You are on page 1of 10

Journal of Gastroenterology and Hepatology (2000) 15, 244–253

REVIEW

Pathophysiology of the intrahepatic biliary epithelium

MARIO STRAZZABOSCO,* CARLO SPIRLÌ* AND LAJOS OKOLICSANYI †

*Department of Medical and Surgical Sciences,University of Padova and Azienda Ospedaliera di Padova,
Padova and †University of Parma, Parma, Italy

Abstract The intrahepatic bile duct epithelium modulates the fluidity and alkalinity of the primary
hepatocellular bile from which it reabsorbs fluids, amino acids, glucose and bile acids, while secreting
water, electrolytes and immunoglobulin A. The transport function of the intrahepatic biliary epithelium
is finely regulated by a number of gastrointestinal hormones, neuropeptides and neurotransmitters that
promote either secretion or absorption. The intrahepatic biliary epithelium appears to be a primary
target in a broad group of chronic cholestatic disorders that represent an important cause of morbid-
ity and mortality. The spectrum of cholangiopathies ranges from conditions in which a normal epithe-
lium is damaged by disordered autoimmunity, infectious agents, toxic compounds or ischaemia, to
genetically determined disorders arising from an abnormal bile duct biology, such as cystic fibrosis or
biliary atresia. Probably as a result of the known heterogeneity in cholangiocyte function, different por-
tions of the biliary tree appear to be preferentially affected in specific cholangiopathies. From a patho-
physiological point of view, cholangiopathies are characterized by the coexistence of cholangiocyte loss
(by apoptotic or lytic cell death) with cholangiocyte proliferation and various degrees of portal inflam-
mation, fibrosis and cholestasis. These basic disease mechanisms are discussed in detail. Better under-
standing of cholangiocyte pathophysiology, in particular the immune regulation of cholangiocyte
function, will help in designing newer genetic or pharmacological approaches to treat cholangiopathies.
© 2000 Blackwell Science Asia Pty Ltd

Key words: apoptosis, cholangiocytes, cholestasis, cystic fibrosis, cytokines.

PHYSIOLOGY OF THE BILIARY ments possessing different functional properties, with


EPITHELIUM hormone-regulated bile secretion being confined to the
interlobular and septal intrahepatic bile ducts.4 Diseases
Bile formation requires the coordinated function of of the bile ducts show a clear predilection for special-
hepatocytes and intrahepatic bile duct epithelial cells ized sections of the biliary tree (Fig. 1); for example,
(cholangiocytes). After its secretion by hepatocytes, primary biliary cirrhosis (PBC) selectively involves the
during its route towards the duodenum, the primary interlobular bile ducts, the section where secretin-
bile is extensively modified by the bile duct epithelium stimulated secretion is supposed to be more active. In
which can secrete fluid, bicarbonate, chloride and comparison, drug-induced ductopenia is restricted to
immunoglobulin (Ig)A or reabsorb glucose, bile acids, the small cholangioles, suggesting that there may be
amino acids and electrolytes.1,2 The intrahepatic ducts functional differences between major ducts and cholan-
form an extensive network inside the liver and are gioles in terms of xenobiotic transport and metabolism.
responsible for the production of approximately 30% of The transport function of the intrahepatic bile duct
bile volume, a percentage that can be rapidly increased epithelium is finely regulated by a complex interplay of
to meet changing physiological demands.3 Alkaliniza- gastrointestinal hormones, neuropeptides and neuro-
tion and fluidification of bile prevails in the intrahepatic transmitters that promote either net secretion or ab-
portion of the biliary tree, while bile concentration and sorption. In response to secretin, the intrahepatic bile
acidification mainly take place in the gall-bladder. The duct epithelium increases the hydration and alkalinity
intrahepatic biliary tree is further separated into seg- of hepatocellular bile by secreting Cl– and HCO3–.

Correspondence: Dr M Strazzabosco, Department of Medical and Surgical Sciences, Clinica Medica I°, University of Padova,
Via Giustiniani, 2, 35100 Padova, Italy. Email: mstrazza@ux1.unipd.it
Accepted for publication 30 September 1999.
14401746, 2000, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1440-1746.2000.02091.x by UFPE - Universidade Federal de Pernambuco, Wiley Online Library on [11/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Pathophysiology of the biliary epithelium 245

