Clinics and Research in Hepatology and Gastroenterology (2012) 36, 536—553
Available online at
www.sciencedirect.com
MINI REVIEW
The bile salt export pump (BSEP) in health and disease Ralf Kubitz ∗, Carola Dröge , Jan Stindt , Katrin Weissenberger , Dieter Häussinger Medical faculty of the Heinrich-Heine-University of Düsseldorf, Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Düsseldorf, Germany Available online 12 July 2012
Summary The bile salt export pump (BSEP) is the major transporter for the secretion of bile acids from hepatocytes into bile in humans. Mutations of BSEP are associated with cholestatic liver diseases of varying severity including progressive familial intrahepatic cholestasis type 2 (PFIC-2), benign recurrent intrahepatic cholestasis type 2 (BRIC-2) and genetic polymorphisms are linked to intrahepatic cholestasis of pregnancy (ICP) and drug-induced liver injury (DILI). Detailed analysis of these diseases has considerably increased our knowledge about physiology and pathophysiology of bile secretion in humans. This review focuses on expression, localization, and function, short- and long-term regulation of BSEP as well as diseases association and treatment options for BSEP-associated diseases. © 2012 Elsevier Masson SAS. All rights reserved.
Introduction A major function of the liver is the transport of bile salts from blood into bile. Bile salt secretion is required for the removal of cholesterol, which is insoluble in water. Within bile cholesterol is incorporated into mixed micelles, which contain large amounts of bile acids (BA) and phospholipids [1]. 12—20 g of bile acids have to be excreted into bile in order to eliminate 500 mg of cholesterol per day. Therefore, effective transport systems have evolved to ensure bile acid excretion. These include the bile salt export pump (BSEP, gene symbol ABCB11) at the
∗
Corresponding author. Tel.: +49 211 8119648. E-mail address:
[email protected] (R. Kubitz).
canalicular (apical) membrane of hepatocytes as well as the sodium-taurocholate cotransporting polypeptide (NTCP) [2] and organic anion transporting polypeptides (OATPs) [3], which mediate uptake of bile salts from blood into hepatocytes. Secretion of bile acids from hepatocytes back into blood may be achieved by the organic solute transporter OST␣/OST [4] and the multidrug resistance associated protein 4 (MRP4) [5,6]. At the canalicular membrane, the multidrug resistance protein 3 (MDR3, gene symbol ABCB4) flops phosphatidylcholine into bile, and forms a functional unit together with the heterodimeric cholesterol transporter ABCG5/8 and BSEP, because their substrates (phosphatidylcholine, cholesterol, bile acids) together constitute mixed micelles (Fig. 1). Canalicular secretion is regarded as the rate-controlling step of the vectorial transport of bile acids through hepatocytes [7—9]. Due to the crucial localization of BSEP and
2210-7401/$ – see front matter © 2012 Elsevier Masson SAS. All rights reserved. http://dx.doi.org/10.1016/j.clinre.2012.06.006
The bile salt export pump (BSEP) in health and disease
PFIC-1 BRIC-1 PFIC-2 BRIC-2 ICP/ DILI DubinJohnson-S.
FIC1
537
ATP8B1
ATP7B
M. Wilson
BSEP
MDR3
PFIC-3 LPAC ICP
ABCB11
MRP2 ABCC2
ABCB4
SitoABCG5/8 sterolemia
Figure 1 Canalicular transporter proteins and associated liver diseases. Model of a hepatocyte couplet with transporter proteins at the canalicular membrane. ATP8B1 (FIC1) is a putative aminophospholipid flippase, mutations are associated with progressive familial intrahepatic cholestasis type 1 (PFIC-1) and benign recurrent intrahepatic cholestasis type 1 (BRIC-1). Its activity probably is a precondition for proper bile salt export pump (BSEP) function. Mutations of BSEP (ABCB11) are related to PFIC-2, BRIC-2 and drug-induced liver injury (DILI). The multidrug resistance protein 3 (MDR3/ABCB4) acts as a floppase for phosphatidylcholine. PFIC-3, LPAC (low phospholipid associated cholelithiasis syndrome) and ICP (intrahepatic cholestasis of pregnancy) are related to mutations of MDR3. Mutations of the heterodimeric cholesterol transporter ABCG5/8 are related to sitosterolemia (characterized by increased serum levels of plant sterols and premature atherosclerosis). The substrates of BSEP (bile acids), MDR3 (phospholipids) and ABCG5/8 (cholesterol) form mixed micelles in bile. Mutations of the bilirubin transporter multidrug resistance associated protein 2 (MRP2/ABCC2) are linked to the Dubin-Johnson-Syndrome, characterized by conjugated hyperbilirubinemia. Morbus Wilson (copper storage disease) is caused by mutations of the copper transporter ATP7B.
the multiple effects of bile acids, alteration of BSEP may directly or indirectly affect processes such as liver regeneration [10], glucose homeostasis [11], energy expenditure [12], atherosclerosis [13], or carcinogenesis [14]. Elevated bile acid concentrations may be toxic due to their detergent properties [15] and hydrophobic bile acids may activate proapoptotic pathways [16]. All these aspects may explain why BSEP plays a central role in many liver diseases.
Expression, localization and function of bile salt export pump (BSEP) The bile salt export pump BSEP (ABCB11) belongs to the MDR/TAP subfamily of ATP binding cassette transporter (ABC transporter) together with P-glycoprotein (P-gp/or MDR1, gene: ABCB1) and MDR3 (ABCB4). ABCB11 is localized on the long arm of chromosome 2 (2q24) in humans [17]. BSEP consists of 27 coding exons following the untranslated first exon. The protein includes 1.321 amino acids with a molecular mass of approximately 160 kDa (Fig. 2). On a molecular level, BSEP is composed of 12 transmembrane helices and two large nucleotide binding domains. BSEP is responsible for the bile salt-dependent bile flow. It mainly transports monovalent bile acids including taurin and glycine conjugates of the primary bile acids cholic acid (CA) and chenodeoxycholic acid (CDCA) as well as the secondary bile acid deoxycholic acid (DCA) [18]. In addition, it secretes ursodeoxycholic acid (UDCA) and its conjugates into bile. Km-values of human BSEP for taurocholate (TC), taurochenodeoxycholate (TCDC),
glycocheno-deoxycholate (GCDC) and glycocholate (GC) are 6.2 M, 6.6 M, 7.5 M and 21.7 M, respectively, as determined in vesicles from transfected HEK293 cells. Calculated intrinsic clearance values (Vmax/Km) resulted in a rank order of TCDC > GCDC > TC > GC [19]. Because CDCA and its conjugates are potentially more toxic for hepatocytes in vivo [20] and in vitro [16,20] a ‘‘better’’ elimination of chenodeoxycholate as compared to cholate derivatives appears favourable. BSEP is exclusively expressed in hepatocytes and is mainly localized at the canalicular membrane. It is not entirely clear, if BSEP reaches the canalicular membrane directly from the Golgi apparatus to the canalicular membrane [21] or indirectly via the basolateral membrane [22]. The latter possibility is favoured by the observation that anti-BSEP antibodies, which may develop in children with PFIC-2 and liver transplantation, selectively reach the canaliculi within less than 30 minutes [23] in the intact organ, suggesting binding of the antibodies by BSEP at the basolateral membrane followed by transcytosis. Targeting of BSEP from the Golgi to the canalicular membrane is stimulated by the p38MAP kinase and protein kinase C [24], which are (together with extracellular-signalregulated kinases) otherwise involved in bile acid induced exocytosis of transporter bearing vesicles [25,26]. Internalisation of BSEP from the canalicular membrane is regulated by HCLS1-associated protein X-1 (Hax1). Depletion of Hax1 leads to an increase of BSEP at the apical membrane in MDCK cells. Likewise, a dominant negative form of cortactin (lacking the C-terminal SH3 domain), an interacting partner of Hax1, increases apical BSEP in MDCK cells [27]. Cortactin
538
R. Kubitz et al. membrane asymmetry. This may be essential for resistance of the canalicular membrane towards detergent effects of bile acids [34] and was suggested to be directly required for BSEP activity [34,35]. Another possible mechanism of interaction between FIC1 and BSEP involves FXR (see below for details concerning FXR) and the atypical protein kinase C zeta (PKC), which has a prominent canalicular localization [36]. It was shown that inhibition of PKC by a pseudosubstrate or its downregulation by siRNA reduced the nuclear localization of FXR [37], which eventually reduces BSEP expression. The interplay between FIC1 and BSEP on different levels (membrane asymmetry/transporter activity or signalling/transporter expression) explains the development of cholestasis in patients with FIC1 mutations similar to patients with BSEP mutations. However, apart from its role in cholestasis FIC1 deficiency causes several symptoms such as diarrhoea or hearing loss, which are not observed in BSEP disease and which are explained by the extrahepatic expression of FIC1 [38,39]. If in turn reduced BSEP activity influences the proposed FIC1-PKC-FXR cascade is not known yet. Furthermore, Bsep function depends on membrane cholesterol content [40,41]. Interference with cholesterol synthesis by pravastatin, a HMG-CoA reductase inhibitor, not only decreases serum cholesterol levels but also ameliorates cholestasis in bile duct-ligated rats along with an increase in Bsep expression [42].
Long-term regulation of bile salt export pump (BSEP) Figure 2 Molecular model of bile salt export pump (BSEP). A model of BSEP was constructed according to [23] using the crystal structure of the multidrug transporter Sav1866 from S. aureus (Protein Data Bank entry: 2HYD) as a template. The putative transmembrane helices (TMH) of the N-terminal half of the BSEP are shown in green, the TMH of the C-terminal half are shown in blue. They were calculated by the transmembrane helices prediction tool from http://www.enzim.hu/hmmtop [229]. The extracellular loops (representing the probable binding sites for anti-BSEP-autoantibodies, see text) of the N-terminal half are given in magenta. Within the nucleotide binding domains the Walker A motifs are shown in red, the Walker B motif in green and the ABC transporter signature in yellow.
phosphorylation is induced by cell shrinkage via a p47phox and Fyn-dependent pathway, which results in endocytosis of Bsep and Mrp2 and subsequent cholestasis [28]. A tyrosinebased motif at the C-terminus was recently shown to be essential for dynamin- and clathrin-dependent endocytosis of BSEP [29] involving the AP2 adaptor complex [30]. Cortactin is known to interact with dynamin [31] and this interaction may thus represent a link between external stimuli and transporter endocytosis. The ‘‘familial intrahepatic cholestasis 1’’ protein 1 (FIC1, ATP8B1) belongs to the large family of P-type ATPase ion pumps with ubiquitous expression [32]. The exact function of FIC1 and especially its downstream signalling is still under debate. FIC1 probably acts as a flippase, which transports phosphatidylserine and phosphatidylethanolamine from the outer to the inner leaflet of cell membranes [33] thereby maintaining plasma
Long-term regulation of BSEP is complex and has to fulfil many demands. It occurs in large parts on the level of transcription. Major regulators of BSEP expression are the farnesoid X receptor (FXR), the liver receptor homolog 1 (Lrh1) and the nuclear factor erythroid 2-related factor 2 (Nrf2) (Fig. 3). The farnesoid X receptor (FXR/Fxr for the human and rodent orthologues) together with its obligate partner RXR␣ directly transactivates the human [43,44] and rat BSEP/Bsep promoter [45] after bile acid binding to FXR/Fxr. In Fxr knockout mice, basal expression of mouse Bsep (mBsep) is diminished and inducibility of Bsep expression by cholic acid feeding is almost completely abolished [46]. Chenodeoxycholate (CDCA), the predominant bile acid in humans, is the most potent natural agonist for FXR [47,48]. CDCA facilitates the recruitment of the coactivator complex ASCOM (including the coactivator NCOA6 and the H3K4 lysine methyltransferase MLL3) leading to methylation of histones within the mBsep promoter region, a process shown to be essential for Fxr-dependent Bsep expression [49]. Bile acids activate the BSEP promoter via FXR in the rank order of CDCA > DCA > CA > UDCA with a ratio of 25:20:18:8 in vitro, similar to the ratios found in HepG2 cells (∼50:17:5:1) [50], whereas lithocholic acid (LCA) was identified as a ‘‘natural’’ antagonist [51]. The steroid receptor coactivator-2 (SRC-2; also known as nuclear receptor coactivator-2 [NCOA2]) was also shown to cooperatively interact with BSEP promoter stimulation by FXR [52]. SRC-2 is activated via phosphorylation by the AMP activated protein kinase (AMPK), which is regarded as an energy depletion sensing kinase. SRC-2 possesses an intrinsic
The bile salt export pump (BSEP) in health and disease
539
Figure 3 Regulation of the bile salt export pump (BSEP) promoter. Bile acids (BA) are major regulators of BSEP expression through activation of the farnesoid X receptor (FXR). The activating complex ASCOM (see text for details) is recruited by BA/FXR for the methylation (Me) of histones within the BSEP promoter. The nuclear factor erythroid 2-related factor 2 (NRF2) is activated by oxidative stress (e.g. by certain ‘‘toxic’’ BA), binds to a Maf recognition element (MARE) and transactivates the BSEP promoter. The steroid receptor coactivator-2 (SRC2) is activated by the liver kinase B1 (LKB1) and the ‘‘energy sensor kinase’’ AMP activated protein kinase (AMPK) and activates gene expression via acetylation (Ac) of histones. Liver receptor homolog 1 (LRH1) transactivates BSEP and CYP7A1, the rate-controlling enzyme of BA synthesis. Increased BSEP transcription eventually increases BSEP protein expression and removal of intracellular BA. (FXRE: FXR response element; LRHRE: LRH response element; NTCP: Na+ -taurocholate cotransporting polypeptide; ROS: reactive oxygen species; RXR: Retinoid X receptor; the arrows indicate stimulation/activation; dotted lines represent BA flux.)
