activation of nuclear factor B. Ann. Clin.
Lab. Sci. 31, 383390
105 Denson, L. A., Bohan, A., Held, M. A. and Boyer, J. L.
(2002) Organ-specific alterations in RAR:RXR
abundance regulate rat Mrp2 (Abcc2) expression in
obstructive cholestasis. Gastroenterology 123, 599607
106 Denson, L. A., Auld, K. L., Schiek, D. S., McClure,
M. H., Mangelsdorf, D. J. and Karpen, S. J. (2000)
Interleukin-1 suppresses retinoid transactivation of
two hepatic transporter genes involved in bile formation.
J. Biol. Chem. 275, 88358843
107 Ghose, R., Zimmerman, T. L., Thevananther, S. and
Karpen, S. J. (2004) Endotoxin leads to rapid subcellular
re-localization of hepatic RXR: a novel mechanism for
reduced hepatic gene expression in inflammation. Nucl.
Recept. 2, 4
108 Kast, H. R., Goodwin, B., Tarr, P. T. et al. (2002)
Regulation of multidrug resistance-associated protein 2
(ABCC2) by the nuclear receptors pregnane X receptor,
farnesoid X-activated receptor, and constitutive
androstane receptor. J. Biol. Chem. 277, 29082915
109 Urquhart, B. L., Tirona, R. G. and Kim, R. B. (2007)
Nuclear receptors and the regulation of
drug-metabolizing enzymes and drug transporters:
implications for interindividual variability in response to
drugs. J. Clin. Pharmacol. 47, 566578
110 Eloranta, J. J. and Kullak-Ublick, G. A. (2005) Coordinate
transcriptional regulation of bile acid homeostasis and
drug metabolism. Arch. Biochem. Biophys. 433,
397412
111 Staudinger, J., Liu, Y., Madan, A., Habeebu, S. and
Klaassen, C. D. (2001) Coordinate regulation of
xenobiotic and bile acid homeostasis by pregnane X
receptor. Drug Metab. Dispos. 29, 14671472
112 Li, T. and Chiang, J. Y. (2005) Mechanism of rifampicin
and pregnane X receptor inhibition of human cholesterol
7-hydroxylase gene transcription. Am. J. Physiol.
Gastrointest. Liver Physiol. 288, G74G84
113 Hunt, M. C., Yang, Y. Z., Eggertsen, G. et al. (2000) The
peroxisome proliferator-activated receptor (PPAR)
regulates bile acid biosynthesis. J. Biol. Chem. 275,
2894728953
114 Patel, D. D., Knight, B. L., Soutar, A. K., Gibbons, G. F.
and Wade, D. P. (2000) The effect of peroxisomeproliferator-activated receptor- on the activity of the
cholesterol 7-hydroxylase gene. Biochem. J. 351,
747753
115 Guo, G. L., Lambert, G., Negishi, M. et al. (2003)
Complementary roles of farnesoid X receptor, pregnane
X receptor, and constitutive androstane receptor in
protection against bile acid toxicity. J. Biol. Chem. 278,
4506245071
116 Lehmann, J. M., McKee, D. D., Watson, M. A., Willson,
T. M., Moore, J. T. and Kliewer, S. A. (1998) The human
orphan nuclear receptor PXR is activated by compounds
that regulate CYP3A4 gene expression and cause drug
interactions. J. Clin. Invest. 102, 10161023