Figure 1 The biliary epithelium is composed of multiple


segments of increasing diameter that possess different func- Figure 2 Mechanisms of fluid and electrolyte transport in
tional properties. Colangiopathies show a clear predilection cholangiocytes. At the basolateral membrane, acid extrusion
for specialized sections of the biliary tree. Primary bilary is carried out by (1) the Na+/H+ exchanger and (2) Na+:HCO3–
cirrhosis selectively involves the interlobular bile duct, drug- symport or (3) Na+-dependent Cl–/HCO3– in humans. The
induced ductopenia is restricted to small cholangioles, while main acid loader, a Cl–/HCO3– exchanger, is located at both
primary biliary cholangitis may affect any section of the biliary the basolateral (4) and apical (5) membrane, where its activ-
epithelium, from the small cholangiocytes to the extrahepatic ity is stimulated by cAMP/protein kinase A (PKA). (6) A
tree. Na+/H+ exchanger (NHE-2) is located at the apical membrane
and is involved in Na+ reabsorption. (7) The cystic fibrosis
transmembrane conductance regulator (CFTR) is localized at
the apical membrane and is stimulated by secretin and cAMP.
Bicarbonate secretion is a major function of the biliary (8) A water-selective channel, aquaporin 1, located at both the
epithelium, being important, not only to meet digestive apical and the lateral membrane, mediates osmotic water
needs, but also to regulate biliary pH and thus the movement and may be regulated by secretin. In addition to
absorption of weakly lipophilic organic acids. CFTR, a Ca2+-activated Cl– channel (9) is present on the
In the last decade our understanding of the molecular apical pole of the cell and may be activated by luminal puriner-
mechanisms underlying secretory/absorptive functions gic nucleotides. (10) A basolateral K+ channel and Na+/K+-
of cholangiocytes has increased tremendously (Fig. 2). ATPase establish the membrane potential and Na+ gradient.
Here we will briefly summarize current knowledge on Chloride uptake is mediated by Na+/K+/2Cl– cotransport (11).
the molecular physiology of cholangiocyte secretion; the The intrahepatic biliary epithelium also plays a role in Na+-
reader is referred to a number of recent reviews for a dependent reabsorption of taurocholate (TCA) via an apical
more detailed description.1,5,6 A number of different ion carrier similar to the ileal bile acid transporter (12). iBAT, ileal
carriers and channels have been identified The basolat- bile acid transporter.
eral Na+/H+ exchanger isoform 1 (NHE1), Na+:HCO3–
symporter7 (or a Na+-dependent Cl–/HCO3– exchanger
in humans)8 intrude HCO3– into the cell; Na+-K+-
ATPase maintains the Na+ gradient and, together with tamate transporter14 and the ileal bile acid transporter
K+ channels, determines the membrane potential differ- (iBAT)15 are expressed on the apical membrane of
ence. A Na+/K+/2Cl– cotransporter9 actively transports cholangiocytes and participate in the reabsorption of
Cl– ions into the cell and appears to be a major deter- glucose, glutathione breakdown products and conju-
minant of fluid secretion. On the apical pole of the cell, gated bile acids. Finally, a water-selective channel, aqua-
a Na+-independent Cl–/HCO3– exchanger (AE2),7 porin (AQP-1)16 is expressed both at the apical and
extrudes bicarbonate into the bile, according to the intra- basolateral cell membrane and mediates water move-
cellular pH and the in-to-out Cl– gradient imposed ment according to the osmotic gradients.
by a cAMP-stimulated low conductance Cl– channel After binding to a G-protein-coupled receptor and
(CFTR).10 Application of patch–clamp techniques has stimulation of the cAMP/protein kinase A (PKA)-
led to the identification of at least two more types of Cl– dependent signalling pathway,17 secretin, the principal
channels in cholangiocytes: a Ca2+-dependent Cl– con- stimulatory hormone, promotes Cl– efflux, HCO3–
ductance and a Ca2+- and cAMP-independent high secretion and membrane vesicular transport, result-
conductance Cl– channel inhibited by pertussis toxin ing in increased ductular choleresis.18 In addition to
which binds guanosine triphosphate (GTP)-binding secretin, a number of gastrointestinal hormones
proteins.11 More recent work has demonstrated the and neuropeptides stimulate (e.g. vasoactive intestinal
expression of apical isoforms of the Na+/H+ exchanger peptide (VIP)19 and bombesin20) or inhibit (somato-
(NHE2),12 consistent with a role for intrahepatic cholan- statin and gastrin) ductal secretion.21 A rich peptider-
giocytes in Na+ reabsorption. Other carriers, such as the gic plexus is present in the liver parenchyma; in
Na+-dependent glucose transporter (SGLT1),13 the glu- particular, bombesin immunoreactivity is localized in
14401746, 2000, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1440-1746.2000.02091.x by UFPE - Universidade Federal de Pernambuco, Wiley Online Library on [11/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
246 M Strazzabosco et al.

the portal triads in close contact with the bile duct


epithelium.22 Nerve fibres are also present in the wall of
bile ducts; vagal (cholinergic) stimulation increases
HCO3– output after acetylcholine binding to M3
muscarinic receptors on the basolateral membrane
of cholangiocytes and potentiates the rise in cAMP
induced by secretin.23 The effects of secretin and acetyl-
choline, including their net effect on secretion, are
additive, an observation that may bear physiological rel-
evance, as the former stimulates bile duct cells during
periods of parasympathetic activity.23
Regulation of ductal secretion also takes place at the
apical membrane of cholangiocytes through paracrine
factors present in the bile. Among them, ATP, a potent
secretagogue in a number of epithelia, is present in
micromolar concentrations in bile24 where it is released
by hepatocytes.25 Luminal ATP binds to P2Y2 puriner-
gic receptors present at the apical pole of cholangiocytes
and activates luminal Cl– efflux through Ca2+-activated
Cl– channels26 and basolateral HCO3– influx via NHE-
1.27 The effects of ATP and cAMP appear to be
additive in terms of stimulation of HCO3– efflux.
Furthermore, cholangiocytes regulate the concentration
of osmotically active compounds in bile. One such sub-
stance is glutathione, a tripeptide that represents both
the major determinant of bile salt-independent bile flow
and a major hepatic detoxifying mechanism.3 The apical
membrane of cholangiocytes is enriched with g-
glutamyltranspeptidase, an ectoenzyme able to catabo-
lize biliary glutathione to glutamate and cysteinyl-
glycine; which are then taken up by a glutamate carrier
and by a dipeptide transporter located at the apical pole
of cholangiocytes, respectively (H Daniel, pers. comm. Figure 3 Theoretical scheme for paracrine communications
1999), thus realizing an important cycle for the con- between cholangiocytes and non-epithelial cells in the liver.
servation of glutathione. Other important biliary con- Cholangiocytes synthesize and secrete a number of peptides
stituents that are recycled by cholangiocytes are the and mediators that are likely to enable the biliary epithelium
conjugated bile acids which are taken up by an apical to communicate extensively with other liver cells, such as
carrier similar to the ileal bile acid transporter.15,28 The hepatic stellate cells, portal fibroblasts and inflammatory cells,
physiological role of this carrier is still unclear, but it by stimulating the fibrogenic response. However, peptides and
may act as a regulator of the final concentration of mediators, either released in the portal spaces by immune
monomeric bile acids in bile. Apical uptake of glucose cells, macrophages (Mf) and mesenchymal cells or produced
from the bile via SGLT1 may also regulate biliary by the epithelium itself, may have profound effects on epithe-
osmolarity;13 in fact, as SGLT-1 transports approxi- lial cell function, inducing expression of molecules of major
mately 210 water molecules for each Na+ and glucose histocompatibility complex (MHC) class II antigen expression
molecule taken up, this carrier may function as a mo- or affecting transport properties. MCP-1, monocyte chemo-
lecular water pump.29 tactic protein-1; NO, nitric oxide; ET-1, endothelin-1; FGF,
Finally, cholangiocytes synthesize and secrete a fibroblast growth factor;TGF-b1, transforming growth factor-
number of peptides and mediators, such as interleukin b1; PDGF, platelet-derived growth factor; HGF, hepatocyte
(IL)-6,30 transforming growth factor-b2 (TGF-b2,31 growth factor; IFN-g, Interferon-g; CIV, collagen IV; CXVII:
endothelin-1,32 monocyte chemotactic protein-1 collagen XVII; IL, interleukin; TNF-a, tumour necrosis
(MCP-1),33,34 platelet-derived growth factor (PDGF)- factor-a; MMP-1 matrix metalloproteinase-1; PMN, poly-
B chain,31 tumour necrosis factor-a (TNF-a35 and morphonuclear cell. Modified and adapted from Shuppan and
nitric oxide (NO),36 that likely enable the bile duct Hahn.85
epithelium to communicate extensively with other liver
cells, including hepatic stellate cells (HSC), portal
fibroblasts and inflammatory cells (Fig. 3). Interest- BASIC DISEASE MECHANISMS IN
ingly, the biliary epithelium does not produce these CHOLANGIOPATHIES
paracrine factors under normal conditions, but actively
synthesizes them in many forms of acute and chronic A derangement in the physiological processes outlined
liver injury. Thus, activated cholangiocytes are likely to constitutes the pathophysiological basis for cholan-
play an important role, not only in reparative processes, giopathies. This is a group of chronic, progressive, liver
but also in the progression of chronic liver damage in disorders, characterized by the clinical syndrome of
chronic cholestatic syndromes. chronic cholestasis, which affects the biliary tree and is
14401746, 2000, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1440-1746.2000.02091.x by UFPE - Universidade Federal de Pernambuco, Wiley Online Library on [11/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Pathophysiology of the biliary epithelium 247