histone acetyltransferase activity and is recruited to promoter sites of genes. Acetylation of histones by SRC-2 in vicinity to the promoter sites increases the accessibility of the related genes for transcription. It was suggested that the AMPK-SRC2/FXR-BSEP cascade eventually would enhance fat absorption from the intestine via increased BSEP-dependent bile salt secretion [52], representing another link between energy and bile acid homeostasis [53]. The importance of SRC-2 was highlighted by the observation that BSEP mRNA and protein expression was significantly reduced in hepatocytes-specific SRC-2 knockout mice, whereas deletion of SRC-1 or SRC-3 has no effect [52]. Interaction of SRC-2 with several other nuclear receptors including the retinoid X receptor [54,55] or the peroxisome proliferator-activated receptor gamma [56] has been demonstrated. The liver kinase B1 (LKB1), also known as serine/threonine kinase 11 (STK11), acts upstream of the AMPK and in addition activates other kinases from the AMPK-related kinase family. LKB1 regulates cell polarity and, via AMPK, cellular energy homeostasis. Interestingly, liver-specific knockout of LKB1 led to defective bile duct formation, impaired bile acid clearance and a marked retention of mBsep in intracellular pools along with reduced NtcpmRNA and protein expression [57]. In Fxr wildtype mice mBsep expression is upregulated or at least maintained after bile duct ligation (BDL). In Fxr knockout mice (Fxr−/− ) mBsep is almost undetectable with or without BDL. Absent mBsep expression leads to reduced pressure within the biliary tree and absence of bile infarcts after BDL [58] suggesting a beneficial effect of Fxr
repression in BDL. In addition, a stronger upregulation of Mrp4 and Mrp3 expression was observed in Fxr−/− mice [58], which otherwise is repressed by Fxr [59]. Upregulation of MRP4 probably represents an overflow mechanism, which protects hepatocytes from increased bile acids concentrations. In line with this, an almost 10-fold induction of MRP4 was observed in humans with progressive familial intrahepatic cholestasis [60]. Whether stimulation or inhibition of Fxr is more beneficial may depend on the species but also on the type of cholestasis. In rats with obstructive cholestasis, stimulation of Fxr by the Fxr agonist GW4064 has a protective effect [61]. In HepG2 cells stigmasterol acetate, a phytosterol component of intravenous nutrition, suppresses FXR-target genes including BSEP, FGF-19, OST␣/, and SHP. It was suggested that stigmasterol is directly involved in total parenteral nutrition-associated cholestasis due to its FXR-antagonistic action [62]. In mice treated with alphanaphthylisothiocyanate (ANIT) liver injury was more evident in Fxr−/− mice as compared to wildtype or pregnane X receptor knockout mice (Pxr−/− ), in line with a protective role of Fxr in ANIT-induced intrahepatic cholestasis [63]. The liver receptor homolog 1 (Lrh1, NR5A2) is a key regulator of cholesterol 7 alpha-hydroxylase (CYP7A1), a rate-controlling enzyme for BA synthesis from cholesterol. Lrh1 is another regulator of Bsep and in hepatocyte-specific Lrh1 knockout mice a reduced Bsep expression was observed [64] along with reduced cholic acid synthesis [65]. Likewise, the nuclear factor erythroid 2-related factor 2 (Nrf2) positively regulates Bsep [66] and in addition
540 Mrp2, Mrp3 and Mrp4 [67,68]. Nrf2 is a sensor for oxidative stress [69,70], which may be relevant for counteracting reactive oxygen species (ROS) induced by toxic bile acids [71]. Nrf2 binds to the Bsep promoter approx. two hundred base pairs in front of the transcription start site to a musculo-aponeurotic fibrosacroma (Maf) recognition element (MARE) [66]. A single application of perfluorodecanoic acid, a known agonist of peroxisome proliferator-activated receptor ␣ (PPAR␣) induces Mrp3 up to four-fold and Mrp4 more than 30-fold in a Nrf2-dependent manner [72], underscoring the hepatoprotective role of Nrf2. It was even suggested that some beneficial effects of UDCA are mediated by Nrf2 [68]. In the regenerating liver after partial hepatectomy, Bsep expression remains unaffected [73], whereas the sinusoidal uptake transporter Ntcp is downregulated by up to 90% [74] along with a sustained upregulation of the bile salt exporter Mrp4 [75]. Likewise, Oatp1 and 2 (rat) are reduced at the mRNA and protein level by 50 to 60% [73], preventing uncontrolled uptake of potentially toxic substrates into hepatocytes. Interestingly, despite reduced net uptake of bile acids (leading to a 10-fold increase in serum bile salts) bile flow and bile acid excretion into bile per gram body weight remains unaltered [76] or even increase after partial hepatectomy [77]. Although increased bile acid concentrations are generally considered to be toxic, elevated bile acid levels and downstream signalling are required for proper liver regeneration [10].
Short-term regulation of bile salt export pump (BSEP) Rapid adaptation of the activity of canalicular transporter proteins including Bsep may be achieved by rapid vesicular insertion and retrieval into and from the canalicular membrane, which represents a major mechanism for short-term regulation of bile formation [8,78]. The number of transporter molecules and consequently the maximum transport rate Vmax may be adjusted within minutes according to actual needs. This mechanism is involved when changes in hepatocellular hydration [79], hormones [80], bile salts [26,81,82] or oxidative stress [83] occur. Cell volume changes are of central importance, irrespective if they are induced by anisotonic exposure, amino acid uptake, insulin, or ethanol [84,85]. Increments in cell volume increase bile salt secretion in a microtubules-dependent manner [86] and involve rapid insertion of intracellular stored Bsep into the canalicular membrane as shown by immunohistochemistry [79]. Similarly, the bilirubin transporter Mrp2 is regulated by osmolarity [87,88], lipopolysaccharide [89], or oxidative stress [90], however, following hyperosmotic exposure, Bsep and Mrp2 are retrieved into different intracellular vesicular compartments [79]. Only 15% of retrieved vesicles contained both, Bsep and Mrp2, which may be due to different retrieval mechanisms for these transporters. Interestingly, within the canalicular membrane, Bsep is found in the same bile salt resistant lipid microdomains like Mrp2 or transporters such as AbcG5 [91] and it is not yet known how endocytosis of Bsep independent of Mrp2 is achieved. In line with transporter specific mechanisms of regulation, Bsep expression was unchanged in radixin knock out mice
R. Kubitz et al. (a cytoskeletal protein of the ezrin-radixin-moesin family), whereas canalicular Mrp2 protein expression was lacking despite normal Mrp2-mRNA levels [92]. Taurocholate induces hepatocyte swelling, therefore swelling-induced Bsep insertion may acts as feed forward regulation of canalicular secretion [85]. Such a response would also accelerate enterohepatic circulation of bile acids after ingestion of a meal, which in turn triggers nutrient-driven hepatocyte swelling by the concentrative uptake of amino acids into hepatocytes. In addition to endo-/exocytosis Bsep may be regulated by covalent modifications. Coexpression of mBsep and mouse PKC alpha in Sf9 insect cells led to phosphorylation of mBsep [93]. Furthermore, short-chain ubiquitination of BSEP has been demonstrated, which may initiate degradation of BSEP [94]. Signalling events in the above mentioned processes are complex but have been unravelled in large parts. For example, hepatocyte swelling is sensed by integrins (␣5 ß1 ), consecutively triggering activation of focal adhesion kinase, c-Src and mitogen activated protein kinases Erks and p38MAPK [26,95—97]. On the other hand, hyperosmotic Bsep (and Mrp2) retrieval involves endosomal osmosensing with activation of acidic sphingomyelinase, followed by ceramide formation, protein kinase C activation, p47phox phosphorylation and NADPH oxidase activation. The resulting oxidative stress signal induces an activation of the Src family kinase Fyn, which then triggers Bsep retrieval probably via cortactin phosphorylation [28]. Tauroursodeoxycholate (TUDC) stimulates bile acid secretion in part via the same pathways as cell swelling (for detailed review see [98]). In addition, TUDC [99] or taurolithocholic acid [100] activate different PKC isoforms. In the intact organ, activation of Ca2+ -dependent PKC isoforms (cPKC) by phorbolesters or thymeleatoxin, a selective agonist for cPKCs, induces cholestasis [101] along with retrieval of Bsep. cPKCs together with phosphoinositide 3-kinase (PI3K) are also involved in mediating the cholestatic effect of estradiol 17-D-glucuronide (E[2]17G) [102,103]. Evidence was presented that Ca2+ via type II inositol 1,4,5-trisphosphate receptor (InsP3R2) is necessary for maintenance of rat Bsep activity, as shown in sandwich cultures of hepatocytes [104].
Pharmacology of bile salt export pump (BSEP) Although BSEP almost exclusively transports bile salts, endogenous substances as well as many drugs can act as BSEP inhibitors [105,106]. A correlation between potency of BSEP inhibition by different pharmaceuticals and risk of cholestatic drug-induced liver injury (DILI) was observed, which was attributed in part to the physicochemical properties of the compound such as molecular weight, nonpolar surface area and the partition coefficient [107]. Compounds known to inhibit BSEP were found to be hepatocytotoxic in a sandwich culture model, when bile acids such as GCDC were added together with these compounds [108], supporting the concept that intracellular accumulation of bile acids represent an important mechanism of drug-induced liver injury [109]. It has been proposed that the potential of BSEP inhibition should be considered in the course of drug development
The bile salt export pump (BSEP) in health and disease just as properties such as reactive metabolite formation, cytotoxicity and mitochondrial toxicity [110]. Z-Guggulsterone (a plant steroid found in the resin of Commiphora wightii) acts as a FXR antagonist. It was shown that it induces BSEP as well as expression of cholesteryl ester hydrolase (CES1), thereby affecting cholesterol metabolism. Z-Guggulsterone is metabolized by CYP3A4 and its metabolites even more increase BSEP but not CES1. These differential effects on BSEP and CES1 expression may explain why different dosages of Z-Guggulsterone either increase or decrease cholesterol levels [111]. Finally, scoparone, a compound from a Chinese herb, potentiated BSEP expression by increasing the effect of CDCA on the human BSEP promoter [112].
Bile salt export pump (BSEP)-related liver diseases Several transporter proteins are localized within the canalicular membrane, which are associated with (monogenetic) liver diseases including progressive familial intrahepatic cholestasis (PFIC) type 1 to 3, benign recurrent intrahepatic cholestasis (BRIC) type 1 and 2, low phospholipid associated cholelithiasis (LPAC), intrahepatic cholestasis of pregnancy (ICP), Wilson’s disease, Sitosterolemia and the Dubin-Johnson-Syndrome. In addition to inherited diseases BSEP has been recognized to be involved in acquired forms of cholestasis such as drug-induced liver injury (DILI) (Fig. 1).