Hepatocellular transport in acquired cholestasis

117 Goodwin, B., Hodgson, E., DCosta, D. J., Robertson,


G. R. and Liddle, C. (2002) Transcriptional regulation of
the human CYP3A4 gene by the constitutive androstane
receptor. Mol. Pharmacol. 62, 359365
118 Kocarek, T. A., Schuetz, E. G., Strom, S. C., Fisher, R. A.
and Guzelian, P. S. (1995) Comparative analysis of
cytochrome P4503A induction in primary cultures of rat,
rabbit, and human hepatocytes. Drug Metab. Dispos. 23,
415421
119 Marschall, H. U., Wagner, M., Zollner, G. et al. (2005)
Complementary stimulation of hepatobiliary transport
and detoxification systems by rifampicin and
ursodeoxycholic acid in humans. Gastroenterology 129,
476485
120 Tirona, R. G., Lee, W., Leake, B. F. et al. (2003) The
orphan nuclear receptor HNF4 determines PXR- and
CAR-mediated xenobiotic induction of CYP3A4.
Nat. Med. 9, 220224
121 Duniec-Dmuchowski, Z., Ellis, E., Strom, S. C. and
Kocarek, T. A. (2007) Regulation of CYP3A4 and
CYP2B6 expression by liver X receptor agonists.
Biochem. Pharmacol. 74, 15351540
122 Gardner-Stephen, D., Heydel, J. M., Goyal, A. et al.
(2004) Human PXR variants and their differential effects
on the regulation of human UDP-glucuronosyltransferase
gene expression. Drug Metab. Dispos. 32, 340347
123 Sugatani, J., Kojima, H., Ueda, A. et al. (2001) The
phenobarbital response enhancer module in the human
bilirubin UDP-glucuronosyltransferase UGT1A1 gene
and regulation by the nuclear receptor CAR. Hepatology
33, 12321238
124 Song, C. S., Echchgadda, I., Baek, B. S. et al. (2001)
Dehydroepiandrosterone sulfotransferase gene induction
by bile acid activated farnesoid X receptor. J. Biol. Chem
276, 4254942556
125 Sonoda, J., Xie, W., Rosenfeld, J. M., Barwick, J. L.,
Guzelian, P. S. and Evans, R. M. (2002) Regulation of a
xenobiotic sulfonation cascade by nuclear pregnane X
receptor (PXR). Proc. Natl. Acad. Sci. U.S.A. 99,
1380113806
126 Duanmu, Z., Locke, D., Smigelski, J. et al. (2002)
Effects of dexamethasone on aryl (SULT1A1)- and
hydroxysteroid (SULT2A1)-sulfotransferase gene
expression in primary cultured human hepatocytes.
Drug Metab. Dispos. 30, 9971004
127 Echchgadda, I., Song, C. S., Roy, A. K. and Chatterjee, B.
(2004) Dehydroepiandrosterone sulfotransferase is a
target for transcriptional induction by the vitamin D
receptor. Mol. Pharmacol. 65, 720729
128 Uppal, H., Saini, S. P., Moschetta, A. et al. (2007)
Activation of LXRs prevents bile acid toxicity and
cholestasis in female mice. Hepatology 45, 422432
129 Barbier, O., Torra, I. P., Sirvent, A. et al. (2003) FXR
induces the UGT2B4 enzyme in hepatocytes: a potential
mechanism of negative feedback control of FXR activity.
Gastroenterology 124, 19261940
130 Barbier, O., Duran-Sandoval, D., Pineda-Torra, I.,
Kosykh, V., Fruchart, J. C. and Staels, B. (2003)
Peroxisome proliferator-activated receptor induces
hepatic expression of the human bile acid glucuronidating
UDP-glucuronosyltransferase 2B4 enzyme.
J. Biol. Chem. 278, 3285232860
131 Berg, P. A., Klein, R. and Rocken, M. (1997) Cytokines in
primary biliary cirrhosis. Semin. Liver Dis. 17, 115123
132 Aoki, C. A., Bowlus, C. L. and Gershwin, M. E. (2005)
The immunobiology of primary sclerosing cholangitis.
Autoimmun. Rev. 4, 137143
133 Trauner, M., Fickert, P. and Stauber, R. E. (1999)
Inflammation-induced cholestasis. J. Gastroenterol.
Hepatol. 14, 946959

