Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Liver International 2003, 23, 315–322 Copyright r Blackwell Munksgaard 2003

Printed in Denmark. All rights reserved

Review Article

Genetic cholestasis, causes and


consequences for hepatobiliary transport

Jansen PLM, Sturm E. Genetic cholestasis, causes and consequences for Peter L. M. Jansen and Ekkehard
hepatobiliary transport. Sturm

Liver International 2003: 23: 315–322. r Blackwell Munksgaard, 2003 Departments of Gastroenterology and

Pediatrics, University Hospital Groningen,
Abstract: Bile salts take part in an efficient enterohepatic circulation in
The Netherlands
which most of the secreted bile salts are reclaimed by absorption in the

terminal ileum. In the liver, the sodium-dependent taurocholate transporter

at the basolateral (sinusoidal) membrane and the bile salt export pump at the

canalicular membrane mediate hepatic uptake and hepatobiliary secretion of

bile salts. Canalicular secretion is the driving force for the enterohepatic

cycling of bile salts and most genetic diseases are caused by defects of

canalicular secretion. Impairment of bile flow leads to adaptive changes in

the expression of transporter proteins and enzymes of the cytochrome P-450

system involved in the metabolism of cholesterol and bile acids. Bile salts act

as ligands for transcription factors. As such, they stimulate or inhibit the

transcription of genes encoding transporters and enzymes involved in their
own metabolism. Together these changes appear to serve mainly a

hepatoprotective function. Progressive familial intrahepatic cholestasis

(PFIC) results from mutations in various genes encoding hepatobiliary

transport proteins. Mutations in the FIC1 gene cause relapsing or permanent

cholestasis. The relapsing type of cholestasis is called benign recurrent

intrahepatic cholestasis, the permanent type of cholestasis PFIC type 1. PFIC

type 2 results from mutations in the bile salt export pump (BSEP) gene. This

is associated with permanent cholestasis since birth. Serum gamma- Key words: benign recurrent intrahepatic cho-

glutamyltransferase (gamma-GT) activity is low to normal in PFIC types 1 lestasis – bile acids – bile acid synthesis – bile
salt export pump – farnesoid X-receptor –
and 2. Bile diversion procedures, causing a decreased bile salt pool, have a
hepatobiliary transport – intrahepatic cholestasis
beneficial effect in a number of patients with these diseases. However, liver
of pregnancy – progressive familial intrahepatic
transplantation is often necessary. PFIC type 3 is caused by mutations in the
cholestasis
MDR3 gene. MDR3 is a phospholipid translocator in the canalicular
Prof. Peter L. M. Jansen, MD, PhD,
membrane. Because of the inability to secrete phospholipids, patients with
Division of Gastroenterology and Hepatology,
PFIC type 3 produce bile acid-rich toxic bile that damages the intrahepatic
Department of Medicine, University Hospital,
bile ducts. Serum gamma-GT activity is elevated in these patients. Hanzeplein 1, 9717 GZ Groningen,

Ursodeoxycholic acid therapy is useful for patients with a partial defect. The Netherlands.

Liver transplantation is a more definitive therapy for these patients. E-mail: p.l.m.jansen@int.azg.nl

Bile salts are the main organic solutes in bile, and port of bile salts by hepatocytes against a
their vectorial secretion from blood to bile concentration gradient. The total bile salt pool
represents the major driving force for bile size in adult humans amounts to 50–60 mmol/kg
formation. Although bile is iso-osmotic in rela- body weight, corresponding to 3–4 g, and is
tion to plasma, bile salts are 100–1000-fold largely stored in the gallbladder during the
concentrated in bile, necessitating active trans- fasting state. The human bile salt pool circulates
6–10 times per 24 h, resulting in a daily bile salt
Abbreviations: PFIC 5 progressive familial intrahepatic choles- secretion of 20–40 g. Despite a high degree of
tasis; BRIC 5 benign recurrent intrahepatic cholestasis; IC- intestinal bile salt conservation, about 0.5 g of
P 5 intrahepatic cholestasis of pregnancy; BSEP 5 bile salt bile salts is lost through fecal excretion. This loss
export pump; OATP 5 organic anion transporting polypeptide; is compensated for by de novo hepatic bile salt
MRP 5 multidrug resistance-related protein; ASBT 5 apical
sodium-dependent bile acid transporter; FXR 5 farnesoid X- synthesis, which contributes less than 3% of bile
receptor; SHP 5 small heterodimer partner; JNK 5 c-jun N- salts secreted with hepatic bile. The intrinsic link
terminal kinase. between intestinal bile salt absorption and

315
Jansen and Sturm
hepatic synthesis has recently been delineated by PFIC 3
MRP3 MRP1 MRP4
the discovery that bile salts are ligands for OATP-A
transcription factors that affect the expression OATP-B
OATP-C
MDR3 MDR1
of a number of genes involved in bile salt OATP8 BSEP
FIC1
NTCP MRP2 PFIC 1/BRIC
synthesis and transport. Disturbances of bile salt OCT1 Liver
transport are important causes of acquired and
genetic forms of cholestatic liver disease in PFIC 2
humans.
Dubin Johnson Synd.
MRP3 MRP1
Physiology
ASBT Intestine
Hepatic transport proteins MRP2 MDR1