responsible for a significant morbidity and mortality addition to being involved in ductal plate structure
among paediatric and adult populations.37,38 The spec- regression,42 apoptotic phenomena are also involved in
trum of cholangiopathies ranges from conditions in tissue regression after induced hyperplasia (e.g. the
which the biliary epithelium is damaged by (i) dis- disappearance of proliferated ducts after the relief of
ordered immunity (PBC, graft vs host disease, biliary obstruction).43 The role of apoptosis in cholan-
primary sclerosing cholangitis); (ii) infectious agents giopathies is, however, unclear and because of impor-
(cytomegalovirus or Cryptosporidium infections); (iii) tant methodological problems (apoptosis is a transient
ischaemia (post-transplant hepatic artery stenosis, phenomenon and apoptotic bodies may be eliminated
chronic transplant rejection); (iv) toxic compounds by shedding into the bile duct lumen), the question is
(many common drugs); or (v) genetically transmitted difficult to address. Available morphometric studies
or developmental diseases arising from an abnormal indicate that in PBC, apoptotic and lytic cell deaths
bile duct biology (e.g. cystic fibrosis, Alagille syndrome coexist. Apoptosis of cholangiocytes can be induced in
and biliary atresia). vitro by Cryptosporidium infection.44,45 The effects of
From a pathophysiological point of view, common to cytokines are also unclear: TNF-a, a major pro-
all cholangiopathies is the coexistence of cholangiocyte apoptotic cytokine, is not toxic to cholangiocytes,46
death (lytic or apoptotic), cholangiolar proliferation, while IL-6, a proinflammatory cytokine and a known
various degrees of portal inflammation and fibrosis and autocrine growth factor,30 protects cultured cholangio-
qualitative/quantitative changes in bile production cytes from ceramide-induced apoptosis by changing the
(cholestasis) (Fig. 4). These basic disease mechanisms balance of apoptosis regulatory genes towards an anti-
are present, albeit to a different extent, in all kinds of apoptotic equilibrium (reduction of Bax, increase of
cholangiopathies. Cytokines and other proinflamma- bcl-2).47
tory mediators released in close proximity to the biliary Hydrophobic bile acids can induce apoptosis in hepa-
epithelium contribute to these processes through (i) tocytes via a Fas ligand-independent CD95 (Fas)
stimulation of cholangiocyte apoptotic and proliferative dimerization, induction of the mitochondrial transition
responses;39 (ii) activation of fibrogenetic processes;40 pore and activation of cathepsin B,48 but whether bile
(iii) induction of damage to the peribiliary circulation; acids can trigger apoptosis in cholangiocytes is pre-
(iv) induction of histocompatibility antigen expres- sently unknown. The bile duct epithelium is a potential
sion;41 and (v) alteration of biliary epithelium transport target for bile acid toxicity, as shown by the presence of
function. extensive ductular damage and proliferation with portal
inflammation and fibrosis in mdr-2-deficient mice, an
animal model for progressive familial cholestasis type
III (MDR3 deficiency).49,50 In these conditions, the
Cholangiocyte death (lytic or apoptotic) absence of phospholipids in the bile exposes the biliary
epithelium to the unrestrained effects of high mono-
A vanishing bile duct syndrome (i.e. a decrease in the meric concentrations of bile acids. Under normal
number of bile ducts per portal tract) is the end result conditions, the biliary epithelium is capable of absorb-
of most cholangiopathies. Ductopenia can be consid- ing (passive, non-ionic diffusion, or active transport
ered as the result of bile duct loss prevailing over bile through iBAT), metabolizing and recirculating bile
duct proliferation. The mechanisms by which cholan- acids through the peribiliary circulation. Bile acids may
giocytes die are not well known, but considerable inter- damage the epithelium, promote bile duct cellular pro-
est is being placed on apoptotic phenomena, the role of liferation, or act as secretory signals. According to
which in bile duct morphogenesis is well established. In Alpini et al., expression of iBAT is restricted to major

Figure 4 Pathobiology of the


biliary tree.
14401746, 2000, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1440-1746.2000.02091.x by UFPE - Universidade Federal de Pernambuco, Wiley Online Library on [11/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
248 M Strazzabosco et al.

