Você está na página 1de 11

REVIEWS

Pathogenesis of biliary atresia: defining


biology to understand clinical phenotypes
Akihiro Asai, Alexander Miethke and Jorge A. Bezerra
Abstract | Biliary atresia is a severe cholangiopathy of early infancy that destroys extrahepatic bile ducts
and disrupts bile flow. With a poorly defined disease pathogenesis, treatment consists of the surgical
removal of duct remnants followed by hepatoportoenterostomy. Although this approach can improve the
short‑term outcome, the liver disease progresses to end-stage cirrhosis in most children. Further improvement
in outcome will require a greater understanding of the mechanisms of biliary injury and fibrosis. Here, we
review progress in the field, which has been fuelled by collaborative studies in larger patient cohorts and
the development of cell culture and animal model systems to directly test hypotheses. Advances include the
identification of phenotypic subgroups and stages of disease based on clinical, pathological and molecular
features. Stronger evidence exists for viruses, toxins and gene sequence variations in the aetiology of
biliary atresia, triggering a proinflammatory response that injures the duct epithelium and produces a rapidly
progressive cholangiopathy. The immune response also activates the expression of type 2 cytokines that
promote epithelial cell proliferation and extracellular matrix production by nonparenchymal cells. These
advances provide insight into phenotype variability and might be relevant to the design of personalized trials
to block progression of liver disease.
Asai, A. et al. Nat. Rev. Gastroenterol. Hepatol. advance online publication 26 May 2015; doi:10.1038/nrgastro.2015.74

Introduction
Biliary atresia, a multifaceted liver disease of complex with features of cholestasis and variable degrees of giant
pathogenesis, has devastating consequences to child multinucleated hepatocytes.6
health with rapid progression to end-stage cirrhosis if
not treated in a timely fashion. Departing from solely Epidemiology
descriptive studies, the past decade has seen a prolifer­ The disease occurs on all continents, with variable geo-
ation of mechanistic experiments that use in vitro and graphical frequency ranging from 1 in 5,000 in Taiwan to
in vivo model systems to directly test disease-related an estimated 1 in 15,000 in the USA and 1 in 19,000 live
hypotheses. Here, we review how these studies advance births per year in the Netherlands.7–11 A seasonal clus-
our understanding of disease pathogenesis by provid- tering of the disease has been reported, with rates three
ing unique insights into how an insult from one or more times higher in infants born between December and
environmental factors triggers an immune response, March,11,12 but the lack of seasonality in a large study of
which is modified by genetic and developmental factors 119 Japanese infants underscores the variable geographi-
to target the bile duct epithelium and produce the clinical cal frequency of disease.13 Biliary atresia has a slight
spectrum of biliary atresia. female predominance (1.25:1), especially in patients who
Biliary atresia results from an inflammatory and fibro- also have splenic malformations and the incidence is sub-
sing obstruction of extrahepatic bile ducts. Along with stantially higher among nonwhite infants.11,14 An associa-
the general term of ‘neonatal hepatitis’, biliary atresia is a tion with advanced maternal age, increased parity and a
leading cause of neonatal cholestasis;1–4 as a single disease, tendency for early foetal losses has been found.11 Biliary
Division of Pediatric
Gastroenterology,
biliary atresia is the number one indication for paediatric atresia has rare familial recurrence, with twin studies
Hepatology, and liver transplantation worldwide.5 By the time of diagno- showing that most sets are discordant for the disease.15–19
Nutrition, Department sis, extrahepatic bile ducts are completely obstructed.
of Pediatrics, Cincinnati
Children’s Hospital At the tissue level, segmental or wholesale loss of the Disease hallmarks and outcomes
Medical Center, epithelial lining of extrahepatic bile ducts exists, with The clinical hallmarks of the disease are simple and repro-
University of Cincinnati,
3333 Burnet Avenue,
extensive fibrosis and occasional foci of inflammation. ducible: pathological jaundice with direct or conjugated
Cincinnati, OH 45229- By contrast, intrahepatic bile ducts are typically hyper- hyperbilirubinemia; acholic stools; variable levels of
3039, USA (A.A., A.M., plastic, embedded in portal tracts that contain variable hepato­splenomegaly; and the onset of symptoms restricted
J.A.B.).
inflammation and fibrosis, and are surrounded by lobules to the first few months of life. The progression to end-
Correspondence to: stage cirrhosis when the disease is untreated is consist-
J.A.B.
jorge.bezerra@ Competing interests ent. The initial goal of clinical management is to arrive at
cchmc.org The authors declare no competing interests. the diagnosis promptly so that surgical intervention can