Hepatic uptake
Fig. 1. Human hepatobiliary transport proteins involved in bile
The Na1/taurocholate co-transporting polypep- formation, secretion and reabsorption. Transporter proteins
tide (NTCP; SLC10A1) is the major bile salt located in the basolateral membrane are responsible for hepatic
uptake system of hepatocytes (1) (Fig. 1). It is uptake of bile salts (NTCP, OATPs), bulky organic anions,
uncharged compounds (OATPs) and cations (OATPs, OCT1).
localized in the basolateral membrane of hepa- Transporter proteins located in the canalicular membrane are
tocytes (2). Ntcp in rats preferentially mediates responsible for the biliary secretion of bile salts, phosphatidyl-
sodium-dependent transport of conjugated bile choline, cholesterol and glutathione and the excretion of drugs
salts, such as taurocholate (capital letters, NTCP, and toxins. These are the bile salt export pump BSEP (ABCB11),
the phosphatidylcholine translocator MDR3 (ABCB4), the
are used to denote the human transporter and multispecific organic anion transporter MRP2 (ABCC2) and
lowercase letters, Ntcp, the rodent protein). This the multidrug transporter MDR1 (ABCB1). The organic anion
transport comprises the major fraction of hepatic transporters MRP3 (ABCC3), MRP4 (ABCC4) and MRP1
bile salt uptake. NTCP is not the only hepatic (ABCC1) are present at very low levels in normal human liver
but their expression is strongly increased during cholestasis.
uptake transporter for bile salts. OATP-C Both MRP3 and MRP4 are able to transport bile acid
(SLC21A6) transports a number of organic conjugates out of the hepatocyte. FIC1 (ATP8B1) has been
anions, including bilirubin and bile salts, in a characterized as an aminophospholipid translocase. In the
sodium-independent manner. OATP-B, OATP-C terminal ileum, the apical sodium-dependent bile acid transpor-
ter (ASBT) is responsible for bile acid reabsorption. Genetic
and OATP8 exhibit broad overlapping substrate defects have been described for FIC1 (PFIC type 1, BRIC),
specificities and together account for drug BSEP (PFIC type 2), MDR3 (PFIC type 3, ICP), MRP2
clearance by human liver (3). (Dubin–Johnson syndrome) and ASBT (bile acid malabsorp-
tion).
Canalicular bile salt transport
The human bile salt export pump (BSEP,
ABCB11) is critical for ATP-dependent transport ing that of bile acid conjugates (8, 9). In normal
of bile acids across the hepatocyte canalicular liver their expression is low, but in cholestasis
membrane and for the generation of bile acid- these proteins are highly expressed (10, 11). It is
dependent bile secretion (4). Murine Bsep was postulated that these proteins play a role as
shown to transport taurocholate, taurocheno- basolateral escape transporters when canalicular
deoxycholate and glycocholate (5, 6). Bsep ( / ) Bsep function is impaired as during sepsis, drug-
knockout mice are cholestatic in the sense induced cholestasis or bile duct obstruction.
that taurocholate accumulates in their plasma
because its secretion into bile is strongly impaired Intestinal reabsorption
(italics denote the gene, non-italics the protein) In the ileum, bile salts are reabsorbed. A sodium-
(7). However, in contrast to patients, the mice dependent apical bile salt transporting protein
excrete substantial amounts of tauromuricholate ASBT (SLC10A2) and an intracellular bile salt
as well as tetrahydroxy bile salts via hitherto binding protein play an important role in
undefined canalicular transporters. Humans are reclaiming bile salts from the intestinal lumen,
not capable of converting bile salts into mur- but other mechanisms for bile salt reabsorption
icholate or tetrahydroxy bile salts to any sig- probably co-exist (12, 13). About 90% of the
nificant extent; therefore, this escape route is not total biliary bile salts are reabsorbed in the ileum.
available to man.
Mechanisms of adaptation
Basolateral escape routes
MRP3 (ABCC3), MRP4 (ABCC4) and MRP1 Hepatocytes are strictly polarized cells. They
(ABCC1) are transporter proteins that support absorb substrates from the blood and secrete
the basolateral export of organic anions, includ- metabolites into the bile. The supply of bile acids

316
Genetic cholestasis, causes and consequences for hepatobiliary transport
is highly variable. Absorption of bile salts by the
liver from the portal venous blood is nearly MRP3
complete and, in the fasting state, mainly occurs CHOLESTEROL
CYP7A1
in periportal hepatocytes. Thus, periportal hepa- CYP 8B1

tocytes are continuously exposed to high bile salt NTCP BSEP


concentrations. Hepatocytes more downstream Bile Acids [1]

into the hepatic acinus are exposed to varying +


+
concentrations of bile salts: low in the fasting [2] FXR:RXR BSEP
FXR:RXR [3]
SHP-1
state, high after a meal. Bile salts are cytotoxic FXR:RXR
JNK TNFα/ILβ
and cellular homeostasis demands maintenance SHP-1
NTCP -
-
of intracellular bile salt concentrations within SHP-1
CYP7A1
+
certain limits. Post-translational regulations with LRH-1
MRP3