ducts;28 if so, cholangioles would be more prone to the when in contact with the undifferentiated mesenchyma
toxic effects of hydrophobic bile acids. Hydrophilic bile surrounding the portal vein terminations. The mecha-
acids (e.g ursodeoxycholic acid, UDCA), however, are nisms responsible for bile duct cell proliferation and/or
of benefit in cholangiopathies such as PBC and CF. induction of metaplasia of portal hepatocytes into
Recent data indicate that UDCA counteracts the apop- cholangiocytes, are not well known and their elucida-
totic effects of hydrophobic bile acids on hepatocytes. tion is an area of major effort. A number of growth
It is unknown whether UDCA possess anti-apoptotic factors, such as epidermal growth factor (EGF), hepa-
effects on cholangiocytes. Interestingly, Koga et al. tocyte growth factor (HGF) and IL-6 in humans and
reported a much lower rate of apoptosis in PBC patients insulin-like growth factor-1, EGF and HGF in rats
treated with UDCA.51 have been shown to stimulate cholangiocyte growth
The mechanisms leading to cell damage in immune- in vitro.30 A role for bile acids has been postulated4,57
mediated cholangiopathies are poorly known. Whether and awaits experimental demonstration. The role of
antimitochondrial auto-antibodies have a pathogenic inflammatory mediators, locally released cytokines and
role in PBC is also unknown. The finding that AMA changes in extracellular matrix should also be inves-
antibodies react against the apical membrane of cho- tigated. These studies will be facilitated by the ability
langiocytes in PBC patients52,53 suggests that their mol- to isolate NCAM-positive cells from diseased livers
ecular target is either a mistargeted/truncated form (L Fabris et al. sumbitted).
of pyruvate-dehydrogenase complex-E2 (PDC-E2),
immune complexes or a cross-reactive epitope derived
from an extrinsic (bacterial?) molecule. If this were the
case, its presentation by major histocompatibility Portal inflammation and fibrosis
complex (MHC) class II molecules would allow activa-
tion of CD4+ T cells and, consequently, of the invading Most cholangiopathies are associated with signifi-
effector CD8+ cells. However, as B7 costimulatory mol- cant amounts of inflammatory infiltrate in the portal
ecules seem not to be expressed during the early stages spaces; few studies, however, have addressed the pat-
of the disease, it is presently unclear to what extent tern of cytokines produced in the liver in the course
cholangiocytes may act as professional antigen-present- of immune-mediated cholangiopathies.58 Results are
ing cells. The presence of IgA antimitochondrial anti- equivocal and the interpretation is hampered by dif-
bodies (AMA) in bile of PBC patients and the ferences in the experimental protocols. Studies in PBC
observation that, while PDC-E2 is an ubiquitous have shown increased production of both T helper type
antigen, damage is restricted to biliary cells (i.e. to cells 1 (Th1) cells (IL-2), interferon-g (IFN-g)) and T helper
able to transport IgA transcellularly) have prompted the type 2 (Th2) cells (IL-4, IL-5 and IL-6) cytokines; the
hypothesis that IgA AMA may penetrate the cell and Th1 pattern being predominant in later stages of the
induce liver damage by preventing the correct targeting disease.59 Tumour necrosis factor-a was also shown to
of PDC-E2 and, thus, interfere with mitochondrial increase in PBC.60 An emerging concept is that bile duct
function, an effect that may also result in apoptotic cell epithelial cells are active participants in inflammatory
death.54 However, this sequence of events remains diseases and, in pathological conditions, secrete proin-
highly speculative. flammatory and chemotactic cytokines, such as IL-6,
TNF-a,35 IL-8 and MCP-161 together with growth
factors able to activate mesenchymal cells and matrix
production (endothelin-1, PDGF, TGF-b2), or to
Cholangiocyte proliferation decrease matrix production and leucocyte adhesion
(NO; Fig. 3). These peptides and mediators, either
Cholangiocytes possess latent mitotic capabilities and released in the portal spaces by immune cells,
proliferate following a number of liver injuries (ductu- macrophages and mesenchymal cells or produced by
lar reaction). This is an important step in the develop- the epithelium itself,41 may have profound effects on
ment of chronic liver damage, as ductular reaction is epithelial cell function; for example, IFN-g promotes
considered as the pace-maker of portal fibrosis.55 In MHC class II antigen expression by biliary cells,41
PBC, as in several other cholangiopathies, destruction affects the transport properties of the epithelium and
of interlobular bile ducts is associated with a vigorous stimulates induction of NO production by cholangio-
proliferation of bile ductules; atypical ductules at the cytes.36 In contrast, IL-6 acts as a potent growth factor
border of the cirrhotic nodules show neuroendocrine for biliary cells. Interestingly, in septic patients, cho-
features56 and transiently express phenotypic features lestasis is associated with the pathological picture of
that are present during fetal bile duct development, cholangitis lenta in which proliferation of marginal duc-
such as expression of the anti-apoptotic protein bcl-2 tules coexists with evidence of inspissated bile in bile
and of the neural adhesion molecule (NCAM; L Fabris ductular structures at the edges of the portal tracts.62
et al. submitted). These cells are negative for the prolif- Compared with hepatocellular cholestasis, which is
erative marker Ki67, which is consistent with the origin not associated with progressive fibrosis, in chronic
of atypical ductules from metaplasic hepatocytes. Fol- cholangiopathies there is an extensive fibrotic response
lowing cholestatic injury, it seems that hepatocytes from in the portal tracts. Fibrogenesis is a dynamic process
the limiting plate undergo ductular metaplasia, in a that depends on the extent and duration of parenchy-
manner similar to the ontogenetic response of hepato- mal damage: the persistence of the original noxa causes
blasts that differentiate into primordial cholangiocytes a prolonged activation of tissue repair mechanisms that
14401746, 2000, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1440-1746.2000.02091.x by UFPE - Universidade Federal de Pernambuco, Wiley Online Library on [11/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Pathophysiology of the biliary epithelium 249