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION  |  1


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points the hepatobiliary disease,30 but need proper attention to


prevent complication­s and improve clinical outcome.
■■ Biliary atresia is an inflammatory and fibrosing cholangiopathy of infancy
caused by viruses, environmental toxins and targeted epithelial injury that
obstructs extrahepatic bile ducts and rapidly progresses to end-stage cirrhosis
Embryonic biliary atresia
■■ Precise clinical phenotyping classifies patients into nonsyndromic and About 10% of affected infants have an earlier onset of
embryonic forms, and into cyst-associated and cytomegalovirus-associated jaundice, often present at birth and have nonhepatic
variants, which might have different disease pathologies congenital malformations.14,28–30 This group of patients is
■■ Liver tissue scoring for inflammation and fibrosis by histological features and also referred to as having the congenital, fetal or prenatal
gene expression profiles identifies different stages of disease at diagnosis form of biliary atresia. Consistent with an early onset of
■■ Analyses of human livers and models of experimental biliary atresia suggest bile duct injury, extrahepatic bile ducts might be absent
that a type 1 immune response has a key role in the pathogenesis of bile duct
(that is, no sign of a fibrous cord at the time of explora-
injury and obstruction
■■ The expression of type 2 cytokines promote cholangiocyte proliferation in
tory laparotomy). Splenic abnormalities (as­p lenia,
extrahepatic bile ducts and hepatic fibrosis double spleen and polysplenia) have been reported in
■■ Clinical, histological and molecular variability of disease at presentation form 8–12% of infants with the embryonic form of disease,
a strong rationale for future trials that take into consideration the predominant which occurs in isolation or in combination with one
biological features of affected infants or more additional defects (for example, preduodenal
portal vein, interrupted inferior vena cava, intestinal
mal­rotation) in a variant known as biliary atresia splenic
Table 1 | Clinical phenotypes of biliary atresia* malformation (BASM) syndrome.14,28 Infants with BASM
Phenotype Incidence and disease features Pathogenesis have a higher association with maternal diabetes mellitus
Perinatal form ~80% of patients, isolated malformations, Type 1 cytokine than those with the nonsyndromic form and might have
jaundice-free period after birth response a worse outcome after hepatoportoenterostomy.
Embryonic 7–10% of patients, BASM, laterality defects, Gene mutations
form early onset of jaundice and variants Cystic biliary atresia
Cystic variant 8% of patients, might co-exist with BASM, poor Type 2 cytokine The presence of a cystic malformation near the site
response when HPE performed after 70 days of age response of the common bile duct obstruction, constitutes an
CMV- Variable incidence based on geography and CMV infection anatomic variant often referred to as cystic biliary
associated methodological detection, poor response to HPE atresia, which might be associated with improved bile
*Key features and a proposal for the main mechanisms of pathogenesis. Abbreviations: BASM, biliary drainage after hepatoportoenterostomy.31 Some of the