134 Geier, A., Fickert, P. and Trauner, M. (2006) Mechanisms


of disease: mechanisms and clinical implications of
cholestasis in sepsis. Nat. Clin. Pract. Gastroenterol.
Hepatol. 3, 574585
135 Gilroy, R. K., Mailliard, M. E. and Gollan, J. L. (2003)
Gastrointestinal disorders of the critically ill. Cholestasis
of sepsis. Best Pract. Res. Clin. Gastroenterol. 17, 357367
136 Hartmann, G., Cheung, A. K. and Piquette-Miller, M.
(2002) Inflammatory cytokines, but not bile acids,
regulate expression of murine hepatic anion transporters
in endotoxemia. J. Pharmacol. Exp. Ther. 303, 273281
137 Elferink, M. G., Olinga, P., Draaisma, A. L. et al. (2004)
LPS-induced downregulation of MRP2 and BSEP in
human liver is due to a posttranscriptional process. Am. J.
Physiol. Gastrointest. Liver Physiol. 287, G1008G1016
138 Lee, J. M., Trauner, M., Soroka, C. J., Stieger, B., Meier,
P. J. and Boyer, J. L. (2000) Expression of the bile salt
export pump is maintained after chronic cholestasis in the
rat. Gastroenterology 118, 163172
139 Green, R. M., Beier, D. and Gollan, J. L. (1996)
Regulation of hepatocyte bile salt transporters by
endotoxin and inflammatory cytokines in rodents.
Gastroenterology 111, 193198
140 Cherrington, N. J., Slitt, A. L., Li, N. and Klaassen, C. D.
(2004) Lipopolysaccharide-mediated regulation of hepatic
transporter mRNA levels in rats. Drug Metab. Dispos. 32,
734741
141 Geier, A., Dietrich, C. G., Voigt, S. et al. (2005)
Cytokine-dependent regulation of hepatic organic anion
transporter gene transactivators in mouse liver. Am. J.
Physiol. Gastrointest. Liver Physiol. 289, G831G841
142 Beigneux, A. P., Moser, A. H., Shigenaga, J. K., Grunfeld,
C. and Feingold, K. R. (2000) The acute phase response is
associated with retinoid X receptor repression in rodent
liver. J. Biol. Chem. 275, 1639016399
143 Sturm, E., Havinga, R., Baller, J. F. et al. (2005) Kupffer
cell depletion with liposomal clodronate prevents
suppression of Ntcp expression in endotoxin-treated rats.
J. Hepatol. 42, 102109
144 Lee, H. Y., Suh, Y. A., Robinson, M. J. et al. (2000) Stress
pathway activation induces phosphorylation of retinoid X
receptor. J. Biol. Chem. 275, 3219332199
145 Jahan, A. and Chiang, J. Y. (2005) Cytokine regulation of
human sterol 12-hydroxylase (CYP8B1) gene. Am. J.
Physiol. Gastrointest. Liver Physiol. 288, G685G695
146 Geier, A., Dietrich, C. G., Voigt, S. et al. (2003) Effects of
proinflammatory cytokines on rat organic anion
transporters during toxic liver injury and cholestasis.
Hepatology 38, 345354
147 Siewert, E., Dietrich, C. G., Lammert, F. et al. (2004)
Interleukin-6 regulates hepatic transporters during
acute-phase response. Biochem. Biophys. Res. Commun.
322, 232238
148 Teng, S. and Piquette-Miller, M. (2005) The involvement
of the pregnane X receptor in hepatic gene regulation
during inflammation in mice. J. Pharmacol. Exp. Ther.
312, 841848
149 Chitturi, S. and Farrell, G. C. (2001) Drug-induced
cholestasis. Semin. Gastrointest. Dis. 12, 113124
150 Arrese, M. and Reyes, H. (2006) Intrahepatic cholestasis
of pregnancy: a past and present riddle. Ann. Hepatol. 5,
202205
151 Dourakis, S. P. and Tolis, G. (1998) Sex hormonal
preparations and the liver. Eur. J. Contracept. Reprod.
Health Care 3, 716
152 Micheline, D., Emmanuel, J. and Serge, E. (2002) Effect of
ursodeoxycholic acid on the expression of the
hepatocellular bile acid transporters (Ntcp and bsep) in
rats with estrogen-induced cholestasis. J. Pediatr.
Gastroenterol. Nutr. 35, 185191