leads to tissue fibrosis. Hepatic stellate cells are the activity and increased choleretic responses have also
main connective tissue producing cells and, during been reported in other conditions associated with
tissue repair, undergo a process of activation from the proliferation of differentiated cholangiocytes, such as
quiescent to the highly proliferative myofibroblast alpha-naphthly-isothiocyanate intoxication69 and partial
phenotype.63 This activation, which is also charac- hepatectomy.67
terized by an increased synthesis of collagen types I The role of altered ion transport systems in the
and III, follows a complex interplay between extracellu- pathogenesis of cholangiopathies is demonstrated in
lar matrix components, polypeptide growth factors, cystic fibrosis, a frequently occurring genetic disease in
cytokines and other soluble mediators,63 (Fig. 3). As which mutations in CFTR lead to impaired cAMP-
outlined, most cholangiopathies are associated with the dependent Cl– transport and to pulmonary, pancreatic
proliferation of marginal ducts.55 Several lines of evi- and liver disease. In contrast, in hepatocytes, in which
dence suggest that cholangiocytes play an active role in a number of genetically determined defects in mem-
stimulating the fibrogenic response, possibly activating brane transport systems (such as Dubin–Johnson syn-
the usually quiescent portal fibroblasts and HSC. In drome, progressive familial intrahepatic cholestasis,
chronic liver diseases, in situ hybridization and immuno- benign recurrent intrahepatic cholestasis, etc.)70 have
histochemical studies show that cholangiocytes produce been identified, CF-associated liver disease is the only
paracrine factors such as TGF-a,30 endothelin-1,32 known liver disorder affecting cholangiocyte transport
MCP-1,33,34 PDGF-B and TGF-b2,31 which are able to proteins. Clinical liver decompensation is infrequent,
stimulate HSC activation and matrix production. but bile ductular cell damage can also be demonstrated
Furthermore, bile duct cells synthesize basement in asymptomatic cases, well before hepatocellular
membrane proteins and collagen type IV.64,65 The close damage appears.71 Cystic fibrosis results from a number
association between bile duct proliferation and of mutations in the gene encoding for CFTR, a low con-
mesenchymal activation is also present in cholangio- ductance Cl– channel which in the liver is restricted to
carcinomas, neoplasias that frequently show a strong cholangiocytes and mediates the choleretic effects of
desmoplastic reaction. It is still unclear whether the secretin.71 In addition to its role as a cAMP/PKA acti-
changing epithelial phenotype directly induces an vated Cl– channel, CFTR also participates in the regu-
alteration of the mesenchymal cells or if the changes lation of other membrane transport proteins, such as
in the extracellular matrix induce the phenotypic shift Na+ channels (EnaC),72 K+ channels,73 outward rectify-
in the biliary epithelium. Modulation of the fibrogenic ing Cl– channels,74 and Cl–/HCO3– exchangers.75 The
response may be helpful in avoiding obliteration of CFTR also appear to regulate cellular secretion of
the bile ducts and in slowing the evolution to biliary ATP,76 intracellular vesicle acidification77 and process-
cirrhosis. ing and trafficking of certain proteins.78 In other epithe-
lia CFTR may be permeable to HCO3– and allow a
substantial bicarbonate conductance. For example,
studies in the duodenal mucosa of CFTR (–/–) mice79
Cholestasis suggest a dual pathway for HCO3– secretion: one
component is electrogenic via CFTR-mediated HCO3–
The role of the biliary epithelium in bile produc- conductance, a second component is electroneutral
tion is such that, while damage to the biliary epithelium via CFTR-dependent Cl–/HCO3– exchange and is
will result in the reduced flow of bile, impaired closely associated with carbonic anhydrase activity, an
fluid secretion by cholangiocytes will induce qualita- enzyme that is highly expressed in duct cells. Indeed,
tive changes in the biliary fluid that may predispose cholangiocytes isolated from CF patients undergoing
the biliary epithelium itself to damage from other liver transplantation show reduced cAMP-stimulated
concurrent events. Cholestasis may originate from the Cl– efflux and impaired bicarbonate secretion via
fibro-inflammatory obliteration of bile ducts, from Cl–/HCO3– exchange (A Zsembery et al., submitted).
mutations in transport proteins, or from altered secre- Although the molecular basis of the CF is reasonably
tory responses induced by inflammatory mediators. well understood, the pathogenesis of liver damage in
Complete biliary obstruction is rarely seen in clinical CF is not entirely clear; we speculate that in the pres-
practice; this condition is associated with vigorous pro- ence of a mutated CFTR, reduced bile hydration and
liferation of well-differentiated cholangiocytes, a phe- alkalinity precipitates a series of events that damages
nomenon that may have a compensatory function. In the biliary epithelium, such as retention of cytotoxic
fact, available information suggests that, in contrast to bile acids and xenobiotics and a reduction in na-
hepatocytes where the basolateral sodium-taurocholate tural defences against microbiological pathogens.80 In
transporter polypeptide (ntcp) is decreased,66 chronic response to epithelial damage, release of inflammatory
extrahepatic cholestasis does not impair ion transport mediators and cytokines leads to chronic portal inflam-
in cholangiocytes.67 Rather, the CFTR Cl– channel is mation, that, depending on the immunogenetic back-
more highly expressed10,67 and the choleretic response ground of the individual and other concurrent factors,
to secretin administration is much higher, partly as a may lead to pathognomonic focal biliary cirrhosis
result of an increased expression of the secretin recep- (Fig. 5).71 Furthermore, the lower incidence of CF liver
tor gene.68 These findings might explain the well-known disease, with respect to pancreatic damage, suggests
occurrence of colourless bile enriched with water and that in the liver, other secretory mechanisms and signals
electrolytes after bile duct ligation.67 Increased secretin may play a vicarious role in conditions in which the
receptor gene expression and Cl–/HCO3– exchanger cAMP-mediated secretory pathway is impaired. As
14401746, 2000, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1440-1746.2000.02091.x by UFPE - Universidade Federal de Pernambuco, Wiley Online Library on [11/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
250 M Strazzabosco et al.

IFN-g and TNF-a) inhibit cAMP-dependent fluid


secretion and Cl–/HCO3– activity in polarized rat
cholangiocytes.86 These data are of interest because they
demonstrate that cholangiocyte secretory function is
deeply affected by the inflammatory microenvironment
in the absence of duct obliteration and advocate the use
of anti-inflammatory agents in cholangiopathies.

CONCLUSIONS
Figure 5 Current working hypothesis for the pathogenesis of
liver diseases in cystic fibrosis patients. Reduced bile hydra- The role of the biliary epithelium in the pathogenesis
tion and alkalinity precipitates a series of events able to of biliary tract disease is just beginning to be unveiled.
damage the biliary epithelium: reduced biliary clearance The emerging idea is that cholangiocytes are active
increases the exposure to pathogens and toxic compounds. participants, rather than innocent bystanders, in the
Furthermore, as hypothesized in the respiratory epithelium,80 development of cholangiopathies. Whether genetically
the altered volume and quality of bile may impair the determined, induced by extrahepatic causes or by
epithelium’s innate immune system. This is the first line of inflammatory mediators, the epithelium will respond
defence against pathogenic insult and includes the integrity to the different noxae via apoptotic or lytic cell death
of the epithelium, the continuous flow of bile and the secre- programmes, by activating mitosis and producing a vast
tion of yet to be identified multiple substances with anti- array of mediators that enable paracrine communica-
inflammatory and antimicrobial properties. *In the airways tion with non-parenchymal and inflammatory cells and
epithelium this includes lysozymes, phospholipase A2, defens, by changing its transport capability. Reduced fluid
cathelicidin, complement and immunoglobulin A. and electrolyte transport by cholangiocytes (ductular
cholestasis) is a central step because it will predispose
the biliary epithelium to further damage. The continu-
already discussed, a candidate mediator is biliary ATP, ing inflammatory/fibrotic/cholestatic response will have
which binds to luminal P2Y2 receptors, activates apical a key role in the progression of the disease. The field of
Ca2+-dependent Cl– channels other than CFTR, and immune regulation of cholangiocyte function will prove
increases the activity of NHE-1, possibly resulting in to be very rewarding in the future, particularly in view
stimulation of HCO3 efflux.27 In fact, our own data of the potential to pharmacologically block the action
show that, in cholangiocytes isolated from CF livers, an of specific cytokines. Furthermore, better understand-
increase in intracellular Ca2+ concentration, induced by ing of cholangiocyte physiology will help in the design
administration of ionomycin leads to parallel activation of newer genetic or pharmacological agents that are able
of Cl– channels and HCO3– extrusion. These results to target specific cholangiocyte functions and are likely
indicate that, in CFTR-deficient cells, secretion of to provide effective and safe therapeutic strategies in the
bicarbonate can be restored by alternative activation of near future.
intracellular Ca2+-dependent Cl– channels, an observa-
tion that may have therapeutical relevance.81
The molecular pathogenesis of cholestasis in ACKNOWLEDGEMENTS
immune-mediated cholangiopathies remains unclear.
Cholestasis may be due to obliteration of bile ducts, but The financial support of Telethon (grant E-873) and
it is usually present well before the onset of ductopenia of Ministero Dell’ Università e della Ricerca Scientifica
and it can be seen even in the absence of canalicular (Cofinanziamento 1998, Grant 9806210866) is grate-
dilatation and bile plugs. It has been suggested that fully acknowledged.
alterations in biliary electrolyte transport may con-
tribute to the pathogenesis of cholestasis in primary dis-
orders of bile ducts. For example, defective expression
of the gene coding for the AE-2 anion exchanger REFERENCES
and reduced immunoreactivity for the apical anion
exchanger have been reported in patients with PBC.82,83 1 Strazzabosco M. New insights into cholangiocyte
Changes in cholangiocyte transport function may also physiology. J. Hepatol. 1997; 27: 945–52.
be secondary to the inflammatory condition. In contrast 2 Tavoloni N. The intrahepatic biliary epithelium: An area
to experimental cholestasis, where inflammation is of growing interest in hepatology. Semin. Liver Dis. 1987;
minimal, most human cholangiopathies are associated 7: 280–92.
with portal inflammation and to the release of a number 3 Nathanson MH, Boyer JL. Mechanisms and regulation of
of proinflammatory cytokines that may have profound bile secretion. Hepatology 1991; 14: 551–66.
effects on epithelial cell function, and even alter its 4 Alpini G, Glaser S, Robertson W et al. Bile acid stimulates
transport properties.84 For example, endotoxin admin- proliferative and secretory events in large but not small
istration decreases bile flow, an effect that is inhibited cholangiocytes. Am. J. Physiol. 1997; 273: G518–29.
by antibodies against TNF-a. Our own data show that 5 Strazzabosco M. Biliary tract physiology. Curr. Opin.
mixtures of different proinflammatory cytokines (IL-6, Gastroenterol. 1998; 14: 395–401.
14401746, 2000, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1440-1746.2000.02091.x by UFPE - Universidade Federal de Pernambuco, Wiley Online Library on [11/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Pathophysiology of the biliary epithelium 251