recruitment of BSEP from intracellular stores to


the canalicular membrane may be operational for Fig. 2. Gene regulation by bile salts. Bile salts are taken up in the
this purpose (14). Obstruction of bile efflux by liver by NTCP and secreted into bile by canalicular BSEP. In
gallstones or tumors will lead to a more chronic cholestasis BSEP activity is reduced the intracellular bile salt
concentration increases (1). Bile salts serve as ligands for FXR,
exposure of liver cells to bile salts. Transcrip- which forms a heterodimer with RXR and translocates to the
tional regulation enables alterations of BSEP nucleus (2). The heterodimer activates the transcription of the
expression in order to maintain cellular home- BSEP and SHP-1 genes. SHP-1 antagonizes the expression of
ostasis. Cytokines also play a role here. Bile salts the bile acid biosynthetic enzymes CYP7A1 and CYP8B1 and
the transporter NTCP. In addition, Kupffer cells produce TNF-
stimulate Kupffer cells to produce TNF-a and a and interleukin-1b during cholestasis and, via the c-Jun N-
interleukin-1b. These act upon hepatocyte recep- terminal kinase-dependent (JNK) pathway, they reduce the
tors that affect the c-jun N-terminal kinase signal expression of NTCP and CYP7A1 (3). Recent evidence indicates
transduction pathway. that Lrh-1 (liver receptor homolog-1)-mediated Mrp3 transcrip-
tion is enhanced via a TNF-a signalling pathway (25). Thus, at
increasing bile salt concentrations, de novo synthesis is reduced,
Regulation of gene expression uptake is impaired and secretion, either across the canalicular or
Nuclear hormone receptors have been identified basolateral membrane, is stimulated. As a consequence, the
as important transcription factors in lipid and intracellular bile salt concentration remains controlled and
limited.
bile salt metabolism. The BSEP gene is under the
transcriptional control of FXR (farnesoid X-
receptor) (Fig. 2) (15). FXR is a ligand-activated
transcription factor. Chenodeoxycholic acid and
cholic acid bind and activate FXR (Fig. 2 (1)). genes (20, 21). In some of these, cytokines play an
Subsequently, FXR forms a heterodimer with important role, but a recent study indicates that
RXR (retinoid X-receptor) and translocates to FXR can also suppress the Cyp7a1 gene in an
the nucleus (2). Here the FXR:RXR heterodimer Shp-1-independent way (21). This again indicates
acts as a transcription factor of e.g. the BSEP the key role of bile acids in these adaptations.
and SHP-1 (small heterodimer protein-1) genes. Also in humans, NTCP expression in cholestatic
SHP-1 antagonizes the transcription of CYP7A1, liver disease is decreased (22). Downregulation of
CYP8B and NTCP (16, 17). Thus, FXR controls the NTCP and CYP7A1 genes, and the cons-
several key steps in bile salt metabolism. Studies quent decreased expression of NTCP and
in mice with a genetic disruption of Fxr showed CYP7A1, in cholestatic liver disease reduces the
that the Fxr-mediated response is particularly entry and the synthesis of bile acids. This
important in dealing with a bile salt load as regulation most probably serves a cytoprotective
occurs when mice are fed a high cholesterol- or function. This is particularly important in the
cholate-containing diet. In Fxr null mice, the case of bile duct obstruction when BSEP, via
expression of the Ntcp, Cyp7a1 and Cyp8b genes FXR-stimulated expression of its gene, remains
fails to be downregulated and the expression of active and keeps transporting bile acids into the
the Bsep and Shp-1 genes is not increased (18). bile canaliculus despite the downstream obstruc-
Severe hepatic damage is the consequence of this tion. This leads to disruption of tight junctions
failure of regulation. and bile infarcts (23). Up-regulation of MRP1, 3
Cholestasis in rats is associated with a and 4 in the basolateral membranes of hepato-
decreased expression of Ntcp (19). This in part cytes probably constitutes an important escape
results from enhanced expression of Shp-1 route for the cellular release of cholestatic
through activation of Fxr by retained bile salts products. Bile salts, bilirubin and cytokines are
(17), but also Shp-1-independent pathways exist involved in the transcriptional activation of their
to suppress transcription of the Ntcp and Cyp7a1 respective genes (11, 24, 25).