C

The Authors Journal compilation


C

2008 Biochemical Society

585

586

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

153 Geier, A., Dietrich, C. G., Gerloff, T. et al. (2003)


Regulation of basolateral organic anion transporters in
ethinylestradiol-induced cholestasis in the rat.
Biochim. Biophys. Acta 1609, 8794
154 Fiorucci, S., Clerici, C., Antonelli, E. et al. (2005)
Protective effects of 6-ethyl chenodeoxycholic acid, a
farnesoid X receptor ligand, in estrogen-induced
cholestasis. J. Pharmacol. Exp. Ther. 313, 604612
155 Yamamoto, Y., Moore, R., Hess, H. A. et al. (2006)
Estrogen receptor alpha mediates 17-ethynylestradiol
causing hepatotoxicity. J. Biol. Chem. 281, 1662516631
156 Lai, K., Harnish, D. C. and Evans, M. J. (2003) Estrogen
receptor alpha regulates expression of the orphan receptor
small heterodimer partner. J. Biol. Chem. 278,
3641836429
157 Henriquez-Hernandez, L. A., Flores-Morales, A.,
Santana-Farre, R. et al. (2007) Role of pituitary hormones
on 17-ethinylestradiol-induced cholestasis in rat.
J. Pharmacol. Exp. Ther. 320, 695705
158 Arrese, M., Trauner, M., Ananthanarayanan, M. et al.
(2003) Down-regulation of the Na+ /taurocholate
cotransporting polypeptide during pregnancy in
the rat. J. Hepatol. 38, 148155
159 Cao, J., Huang, L., Liu, Y. et al. (2001) Differential
regulation of hepatic bile salt and organic anion
transporters in pregnant and postpartum rats and the role
of prolactin. Hepatology 33, 140147
160 Ganguly, T. C., OBrien, M. L., Karpen, S. J., Hyde, J. F.,
Suchy, F. J. and Vore, M. (1997) Regulation of the rat liver
sodium-dependent bile acid cotransporter gene by
prolactin. Mediation of transcriptional activation by Stat5.
J. Clin. Invest. 99, 29062914
161 Wood, M., Ananthanarayanan, M., Jones, B. et al. (2005)
Hormonal regulation of hepatic organic anion
transporting polypeptides. Mol. Pharmacol. 68, 218225
162 Cao, J., Stieger, B., Meier, P. J. and Vore, M. (2002)
Expression of rat hepatic multidrug resistance-associated
proteins and organic anion transporters in pregnancy. Am.
J. Physiol. Gastrointest. Liver Physiol. 283, G757-G766
163 Cai, S. Y. and Boyer, J. L. (2006) FXR: a target for
cholestatic syndromes? Expert Opin. Ther. Targets 10,
409421
164 Boyer, J. L. (2007) New perspectives for the treatment of
cholestasis: lessons from basic science applied clinically.
J. Hepatol. 46, 365371
165 Boyer, J. L. (2005) Nuclear receptor ligands: rational and
effective therapy for chronic cholestatic liver disease?
Gastroenterology 129, 735740
166 Fickert, P., Zollner, G., Fuchsbichler, A. et al. (2001)
Effects of ursodeoxycholic and cholic acid feeding on
hepatocellular transporter expression in mouse liver.
Gastroenterology 121, 170183
167 Zollner, G., Fickert, P., Fuchsbichler, A. et al. (2003) Role
of nuclear bile acid receptor, FXR, in adaptive ABC
transporter regulation by cholic and ursodeoxycholic acid
in mouse liver, kidney and intestine. J. Hepatol. 39,
480488
168 Puglielli, L., Amigo, L., Arrese, M. et al. (1994) Protective
role of biliary cholesterol and phospholipid lamellae
against bile acid-induced cell damage. Gastroenterology
107, 244254
169 Kim, M. S., Shigenaga, J., Moser, A., Feingold, K. and
Grunfeld, C. (2003) Repression of farnesoid X receptor
during the acute phase response. J. Biol. Chem. 278,
89888995
170 Denson, L. A., Bohan, A., Bajwa, H. J., Held, M. A. and
Boyer, J. L. (2002) Alterations in nuclear hormone
receptors (NHR) are associated with changes in the
expression of multidrug resistance proteins Mrp2 and
Mrp3 in liver and kidney in cholestasis. J. Hepatol. 34,
A264