Progressive familial intrahepatic cholestasis type 2 (PFIC-2) The term ‘‘progressive familial intrahepatic cholestasis’’ (PFIC) was used in order to distinguish cholestasis-induced end stage liver diseases in childhood, which could not be attributed to neonatal or paediatric cholestasis such as biliary atresia, Alagille syndrome or Aagenaes syndrome [113,114]. These criteria were for example consistent with Byler’s disease, a severe form of cholestasis with low gamma-glutamyltranspeptidase (␥GT) levels, which was first observed in an Amish family descending from Jacob and Nancy Byler [115]. Mutations of FIC1 were shown to be responsible for Byler’s disease [116—118], which was termed PFIC-1, accordingly. A second, FIC1-independent low ␥GTcholestasis, formerly termed ‘‘Byler-like syndrome’’ [119] could be linked to mutations of ABCB11/BSEP and was termed PFIC-2 [17,116]. In contrast to PFIC-1 and -2, PFIC3 (due to mutations of ABCB4/MDR3) is characterized by increased ␥GT levels. Commonly, normal ␥GT levels in PFIC1/-2 are ascribed to the low levels of (free) bile salts in bile, and consequently less damage of cholangiocytes [120]. PFIC-2 patients usually present within the first 6 months of life. Jaundice, itching, and growth failure are typical findings in PFIC-2, but some children may present with severe haemorrhage due to vitamin K deficiency. Pruritus usually is more severe in PFIC-2 (and PFIC-1) as compared to PFIC-3 [121]. All PFIC-patients commonly have significantly lower serum cholesterol levels as compared to other forms of paediatric cholestasis [122]. PFIC-2 patients have a higher risk to develop hepatocellular carcinoma [123—125] or cholangiocarcinoma (CC)
541 [126]. A role of BSEP for the development of CC was also suggested on the basis of an association to the BSEP polymorphism p.V444A in a study of 172 CC-patients [127]. The occurrence of pancreatic adenocarcinoma was reported in a patient with PFIC-2, who was treated by biliary diversion [128]. Reduced or even absent expression of canalicular BSEP is a typical feature of PFIC-2 and is of diagnostic value [60,126,129—131], whereas light and electron microscopy findings are variable and non-specific in PFIC-2 [131]. Absent BSEP expression is self-evident for premature stop-codons or frame shift-mutations. However, BSEP deficiency in the context of missense mutations is more difficult to explain. Possible causes include: firstly, direct interference with splicing efficiency as shown for several PFIC-2-relevant missense mutations [132]. Single nucleotide exchanges may reside within so-called exonic splicing enhancers (ESE). ESEs are hexanucleotides that are involved in the guidance of the splicing machinery to splice sites. By the use of an in vitro minigene approach, interference with splicing was shown for c.1445A > G (p.D482G), c.1757C > T (p.T586I), c.3432C > A (p.S1144R), c.3458G > A (p.R1153H), c.3460T > C (p.S1154P), c.3691C > T (p.R1231 W) and c.3692G > A (p.R1231Q) [132]. If these mutations affect splicing in vivo as in vitro needs to be confirmed; secondly, up to 30% of proteins are degraded by the ER-associated degradation (ERAD) pathway [133]. Induction of protein misfolding by missense mutations and consecutive elimination by ERAD may be another reason for decreased BSEP expression. Interestingly, the very common polymorphism p.V444A of BSEP (allele frequency > 50%), which may occur isoallelic to mutations, strongly increases ERAD, as shown in expression studies of cloned BSEP [23]; thirdly, a decrease in half-life may shorten residency of BSEP at the canalicular membrane as shown for the two common PFIC-2 mutations p.E297G and p.D482G in Madin-Darby canine kidney (MDCK) cells [134]. Ubiquitination may induce endocytosis and thereby can initiate degradation of membrane proteins [135,136]. A higher proportion of short-chain ubiquitination was observed in BSEPE297G and BSEPD482G as compared to wildtype BSEP suggesting that ubiquitin dependent degradation is involved in the reduction of transporter half live [94].
Benign recurrent intrahepatic cholestasis type 2 (BRIC-2) Milder forms of BSEP-associated cholestatic liver diseases have been recognized recently and were termed ‘‘benign recurrent intrahepatic cholestasis type 2’’ (BRIC-2) [137]. The clinical presentation of BRIC-2 is similar to BRIC-1 [138], which is caused by ‘‘mild’’ mutations of FIC1 [117,139]. A higher incidence of gall stones was observed in BRIC-2 as compared to BRIC-1 patients [137], a clinical finding, which may guide the diagnostic approach in patients with recurrent, self-limiting cholestasis and low ␥GT levels. Mutations such as p.E186G, p.E297G, p.R432T, p.A570T, p.T923P, p.A926P, p.G1004D, p.R1050C and p.R1128H have been described in BRIC-2 patients [137,140—142] (Table 1). While some BRIC-2 patients have only one (heterozygous) PFIC-2 mutation, others have mutations on both alleles (compound heterozygous or homozygous). There might also
542
Table 1
Genetic variants of the bile salt export pump (bile salt export pump [BSEP]/ABCB11). PFIC M1V G19R L50S M62K C68Y C107R I112T W114R Y157C A167T A167V I182K M183T M183V G188W M217R R223C S226L G238V T242I A257G V284L E297G R303G R303K Q312H R313S G327E W330R
C336S W342G A382G R387H A390P G410D L413W I420T D440E G455E K461E T463I Q466K R470Q Y472C V481E D482G R487H R487P N490D I498T G499E I512T N515T R517H F540L I541L I541T F548Y
D549V G556R G562D A570T L581F A588V S593R I627T E636G R698C S699P E709K G758R G766R Y818F R832C R832H T859R A865V Q869P G877R S901R R948C N979D G982R G1004D T1029K G1032R A1044P
L1055P C1083Y A1110E S1114R G1116E G1116F G1116R S1120N R1128C S1144R R1153C R1153H S1154P N1173D T1210P N1211D V1212F R1231Q R1231W L1242I D1243G R1268Q A1283V G1292V G1298R
Nonsense mutations (premature stop-codons)
S25X E96X W330X Y354X R415X R470X
Y472X W493X R520X I528X R575X Q702X
Y772X Q791X R928X Y1041X R1057X Q1058X
R1090X V1147X Q1215X R1235X E1302X
ICP
Other liver diseases
Genetic variants without disease association
E135K E137K E186G L198P E297G G374S A390P R432T V444A I498T A570T T586I G648V T655I T923P A926P R948C G1004D R1050C G1116R R1128H L1197G R1231Q
E137K L198P E297G R415Q V444A D482G N591S T655I
T87R M123T S194P L198P G260D E297K V444A T510T T655I D676Y P710P L827I G855R E1186K
V43I S56L Q121K R128H I206V V284A G295C G295R G295S R299K R303K L339V H423R V444A V444D V444G A459V I468I R487L Q546K Q558H E592Q V597M R616G T619A M677L M677V R696Q R698H
S701P L712L A865D A865G S874P I939M R958Q F959C F959V T965S F971L F971Y L1006F N1009H K1145N I1183T
R. Kubitz et al.
Missense mutations
BRIC/NFC
PFIC
BRIC/NFC
Splice site mutations
76 + 3G > T 77-1G > C 99-1G > T 150 + 3A > C 390-1G > A 611 + 1G > A
908 + 1delG 908 + 1G > T 908 + 1G > A 1435-13 -8del 2012-8T > G 2178 + 1G > A
2178 + 1G > T 2179-2A > G 2343 + 1G > T 2343 + 2T > C 2611-2A > T R1001R
3057-2A > G 3213 + 1delG 3213 + 4A > G 3213 + 5G > A
Deletions/insertions/frame shifts
Q101Dfs8X T127Hfs6X N199Ifs14X L232Cfs9X R303Sfs17X V368Rfs27X
L380Wfs18X A382 A388del P456Pfs24X H484Rfs5X I528Sfs21X I610Qfs45X
G648Vfs5X K700Sfs12X T919del K930Efs92X K930Efs79X K969 K972del
Q1058Hfs38X I1061Vfs34X L1165del A1192Efs50X T1256Tfs40X
Synonymous variants without disease association
R33R D36D R52R D58D
F90F I134I S136S V195V
L232L Y269Y Q312Q G319G E395E
I416I G418G F427F A535A
ICP
Other liver diseases
Genetic variants without disease association Q159Q Q361Q
F959Hfs1X F959Gfs48X
G557G V597V A804A G817G
I876I G937G Y981Y G1004G
A1028A K1070K T1086T A1110A
The bile salt export pump (BSEP) in health and disease
Table 1 (Continued)
K1145K
The overview shows ≈ 290 known variants of BSEP on the protein level, except splice site mutations, which are shown on cDNA level. The taxonomy and denomination of genetic variations follows the guidelines of the Human Variation Nomenclature Society (http://www.hgvs.org/mutnomen/). The variants are clustered according to the genetic type of mutation/polymorphism and to their connection with diseases (PFIC: progressive familial intrahepatic cholestasis; BRIC: benign recurrent intrahepatic cholestasis; NFC: non-fibrosing cholestasis; ICP: intrahepatic cholestasis of pregnancy). Some mutations and synonymous variants are not (yet) associated with diseases. For a number of mutations/variants the formal proof for a causal relationship to a distinct phenotype is pending. The variants are collected from different databases (http://www.lovd.nl/2.0/; http://www.ncbi.nlm.nih.gov/SNP/; http://pharmacogenetics.ucsf.edu/; http://abcmutations.hegelab.org/; http://www.hgmd.org/) and from literature [116,123,126,129,131,132,137,141—143,152,159,162,165,195,211—228]. The variants printed in bold have been detected in patients analysed in Düsseldorf and are not yet implemented in databases.
543
544
R. Kubitz et al.
Figure 4 Membrane expression of wildtype bile salt export pump (BSEP) and BSEP-G374S in HEK293 cells. (A) Wildtype BSEP-EYFP (BSEP-wt) and (B) BSEP-G374S-EYFP (green) were transfected in HEK293 cells. Endogenous Na+ -K+ -ATPase was stained in red and colocalization between BSEP and Na+ -K+ -ATPase was measured by fluorescence densitometry in single cells. While expression of Na+ -K+ -ATPase was not significantly different (C), total (D) and plasma membrane expression (E) of BSEP was significantly higher (*) in cells transfected with mutant BSEP-G374S as compared to wildtype BSEP. Although G374S is associated with a severe cholestatic phenotype, there is no obvious trafficking defect.
be a transition between BRIC-2 and PFIC-2 as shown for a patient compound heterozygous for p.I498T and c.2098delA [143] or siblings with the homozygous mutation p.G374S. In the latter patients, development of liver cirrhosis was delayed and occurred after childhood. Amino acid 374 is part of the putative sixth transmembrane helix and lies in vicinity to the channel pore, which may explain the severely reduced transport capacity of BSEPG374S (Stindt, Häussinger, Kubitz, unpublished). While an inverse correlation between the amount of BSEP at the plasma membrane and the severity of phenotypes has been described for other mutations [144] p.G374S showed an increased membrane expression of BSEP (as compared to wildtype BSEP) despite the severity of the phenotype (Weissenberger, Häussinger, Kubitz, unpublished) (Fig. 4).
Intrahepatic cholestasis of pregnancy and bile salt export pump (BSEP) Pruritus is the principal symptom of intrahepatic cholestasis of pregnancy (ICP), whereas jaundice occurs in only 10% of patients [145]. It most often starts during the third trimester of pregnancy [146]. ICP has a distinct geographical prevalence and affects 0,1% of pregnancies in Canada and more than 25% of Araucanians in Chile [147]. Increased
serum bile acid concentrations are almost diagnostic for ICP. Bile acid levels above 40 mol/l are predictive for ICP-related complications [147] such as preterm birth or asphyxia [145,147,148]. Most ICP-patients experience spontaneous recovery within 2 weeks after delivery. In order to exclude that transaminases are elevated independent of ICP, follow up until complete normalization of liver enzymes should be assured [149]. A few ‘‘common’’ BSEP mutations (including p.E297G, p.D482G and p.N591S) have been detected in ICP-patients in heterozygous form and account for about 1.4% (7/491) of ICP-patients [150]. Canalicular expression of BSEP with the ‘‘typical’’ ICP mutation p.N591S was higher than that of the BRIC-2 (p.A570 T, p.R1050 C) or PFIC-2 mutations (p.D482G, p.E297G) [144]. Recently, a neonate has been described with homozygousity for p.N591S and, in addition, a homozygous mutation (p.H338Y) in the SLC27A5 gene. SLC27A5 encodes the bile acyl-CoA synthetase, an enzyme responsible for re-conjugation of de-conjugated BA (as part of the enterohepatic circulation) [151]. This child presented with liver fibrosis within the first year of life, while a sibling was free of symptoms although it had the same homozygous SLC27A5 mutation, while it was heterozygous for p.N591S [151], demonstrating a ‘‘dose effect’’ of N591S. Apart from rare mutations, the common BSEP polymorphism p.V444A (c.1331T > C, rs2287622, valine to alanine at
The bile salt export pump (BSEP) in health and disease position 444) has been linked to ICP. The C-allele is more frequent in ICP-patients as compared to healthy pregnant women [150,152,153]. Contraceptive-induced cholestasis (CIC) is similar to ICP and presents with pruritus, and may be more strongly associated with p.V444A [153]. Estrogens are glucuronidated and consecutively secreted into bile by the multidrug resistance related protein 2 (Mrp2). Within bile glucuronidated estrogens transinhibit Bsep [154]. In addition, estrogens induce vesicular retrieval of Bsep in rat liver [80] and 17␣-ethinylestradiol was shown to downregulate Bsep expression in mice [155]. Apart from estrogens metabolites also sulphated progesterone metabolites are increased in ICP [156,157] and may add to BSEP inhibition [158].