317
Jansen and Sturm
Pathophysiology BRIC
Genetic transport defects Recurrent familial intrahepatic cholestasis is also
known under the name BRIC or Summerskil
The spectrum of diseases caused by defects of
syndrome. Despite recurrent attacks of cholesta-
ABC transporter proteins is diverse and includes
sis, there is no progression to chronic liver disease
the liver diseases: progressive familial intrahepa-
in a majority of patients. During the attacks, the
tic cholestasis (PFIC) (26) (Table 1), benign
patients are severely jaundiced and have pruritus,
recurrent intrahepatic cholestasis (BRIC) (27),
steatorrhoea and weight loss. As in PFIC 1 the
intrahepatic cholestasis of pregnancy (28, 29),
serum gamma-GT is not elevated. Some patients
intrahepatic gallstone formation (30), cystic
also have renal stones, pancreatitis and diabetes.
fibrosis (31), adrenoleukodystrophy (32) and
The gene involved in recurrent familial intrahe-
Dubin–Johnson syndrome (33, 34).
patic cholestasis has been mapped to the FIC1
PFIC constitutes a group of autosomal recessive
locus (27). This suggests that recurrent familial
diseases characterized by cholestasis starting in
intrahepatic cholestasis and PFIC type I are
infancy. For a first differentiation of various PFIC
genetically related. However, in not all BRIC
subtypes, measurement of the serum gamma-
patients could the defect be traced to chromo-
glutamyltransferase (gamma-GT) activity is useful.
some 18 mutations (41). Ursodeoxycholic acid is
Diseases associated with a low bile salt concentra-
of no benefit in BRIC (42). Case reports indicate
tion in bile have a low serum gamma-GT activity.
that rifampicine may reduce the number of
These diseases have an intrahepatocellular block-
cholestatic episodes (43, 44).
ade of bile salt secretion in common. Gamma-GT
in human liver is mainly located in the membranes
lining the biliary tree. Elevation of serum gamma- PFIC type 2
GT results from a detergent, membranolytic effect Genetic studies revealed a number of PFIC
of bile salts on these membranes. Thus an intra- or patients in whom the FIC1 locus does not seem
extrahepatic obstruction of bile flow, or bile devoid to be involved. In a large number of non-Amish
of phosphatidylcholine (as in PFIC type 3, see patients, the disease was mapped to a locus on
below), causes gamma-GT to be released in the chromosome 2q24, which later proved to be the
circulation. ABCB11 (BSEP) gene (45). Antibodies directed
against BSEP showed that the protein is located
PFIC type 1 in the canalicular domain of the hepatocyte
plasma membrane. Liver specimens of patients
PFIC type 1 or Byler disease often begins with with PFIC type 2 stain negative for canalicular
cholestatic episodes progressing to permanent BSEP on immunohistochemistry using BSEP
cholestasis with fibrosis, cirrhosis and liver failure antibodies (46). As in PFIC type 1, the serum
in the first two decades of life (35). Children with gamma-GT activity in these patients is not
PFIC type 1 are small for their age and, in elevated and bile duct proliferation is absent.
addition to cholestasis and pruritus, they often However, there are also some differences with
have diarrhea and occasionally pancreatitis. The PFIC type 1: in PFIC2 the disease often starts as
larger bile ducts are anatomically normal and nonspecific giant cell hepatitis, which is indis-
liver histology shows bland canalicular cholesta- tinguishable from idiopathic neonatal giant cell
sis without much bile duct proliferation, inflam- hepatitis; patients are frequently jaundiced and
mation, fibrosis or cirrhosis (35, 36). On electron the disease rapidly progresses to persistent and
microscopy, there is a paucity of canalicular progressive cholestasis requiring liver transplan-
microvilli and a thickened pericanalicular net- tation within the first decade. The liver histology
work of microfilaments. The coarse granular bile shows more inflammation than in PFIC type 1,
in the canaliculi is called ‘Byler bile’. Character- with giant cell transformation, lobular and portal
istically, the serum gamma-GT activity is not fibrosis (35). The bile of PFIC type 2 patients is
elevated while primary bile salt levels, in parti- amorphous or filamentous on transmission elec-
cular chenodeoxycholic acid, are increased. Ser- tron microscopy. This contrasts with the coarsely
um cholesterol is usually normal. Patients granular bile of PFIC type 1 patients. Extra-
belonging to the Byler kindred are descendants hepatic manifestations are uncommon.
of Jacob and Nancy Byler, who emigrated in the
late 18th century from Germany to the United
PFIC type 3
States. The PFIC syndrome has also been
described in families in the Netherlands, Sweden, The third PFIC subtype, PFIC type 3, is quite
Greenland and an Arab population (35, 37–40). different from the other PFIC subtypes. In patients

318
Genetic cholestasis, causes and consequences for hepatobiliary transport
Table 1. Genetic cholestasis

Disease Chromosome Gene Phenotype Therapy

PFIC type 1 18q21 FIC 1 (ATP8B1) P-type ATPase, First recurrent, later permanent Ursodeoxycholic acid, bile
acts as an aminophospholipid cholestasis, bile duct proliferation diversion, liver transplantation
translocator is a late phenomenon. Diarrhea,
pancreatitis, pruritus, short
stature. Coarse granular bile on
EM. Normal gamma-GT
Benign recurrent 18q21 FIC1 (ATP8B1) Recurrent episodes of cholestasis Cholestyramine and/or
intrahepatic with severe pruritus, steatorrhea rifampicine as symptomatic
cholestasis and weight loss. Normal gamma- antipruritus therapy
GT
PFIC type 2 2q24 BSEP (ABCB11), bile salt export Neonatal hepatitis, progressive Ursodeoxycholic acid bile
pump cholestasis, pruritus, short diversion, liver transplantation
stature, bile duct proliferation is a
late phenomenon, lobular and
portal fibrosis. BSEP protein
absent. Amorphous bile on EM.
Normal gamma-GT
PFIC type 3 7q21 PGY3 (ABCB4, MDR 3), P- Cholestasis, portal hypertension, Ursodeoxycholic acid, liver
glycoprotein 3 extensive bile duct proliferation transplantation
and periportal fibrosis. MDR3 is
not expressed. Elevated gamma-
GT
Intrahepatic e.g. 7q21 e.g. MDR3 Cholestasis in third trimester of Ursodeoxycholic acid causes
cholestasis of pregnancy. High gamma-GT in symptomatic relief in the mother
pregnancy case of MDR3 defect; low and decreases fetal loss
gamma-GT cases may be caused
by genetic defects of other
transporter proteins. High
incidence of fetal loss
Aagenaes syndrome 15q LCS1, LCS2 Episodic cholestasis, Liver transplantation but
lymphedema, normal gamma-GT persistence of lymphedema
Familial 9q12–q13 9q22– TJP2/ZO-2 BAAT Elevated bile acids, severe Liver transplantation
Hypercholanemia q32 pruritus, fat malabsorption,
failure to thrive, rickets, vitamin K
coagulopathy
Bile acid synthesis e.g. 8q2.3 3b-D5-C27-hydroxysteroid Intrahepatic cholestasis, neonatal Ursodeoxycholic acid,
defects oxidoreductase; D4-3- giant cell hepatitis. Normal or chenodeoxycholic acid or cholic
oxosteroid-5b reductase; 3b- elevated gamma-GT, low or acid alone or in combination,
hydroxy C27 steroid elevated serum total bile acids depending on subtype
dehydrogenase/isomerase;
oxysterol 7a-hydroxylase; 24,25-
dihydroxy-cholanoic cleavage
enzyme.