atresia splenic malformation; CMV, cytomegalovirus; HPE, hepatoportoenterostomy.
infants with biliary cysts are detected prenatally during
routine ultrasonography; jaundice and acholic stools
remove the atretic biliary remnants and create a Roux- might present soon after birth or after an asympto-
en‑Y intestinal conduit for bile drainage, also known as the matic period of 1–3 months of life. A review of a large
Kasai hepatoportoenterostomy.20 Unfortunately, an unac- cohort of infants with biliary atresia reported the pres-
ceptable 40–50% of patients still do not have improved ence of biliary cysts in ~8% of patients.32 Infants with
bile drainage after the hepatoportoenterostomy;14,21–23 even this cystic variant were younger at presentation in this
when they do, the majority of the patients require liver cohort, but a delay in performing a portoenterostomy
t­ransplantation for prolonged survival.5 beyond 70 days of age was associated with poor long-
term survival with the native liver. In addition, some of
Precise phenotyping of disease the infants with the cystic variant of biliary atresia shared
Jaundice and acholic stools are uniformly present in features of BASM. Detection of prenatal cystic changes in
infants with biliary atresia. However, increasing evidence some patients suggests an intra-uterine onset of disease;
supports the existence of specific clinical forms and vari- perhaps also s­uggesting that the mechanisms of intra-
ants based on the presence of nonhepatic malformations, uterine tissue injury differs from the other clinical forms.
time of disease onset and morphological and molecular A promin­ent type 2 T helper (TH2)-cell-mediated inflam-
analysis of the hepatobiliary system (Table 1).24–26 matory response has been associated with cystic biliary
atresia in mice.33 Mice engineered to have a prominent
Perinatal or nonsyndromic biliary atresia TH2 response, in a model of rotavirus-induced biliary
Most infants fall into this clinical form, which is com- atresia, was associated with an increased frequency of
monly referred to as ‘perinatal’ based on the isolation of cystic changes in extrahepatic bile ducts.33
viruses in affected livers and on the detection of serum
bilirubin levels (either conjugated or unconjugated) in Cytomegalovirus-associated biliary atresia
the first 1–2 days of life in infants later diagnosed with Reports of an association between cytomegalovirus detec-
biliary atresia.27 However, in the absence of reproduc- tion and poor biliary drainage after hepatoportoentero­
ible data demonstrating a perinatal insult, this group of stomy could be of particular relevance in designing
patients might be more appropriately referred to as ‘non- clinical care protocols and future trials. One study of
syndromic’. A subgroup of these patients might have single disease aetiology in 85 Chinese patients identified cyto-
or multiple nonhepatic malformations such as cardio­ megalovirus DNA and/or protein in the livers of 51
vascular abnormalities and intestinal malrotation.14,28–30 infants,34 and a second study of a smaller cohort showed a
Coexisting malformations do not necessarily worsen low percentage of patients with normalization of bilirubin