6 Boyer JL. Bile duct epithelium: Frontiers in transport 24 Chari RS, Schutz SM, Haebig JE et al. Adenosine
physiology. Am. J. Physiol. 1996; 270: G1–5. nucleotides in bile. Am. J. Physiol. 1996; 270: G246–
7 Strazzabosco M, Mennone A, Boyer JL. Intracellular pHi 52.
regulation in bile ductular cells isolated from bile duct 25 Schlosser SF, Burgstahler AD, Nathanson MH. Isolated
ligated rats. J. Clin. Invest. 1991; 87: 1503–12. rat hepatocytes can signal to other hepatocytes and bile
8 Strazzabosco M, Joplin R, Zsembery A et al. Na+- duct cells by release of nucleotides. Proc. Natl Acad. Sci.
dependent and -independent Cl–/HCO3– exchange USA 1996; 63: 9948–53.
mediate cellular HCO3– transport in cultured human 26 Schlenker T, Roman JMJ, Sharara AC et al. Regulation of
intrahepatic bile duct cells. Hepatology 1997; 25: 976–85. biliary secretion through apical purinergic receptors in
9 Singh SK, Mennone A, Boyer JL. Na+-K+-2Cl– cotrans- cultured rat cholangiocytes. Am. J. Physiol. 1997; 273:
port participates in stimulated fluid secretion in isolated G1108–17.
polarized bile duct units from rat liver. Hepatology 1996; 27 Zsembery A, Spirlì C, Granato A et al. Purinergic
26: 279A (Abstract). regulation of acid/base transport in human and rat biliary
10 Cohn JA, Strong TV, Picciotto MR, Nairn AC, Collins FS, epithelial cell lines. Hepatology 1998; 28: 914–20.
Fitz JG. Localization of the cystic fibrosis transmembrane 28 Alpini G, Glaser SS, Rodgers R et al. Functional expres-
conductance regulator in human bile duct epithelial cells. sion of the apical Na+-dependent bile acid transporter in
Gastroenterology 1993; 105: 1857–64. large but not small rat cholangiocytes. Gastroenterology
11 McGill JM, Gettys TW, Basavappa S, Fitz JG. GTP- 1997; 113: 1734–40.
binding proteins regulate high conductance anion chan- 29 Wolkoff AW, Suchy FJ, Moseley RH et al. Advances in
nels in rat bile duct epithelial cells. J. Membr. Biol. 1993; hepatic transport: Molecular mechanisms, genetic
133: 253–61. disorders and treatment. A summmary of the 1998
12 Spirlì C, Granato A, Zsembery A, Anglani F et al. Func- AASLD single topic conference. Hepatology 1998; 28:
tional polarity of Na+/H+ and Cl–/HCO3– exchangers in a 1713–19.
rat cholangiocyte cell line. Am. J. Physiol. 1998; 275: 30 Matsumoto K, Fujii H, Michalopoulos G, Fung JJ,
G1236–45. Demetris AJ. Human biliary epithelial cells secrete and
13 Lazaridis KN, Pham L,Vroman B, de Groen PC, LaRusso respond to cytokines and hepatocyte growth factors in
NF. Kinetic and molecular identification of sodium- vitro: Interleukin-6, hepatocyte growth factor and epider-
dependent glucose transporter in normal rat cholangio- mal growth factor promote DNA synthesis in vitro. Hepa-
cytes. Am. J. Physiol. 1997; 272: G1168–74. tology 1994; 20: 376–82.
14 Eisemann-Tappe I, Wizigmann S, Gebhardt R. Glutamate 31 Milani S, Herbst H, Schuppan D, Stein H, Surrenti C.
uptake in primary cultures of biliary epithelial cells from Transforming growth factors b1 and b2 are differentially
normal rat liver. Cell Biol. Toxicol. 1991; 7: 315–25. expressed in fibrotic liver disease. Am. J. Pathol. 1991; 139:
15 Lazaridis KN, Pham L, Tiets P et al. Rat cholangiocytes 1221–9.
adsorb bile acids at their apical domain via the ileal 32 Caligiuri A, Glaser S, Rodgers RE et al. Endothelin-1
sodium-dependent bile acid transporter. J. Clin. Invest. inhibits secretin-stimulated ductal secretion by interacting
1997; 100: 2714–21. with ETA receptors on large cholangiocytes. Am. J.
16 Marinelli RA, Pham L, Agre P, LaRusso NF. Secretin Physiol. 1998; 275: G835–46.
promotes osmotic water transport in rat cholangiocytes 33 Marra F, De Franco R, Grappone C et al. Increased
by increasing aquaporin-1 water channels in plasma expression of monocyte chemotactic protein-1 during
membranes. Evidence for a secretin-induced vesicular active hepatic fibrogenesis: Correlation with monocyte
translocation of aquaporin-1. J. Biol. Chem. 1997; 272: 12 infiltration. Am. J. Pathol. 1998; 152: 423–30.
984–8. 34 Morland CM, Fear J, McNab G et al. Promotion of leuko-
17 Alvaro D, Mennone A, Boyer JL. Role of kinases and cyte transendothelial cell migration by chemokines
phosphatases in the regulation of fluid secretion and derived from human biliary epithelial cells in vitro. Proc.
Cl–/HCO3– exchange in cholangiocytes. Am. J. Physiol. AAP 1997; 109: 372–82.
1997; 273: G303–13. 35 Yasoshima M, Kono N, Sugawara H, Katayanagi K,
18 Lenzen R, Alpini G, Tavoloni N. Secretin stimulates bile Harada K, Nakanuma Y. Increased expression of
ductular secretory activity through the cAMP system. Am. interleukin-6 and tumor necrosis factor-alpha in patho-
J. Physiol. 1992; 263: G527–32. logic biliary epithelial cells: In situ and culture study. Lab.
19 Cho WK, Mennone A, Boyer JL. VIP stimulates bile Invest. 1998; 78: 89–100.
secretion in cholangiocytes by cAMP-PKA independent 36 Vos TA, Gouw ASH, Klok PA et al. Differential effects of
mechanisms. Hepatology 1997; 26: 397A (Abstract). nitric oxide synthase inhibitors on endotoxin-induced liver
20 Cho WK, Mennone A, Rydberg SA, Boyer JL. Bombesin damage in rats. Gastroenterology 1997; 113: 1323–33.
stimulates bicarbonate secretion from rat cholangiocytes: 37 Roberts SK, LaRusso NF. Pathobiology of biliary
Implications for neural regulation of bile secretion. epithelia. Curr. Opin. Gastroenterol. 1994; 10: 526–33.
Gastroenterology 1997; 113: 311–21. 38 Woolf GM, Vierling JM. Disappearing intrahepatic bile
21 Glass GB. Gastrointestinal peptide hormones as modula- ducts: The syndromes and their mechanisms. Semin. Liver
tors of bile secretion. Prog. Liver Dis. 1982; 7: 243–60. Dis. 1993; 13: 261–75.
22 Cai W, Gu J, Huang W et al. Peptide immunoreactive 39 Sakamoto T, Liu Z, Murase N et al. Mitosis and apopto-
nerves and cells of the guinea pig gallbladder and biliary sis in the liver of interleukin-6-deficient mice after partial
pathways. Gut 1983; 24: 1186–93. hepatectomy. Hepatology 1999; 29: 403–11.
23 Alvaro D. Biliary epithelium: A new chapter in cell biology. 40 Friedman SL. Cytokines and fibrogenesis. Semin. Liver
Ital. J. Gastroenterol. Hepatol. 1999; 31: 78–83. Dis. 1999; 19: 129–40.
14401746, 2000, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1440-1746.2000.02091.x by UFPE - Universidade Federal de Pernambuco, Wiley Online Library on [11/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
252 M Strazzabosco et al.