gamma-GT 5 gamma glutamyl tranferase.

with PFIC type 3, symptoms present somewhat of disease and insufficient control of symptoms
later in life than in PFIC types 1 and 2, and liver may necessitate further intervention such as liver
failure also occurs at a later age. Jaundice may be transplantation. Ursodeoxycholic may also lead
less apparent during the early stages of disease. The to acceleration of disease (50) or to very high
serum gamma-GT activity is usually markedly serum bile acid levels (41 mmol/l) without any
elevated in these patients and the liver histology increase of bile salt secretion (46). PFIC 3
shows extensive bile duct proliferation, portal and patients are more likely to respond to ursodeoxy-
periportal fibrosis (47, 48). In humans, the MDR3 cholic acid therapy if they carry mild mutations
gene is mutated in this disease (28, 29, 47, 48). of the MDR3 gene (48, 49).
Patients with a partial MDR3 defect often respond For PFIC types 1 and 2, partial external biliary
to ursodeoxycholic acid therapy (49). The majority diversion is an accepted mode of therapy (51, 52).
of patients, particularly those with a complete The majority of patients respond with a significant
defect, have to be transplanted. improvement of symptoms (52–54). When per-
formed early, ongoing hepatic injury may be
interrupted with resolution of histological abnorm-
Therapy
alities including reversal of fibrosis (54). Clinically,
Subgroups of PFIC types 1–3 may respond to the patients may experience long-term amelioration
ursodeoxycholic acid (50). However, progression of pruritus and induction of catch-up growth (52).