C

The Authors Journal compilation


C

2008 Biochemical Society

171 Trauner, M., Wagner, M., Fickert, P. and Zollner, G.


(2005) Molecular regulation of hepatobiliary transport
systems: clinical implications for understanding
and treating cholestasis. J. Clin. Gastroenterol. 39,
S111S124
172 Crocenzi, F. A., DAndrea, V., Catania, V. A. et al. (2005)
Prevention of Mrp2 activity impairment in
ethinylestradiol-induced cholestasis by
ursodeoxycholate in the rat. Drug Metab. Dispos.
33, 888891
173 Crocenzi, F. A., Sanchez Pozzi, E. J., Pellegrino, J. M.
et al. (2001) Beneficial effects of silymarin on estrogeninduced cholestasis in the rat: a study in vivo
and in isolated hepatocyte couplets. Hepatology 34,
329339
174 Pauli-Magnus, C. and Meier, P. J. (2006) Hepatobiliary
transporters and drug-induced cholestasis. Hepatology
44, 778787
175 Crocenzi, F., Mottino, A. and Roma, M. (2004)
Regulation of synthesis and trafficking of canalicular
transporters and its alteration in acquired
hepatocellular cholestasis: experimental therapeutic
strategies for its prevention. Curr. Med. Chem. 11,
501524
176 Stieger, B., Fattinger, K., Madon, J., Kullak-Ublick, G. A.
and Meier, P. J. (2000) Drug- and estrogen-induced
cholestasis through inhibition of the hepatocellular bile
salt export pump (Bsep) of rat liver. Gastroenterology
118, 422430
177 Fattinger, K., Funk, C., Pantze, M. et al. (2001) The
endothelin antagonist bosentan inhibits the canalicular
bile salt export pump: a potential mechanism for
hepatic adverse reactions. Clin. Pharmacol. Ther. 69,
223231
178 Mano, Y., Usui, T. and Kamimura, H. (2007) Effects of
bosentan, an endothelin receptor antagonist, on bile salt
export pump and multidrug resistance-associated protein
2. Biopharm. Drug Dispos. 28, 1318
179 Funk, C., Ponelle, C., Scheuermann, G. and Pantze, M.
(2001) Cholestatic potential of troglitazone as a possible
factor contributing to troglitazone-induced
hepatotoxicity: in vivo and in vitro interaction at the
canalicular bile salt export pump (Bsep) in the rat.
Mol. Pharmacol. 59, 627635
180 Snow, K. L. and Moseley, R. H. (2007) Effect of
thiazolidinediones on bile acid transport in rat liver.
Life Sci. 80, 732740
181 Byrne, J. A., Strautnieks, S. S., Mieli-Vergani, G., Higgins,
C. F., Linton, K. J. and Thompson, R. J. (2002) The
human bile salt export pump: characterization of
substrate specificity and identification of inhibitors.
Gastroenterology 123, 16491658
182 Iwanaga, T., Nakakariya, M., Yabuuchi, H., Maeda, T. and
Tamai, I. (2007) Involvement of bile salt export pump in
flutamide-induced cholestatic hepatitis. Biol. Pharm. Bull.
30, 739744
183 Vallejo, M., Briz, O., Serrano, M. A., Monte, M. J. and
Marin, J. J. (2006) Potential role of trans-inhibition of the
bile salt export pump by progesterone metabolites in the
etiopathogenesis of intrahepatic cholestasis of pregnancy.
J. Hepatol. 44, 11501157
184 Adinolfi, L. E., Utili, R., Gaeta, G. B., Abernathy, C. O.
and Zimmerman, H. J. (1984) Cholestasis induced by
estradiol 17-d-glucuronide: mechanisms and prevention
by sodium taurocholate. Hepatology 1, 3037
185 Takikawa, H., Sano, N., Aiso, M., Takamori, Y. and
Yamanaka, M. (1997) Effect of tauro--muricholate and
tauro--muricholate on oestradiol-17-glucuronideinduced cholestasis in rats. J. Gastroenterol. Hepatol. 12,
8486