41 Ayres RCS, Neuberger JM, Shaw J, Adams DH. Intercel- sis: Predominance of Th1 subset. Hepatology 1997; 25:
lular adhesion molecule-1 and MHC antigens on human 791–6.
intrahepatic bile duct cells: Effect of proinflammatory 60 Berg PA, Klein R, Rocken M. Cytokines in primary biliary
cytokines. Gut 1993; 34: 1245–9. cirrhosis. Semin. Liver Dis. 1997; 17: 115–23.
42 Terada T, Nakanuma Y. Detection of apoptosis and expres- 61 Adams DH. Biliary epithelial cells: Innocent victims or
sion of apoptosis-related proteins during human intra- active participants in immune-mediated liver disease? J.
hepatic bile duct development. Am. J. Pathol 1995; 146: Lab Clin. Med. 1996; 128: 528–30.
67–74. 62 Lefkovitch JH. Bile ductular cholestasis: An ominous
43 Bhathal PS, Gall JA. Deletion of hyperplastic biliary histopathologic sign related to sepsis and ‘cholangitis
epithelial cells by apoptosis following removal of the lenta’. Human Pathol. 1981; 13: 19–24.
proliferative stimulus. Liver 1985; 5: 311–25. 63 Pinzani M, Marra F, Carloni V. Signal transduction in
44 Chen XM, Levine SA, Tietz P et al. Cryptosporidium hepatic stellate cells. Liver 1998; 18: 2–13.
parvum is cytopathic for cultured human biliary epithelia 64 Herbst H, Frey A, Heinrichs O et al. Heterogeneity of
via an apoptotic mechanism. Hepatology 1998; 28: 907–13. liver cells expressing procollagen type I and IV in vivo.
45 Chen XM, Gores GJ, Paya CV, LaRusso NF. Cryp- Histochem. Cell Biol. 1997; 107: 399–409.
tosporidium parvum induces apoptosis in biliary epithelia 65 Terada T, Nakanuma Y. Expression of tenascin, type IV
by a Fas/Fas ligand-dependent mechanism. Am. J. Physiol. collagen and laminin during human intrahepatic bile duct
1999; 277: G599–608. development and intrahepatic cholangiocarcinoma.
46 Mano Y, Ishii M, Okamoto H, Igarashi T, Kobayashi K, Histopathology 1994; 25: 143–50.
Toyota T. Effect of tumor necrosis factor alpha on intra- 66 Green RM, Whiting JF, Rosenbluth AB, Beier D, Gollan
hepatic bile duct epithelial cell of rat liver. Hepatology JL. Interleukin-6 inhibits hepatocyte taurocholate uptake
1996; 23: 1602–7. and sodium-potassium-adenosinetriphosphate activity.
47 Patel T, LaRusso NF, Gores JG. IL-6 suppresses cholan- Am. J. Physiol. 1994; 267: G1094–100.
giocyte apoptosis by down-regulation of Bax. Hepatology 67 Alpini G, Lenzi R, Zhai WR, Slott PA, Lin MH, Tavoloni
1997; 26: 226A (Abstract). N. Bile secretory function of intrahepatic biliary epithe-
48 Rodrigues CMP, Fan G, Ma X, Kren BT, Steer CJ. A lium in the rat. Am. J. Physiol. 1989; 257: G124–33.
novel role for ursodeoxycholic acid in inhibiting apopto- 68 Alpini G, Ulrich II CD, Phillips JO, Pham LD, Miller JL,
sis by modulating mitochondrial membrane perturbation. LaRusso NF. Upregulation of secretin receptor gene
J. Clin. Invest. 1998; 101: 2790–9. expression in rat cholangiocytes after bile duct ligation.
49 De Vree JML, Jacquemin E, Sturm E et al. Mutations in Am. J. Physiol. 1994; 266: G922–8.
the MDR3 gene cause progressive familial intrahepatic 69 Slott PA, Liu MH,Tavoloni N. Origin, pattern, and mech-
cholestasis. Proc. Natl Acad. Sci. USA 1998; 95: 282–7. anism of bile duct proliferation following biliary obstruc-
50 Smit JJ, Schinkel AH, Oude Elferink RP et al. Homozy- tion in the rat. Gastroenterology 1990; 99: 466–77.
gous disruption of the murine mdr2 P-glycoprotein gene 70 Arias IM. New genetics of inheritable jaundice and
leads to a complete absence of phospholipid from bile and cholestatic liver disease. Lancet 1998; 352: 82–3.
to liver disease. Cell 1993; 75: 451–62. 71 Colombo C, Battezzati PM, Strazzabosco M, Podda M.
51 Koga H, Sakisaka S, Ohishi M, Sata M, Tanikawa K. Liver and biliary problems in cystic fibrosis. Semin. Liver
Nuclear DNA fragmentation and expression of Bcl-2 in Dis. 1998; 18: 227–35.
primary biliary cirrhosis. Hepatology 1997; 25: 1077–84. 72 Stutts MJ, Canessa CM, Olsen JC. CFTR as a cAMP-
52 Joplin R, Lindsay JG, Johnson GD, Strain A, Neuberger dependent regulator of sodium channels. Science 1995;
J. Membrane dihydrolipoamideacetyltransferase (E2) on 269: 847–50.
human biliary cirrhosis. Lancet 1992; 339: 93–4. 73 McNicholas CM, Guggino WB, Schwiebert EM, Hebert
53 Van de Water J, Turchany J, Leung PSC et al. Molecular SC, Giebish G, Egan ME. Sensitivity of a renal K+ channel
mimicry in primary biliary cirrhosis. J. Clin. Invest. 1993; (ROMK2) to the inhibitory sulfonylurea compound
91: 2653–64. glybenclamide is enhanced by coexpression with the
54 Malborg AC, Schultz DB, Luton F et al. Penetration and ATP-binding cassette transporter cystic fibrosis trans-
co-localization in MDCK cell mitochondria of IgA membrane regulator. Proc. Natl Acad. Sci. USA 1996; 93:
derived from patients with primary biliary cirrhosis. J. 8083–8.
Autoimmun. 1998; 11: 573–80. 74 Egan M, Flotte T, Afion S. Defective regulation of out-
55 Desmet VJ, Roskams T, Van Eyken P. Ductular reaction in wardly rectifiying Cl– channels by protein kinase A
the liver. Pathol. Res. Pract. 1995; 191: 513–24. corrected by insertion of CFTR. Nature 1992; 358:
56 Roskams T, van Der Oord JJ, DeVos R, Desmet V. 581–4.
Neuroendocrine features of reactive bile ductules in 75 Kopelman HR, Corey M, Gaskin KJ, Durie P, Forstner
cholestatic liver diseases. Am.J.Pathol. 1990; 137: 1019–25. GG. Impaired chloride secretion, as well as bicarbonate
57 Alpini G, Glaser SS, Ueno Y et al. Bile acid feeding secretion, underlies the fluid and secretory defect in the
induces cholangiocytes proliferation and secretion: Evi- cystic fibrosis pancreas. Gastroenterology 1988; 98: 349–55.
dence for bile acid-regulated ductal secretion. Gastroen- 76 Schwiebert EM, Egan E, Hwang TH. CFTR regulates
terology 1999; 116: 179–86. outwardly rectifying chloride channels through an
58 Martinez OM, Villanueva JC, Gershwin ME, Krams SM. autocrine mechanism involving ATP. Cell 1995; 81:
Cytokine patterns and cytotoxic mediators in primary 1063–73.
biliary cirrhosis. Hepatology 1995; 21: 113–19. 77 Barasch J, Kiss B, Prince A, Saiman L, Gruenert D,
59 Harada K, Van de Water J, Leung PSC et al. In situ nucleic Al-Awqati Q. Defective acidification of intracellular
acid hybridization of cytokines in primary biliary cirrho- organelles in cystic fibrosis. Nature 1991; 352: 70–3.
14401746, 2000, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1440-1746.2000.02091.x by UFPE - Universidade Federal de Pernambuco, Wiley Online Library on [11/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Pathophysiology of the biliary epithelium 253