319
Jansen and Sturm
Liver transplantation has been performed for 2. Stieger B, Hagenbuch B, Landmann L, Hochli M,
patients with PFIC types 1–3. In general, these Schroeder A, Meier P J. In situ localization of the
hepatocytic Na1/taurocholate cotransporting polypeptide
patients have a good prognosis after successful in rat liver. Gastroenterology 1994; 107(6): 1781–7.
transplantation (55). Since PFIC type 1 is a 3. Kullak-Ublick G A, Ismair M G, Stieger B, Land-
multiorgan disease, diarrhea may persist and mann L, Huber R, Pizzagalli F, et al. Organic anion-
pancreatitis may occur after transplantation. transporting polypeptide B (OATP-B) and its functional
Diarrhea in these patients is associated with comparison with three other OATPs of human liver.
Gastroenterology 2001; 120(2): 525–33.
malabsorption; therefore, catch-up growth after 4. Gerloff T, Stieger B, Hagenbuch B, Madon J, Land-
transplantation may be disappointing. mann L, Roth J, et al. The sister of P-glycoprotein
represents the canalicular bile salt export pump of
Intrahepatic cholestasis of pregnancy (ICP) mammalian liver. J Biol Chem 1998; 273(16): 10046–50.
5. Green R M, Hoda F, Ward K L. Molecular cloning and
Jacquemin et al. (29) reported a high incidence of characterization of the murine bile salt export pump. Gene
ICP in families with PFIC type 3. This suggests 2000; 241(1): 117–23.
that in persons carrying one mutated MDR3 6. Lecureur V, Sun D, Hargrove P, Schuetz E G, Kim R
gene, cholestasis may occur during pregnancy. B, Lan L B, et al. Cloning and expression of murine sister
of P-glycoprotein reveals a more discriminating transporter
Mutations leading to single amino acid substitu- than MDR1/P-glycoprotein. Mol Pharmacol 2000; 57(1):
tions of the MDR3 protein may cause intracel- 24–35.
lular traffic mutants, i.e. the protein is 7. Wang R, Salem M, Yousef I M, Tuchweber B, Lam P,
synthesized but does not reach its destination in Childs S J, et al. Targeted inactivation of sister of P-
the canalicular membrane (56). ICP has also been glycoprotein gene (spgp) in mice results in nonprogressive
but persistent intrahepatic cholestasis. Proc Natl Acad Sci
described in families with other PFIC subtypes USA 2001; 98(4): 2011–6.
(57). Thus, ICP may be associated either with 8. Hirohashi T, Suzuki H, Takikawa H, Sugiyama Y.
elevated serum gamma-GT or with a normal ATP-dependent transport of bile salts by rat multidrug
enzyme activity. Ursodeoxycholic acid has been resistance-associated protein 3 (Mrp3). J Biol Chem 2000;
shown to be of benefit in these patients, with also 275(4): 2905–10.
9. Zelcer N, Reid G, Wielinga P, Kuil A, van D H I,
a decrease of fetal loss (58, 59). Schuetz J D, et al. Steroid and bile acid conjugates are
substrates of human multidrug-resistance protein (MRP) 4
Other forms of intrahepatic cholestasis (ATP-binding cassette C4). Biochem J 2003; 371(Part 2):
361–7.
Aageneas syndrome is a combination of severe 10. Ogawa K, Suzuki H, Hirohashi T, Ishikawa T, Meier
progressive lymphedema and episodic intrahepa- P J, Hirose K, et al. Characterization of inducible nature of
tic cholestasis (60). The locus for this disease has MRP3 in rat liver. Am J Physiol Gastrointest Liver Physiol
been mapped to one locus, or perhaps two loci, 2000; 278(3): G438–46.
11. Schuetz E G, Strom S, Yasuda K, Lecureur V, Assem
on chromosome 15q (61, 62). Recently, Carlton M, Brimer C, et al. Disrupted bile acid homeostasis reveals
et al. (63) described patients with familial an unexpected interaction among nuclear hormone recep-
hypercholanemia characterized by elevated ser- tors, transporters, and cytochrome P450. J Biol Chem 2001;
um bile acids, pruritus, fat malabsorption, failure 276(42): 39411–8.
to thrive, rickets and vitamin K coagulopathy. 12. Oelkers P, Kirby L C, Heubi J E, Dawson P A. Primary
bile acid malabsorption caused by mutations in the ileal
This disease is associated with a mutation of the sodium-dependent bile acid transporter gene (SLC10A2). J
tight-junction protein 2 gene (TJP2 or ZO-2) Clin Invest 1997; 99(8): 1880–7.
with or without additional mutations in a gene 13. Wong M H, Oelkers P, Dawson P A. Identification of a
encoding the bile acid coenzyme A:amino acid N- mutation in the ileal sodium-dependent bile acid transporter
acyltransferase gene (BAAT). Patients with TJP2 gene that abolishes transport activity. J Biol Chem 1995;
270(45): 27228–34.
mutations had higher gamma-GT serum levels 14. Kipp H, Pichetshote N, Arias I M. Transporters on
than patients with BAAT gene mutations only. demand: intrahepatic pools of canalicular ATP binding
The serum of patients with BAAT mutations did cassette transporters in rat liver. J Biol Chem 2001; 276(10):
not contain bile acid amino acid conjugates. 7218–24.
15. Ananthanarayanan M, Balasubramanian N, Ma-
kishima M, Mangelsdorf D J, Suchy F J. Human bile
Acknowledgements salt export pump promoter is transactivated by the
This research was supported by the Netherlands Organization farnesoid X receptor/bile acid receptor. J Biol Chem 2001;
for Scientific Research, NWO program grant 902-23-191 and 276(31): 28857–65.
NWO project grants 902-23-253, 902-23-257 and 903-39-188. 16. Lu T T, Makishima M, Repa J J, Schoonjans K, Kerr T
A, Auwerx J, et al. Molecular basis for feedback
regulation of bile acid synthesis by nuclear receptors. Mol
References Cell 2000; 6(3): 507–15.
1. Kullak-Ublick G A, Stieger B, Hagenbuch B, Meier 17. Denson L A, Sturm E, Echevarria W, Zimmerman T L,
P J. Hepatic transport of bile salts. Semin Liver Dis 2000; Makishima M, Mangelsdorf D J, et al. The orphan
20(3): 273–92. nuclear receptor, shp, mediates bile acid-induced inhibition