Anti-bile salt export pump (BSEP) antibodies and cholestasis When medical or surgical treatment (see below) of PFIC2 patients fails, liver transplantation is often required. It has been observed, that some of these patients suffer from otherwise unexplained recurrent cholestasis similar to preceding PFIC-2. In some of these patients de novo formation of anti-BSEP antibodies could be demonstrated [23,159,160]. Due to our experience (five cases), these antibodies always bind to an extracellular loop of the N-terminal half of BSEP (Fig. 2). It is speculated that the complete absence of BSEP expression (e.g. in the presence of Stopor frame shift-mutations) in the native liver is responsible for the failure to develop auto-tolerance towards BSEP, and therefore BSEP is recognized as a foreign antigen after transplantation. Treatment of children with anti-BSEP antibodies includes a change of immune-suppressants (e.g. from a calcineurin inhibitor to sirolimus [161]), plasmapheresis or immune-adsorption [23]. B-cell depletion by rituximab (directed against the CD20 epitope) represents another treatment option.
Drug-induced liver injury The common BSEP polymorphism p.V444A (c.1331T > C) is not only associated to ICP (see above), but predisposes for the development of drug-induced liver injury (DILI). Allele frequency of the C-allele in patients with DILI was 76% as compared to 59% in controls corresponding to an Odds ratio of 3 [162]. The common explanation for the relation between p.V444A and ICP or DILI is a decreased expression of BSEP in the presence of the polymorphism as shown in a small cohort [132,163], whereas no differences in the transport kinetics of the two variants (valine or alanine at position 444 of BSEP) were observed [162]. Morgan et al. determined IC50 values from 200 model compounds in a vesicle based assay and could identify 38 compounds, which inhibited BSEP-mediated TC transport with IC50 values below 25 M [164]. Ogimura et al. investigated potential BSEP-related toxicity of 26 compounds in an in vitro cell model. Drugs such as Ritonavir, Cyclosporin A, Simvastatin and other became toxic in the presence, but not in the absence of bile acids. Apart from some substances (e.g. verapamil) there was a good correlation between the
545 results of the two studies, suggesting that certain drugs cause cholestasis through direct inhibition of BSEP function.
Other liver diseases in relation to bile salt export pump (BSEP) There is no association between BSEP polymorphisms/mutations and primary biliary cirrhosis (PBC) or primary sclerosing cholangitis (PSC) [165,166]. However, BSEP expression may be severely reduced in MDR3dependent ICP when p.V444A is present [167]. Furthermore, p.V444A may influence development of liver fibrosis in patients with chronic hepatitis C infection. The C-allele (corresponding to alanine) was associated with more severe liver fibrosis [168]. In a single cohort of 352 patients the C-allele (heterozygous or homozygous) was associated to a lower end of treatment response (ETR) and sustained virological response (SVR) [169]. The polymorphism p.V444A has a less stringent impact on HCV treatment as compared to IL-28B polymorphisms [170—173] and further studies are needed [174] to more precisely define the role of BSEP in interferon-based therapy regimens of HCV. There is evidence for an association between BSEP and cholangiocarcinoma (CC). In a recent study a higher prevalence of p.V444A was observed in CC-patients [127]. Restricted BSEP expression due to the genetic polymorphism p.V444A or due to BSEP mutations may result in altered biliary bile salt concentrations, which in turn may have an impact of cholangiocyte proliferation or apoptosis [175]. In the course of liver surgery, when a Pringle manoeuvre is carried out (interruption of blood flow through the hepatic artery and portal vein) ischemia-reperfusion injury may develop. Higher presurgical heat shock protein 70 (Hsp70) and lower Bsep mRNA levels have been associated with lower incidence of postoperative jaundice and a protective role of Hsp70 has been discussed [176]. In critically ill patients from the intensive care unit, elevation of serum bile acids levels were observed and were correlated with a reversal of BA transport due to decreased BSEP and increased basolateral MRP3 expression [177]. Finally, sepsis-associated cholestasis is regularly observed in critically ill patients. It is mediated by lipopolysaccharides (LPS) that trigger the downregulation of Bsep and Mrp2 [89,178,179] involving cytokines such as IL-6, IL-1␣ or TNF␣ [180—182]. In human liver, LPSdependent downregulation of BSEP may largely occur on a posttranscriptional level [183]. LPS-induced endocytosis of Bsep occurs in a zonal pattern with the strongest internalization in periportal hepatocytes [182]. Bsep and the bilirubin transporter Mrp2 may be endocytosed in different compartments [184]. Coadministration of dexamethasone along with LPS prevented cholestasis [89] as did feeding of rats with taurine [185]. Likewise, applying heat shock to livers prior to LPS treatment results in earlier recovery of transporter mRNA. Under these conditions physical interaction between heat shock protein 70 and Bsep was observed, which may protect Bsep from degradation [186].
546
Treatment of bile salt export pump (BSEP)-associated liver diseases In patients with PFIC-2 surgical and medical treatment options intent to prevent growth retardation and complication of chronic liver diseases [187]. Supportive treatment includes supplementation of fat-soluble vitamin K and D in order to prevent haemorrhage and rickets. Administration of medium chain triglycerides, which are absorbed independent of bile acids [187,188] may circumvent malabsorption. Partial biliary external diversion (PEBD) [189—191] aims to bypass the terminal ileum in order to remove bile acids from the enterohepatic circulation. PEBD improves clinical symptoms, growth, histology and liver function tests [191] and a success rate (drop of serum bile acid levels below 10 mol/l, normalization of transaminases) of about 75% can be expected [190]. A laparoscopic approach of PEBD is possible and less invasive [192,193]. Standard treatment of PFIC includes ursodeoxycholic acid (UDCA) in a dose of 20 to 30 mg/kg/day [194], which was shown to be effective in PFIC-2 [39,169,195], PFIC-3 [149,196,197] or PFIC-1 patients [198], although failure of UDCA has also been reported [199]. The underlying genetic variants determine the responsiveness to UDCA therapy; in general treatment is more likely to be effective in patients with missense than with premature stop-codon mutations [196]. UDCA replaces toxic bile salts [200] and may amount to 40% of total serum bile salt concentration. In addition, UDCA may induce choleresis by vesicular insertion of Bsep [26,82] and may counteract bile acid induced apoptosis [201]. UDCA was shown to increase the expression of BSEP in human liver [202] and to restore rat Bsep expression in estrogen-induced cholestasis [203]. Furthermore, although UDCA is less active as an FXR agonist as compared to CDCA or CA, it may effectively induce FXR protein expression and thereby could influence BSEP expression [50]. BRIC patients do not seem to benefit from phenobarbital or cholestyramin [138], two compounds usually used in cholestatic patients [204]. Although characterized by spontaneous resolution of cholestasis, cholestatic episodes in BRIC may be disabling especially due to severe pruritus. Treatment with liver support systems has been shown to be effective and may relieve itching for months [205—207]. Rifampicin has been reported to be effective for treating pruritus in PFIC-patients [208]. Rifampicin not only induces expression of drug metabolising enzymes but also increases BSEP and MRP4 expression [209] along with a reduction of NTCP, altogether lowering intra-hepatocytic bile salt concentrations. It has been suggested that induction of MRP4/Mrp4 acts as an overflow mechanism in order to prevent bile acid overload of hepatocytes [7,60,202,210]. Chaperons may gain a role in the treatment of inherited diseases such as PFIC. At least, 4-phenylbutyrate, which is used for the treatment of urea cycle defects and which may act as a chemical chaperon, was shown to increase membrane expression of BSEP [134].
Concluding remarks The bile salt export pump is crucial for canalicular secretion of bile acids. Mutations of BSEP are associated from
R. Kubitz et al. moderate to severe forms of cholestasis including progressive familial intrahepatic cholestasis (PFIC) type 2, benign recurrent cholestasis (BRIC) type 2 and intrahepatic cholestasis of pregnancy (ICP). Detailed analyses of mutations and genetic variants in humans have promoted our knowledge about bile formation and will in future open new therapeutic strategies for (cholestatic) liver diseases.
Disclosure of interest The authors declare that they have no conflicts of interest concerning this article.
References [1] Admirand WH, Small DM. The physicochemical basis of cholesterol gallstone formation in man. J Clin Invest 1968;47:1043—52. [2] Koopen NR, Wolters H, Muller M, Schippers IJ, Havinga R, Roelofsen H, et al. Hepatic bile salt flux does not modulate level and activity of the sinusoidal Na+ -taurocholate cotransporter (Ntcp) in rats. J Hepatol 1997;27:699—706. [3] Hagenbuch B, Meier PJ. The superfamily of organic anion transporting polypeptides. Biochim Biophys Acta 2003;1609:1—18. [4] Ballatori N, Christian WV, Lee JY, Dawson PA, Soroka CJ, Boyer JL, et al. OSTalpha-OSTbeta: a major basolateral bile acid and steroid transporter in human intestinal, renal, and biliary epithelia. Hepatology 2005;42:1270—9. [5] Rius M, Nies AT, Hummel-Eisenbeiss J, Jedlitschky G, Keppler D. Cotransport of reduced glutathione with bile salts by MRP4 (ABCC4) localized to the basolateral hepatocyte membrane. Hepatology 2003;38:374—84. [6] Rius M, Hummel-Eisenbeiss J, Hofmann AF, Keppler D. Substrate specificity of human ABCC4 (MRP4)-mediated cotransport of bile acids and reduced glutathione. Am J Physiol Gastrointest Liver Physiol 2006;290:G640—9. [7] Trauner M, Boyer JL. Bile salt transporters: molecular characterization, function, and regulation. Physiol Rev 2003;83:633—71. [8] Häussinger D, Schmitt M, Weiergräber O, Kubitz R. Shortterm regulation of canalicular transport. Semin Liver Dis 2000;20:307—21. [9] Häussinger D, Kubitz R, Reinehr R, Bode JG, Schliess F. Molecular aspects of medicine: from experimental to clinical hepatology. Mol Aspects Med 2004;25:221—360. [10] Huang W, Ma K, Zhang J, Qatanani M, Cuvillier J, Liu J, et al. Nuclear receptor-dependent bile acid signaling is required for normal liver regeneration. Science 2006;312:233—6. [11] Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab 2009;10:167—77. [12] Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 2006;439:484—9. [13] Pols TW, Nomura M, Harach T, Lo SG, Oosterveer MH, Thomas C, et al. TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading. Cell Metab 2011;14:747—57. [14] Bernstein H, Bernstein C, Payne CM, Dvorak K. Bile acids as endogenous etiologic agents in gastrointestinal cancer. World J Gastroenterol 2009;15:3329—40. [15] Perez MJ, Briz O. Bile acid induced cell injury protection. World J Gastroenterol 2009;15:1677—89.