Hepatocellular transport in acquired cholestasis

186 Utili, R., Tripodi, M. F., Adinolfi, L. E., Gaeta, G. B.,


Abernathy, C. O. and Zimmerman, H. J. (1990)
Estradiol-17-d-glucuronide (E-17G) cholestasis in
perfused rat liver: fate of E-17G and choleretic responses
to bile salts. Hepatology 11, 735742
187 Huang, L., Smit, J. W., Meijer, D. K. F. and Vore, M.
(2000) Mrp2 is essential for estradiol
17-(-d-glucuronide)-induced cholestasis in rats.
Hepatology 32, 6672
188 Paulusma, C. C., Kothe, M. J., Bakker, C. T. et al. (2000)
Zonal down-regulation and redistribution of the
multidrug resistance protein 2 during bile duct ligation in
rat liver. Hepatology 31, 684693
189 Kubitz, R., DUrso, D., Keppler, D. and Haussinger, D.
(1997) Osmodependent dynamic localization of the
multidrug resistance protein 2 in the rat hepatocyte
canalicular membrane. Gastroenterology 113, 14381442
190 Dombrowski, F., Kubitz, R., Chittattu, A., Wettstein, M.,
Saha, N. and Haussinger, D. (2000) Electromicroscopic
demonstration of multidrug resistance protein 2 (Mrp2)
retrieval from the canalicular membrane in response to
hyperosmolarity and lipopolysaccharide. Biochem. J. 348,
183188
191 Schmitt, M., Kubitz, R., Wettstein, M., vom Dahl, S. and
Haussinger, D. (2000) Retrieval of the mrp2 gene encoded
conjugate export pump from the canalicular membrane
contributes to cholestasis induced by tert-butyl
hydroperoxide and chloro-dinitrobenzene. Biol. Chem.
381, 487495
192 Sekine, S., Ito, K. and Horie, T. (2006) Oxidative stress
and Mrp2 internalization. Free Radical Biol. Med. 40,
21662174
193 Perez, L. M., Milkiewicz, P., Elias, E., Coleman, R.,
Sanchez Pozzi, E. J. and Roma, M. G. (2006) Oxidative
stress induces internalization of the bile salt export pump,
Bsep, and bile salt secretory failure in isolated rat
hepatocyte couplets: a role for protein kinase C and
prevention by protein kinase A. Toxicol. Sci. 91, 150158
194 Mottino, A. D., Cao, J., Veggi, L. M., Crocenzi, F. A.,
Roma, M. G. and Vore, M. (2002) Altered localization and
activity of canalicular Mrp2 in estradiol-17-dglucuronide-induced cholestasis. Hepatology 35,
14091419
195 Crocenzi, F. A., Mottino, A. D., Cao, J. et al. (2003)
Estradiol-17-d-glucuronide induces endocytic
internalization of Bsep in rats. Am. J. Physiol.
Gastrointest. Liver Physiol. 285, G449G459
196 Beuers, U., Bilzer, M., Chittattu, A. et al. (2001)
Tauroursodeoxycholic acid inserts the apical conjugate
export pump, Mrp2, into canalicular membranes and
stimulates organic anion secretion by protein kinase
C-dependent mechanisms in cholestatic rat liver.
Hepatology 33, 12061216
197 Crocenzi, F. A., Mottino, A. D., Sanchez Pozzi, E. J. et al.
(2003) Impaired localisation and transport function of
canalicular Bsep in taurolithocholate-induced cholestasis
in the rat. Gut 52, 11701177
198 Roman, I. D., Fernandez-Moreno, M. D., Fueyo, J. A.,
Roma, M. G. and Coleman, R. (2003) Cyclosporin A
induced internalization of the bile salt export pump in
isolated rat hepatocyte couplets. Toxicol. Sci. 71, 276281
199 Kudo, A., Kashiwagi, S., Kajimura, M. et al. (2004)
Kupffer cells alter organic anion transport through
multidrug resistance protein 2 in the post-cold ischemic
rat liver. Hepatology 39, 10991109
200 Mottino, A. D., Hoffman, T., Crocenzi, F. A. et al. (2007)
Disruption of function and localization of
tight-junctional structures and Mrp2 in sustained
estradiol-17-d-glucuronide-induced cholestasis. Am. J.
Physiol. Gastrointest. Liver Physiol. 293, G391G402