320
Genetic cholestasis, causes and consequences for hepatobiliary transport
of the rat bile acid transporter, ntcp. Gastroenterology 34. Kartenbeck J, Leuschner U, Mayer R, Keppler D.
2001; 121(1): 140–7. Absence of the canalicular isoform of the MRP gene-
18. Sinal C J, Tohkin M, Miyata M, Ward J M, Lambert encoded conjugate export pump from the hepatocytes
G, Gonzalez F J. Targeted disruption of the nuclear in Dubin–Johnson syndrome. Hepatology 1996; 23(5):
receptor FXR/BAR impairs bile acid and lipid homeostasis. 1061–6.
Cell 2000; 102(6): 731–44. 35. Bull L N, Carlton V E, Stricker N L, Baharloo S,
19. Gartung C, Ananthanarayanan M, Rahman M A, DeYoung J A, Freimer N B, et al. Genetic and
Schuele S, Nundy S, Soroka C J, et al. Down-regulation morphological findings in progressive familial intrahepatic
of expression and function of the rat liver Na1/bile acid cholestasis (Byler disease [PFIC-1] and Byler syndrome):
cotransporter in extrahepatic cholestasis. Gastroenterology evidence for heterogeneity. Hepatology 1997; 26(1):
1996; 110(1): 199–209. 155–64.
20. Kerr T A, Saeki S, Schneider M, Schaefer K, Berdy 36. Alonso E M, Snover D C, Montag A, Freese D K,
S, Redder T, et al. Loss of nuclear receptor SHP impairs Whitington P F. Histologic pathology of the liver in
but does not eliminate negative feedback regulation of bile progressive familial intrahepatic cholestasis. J Pediatr
acid synthesis. Dev Cell 2002; 2(6): 713–20. Gastroenterol Nutr 1994; 18(2): 128–33.
21. Holt J A, Luo G, Billin A N, Bisi J, McNeill Y Y, Ko- 37. Bourke B, Goggin N, Walsh D, Kennedy S, Setchell
zarsky K F, et al. Definition of a novel growth factor- K D, Drumm B. Byler-like familial cholestasis in an
dependent signal cascade for the suppression of bile acid extended kindred. Arch Dis Child 1996; 75(3): 223–7.
synthesis. Genes Dev 2003; 17: 1581–91. 38. Arnell H, Nemeth A, Anneren G, Dahl N. Progressive
22. Zollner G, Fickert P, Zenz R, Fuchsbichler A, familial intrahepatic cholestasis (PFIC): evidence for genetic
Stumptner C, Kenner L, et al. Hepatobiliary trans- heterogeneity by exclusion of linkage to chromosome
porter expression in percutaneous liver biopsies of patients 18q21–q22. Hum Genet 1997; 100(3–4): 378–81.
with cholestatic liver diseases. Hepatology 2001; 33(3): 39. Naveh Y, Bassan L, Rosenthal E, Berkowitz D, Jaffe
633–46. M, Mandel H, et al. Progressive familial intrahepatic
23. Fickert P, Zollner G, Fuchsbichler A, Stumptner C, cholestasis among the Arab population in Israel. J Pediatr
Weiglein A H, Lammert F, et al. Ursodeoxycholic acid Gastroenterol Nutr 1997; 24(5): 548–54.
aggravates bile infarcts in bile duct-ligated and Mdr2 40. Kagalwalla A F, Al Amir A R, Khalifa A, Sylven M,
knockout mice via disruption of cholangioles. Gastroenter- Al Ajaji S, Kagalwalla Y A. Progressive familial
ology 2002; 123(4): 1238–51. intrahepatic cholestasis (Byler’s disease) in Arab children.
24. Inokuchi A, Hinoshita E, Iwamoto Y, Kohno K, Ku- Ann Trop Paediatr 1995; 15(4): 321–7.
wano M, Uchiumi T. Enhanced expression of the human 41. Floreani A, Molaro M, Mottes M, Sangalli A, Bar-
multidrug resistance protein 3 by bile salt in human agiotta A, Roda A, et al. Autosomal dominant benign
enterocytes. A transcriptional control of a plausible bile recurrent intrahepatic cholestasis (BRIC) unlinked to 18q21
acid transporter. J Biol Chem 2001; 276(50): 46822–9. and 2q24. Am J Med Genet 2000; 95(5): 450–3.
25. Bohan A, Chen S-W, Denson L A, Held M A, Boyer J 42. Brenard R, Geubel A P, Benhamou J P. Benign recurrent
L. TNF alpha dependent up-regulation of Lrh-1 and intrahepatic cholestasis. A report of 26 cases. J Clin
Mrp3(Abcc3) reduces liver injury in obstructive cholestasis. Gastroenterol 1989; 11(5): 546–51.
J Biol Chem 2003, in press. 43. Cancado E L, Leitao R M, Carrilho F J, Laudanna A
26. Thompson R, Jansen P L. Genetic defects in hepatocana- A. Unexpected clinical remission of cholestasis after
licular transport. Semin Liver Dis 2000; 20(3): 365–72. rifampicin therapy in patients with normal or slightly
27. Bull L N, van Eijk M J, Pawlikowska L, DeYoung J A, increased levels of gamma-glutamyl transpeptidase. Am J
Juijn J A, Liao M, et al. A gene encoding a P-type ATPase Gastroenterol 1998; 93(9): 1510–7.
mutated in two forms of hereditary cholestasis. Nat Genet 44. Balsells F, Wyllie R, Steffen R, Kay M. Benign
1998; 18(3): 219–24. recurrent intrahepatic cholestasis: improvement of pruritus
28. De Vree J M, Jacquemin E, Sturm E, Cresteil D, Bos- and shortening of the symptomatic phase with rifampin
ma P J, Aten J, et al. Mutations in the MDR3 gene cause therapy: a case report. Clin Pediatr (Phila) 1997; 36(8): 483–
progressive familial intrahepatic cholestasis. Proc Natl 5.
Acad Sci USA 1998; 95(1): 282–7. 45. Strautnieks S S, Bull L N, Knisely A S, Kocoshis S A,
29. Jacquemin E, Cresteil D, Manouvrier S, Boute O, Dahl N, Arnell H, et al. A gene encoding a liver-specific
Hadchouel M. Heterozygous non-sense mutation of the ABC transporter is mutated in progressive familial intra-
MDR3 gene in familial intrahepatic cholestasis of preg- hepatic cholestasis. Nat Genet 1998; 20(3): 233–8.
nancy. Lancet 1999; 353(9148): 210–1. 46. Jansen P L, Strautnieks S S, Jacquemin E, Hadchouel
30. Rosmorduc O, Hermelin B, Poupon R. MDR3 gene M, Sokal E M, Hooiveld G J, et al. Hepatocanalicular
defect in adults with symptomatic intrahepatic and gall- bile salt export pump deficiency in patients with progressive
bladder cholesterol cholelithiasis. Gastroenterology 2001; familial intrahepatic cholestasis. Gastroenterology 1999;
120(6): 1459–67. 117(6): 1370–9.
31. Zielenski J, Tsui L C. Cystic fibrosis: genotypic and 47. Deleuze J F, Jacquemin E, Dubuisson C, Cresteil D,
phenotypic variations. Annu Rev Genet 1995; 29: Dumont M, Erlinger S, et al. Defect of multidrug-
777–807. resistance 3 gene expression in a subtype of progressive
32. Mosser J, Lutz Y, Stoeckel M E, Sarde C O, Kretz familial intrahepatic cholestasis. Hepatology 1996; 23(4):
C, Douar A M, et al. The gene responsible for 904–8.
adrenoleukodystrophy encodes a peroxisomal membrane 48. Jacquemin E, De Vree J M, Cresteil D, Sokal E M,
protein. Hum Mol Genet 1994; 3(2): 265–71. Sturm E, Dumont M, et al. The wide spectrum of
33. Paulusma C C, Kool M, Bosma P J, Scheffer G L, ter multidrug resistance 3 deficiency: from neonatal cholestasis
Borg F, Scheper R J, et al. A mutation in the human to cirrhosis of adulthood. Gastroenterology 2001; 120(6):
canalicular multispecific organic anion transporter gene 1448–58.
causes the Dubin–Johnson syndrome. Hepatology 1997; 49. Jacquemin E, Hermans D, Myara A, Habes D, Debray
25(6): 1539–42. D, Hadchouel M, et al. Ursodeoxycholic acid therapy in