The bile salt export pump (BSEP) in health and disease [16] Reinehr R, Graf D, Häussinger D. Bile salt-induced hepatocyte apoptosis involves epidermal growth factor receptordependent CD95 tyrosine phosphorylation. Gastroenterology 2003;125:839—53. [17] Strautnieks SS, Kagalwalla AF, Tanner MS, Knisely AS, Bull L, Freimer N, et al. Identification of a locus for progressive familial intrahepatic cholestasis PFIC2 on chromosome 2q24. Am J Hum Genet 1997;61:630—3. [18] Stieger B. Recent insights into the function and regulation of the bile salt export pump (ABCB11). Curr Opin Lipidol 2009;20:176—81. [19] Hayashi H, Takada T, Suzuki H, Onuki R, Hofmann AF, Sugiyama Y. Transport by vesicles of glycine- and taurineconjugated bile salts and taurolithocholate 3 sulfate: a comparison of human BSEP with rat Bsep. Biochim Biophys Acta 2005;1738:54—62. [20] Song P, Zhang Y, Klaassen CD. Dose-response of five bile acids on serum and liver bile acid concentrations and hepatotoxicty in mice. Toxicol Sci 2011;123:359—67. [21] Kipp H, Arias IM. Newly synthesized canalicular ABC transporters are directly targeted from the Golgi to the hepatocyte apical domain in rat liver. J Biol Chem 2000;275:15917—25. [22] Soroka CJ, Pate MK, Boyer JL. Canalicular export pumps traffic with polymeric immunoglobulin A receptor on the same microtubule-associated vesicle in rat liver. J Biol Chem 1999;274:26416—24. [23] Keitel V, Burdelski M, Vojnisek Z, Schmitt L, Häussinger D, Kubitz R. De novo bile salt transporter antibodies as a possible cause of recurrent graft failure after liver transplantation: a novel mechanism of cholestasis. Hepatology 2009;50:510—7. [24] Kubitz R, Sutfels G, Kuhlkamp T, Kolling R, Haussinger D. Trafficking of the bile salt export pump from the Golgi to the canalicular membrane is regulated by the p38 MAP kinase. Gastroenterology 2004;126:541—53. [25] Beuers U, Bilzer M, Chittattu A, Kullak-Ublick GA, Keppler D, Paumgartner G, et al. 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 2001;33:1206—16. [26] Kurz AK, Graf D, Schmitt M, vom Dahl S, Häussinger D. Tauroursodesoxycholate-induced choleresis involves p38(MAPK) activation and translocation of the bile salt export pump in rats. Gastroenterology 2001;121:407—19. [27] Ortiz DF, Moseley J, Calderon G, Swift AL, Li S, Arias IM. Identification of HAX-1 as a protein that binds bile salt export protein and regulates its abundance in the apical membrane of Madin-Darby canine kidney cells. J Biol Chem 2004;279:32761—70. [28] Cantore M, Reinehr R, Sommerfeld A, Becker M, Häussinger D. The Src Family Kinase Fyn Mediates Hyperosmolarity-induced Mrp2 and Bsep Retrieval from Canalicular Membrane. J Biol Chem 2011;286:45014—29. [29] Lam P, Xu S, Soroka CJ, Boyer JL. A c-terminal tyrosine-based motif in the bile salt export pump directs clathrin-dependent endocytosis. Hepatology 2012;55:1901—11. [30] Hayashi H, Inamura K, Aida K, Naoi S, Horikawa R, Nagasaka H, et al. AP2 mediates BSEP internalization and modulates its hepatocanalicular expression and transport function. Hepatology 2012;55:1889—900. [31] McNiven MA, Kim L, Krueger EW, Orth JD, Cao H, Wong TW. Regulated interactions between dynamin and the actinbinding protein cortactin modulate cell shape. J Cell Biol 2000;151:187—98. [32] Chan H, Babayan V, Blyumin E, Gandhi C, Hak K, Harake D, et al. The p-type ATPase superfamily. J Mol Microbiol Biotechnol 2010;19:5—104.
547 [33] Ujhazy P, Ortiz D, Misra S, Li S, Moseley J, Jones H, et al. Familial intrahepatic cholestasis 1: studies of localization and function. Hepatology 2001;34:768—75. [34] Paulusma CC, Groen A, Kunne C, Ho-Mok KS, Spijkerboer AL, Rudi de WD, et al. Atp8b1 deficiency in mice reduces resistance of the canalicular membrane to hydrophobic bile salts and impairs bile salt transport. Hepatology 2006;44:195—204. [35] Cai SY, Gautam S, Nguyen T, Soroka CJ, Rahner C, Boyer JL. ATP8B1 deficiency disrupts the bile canalicular membrane bilayer structure in hepatocytes, but FXR expression and activity are maintained. Gastroenterology 2009;136:1060—9. [36] Stross C, Keitel V, Winands E, Häussinger D, Kubitz R. Expression and localization of atypical PKC isoforms in liver parenchymal cells. Biol Chem 2009;390:235—44. [37] Chen F, Ellis E, Strom SC, Shneider BL, ATPase Class I. Type 8B Member 1 and protein kinase C zeta induce the expression of the canalicular bile salt export pump in human hepatocytes. Pediatr Res 2010;67:183—7. [38] van Mil SW, Klomp LW, Bull LN, Houwen RH. FIC1 disease: a spectrum of intrahepatic cholestatic disorders. Semin Liver Dis 2001;21:535—44. [39] Pawlikowska L, Strautnieks S, Jankowska I, Czubkowski P, Emerick K, Antoniou A, et al. Differences in presentation and progression between severe FIC1 and BSEP deficiencies. J Hepatol 2010;53:170—8. [40] Paulusma CC, De Waart DR, Kunne C, Mok KS, Elferink RP. Activity of the bile salt export pump (ABCB11) is critically dependent on canalicular membrane cholesterol content. J Biol Chem 2009;284:9947—54. [41] Kis E, Ioja E, Nagy T, Szente L, Heredi-Szabo K, Krajcsi P. Effect of membrane cholesterol on BSEP/Bsep activity: species specificity studies for substrates and inhibitors. Drug Metab Dispos 2009;37:1878—86. [42] Kolouchova G, Brcakova E, Hirsova P, Sispera L, Tomsik P, Cermanova J, et al. Pravastatin modulates liver bile acid and cholesterol homeostasis in rats with chronic cholestasis. J Gastroenterol Hepatol 2011;26:1544—51. [43] Ananthanarayanan M, Balasubramanian N, Makishima M, Mangelsdorf DJ, Suchy FJ. Human bile salt export pump promoter is transactivated by the farnesoid X receptor/bile acid receptor. J Biol Chem 2001;276:28857—65. [44] Plass JR, Mol O, Heegsma J, Geuken M, Faber KN, Jansen PL, et al. Farnesoid X receptor and bile salts are involved in transcriptional regulation of the gene encoding the human bile salt export pump. Hepatology 2002;35:589—96. [45] Gerloff T, Geier A, Roots I, Meier PJ, Gartung C. Functional analysis of the rat bile salt export pump gene promoter. Eur J Biochem 2002;269:3495—503. [46] Sinal CJ, Tohkin M, Miyata M, Ward JM, Lambert G, Gonzalez FJ. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 2000;102:731—44. [47] Parks DJ, Blanchard SG, Bledsoe RK, Chandra G, Consler TG, Kliewer SA, et al. Bile acids: natural ligands for an orphan nuclear receptor. Science 1999;284:1365—8. [48] Makishima M, Okamoto AY, Repa JJ, Tu H, Learned RM, Luk A, et al. Identification of a nuclear receptor for bile acids. Science 1999;284:1362—5. [49] Ananthanarayanan M, Li Y, Surapureddi S, Balasubramaniyan N, Ahn J, Goldstein JA, et al. Histone H3K4 trimethylation by MLL3 as part of ASCOM complex is critical for NR activation of bile acid transporter genes and is downregulated in cholestasis. Am J Physiol Gastrointest Liver Physiol 2011;300:G771—81. [50] Lew JL, Zhao A, Yu J, Huang L, De Pedro N, Pelaez F, et al. The farnesoid X receptor controls gene expression in a ligand- and promoter-selective fashion. J Biol Chem 2004;279:8856—61.
548 [51] Yu J, Lo JL, Huang L, Zhao A, Metzger E, Adams A, et al. Lithocholic acid decreases expression of bile salt export pump through farnesoid X receptor antagonist activity. J Biol Chem 2002;277:31441—7. [52] Chopra AR, Kommagani R, Saha P, Louet JF, Salazar C, Song J, et al. Cellular energy depletion resets whole-body energy by promoting coactivator-mediated dietary fuel absorption. Cell Metab 2011;13:35—43. [53] Ferre P, Foufelle F. A new role for a metabolic star: AMP activated protein kinase stimulates fat absorption. Cell Metab 2011;13:1—2. [54] Wansa KD, Harris JM, Muscat GE. The activation function1 domain of Nur77/NR4A1 mediates transactivation, cell specificity, and coactivator recruitment. J Biol Chem 2002;277:33001—11. [55] Lee WY, Noy N. Interactions of RXR with coactivators are differentially mediated by helix 11 of the receptor’s ligand binding domain. Biochemistry 2002;41:2500—8. [56] Hartig SM, He B, Long W, Buehrer BM, Mancini MA. Homeostatic levels of SRC-2 and SRC-3 promote early human adipogenesis. J Cell Biol 2011;192:55—67. [57] Woods A, Heslegrave AJ, Muckett PJ, Levene AP, Clements M, Mobberley M, et al. LKB1 is required for hepatic bile acid transport and canalicular membrane integrity in mice. Biochem J 2011;434:49—60. [58] Wagner M, Fickert P, Zollner G, Fuchsbichler A, Silbert D, Tsybrovskyy O, et al. Role of farnesoid X receptor in determining hepatic ABC transporter expression and liver injury in bile duct-ligated mice. Gastroenterology 2003;125:825—38. [59] Stedman C, Liddle C, Coulter S, Sonoda J, Alvarez JG, Evans RM, et al. Benefit of farnesoid X receptor inhibition in obstructive cholestasis. Proc Natl Acad Sci U S A 2006;103: 11323—8. [60] Keitel V, Burdelski M, Warskulat U, Kühlkamp T, Keppler D, Häussinger D, et al. Expression and localization of hepatobiliary transport proteins in progressive familial intrahepatic cholestasis. Hepatology 2005;41:1160—72. [61] Liu Y, Binz J, Numerick MJ, Dennis S, Luo G, Desai B, et al. Hepatoprotection by the farnesoid X receptor agonist GW4064 in rat models of intra- and extrahepatic cholestasis. J Clin Invest 2003;112:1678—87. [62] Carter BA, Taylor OA, Prendergast DR, Zimmerman TL, Von FR, Moore DD, et al. Stigmasterol, a soy lipid-derived phytosterol, is an antagonist of the bile acid nuclear receptor FXR. Pediatr Res 2007;62:301—6. [63] Cui YJ, Aleksunes LM, Tanaka Y, Goedken MJ, Klaassen CD. Compensatory induction of liver efflux transporters in response to ANIT-induced liver injury is impaired in FXR-null mice. Toxicol Sci 2009;110:47—60. [64] Song X, Kaimal R, Yan B, Deng R. Liver receptor homolog 1 transcriptionally regulates human bile salt export pump expression. J Lipid Res 2008;49:973—84. [65] Mataki C, Magnier BC, Houten SM, Annicotte JS, Argmann C, Thomas C, et al. Compromised intestinal lipid absorption in mice with a liver-specific deficiency of liver receptor homolog 1. Mol Cell Biol 2007;27:8330—9. [66] Weerachayaphorn J, Cai SY, Soroka CJ, Boyer JL. Nuclear factor erythroid 2-related factor 2 is a positive regulator of human bile salt export pump expression. Hepatology 2009;50:1588—96. [67] Maher JM, Dieter MZ, Aleksunes LM, Slitt AL, Guo G, Tanaka Y, et al. Oxidative and electrophilic stress induces multidrug resistance associated protein transporters via the nuclear factor E2-related factor 2 transcriptional pathway. Hepatology 2007;46:1597—610. [68] Okada K, Shoda J, Taguchi K, Maher JM, Ishizaki K, Inoue Y, et al. Ursodeoxycholic acid stimulates Nrf2-mediated hepatocellular transport, detoxification, and antioxidative stress