201 Kojima, H., Nies, A. T., Konig, J. et al. (2003) Changes in


the expression and localization of hepatocellular
transporters and radixin in primary biliary cirrhosis.
J. Hepatol. 39, 693702
202 Kojima, H., Sakurai, S., Yoshiji, H., Uemura, M.,
Yoshikawa, M. and Fukui, H. (2008) The role of radixin in
altered localization of canalicular conjugate export pump
Mrp2 in cholestatic rat liver. Hepatol. Res. 38, 202210
203 Watanabe, N., Takashimizu, S., Kojima, S. et al. (2007)
Clinical and pathological features of a prolonged type
of acute intrahepatic cholestasis. Hepatol. Res. 37,
598607
204 Milkiewicz, P., Chilton, A. P., Hubscher, S. G. and Elias,
E. (2003) Antidepressant induced cholestasis:
hepatocellular redistribution of multidrug resistant
protein (MRP2). Gut 52, 300303
205 Mottino, A. D., Crocenzi, F. A., Pozzi, E. J., Veggi,
L. M., Roma, M. G. and Vore, M. (2005) Role of
microtubules in estradiol-17-d-glucuronide-induced
alteration of canalicular Mrp2 localization and activity.
Am. J. Physiol. Gastrointest. Liver Physiol. 288,
G327G336
206 Rost, D., Kartenbeck, J. and Keppler, D. (1999) Changes
in the localization of the rat canalicular conjugate export
pump Mrp2 in phalloidin-induced cholestasis.
Hepatology 29, 814821
207 Kikuchi, S., Hata, M., Fukumoto, K. et al. (2002) Radixin
deficiency causes conjugated hyperbilirubinemia with loss
of Mrp2 from bile canalicular membranes. Nat. Genet. 31,
320325
208 Wang, W., Soroka, C. J., Mennone, A. et al. (2006) Radixin
is required to maintain apical canalicular membrane
structure and function in rat hepatocytes.
Gastroenterology 131, 878884
209 Kubitz, R., Saha, N., Kuhlkamp, T. et al. (2004)
Ca2+ -dependent protein kinase C-isoforms induce
cholestasis in rat liver. J. Biol. Chem. 279, 1032310330
210 Kubitz, R., Huth, C., Schmitt, M., Horbach, A.,
Kullak-Ublick, G. and Haussinger, D. (2001) Protein
kinase C-dependent distribution of the multidrug
resistance protein 2 from the canalicular to the basolateral
membrane in human HepG2 cells. Hepatology 34,
340350
211 Beuers, U., Probst, I., Soroka, C., Boyer, J. L.,
Kullak-Ublick, G. A. and Paumgartner, G. (1999)
Modulation of protein kinase C by taurolithocholic
acid in isolated rat hepatocytes. Hepatology 29,
477482
212 Beuers, U., Denk, G. U., Soroka, C. J. et al. (2003)
Taurolithocholic acid exerts cholestatic effects via
phosphatidylinositol 3-kinase-dependent mechanisms
in perfused rat livers and rat hepatocyte couplets.
J. Biol. Chem. 278, 1781017818
213 Trauner, M. and Boyer, J. L. (2001) Cholestatic
syndromes. Curr. Opin. Gastroenterol. 17, 242256
214 Mottino, A. D., Crocenzi, F. A., Pozzi, E. J., Veggi,
L. M., Roma, M. G. and Vore, M. (2005) Role of
microtubules in estradiol-17-d-glucuronide-induced
alteration of canalicular Mrp2 localization and activity.
Am. J. Physiol. Gastrointest. Liver Physiol. 288,
G327G336
215 Hayakawa, T., Bruck, R., Ng, O. C. and Boyer, J. L.
(1990) DBcAMP stimulates vesicle transport and HRP
excretion in isolated perfused rat liver. Am. J. Physiol.
259, G727G735
216 Crocenzi, F., Basiglio, C., Perez, L., Portesio, M., Sanchez
Pozzi, E. and Roma, M. (2005) Silibinin prevents
cholestasis-associated retrieval of the bile salt export
pump, Bsep, in isolated rat hepatocyte couplets: possible
involvement of cAMP. Biochem. Pharmacol. 69,
11131120


C

The Authors Journal compilation


C

2008 Biochemical Society

587

588

M. G. Roma, F. A. Crocenzi and E. A. Sanchez Pozzi

217 Beuers, U., Nathanson, M. H., Isales, C. M. and Boyer,


J. L. (1993) Tauroursodeoxycholic acid stimulates
hepatocellular exocytosis and mobilizes extracellular
Ca++ mechanisms defective in cholestasis. J. Clin. Invest.
92, 29842993
218 Dombrowski, F., Stieger, B. and Beuers, U. (2006)
Tauroursodeoxycholic acid inserts the bile salt export
pump into canalicular membranes of cholestatic rat liver.
Lab. Invest. 86, 166174

219 Schliess, F., Kurz, A. K., vom Dahl, S. and Haussinger, D.


(1997) Mitogen-activated protein kinases mediate the
stimulation of bile acid secretion by tauroursodeoxycholate in rat liver. Gastroenterology 113,
13061314
220 Kurz, A. K., Graf, D., Schmitt, M. et al. (2001)
Tauroursodeoxycholate-induced choleresis involves
p38MAPK activation and translocation of the bile salt
export pump in rats. Gastroenterology 121, 407419

Received 3 July 2007/30 October 2007; accepted 7 November 2007


Published on the Internet 2 April 2008, doi:10.1042/CS20070227


C

The Authors Journal compilation


C

2008 Biochemical Society

You might also like