321
Jansen and Sturm
pediatric patients with progressive familial intrahepatic intrahepatic cholestasis. Interrelation with intrahepatic
cholestasis. Hepatology 1997; 25(3): 519–23. cholestasis of pregnancy and from oral contraceptives.
50. Jacquemin E. Progressive familial intrahepatic cholestasis. Gastroenterology 1976; 71(2): 202–7.
J Gastroenterol Hepatol 1999; 14(6): 594–9. 58. Mazzella G, Nicola R, Francesco A, Patrizia S, Lu-
51. Emond J C, Whitington P F. Selective surgical manage- ciano B, Anna M, et al. Ursodeoxycholic acid adminis-
ment of progressive familial intrahepatic cholestasis (Byler’s tration in patients with cholestasis of pregnancy: effects on
disease). J Pediatr Surg 1995; 30(12): 1635–41. primary bile acids in babies and mothers. Hepatology 2001;
52. Melter M, Rodeck B, Kardorff R, Hoyer P F, Peter- 33(3): 504–8.
sen C, Ballauff A, et al. Progressive familial intrahepatic 59. Palma J, Reyes H, Ribalta J, Hernandez I, Sandoval
cholestasis: partial biliary diversion normalizes serum lipids L, Almuna R, et al. Ursodeoxycholic acid in the treatment
and improves growth in noncirrhotic patients. Am J of cholestasis of pregnancy: a randomized, double-blind
Gastroenterol 2000; 95(12): 3522–8. study controlled with placebo. J Hepatol 1997; 27(6):
53. Rebhandl W, Felberbauer F X, Turnbull J, Paya K, 1022–8.
Barcik U, Huber W D, et al. Biliary diversion by use of 60. Aagenaes O. Hereditary cholestasis with lymphoedema
the appendix (cholecystoappendicostomy) in progressive (Aagenaes syndrome, cholestasis-lymphoedema syndrome).
familial intrahepatic cholestasis. J Pediatr Gastroenterol New cases and follow-up from infancy to adult age. Scand J
Nutr 1999; 28(2): 217–9. Gastroenterol 1998; 33(4): 335–45.
54. Whitington P F, Whitington G L. Partial external 61. Bull L N, Roche E, Song E J, Pedersen J, Knisely A S,
diversion of bile for the treatment of intractable pruritus Der Hagen C B, et al. Mapping of the locus for
associated with intrahepatic cholestasis. Gastroenterology cholestasis-lymphedema syndrome (Aagenaes syndrome)
1988; 95(1): 130–6. to a 6.6-cM interval on chromosome 15q. Am J Hum
55. Soubrane O, Gauthier F, Devictor D, Bernard O, Genet 2000; 67(4): 994–9.
Valayer J, Houssin D, et al. Orthotopic liver trans- 62. Fruhwirth M, Janecke A R, Muller T, Carlton V E,
plantation for Byler disease. Transplantation 1990; 50(5): Kronenberg F, Offner F, et al. Evidence for genetic
804–6. heterogeneity in lymphedema-cholestasis syndrome. J Pe-
56. Dixon P H, Weerasekera N, Linton K J, Donaldson diatr 2003; 142(4): 441–7.
O, Chambers J, Egginton E, et al. Heterozygous MDR3 63. Carlton V E, Harris B Z, Puffenberger E G, Batta A
missense mutation associated with intrahepatic cholestasis K, Knisely A S, Robinson D L, et al. Complex inheritance
of pregnancy: evidence for a defect in protein trafficking. of familial hypercholanemia with associated mutations in
Hum Mol Genet 2000; 9(8): 1209–17. TJP2 and BAAT. Nat Genet 2003; 34(1): 91–6.
57. de Pagter A G, Berge Henegouwen G P, Bokkel Hui-
nink J A, Brandt K H. Familial benign recurrent

322

You might also like