78 Bradbury NA, Bridges RJ. Role of membrane trafficking 83 Medina JF, Martinez A, Vasquez JJ, Prieto J. Decreased
in plasma membrane solute transport. Am. J. Physiol. anion exchanger-2 immunoreactivity in the liver of
1994; 267: C1–24. patients with primary biliary cirrhosis. Hepatology 1997;
79 Clarke LL, Harline MC. Dual role of CFTR in cAMP- 25: 12–17.
stimulated HCO3– secretion across murine duodenum. 84 Balfour Sartor R. Cytokines in intestinal inflammation:
Am. J. Physiol. 1998; 274: G718–26. Pathophysiological and clinical considerations. Gastroen-
80 Bals R, Weiner DJ, Wilson JM. The innate immune system terology 1994; 106: 533–9.
in cystic fibrosis lung disease. J. Clin. Invest. 1999; 103: 85 Schuppan D, Hahn EG. Fibrogenesis in PBC. In: Lindor
303–7. DK, Heathcote EJ, Poupon R. eds. Primary Biliary Cir-
81 Zsembery A, Spirlì C, Sitter G et al. Ca2+-activated Cl– rhosis: From Pathogenesis to Clinical Treatment. Dordrecht:
channels can substitute for CFTR in stimulation of Kluwer Academic Publishers, 1998.
anion-exchanger in human isolated CF cholangiocytes. J. 86 Spirlì C, Nathanson MH, Burgstahler AD et al. Pro-
Hepatol. 1999; 30: 141 (Abstract). inflammatory cytokines (IL-6, IFN-g, TNF-a) inhibit
82 Prieto J, Qian C, Garcia N, Diez J, Medina JF. Abnormal cholangiocyte secretion by downregulation of Cl-/HCO3-
expression of anion exchanger genes in primary biliary cir- exchanger activity and expression. Hepatology 1999; 30:
rhosis. Gastroenterology 1993; 105: 572–8. 417A (Abstract).

You might also like