R. Kubitz et al.
[69]
[70]
[71]
[72]
[73]
[74]
[75]
[76]
[77]
[78] [79]
[80]
[81]
[82]
[83]
[84]
[85]
[86]
systems in mice. Am J Physiol Gastrointest Liver Physiol 2008;295:G735—47. Klaassen CD, Reisman SA. Nrf2 the rescue: effects of the antioxidative/electrophilic response on the liver. Toxicol Appl Pharmacol 2010;244:57—65. Taguchi K, Motohashi H, Yamamoto M. Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution. Genes Cells 2011;16:123—40. Tan KP, Yang M, Ito S. Activation of nuclear factor (erythroid2 like) factor 2 by toxic bile acids provokes adaptive defense responses to enhance cell survival at the emergence of oxidative stress. Mol Pharmacol 2007;72:1380—90. Maher JM, Aleksunes LM, Dieter MZ, Tanaka Y, Peters JM, Manautou JE, et al. Nrf2- and PPAR alpha-mediated regulation of hepatic Mrp transporters after exposure to perfluorooctanoic acid and perfluorodecanoic acid. Toxicol Sci 2008;106:319—28. Gerloff T, Geier A, Stieger B, Hagenbuch B, Meier PJ, Matern S, et al. Differential expression of basolateral and canalicular organic anion transporters during regeneration of rat liver. Gastroenterology 1999;117:1408—15. Green RM, Gollan JL, Hagenbuch B, Meier PJ, Beier DR. Regulation of hepatocyte bile salt transporters during hepatic regeneration. Am J Physiol 1997;273:G621—7. Miura T, Kimura N, Yamada T, Shimizu T, Nanashima N, Yamana D, et al. Sustained repression and translocation of Ntcp and expression of Mrp4 for cholestasis after rat 90% partial hepatectomy. J Hepatol 2011;55:407—14. Vos TA, Ros JE, Havinga R, Moshage H, Kuipers F, Jansen PL, et al. Regulation of hepatic transport systems involved in bile secretion during liver regeneration in rats. Hepatology 1999;29:1833—9. Csanaky IL, Aleksunes LM, Tanaka Y, Klaassen CD. Role of hepatic transporters in prevention of bile acid toxicity after partial hepatectomy in mice. Am J Physiol Gastrointest Liver Physiol 2009;297:G419—33. Kubitz R, Helmer A, Häussinger D. Biliary transport systems: short-term regulation. Methods Enzymol 2005;400:542—57. Schmitt M, Kubitz R, Lizun S, Wettstein M, Häussinger D. Regulation of the dynamic localization of the rat Bsep gene encoded bile salt export pump by anisoosmolarity. Hepatology 2001;33:509—18. Crocenzi FA, Mottino AD, Cao J, Veggi LM, Pozzi EJ, Vore M, et al. Estradiol-17beta-D-glucuronide induces endocytic internalization of Bsep in rats. Am J Physiol Gastrointest Liver Physiol 2003;285:G449—59. Crocenzi FA, Mottino AD, Sanchez Pozzi EJ, Pellegrino JM, Rodriguez Garay EA, Milkiewicz P, et al. Impaired localisation and transport function of canalicular Bsep in taurolithocholate induced cholestasis in the rat. Gut 2003;52:1170—7. Dombrowski F, Stieger B, Beuers U. Tauroursodeoxycholic acid inserts the bile salt export pump into canalicular membranes of cholestatic rat liver. Lab Invest 2006;86:166—74. Perez LM, Milkiewicz P, Elias E, Coleman R, Sanchez Pozzi EJ, Roma MG. 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 2006;91:150—8. Hallbrucker C, Lang F, Gerok W, Häussinger D. Cell swelling increases bile flow and taurocholate excretion into bile in isolated perfused rat liver. Biochem J 1992;281(Pt 3):593—5. Häussinger D, Hallbrucker C, Saha N, Lang F, Gerok W. Cell volume and bile acid excretion. Biochem J 1992;288(Pt 2):681—9. Häussinger D, Saha N, Hallbrucker C, Lang F, Gerok W. Involvement of microtubules in the swelling-induced stimulation of transcellular taurocholate transport in perfused rat liver. Biochem J 1993;291(Pt 2):355—60.
The bile salt export pump (BSEP) in health and disease [87] Kubitz R, D’ Urso D, Keppler D, Häussinger D. Osmodependent dynamic localization of the multidrug resistance protein 2 in the rat hepatocyte canalicular membrane. Gastroenterology 1997;113:1438—42. [88] Dombrowski F, Kubitz R, Chittattu A, Wettstein M, Saha N, Häussinger D. Electron-microscopic demonstration of multidrug resistance protein 2 (Mrp2) retrieval from the canalicular membrane in response to hyperosmolarity and lipopolysaccharide. Biochem J 2000;348(Pt 1):183—8. [89] Kubitz R, Wettstein M, Warskulat U, Häussinger D. Regulation of the multidrug resistance protein 2 in the rat liver by lipopolysaccharide and dexamethasone. Gastroenterology 1999;116:401—10. [90] Schmitt M, Kubitz R, Wettstein M, vom Dahl S, Häussinger D. Retrieval of the mrp2 gene encoded conjugate export pump from the canalicular membrane contributes to cholestasis-induced by tert-butyl hydroperoxide and chlorodinitrobenzene. Biol Chem 2000;381:487—95. [91] Guyot C, Stieger B. Interaction of bile salts with rat canalicular membrane vesicles: evidence for bile salt resistant microdomains. J Hepatol 2011;55:1368—76. [92] Kikuchi S, Hata M, Fukumoto K, Yamane Y, Matsui T, Tamura A, et al. Radixin deficiency causes conjugated hyperbilirubinemia with loss of Mrp2 from bile canalicular membranes. Nat Genet 2002;31:320—5. [93] Noé J, Hagenbuch B, Meier PJ, St Pierre MV. Characterization of the mouse bile salt export pump overexpressed in the baculovirus system. Hepatology 2001;33:1223—31. [94] Hayashi H, Sugiyama Y. Short-chain ubiquitination is associated with the degradation rate of a cell surface resident bile salt export pump (BSEP/ABCB11). Mol Pharmacol 2009;75:143—50. [95] Häussinger D, Kurz AK, Wettstein M, Graf D, vom Dahl S, Schliess F. Involvement of integrins and Src in tauroursodeoxycholate-induced and swelling-induced choleresis. Gastroenterology 2003;124:1476—87. [96] Schliess F, Reissmann R, Reinehr R, vom DS, Haussinger D. Involvement of integrins and Src in insulin signaling toward autophagic proteolysis in rat liver. J Biol Chem 2004;279:21294—301. [97] vom Dahl S, Schliess F, Reissmann R, Gorg B, Weiergraber O, Kocalkova M, et al. Involvement of integrins in osmosensing and signaling toward autophagic proteolysis in rat liver. J Biol Chem 2003;278:27088—95. [98] Häussinger D, Reinehr R. Osmotic regulation of bile acid transport, apoptosis and proliferation in rat liver. Cell Physiol Biochem 2011;28:1089—98. [99] Beuers U, Throckmorton DC, Anderson MS, Isales CM, Thasler W, Kullak-Ublick GA, et al. Tauroursodeoxycholic acid activates protein kinase C in isolated rat hepatocytes. Gastroenterology 1996;110:1553—63. [100] Beuers U, Probst I, Soroka C, Boyer JL, Kullak-Ublick GA, Paumgartner G. Modulation of protein kinase C by taurolithocholic acid in isolated rat hepatocytes. Hepatology 1999;29:477—82. [101] Kubitz R, Saha N, Kühlkamp T, Dutta S, vom Dahl S, Wettstein M, et al. Ca2+ -dependent protein kinase C isoforms induce cholestasis in rat liver. J Biol Chem 2004;279:10323—30. [102] Crocenzi FA, Sanchez Pozzi EJ, Ruiz ML, Zucchetti AE, Roma MG, Mottino AD, et al. Ca(2+)-dependent protein kinase C isoforms are critical to estradiol 17beta-D-glucuronide-induced cholestasis in the rat. Hepatology 2008;48:1885—95. [103] Boaglio AC, Zucchetti AE, Sanchez Pozzi EJ, Pellegrino JM, Ochoa JE, Mottino AD, et al. Phosphoinositide 3kinase/protein kinase B signaling pathway is involved in estradiol 17beta-D-glucuronide-induced cholestasis: complementarity with classical protein kinase C. Hepatology 2010;52:1465—76.
549 [104] Kruglov EA, Gautam S, Guerra MT, Nathanson MH. Type 2 inositol 1,4,5-trisphosphate receptor modulates bile salt export pump activity in rat hepatocytes. Hepatology 2011;54:1790—9. [105] Byrne JA, Strautnieks SS, Mieli-Vergani G, Higgins CF, Linton KJ, Thompson RJ. The human bile salt export pump: characterization of substrate specificity and identification of inhibitors. Gastroenterology 2002;123:1649—58. [106] Stieger B. The role of the sodium-taurocholate cotransporting polypeptide (NTCP) and of the bile salt export pump (BSEP) in physiology and pathophysiology of bile formation. Handb Exp Pharmacol 2011:205—59. [107] Dawson S, Stahl S, Paul N, Barber J, Kenna JG. In vitro inhibition of the bile salt export pump correlates with risk of cholestatic drug-induced liver injury in humans. Drug Metab Dispos 2012;40:130—8. [108] Ogimura E, Sekine S, Horie T. Bile salt export pump inhibitors are associated with bile acid-dependent drug-induced toxicity in sandwich-cultured hepatocytes. Biochem Biophys Res Commun 2011;416:313—7. [109] Kis E, Ioja E, Rajnai Z, Jani M, Mehn D, Heredi-Szabo K, et al. BSEP inhibition - In vitro screens to assess cholestatic potential of drugs. Toxicol In Vitro 2011 Nov 18 [Epub ahead of print]. [110] Stepan AF, Walker DP, Bauman J, Price DA, Baillie TA, Kalgutkar AS, et al. Structural alert/reactive metabolite concept as applied in medicinal chemistry to mitigate the risk of idiosyncratic drug toxicity: a perspective based on the critical examination of trends in the top 200 drugs marketed in the United States. Chem Res Toxicol 2011;24:1345—410. [111] Yang D, Yang J, Shi D, Xiao D, Chen YT, Black C, et al. Hypolipidemic agent Z-guggulsterone: metabolism interplays with induction of cholesteryl ester hydrolase ces1 and bile salt export pump. J Lipid Res 2012;53:529—39. [112] Yang D, Yang J, Shi D, Deng R, Yan B. Scoparone potentiates transactivation of the bile salt export pump gene and this effect is enhanced by cytochrome P450 metabolism but abolished by a PKC inhibitor. Br J Pharmacol 2011;164:1547—57. [113] Ballow M, Margolis CZ, Schachtel B, Hsia YE. Progressive familial intrahepatic cholestasis. Pediatrics 1973;51:998—1007. [114] Knisely AS. Progressive familial intrahepatic cholestasis: a personal perspective. Pediatr Dev Pathol 2000;3:113—25. [115] Clayton RJ, Iber FL, Ruebner BH, McKusick VA. Byler disease. Fatal familial intrahepatic cholestasis in an Amish kindred. Am J Dis Child 1969;117:112—24. [116] Bull LN, Carlton VE, Stricker NL, Baharloo S, DeYoung JA, Freimer NB, et al. Genetic and morphological findings in progressive familial intrahepatic cholestasis (Byler disease [PFIC-1] and Byler syndrome): evidence for heterogeneity. Hepatology 1997;26:155—64. [117] Bull LN, van Eijk MJ, Pawlikowska L, DeYoung JA, Juijn JA, Liao M, et al. A gene encoding a P-type ATPase mutated in two forms of hereditary cholestasis. Nat Genet 1998;18:219—24. [118] Klomp LW, Vargas JC, van Mil SW, Pawlikowska L, Strautnieks SS, van Eijk MJ, et al. Characterization of mutations in ATP8B1 associated with hereditary cholestasis. Hepatology 2004;40:27—38. [119] Bourke B, Goggin N, Walsh D, Kennedy S, Setchell KD, Drumm B. Byler-like familial cholestasis in an extended kindred. Arch Dis Child 1996;75:223—7. [120] Jacquemin E. Role of multidrug resistance 3 deficiency in pediatric and adult liver disease: one gene for three diseases. Semin Liver Dis 2001;21:551—62. [121] Davit-Spraul A, Gonzales E, Baussan C, Jacquemin E. Progressive familial intrahepatic cholestasis. Orphanet J Rare Dis 2009;4:1. [122] Whitington PF, Freese DK, Alonso EM, Schwarzenberg SJ, Sharp HL. Clinical and biochemical findings in progressive
550
[123]
[124] [125]
[126]
[127]
[128]
[129]
[130]
[131]
[132]
[133]
[134]
[135]
[136]
[137]
[138]
[139]
[140]
R. Kubitz et al. familial intrahepatic cholestasis. J Pediatr Gastroenterol Nutr 1994;18:134—41. Knisely AS, Strautnieks SS, Meier Y, Stieger B, Byrne JA, Portmann BC, et al. Hepatocellular carcinoma in ten children under five years of age with bile salt export pump deficiency. Hepatology 2006;44:478—86. Knisely AS, Portmann BC. Deficiency of BSEP in PFIC with hepatocellular malignancy. Pediatr Transplant 2006;10:644—5. Romano F, Stroppa P, Bravi M, Casotti V, Lucianetti A, Guizzetti M, et al. Favorable outcome of primary liver transplantation in children with cirrhosis and hepatocellular carcinoma. Pediatr Transplant 2011;15:573—9. Strautnieks SS, Byrne JA, Pawlikowska L, Cebecauerova D, Rayner A, Dutton L, et al. Severe bile salt export pump deficiency: 82 different ABCB11 mutations in 109 families. Gastroenterology 2008;134:1203—14. Wadsworth CA, Dixon PH, Wong JH, Chapman MH, McKay SC, Sharif A, et al. Genetic factors in the pathogenesis of cholangiocarcinoma. Dig Dis 2011;29:93—7. Bass LM, Patil D, Rao MS, Green RM, Whitington PF. Pancreatic adenocarcinoma in type 2 progressive familial intrahepatic cholestasis. BMC Gastroenterol 2010;10:30. Jansen PL, Strautnieks SS, Jacquemin E, Hadchouel M, Sokal EM, Hooiveld GJ, et al. Hepatocanalicular bile salt export pump deficiency in patients with progressive familial intrahepatic cholestasis. Gastroenterology 1999;117:1370—9. Wang L, Soroka CJ, Boyer JL. The role of bile salt export pump mutations in progressive familial intrahepatic cholestasis type II. J Clin Invest 2002;110:965—72. Evason K, Bove KE, Finegold MJ, Knisely AS, Rhee S, Rosenthal P, et al. Morphologic findings in progressive familial intrahepatic cholestasis 2 (PFIC2): correlation with genetic and immunohistochemical studies. Am J Surg Pathol 2011;35:687—96. Byrne JA, Strautnieks SS, Ihrke G, Pagani F, Knisely AS, Linton KJ, et al. Missense mutations and single nucleotide polymorphisms in ABCB11 impair bile salt export pump processing and function or disrupt premessenger RNA splicing. Hepatology 2009;49:553—67. Schubert U, Anton LC, Gibbs J, Norbury CC, Yewdell JW, Bennink JR. Rapid degradation of a large fraction of newly synthesized proteins by proteasomes. Nature 2000;404:770—4. Hayashi H, Sugiyama Y. 4-phenylbutyrate enhances the cell surface expression and the transport capacity of wildtype and mutated bile salt export pumps. Hepatology 2007;45:1506—16. Mosesson Y, Yarden Y. Monoubiquitylation: a recurrent theme in membrane protein transport. Isr Med Assoc J 2006;8: 233—7. d’Azzo A, Bongiovanni A, Nastasi T. E3 ubiquitin ligases as regulators of membrane protein trafficking and degradation. Traffic 2005;6:429—41. van Mil SW, Van Der Woerd WL, Van Der BG, Sturm E, Jansen PL, Bull LN, et al. Benign recurrent intrahepatic cholestasis type 2 is caused by mutations in ABCB11. Gastroenterology 2004;127:379—84. Brenard R, Geubel AP, Benhamou JP. Benign recurrent intrahepatic cholestasis. A report of 26 cases. J Clin Gastroenterol 1989;11:546—51. Tygstrup N, Steig BA, Juijn JA, Bull LN, Houwen RH. Recurrent familial intrahepatic cholestasis in the Faeroe Islands. Phenotypic heterogeneity but genetic homogeneity. Hepatology 1999;29:506—8. Kubitz R, Keitel V, Scheuring S, Köhrer K, Häussinger D. Benign recurrent intrahepatic cholestasis associated with mutations of the bile salt export pump. J Clin Gastroenterol 2006;40:171—5.
[141] Chen HL, Liu YJ, Su YN, Wang NY, Wu SH, Ni YH, et al. Diagnosis of BSEP/ABCB11 mutations in Asian patients with cholestasis using denaturing high performance liquid chromatography. J Pediatr 2008;153:825—32. [142] Noe J, Kullak-Ublick GA, Jochum W, Stieger B, Kerb R, Haberl M, et al. Impaired expression and function of the bile salt export pump due to three novel ABCB11 mutations in intrahepatic cholestasis. J Hepatol 2005;43:536—43. [143] Lam CW, Cheung KM, Tsui MS, Yan MS, Lee CY, Tong SF. A patient with novel ABCB11 gene mutations with phenotypic transition between BRIC2 and PFIC2. J Hepatol 2006;44:240—2. [144] Lam P, Pearson CL, Soroka CJ, Xu S, Mennone A, Boyer JL. Levels of plasma membrane expression in progressive and benign mutations of the bile salt export pump (Bsep/Abcb11) correlate with severity of cholestatic diseases. Am J Physiol Cell Physiol 2007;293:C1709—16. [145] Rioseco AJ, Ivankovic MB, Manzur A, Hamed F, Kato SR, Parer JT, et al. Intrahepatic cholestasis of pregnancy: a retrospective case-control study of perinatal outcome. Am J Obstet Gynecol 1994;170:890—5. [146] Lammert F, Marschall HU, Glantz A, Matern S. Intrahepatic cholestasis of pregnancy: molecular pathogenesis, diagnosis and management. J Hepatol 2000;33:1012—21. [147] Glantz A, Marschall HU, Mattsson LA. Intrahepatic cholestasis of pregnancy: relationships between bile acid levels and fetal complication rates. Hepatology 2004;40:467—74. [148] Bacq Y, Sapey T, Brechot MC, Pierre F, Fignon A, Dubois F. Intrahepatic cholestasis of pregnancy: a French prospective study. Hepatology 1997;26:358—64. [149] Kubitz R, Bode J, Erhardt A, Graf D, Kircheis G, Muller-Stover I, et al. Cholestatic liver diseases from child to adult: the diversity of MDR3 disease. Z Gastroenterol 2011;49:728—36. [150] Dixon PH, van MS, Chambers J, Strautnieks S, Thompson R, Lammert F, et al. Contribution of variant alleles of ABCB11 to susceptibility to intrahepatic cholestasis of pregnancy. Gut 2009;58:537—44. [151] Chong CP, Mills PB, McClean P, Gissen P, Bruce C, Stahlschmidt J, et al. Bile acid-CoA ligase deficiency: a new inborn error of bile acid metabolism. J Inherit Metab Dis 2012;35:521—30. [152] Pauli-Magnus C, Lang T, Meier Y, Zodan-Marin T, Jung D, Breymann C, et al. Sequence analysis of bile salt export pump (ABCB11) and multidrug resistance p-glycoprotein 3 (ABCB4, MDR3) in patients with intrahepatic cholestasis of pregnancy. Pharmacogenetics 2004;14:91—102. [153] Meier Y, Zodan T, Lang C, Zimmermann R, Kullak-Ublick GA, Meier PJ, et al. Increased susceptibility for intrahepatic cholestasis of pregnancy and contraceptive-induced cholestasis in carriers of the 1331T > C polymorphism in the bile salt export pump. World J Gastroenterol 2008;14:38—45. [154] Stieger B, Fattinger K, Madon J, Kullak-Ublick GA, Meier PJ. Drug- and estrogen-induced cholestasis through inhibition of the hepatocellular bile salt export pump (Bsep) of rat liver. Gastroenterology 2000;118:422—30. [155] Green RM, Hoda F, Ward KL. Molecular cloning and characterization of the murine bile salt export pump. Gene 2000;241:117—23. [156] Meng LJ, Reyes H, Axelson M, Palma J, Hernandez I, Ribalta J, et al. Progesterone metabolites and bile acids in serum of patients with intrahepatic cholestasis of pregnancy: effect of ursodeoxycholic acid therapy. Hepatology 1997;26:1573—9. [157] Meng LJ, Reyes H, Palma J, Hernandez I, Ribalta J, Sjovall J. Profiles of bile acids and progesterone metabolites in the urine and serum of women with intrahepatic cholestasis of pregnancy. J Hepatol 1997;27:346—57. [158] Vallejo M, Briz O, Serrano MA, Monte MJ, Marin JJ. Potential role of transinhibition of the bile salt export pump by progesterone metabolites in the etiopathogenesis of
The bile salt export pump (BSEP) in health and disease
[159]
[160]
[161]
[162]
[163]
[164]
[165]
[166]
[167]
[168]
[169]
[170]
[171]
[172]
[173]
[174]
intrahepatic cholestasis of pregnancy. J Hepatol 2006;44: 1150—7. Jara P, Hierro L, Martinez-Fernandez P, varez-Doforno R, Yanez F, Diaz MC, et al. Recurrence of bile salt export pump deficiency after liver transplantation. N Engl J Med 2009;361:1359—67. Maggiore G, Gonzales E, Sciveres M, Redon MJ, Grosse B, Stieger B, et al. Relapsing features of bile salt export pump deficiency after liver transplantation in two patients with progressive familial intrahepatic cholestasis type 2. J Hepatol 2010;53:981—6. Basso MS, Subramaniam P, Tredger M, Verma A, Heaton N, Rela M, et al. Sirolimus as renal and immunological rescue agent in pediatric liver transplant recipients. Pediatr Transplant 2011;15:722—7. Lang C, Meier Y, Stieger B, Beuers U, Lang T, Kerb R, et al. Mutations and polymorphisms in the bile salt export pump and the multidrug resistance protein 3 associated with drug-induced liver injury. Pharmacogenet Genomics 2007;17:47—60. Meier Y, Pauli-Magnus C, Zanger UM, Klein K, Schaeffeler E, Nussler AK, et al. Interindividual variability of canalicular ATP binding cassette (ABC)-transporter expression in human liver. Hepatology 2006;44:62—74. Morgan RE, Trauner M, van Staden CJ, Lee PH, Ramachandran B, Eschenberg M, et al. Interference with bile salt export pump function is a susceptibility factor for human liver injury in drug development. Toxicol Sci 2010;118:485—500. Pauli-Magnus C, Kerb R, Fattinger K, Lang T, Anwald B, KullakUblick GA, et al. BSEP and MDR3 haplotype structure in healthy Caucasians, primary biliary cirrhosis and primary sclerosing cholangitis. Hepatology 2004;39:779—91. Poupon R, Ping C, Chretien Y, Corpechot C, Chazouilleres O, Simon T, et al. Genetic factors of susceptibility and of severity in primary biliary cirrhosis. J Hepatol 2008;49:1038—45. Keitel V, Vogt C, Häussinger D, Kubitz R. Combined mutations of canalicular transporter proteins cause severe intrahepatic cholestasis of pregnancy. Gastroenterology 2006;131: 624—9. Iwata R, Stieger B, Mertens JC, Muller T, Baur K, Frei P, et al. The role of bile acid retention and a common polymorphism in the ABCB11 gene as host factors affecting antiviral treatment response in chronic hepatitis C. J Viral Hepat 2011;18:768—78. Kubitz R, Brinkmeyer C, Sagir A, Herebian D, Haussinger D. Genetic variants of the bile salt export pump: inducers and modifiers of liver diseases. Dig Dis 2011;29:89—92. Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, Urban TJ, et al. Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance. Nature 2009;461:399—401. Suppiah V, Moldovan M, Ahlenstiel G, Berg T, Weltman M, Abate ML, et al. IL28B is associated with response to chronic hepatitis C interferon-alpha and ribavirin therapy. Nat Genet 2009;41:1100—4. Tanaka Y, Nishida N, Sugiyama M, Kurosaki M, Matsuura K, Sakamoto N, et al. Genome-wide association of IL28B with response to pegylated interferon-alpha and ribavirin therapy for chronic hepatitis C. Nat Genet 2009;41:1105—9. Rauch A, Kutalik Z, Descombes P, Cai T, Di IJ, Mueller T, et al. Genetic variation in IL28B is associated with chronic hepatitis C and treatment failure: a genome-wide association study. Gastroenterology 2010;138:1338—45. Stieger B, Geier A. Genetic variations of bile salt transporters as predisposing factors for drug-induced cholestasis, intrahepatic cholestasis of pregnancy and therapeutic response of viral hepatitis. Expert Opin Drug Metab Toxicol 2011;7:411—25.
551 [175] Keitel V, Ullmer C, Häussinger D. The membrane-bound bile acid receptor TGR5 (Gpbar-1) is localized in the primary cilium of cholangiocytes. Biol Chem 2010;391:785—9. [176] Bernhardt GA, Zollner G, Cerwenka H, Kornprat P, Fickert P, Bacher H, et al. Hepatobiliary transporter expression and postoperative jaundice in patients undergoing partial hepatectomy. Liver Int 2012;32:119—27. [177] Vanwijngaerden YM, Wauters J, Langouche L, Vander PS, Liddle C, Coulter S, et al. Critical illness evokes elevated circulating bile acids related to altered hepatic transporter and nuclear receptor expression. Hepatology 2011;54:1741—52. [178] Warskulat U, Kubitz R, Wettstein M, Stieger B, Meier PJ, Häussinger D. Regulation of bile salt export pump mRNA levels by dexamethasone and osmolarity in cultured rat hepatocytes. Biol Chem 1999;380:1273—9. [179] Trauner M, Arrese M, Lee H, Boyer JL, Karpen SJ. Endotoxin downregulates rat hepatic Ntcp gene expression via decreased activity of critical transcription factors. J Clin Invest 1998;101:2092—100. [180] Hartmann G, Cheung AK, Piquette-Miller M. Inflammatory cytokines, but not bile acids, regulate expression of murine hepatic anion transporters in endotoxemia. J Pharmacol Exp Ther 2002;303:273—81. [181] Siewert E, Dietrich CG, Lammert F, Heinrich PC, Matern S, Gartung C, et al. Interleukin-6 regulates hepatic transporters during acute-phase response. Biochem Biophys Res Commun 2004;322:232—8. [182] Donner MG, Schumacher S, Warskulat U, Heinemann J, Haussinger D. Obstructive cholestasis induces TNF-alpha- and IL-1 -mediated periportal downregulation of Bsep and zonal regulation of Ntcp, Oatp1a4, and Oatp1b2. Am J Physiol Gastrointest Liver Physiol 2007;293:G1134—46. [183] Elferink MG, Olinga P, Draaisma AL, Merema MT, Faber KN, Slooff MJ, et al. LPS-induced downregulation of MRP2 and BSEP in human liver is due to a posttranscriptional process. Am J Physiol Gastrointest Liver Physiol 2004;287:G1008—16. [184] Zinchuk V, Zinchuk O, Okada T. Experimental LPS-induced cholestasis alters subcellular distribution and affects colocalization of Mrp2 and Bsep proteins: a quantitative colocalization study. Microsc Res Tech 2005;67:65—70. [185] Mühlfeld A, Kubitz R, Dransfeld O, Häussinger D, Wettstein M. Taurine supplementation induces Mrp2 and Bsep expression in rats and prevents endotoxin-induced cholestasis. Arch Biochem Biophys 2003;413:32—40. [186] Bolder U, Jeschke MG, Landmann L, Wolf F, de SC, Schlitt HJ, et al. Heat stress enhances recovery of hepatocyte bile acid and organic anion transporters in endotoxemic rats by multiple mechanisms. Cell Stress Chaperones 2006;11:89—100. [187] De Bruyne R, Van Biervliet S, Vande Velde S, van Zanten BA. Clinical practice: neonatal cholestasis. Eur J Pediatr 2011;170:279—84. [188] Alissa FT, Jaffe R, Shneider BL. Update on progressive familial intrahepatic cholestasis. J Pediatr Gastroenterol Nutr 2008;46:241—52. [189] Whitington PF, Whitington GL. Partial external diversion of bile for the treatment of intractable pruritus associated with intrahepatic cholestasis. Gastroenterology 1988;95:130—6. [190] Emond JC, Whitington PF. Selective surgical management of progressive familial intrahepatic cholestasis (Byler’s disease). J Pediatr Surg 1995;30:1635—41. [191] Kurbegov AC, Setchell KD, Haas JE, Mierau GW, Narkewicz M, Bancroft JD, et al. Biliary diversion for progressive familial intrahepatic cholestasis: improved liver morphology and bile acid profile. Gastroenterology 2003;125:1227—34. [192] Metzelder ML, Bottlander M, Melter M, Petersen C, Ure BM. Laparoscopic partial external biliary diversion procedure in progressive familial intrahepatic cholestasis: a new approach. Surg Endosc 2005;19:1641—3.
552 [193] Metzelder ML, Petersen C, Melter M, Ure BM. Modified laparoscopic external biliary diversion for benign recurrent intrahepatic cholestasis in obese adolescents. Pediatr Surg Int 2006;22:551—3. [194] Paumgartner G, Beuers U. Mechanisms of action and therapeutic efficacy of ursodeoxycholic acid in cholestatic liver disease. Clin Liver Dis 2004;8:67—81 [vi]. [195] Takahashi A, Hasegawa M, Sumazaki R, Suzuki M, Toki F, Suehiro T, et al. Gradual improvement of liver function after administration of ursodeoxycholic acid in an infant with a novel ABCB11 gene mutation with phenotypic continuum between BRIC2 and PFIC2. Eur J Gastroenterol Hepatol 2007;19:942—6. [196] Jacquemin E, De Vree JM, Cresteil D, Sokal EM, Sturm E, Dumont M, et al. The wide spectrum of multidrug resistance 3 deficiency: from neonatal cholestasis to cirrhosis of adulthood. Gastroenterology 2001;120:1448—58. [197] Jacquemin E, Hermans D, Myara A, Habes D, Debray D, Hadchouel M, et al. Ursodeoxycholic acid therapy in pediatric patients with progressive familial intrahepatic cholestasis. Hepatology 1997;25:519—23. [198] Dinler G, Kocak N, Ozen H, Yuce A, Gurakan F. Ursodeoxycholic acid treatment in children with Byler disease. Pediatr Int 1999;41:662—5. [199] Crosignani A, Podda M, Bertolini E, Battezzati PM, Zuin M, Setchell KD. Failure of ursodeoxycholic acid to prevent a cholestatic episode in a patient with benign recurrent intrahepatic cholestasis: a study of bile acid metabolism. Hepatology 1991;13:1076—83. [200] Oude Elferink RP, Paulusma CC. Function and pathophysiological importance of ABCB4 (MDR3 P-glycoprotein). Pflugers Arch 2007;453:601—10. [201] Graf D, Kurz AK, Fischer R, Reinehr R, Häussinger D. Taurolithocholic acid-3 sulfate induces CD95 trafficking and apoptosis in a c-Jun N-terminal kinase-dependent manner. Gastroenterology 2002;122:1411—27. [202] Marschall HU, Wagner M, Zollner G, Fickert P, Diczfalusy U, Gumhold J, et al. Complementary stimulation of hepatobiliary transport and detoxification systems by rifampicin and ursodeoxycholic acid in humans. Gastroenterology 2005;129:476—85. [203] Dumont M, Jacquemin E, Erlinger S. 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 2002;35:185—91. [204] Beuers U, Boberg KM, Chapman RW, Chazouillères O, Invernizzi P, Jones DE, et al. EASL clinical practice guidelines: management of cholestatic liver diseases. J Hepatol 2009;51:237—67. [205] Saich R, Collins P, Ala A, Standish R, Hodgson H. Benign recurrent intrahepatic cholestasis with secondary renal impairment treated with extracorporeal albumin dialysis. Eur J Gastroenterol Hepatol 2005;17:585—8. [206] Sturm E, Franssen CF, Gouw A, Staels B, Boverhof R, De Knegt RJ, et al. Extracorporal albumin dialysis (MARS) improves cholestasis and normalizes low apo A-I levels in a patient with benign recurrent intrahepatic cholestasis (BRIC). Liver 2002;22(Suppl 2):72—5. [207] Huster D, Schubert C, Achenbach H, Caca K, Mossner J, Berr F. Successful clinical application of extracorporal albumin dialysis in a patient with benign recurrent intrahepatic cholestasis (BRIC). Z Gastroenterol 2001;39(Suppl 2):13—4. [208] Yerushalmi B, Sokol RJ, Narkewicz MR, Smith D, Karrer FM. Use of rifampin for severe pruritus in children with chronic cholestasis. J Pediatr Gastroenterol Nutr 1999;29:442—7. [209] Gonzalez R, Cruz A, Ferrin G, Lopez-Cillero P, Briceno J, Gomez MA, et al. Cytoprotective properties of rifampicin are related to the regulation of detoxification system and
R. Kubitz et al.
[210]
[211]
[212]
[213]
[214]
[215]
[216]
[217]
[218]
[219]
[220]
[221]
[222]
[223]
[224]
bile acid transporter expression during hepatocellular injury induced by hydrophobic bile acids. J Hepatobiliary Pancreat Sci 2011;18:740—50. Wang R, Sheps JA, Ling V. ABC transporters, bile acids, and inflammatory stress in liver cancer. Curr Pharm Biotechnol 2011;12:636—46. Alvarez L, Jara P, Sanchez-Sabate E, Hierro L, Larrauri J, Diaz MC, et al. Reduced hepatic expression of Farnesoid X Receptor in hereditary cholestasis associated to mutation in ATP8B1. Hum Mol Genet 2004;13:2451—60. Chen HL, Chang PS, Hsu HC, Ni YH, Hsu HY, Lee JH, et al. FIC1 and BSEP defects in Taiwanese patients with chronic intrahepatic cholestasis with low gamma-glutamyltranspeptidase levels. J Pediatr 2002;140:119—24. Davit-Spraul A, Fabre M, Branchereau S, Baussan C, Gonzales E, Stieger B, et al. ATP8B1 and ABCB11 analysis in 62 children with normal gamma-glutamyl transferase progressive familial intrahepatic cholestasis (PFIC): phenotypic differences between PFIC1 and PFIC2 and natural history. Hepatology 2010;51:1645—55. Goto K, Sugiyama K, Sugiura T, Ando T, Mizutani F, Terabe K, et al. Bile salt export pump gene mutations in two Japanese patients with progressive familial intrahepatic cholestasis. J Pediatr Gastroenterol Nutr 2003;36:647—50. Ho RH, Leake BF, Kilkenny DM, Meyer Zu Schwabedissen HE, Glaeser H, Kroetz DL, et al. Polymorphic variants in the human bile salt export pump (BSEP; ABCB11): functional characterization and interindividual variability. Pharmacogenet Genomics 2010;20:45—57. Kim SR, Saito Y, Itoda M, Maekawa K, Kawamoto M, Kamatani N, et al. Genetic variations of the ABC transporter gene ABCB11 encoding the human bile salt export pump (BSEP) in a Japanese population. Drug Metab Pharmacokinet 2009;24:277—81. Lang T, Haberl M, Jung D, Drescher A, Schlagenhaufer R, Keil A, et al. Genetic variability, haplotype structures, and ethnic diversity of hepatic transporters MDR3 (ABCB4) and bile salt export pump (ABCB11). Drug Metab Dispos 2006;34: 1582—99. Liu C, Aronow BJ, Jegga AG, Wang N, Miethke A, Mourya R, et al. Novel resequencing chip customized to diagnose mutations in patients with inherited syndromes of intrahepatic cholestasis. Gastroenterology 2007;132:119—26. Liu LY, Wang ZL, Wang XH, Zhu QR, Wang JS. ABCB11 gene mutations in Chinese children with progressive intrahepatic cholestasis and low gamma-glutamyltransferase. Liver Int 2010;30:809—15. Matte U, Mourya R, Miethke A, Liu C, Kauffmann G, Moyer K, et al. Analysis of gene mutations in children with cholestasis of undefined etiology. J Pediatr Gastroenterol Nutr 2010;51:488—93. Nobili V, Di GS, Francalanci P, Callea F, Marcellini M, Santorelli FM. A new ABCB11 mutation in two Italian children with familial intrahepatic cholestasis. J Gastroenterol 2006;41:598—603. Sakurai A, Kurata A, Onishi Y, Hirano H, Ishikawa T. Prediction of drug-induced intrahepatic cholestasis: in vitro screening and QSAR analysis of drugs inhibiting the human bile salt export pump. Expert Opin Drug Saf 2007;6:71—86. Scheimann AO, Strautnieks SS, Knisely AS, Byrne JA, Thompson RJ, Finegold MJ. Mutations in bile salt export pump (ABCB11) in two children with progressive familial intrahepatic cholestasis and cholangiocarcinoma. J Pediatr 2007;150:556—9. Shapiro R, Anikster Y, Yardeni T, Korem S, Hartman K, Shamir R, et al. DHPLC screening for mutations in progressive familial intrahepatic cholestasis patients. J Hum Genet 2010;55:308—13.
The bile salt export pump (BSEP) in health and disease [225] Shimizu H, Migita O, Kosaki R, Kasahara M, Fukuda A, Sakamoto S, et al. Living-related liver transplantation for siblings with progressive familial intrahepatic cholestasis 2, with novel genetic findings. Am J Transplant 2011;11:394—8. [226] Siebold L, Dick AA, Thompson R, Maggiore G, Jacquemin E, Jaffe R, et al. Recurrent low gamma-glutamyl transpeptidase cholestasis following liver transplantation for bile salt export pump (BSEP) disease (posttransplant recurrent BSEP disease). Liver Transpl 2010;16:856—63. [227] Strautnieks SS, Bull LN, Knisely AS, Kocoshis SA, Dahl N, Arnell H, et al. A gene encoding a liver-specific ABC transporter is
553 mutated in progressive familial intrahepatic cholestasis. Nat Genet 1998;20:233—8. [228] Walkowiak J, Jankowska I, Pawlowska J, Strautnieks S, Bull L, Thompson R, et al. Exocrine pancreatic function in children with progressive familial intrahepatic cholestasis type 2. J Pediatr Gastroenterol Nutr 2006;42: 416—8. [229] Tusnady GE, Simon I. Principles governing amino acid composition of integral membrane proteins: application to topology prediction. J Mol Biol 1998;283: 489—506.