2  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

levels after hepatoportoenterostomy.35 Poor bile drainage fibrosis). None of these histo­logical groups correlated with
and highest risk of death were also linked to the presence age at diagnosis, clinical par­ameters, or 2-year survival
of cytomegalovirus IgM antibodies in a cohort of infants with the native liver. Reasoning that morphological quan-
at King’s College Hospital, London, UK, who had a 10% tification methods might be limited by sampling artefacts
survival with the native liver at 2 years of age.36 In a larger or variability in injury patterns, the investigators generated
cohort of 260 patients, these investigators also found that gene expression signatures of biopsy fragments from the
infants with cytomegalovirus-associated biliary atresia same patients. By differential profiling of gene expression
had the highest aspartate aminotransferase:platelet ratio and prediction analysis models, most of the individuals
index when compared with patients with the cystic, were grouped into inflammation (36%) or fibrosis (55%),
BASM and perinatal forms of biliary atresia.37 However, with only 9% of the specimens displaying mixed molec­ular
geographical variations in cytomegalovirus-associated profiling. Infants with an inflammatory signature were
biliary atresia exist. 38,39 Some of the inconsistencies younger than those with fibrosis; however, age alone could
between studies might result from methodological differ- not predict molecular grouping based on the observation
ences used to study the patient population. For example, that several infants younger than 8 weeks were classified
using an assay that tests lymphocyte activation to cyto- into the fibrosis group. The clinical relevance of the molec-
megalovirus proteins identified a positive response in ular classification was supported by a decreased survival
56% of infants in the USA, even when no other biochemi- without transplantation at 2 years of age in infants with the
cal markers of cytomegalovirus infection were present.40 fibrosis signature.26 If these studies are validated in new
Altogether, efforts to precisely define the status of cyto- prospective cohorts, molecular staging (with or without
megalovirus infection in patient cohorts are justified by histological staging) of liver disease should be considered
this data, as it might trigger unique pathogenic mecha- when designing new clinical trials.
nisms and/or constitute a severity factor influencing the
clinical outcome in biliary atresia. Pathogenic mechanisms of disease
Studies into the epidemiology, pathology and clinical
Staging the disease at diagnosis forms of biliary atresia implicate numerous factors in the
Several clinical reports suggest that an age <60 days at pathogenesis of disease, including: defects in embryo-
the time of hepatoportoenterostomy is associated with genesis; abnormal fetal or prenatal circulation; genetic
improved bile flow and long-term outcome.41 This asso- factors; environmental toxins; viral infection; abnormal
ciation implies that a younger patient will have an earlier inflammatory response; autoimmunity; and susceptibil-
phase of disease and perhaps a lesser degree of liver ity factors.47,48 These seemingly different factors can be
injury. However, age alone is not a uniform predictor grouped into the broadly defined categories of abnormal
of outcome as demonstrated by the poor survival with morphogenesis, environmental factors and inflamma-
the native liver of a cohort of young infants with biliary tory dysregulation, which could interplay to produce a
atresia after hepatoportoenterostomy,42 suggesting that particular phenotype of biliary atresia (Box 1).
other factors influence the clinical course of disease, such
as the coexistence of severe cardiovascular disease, cyto- Defective embryogenesis
megalovirus infection, and the type and extent of liver Developmental defects of the liver and biliary tract might
pathology at diagnosis. Prominent histological findings of be genetically driven or might result from an insult that
inflammation in the liver at diagnosis has been reported disrupts biological circuits critical to organogenesis.
to correlate with a poor response to hepatoportoentero­ Genetic mutations and/or variants relevant to embryo-
stomy.43 If one assumes that prominent inflammation genesis might be linked to biliary atresia pathogenesis,
reflects the severity of tissue injury this association would as suggested by BASM and laterality defects (anomalies
make sense, as would the poor outcome despite surgery. of left-right determination of visceral organs), among
Alternatively, if one assumes that prominent inflamma- others. Disruption of complex molecular circuits during
tion reflects an early phase of tissue involvement or injury vulnerable periods of embryogenesis could also result
this association would be unexpected and an improved from an insult at a precise time in embryogenesis, with
outcome after surgical intervention would be expected. a potential for reprogramming cellular differentiation
Our lack of understanding of the relationship between and an abnormal developmental outcome. Although no
histopathology and clinical response is underscored by specific insult during embryogenesis has yet been linked
other studies reporting a poor outcome in children with directly to biliary atresia, abnormal cell fate in the devel-
advanced hepatic fibrosis,44,45 or the lack of a correlation oping bile duct has been proposed to produce ductal
between fibrosis and clinical outcome.46 plate malformations that persist postnatally in some
Seeking additional insight into the relationship between patients.49 The ductal plate is a bilayered tubular struc-
the degree of liver injury with age at diagnosis and the clin- ture surrounded by thick mesenchyme that is formed
ical course after hepatoportoenterostomy, Moyer et al.26 near the portal vein between 11 and 13 weeks of gesta-
scored liver biopsy samples from infants for inflamma- tion.50 The plate gradually disappears, except for a focal
tion and fibrosis at the time of diagnosis. Only 19% and area where it forms a lumen and gives rise to intrahepatic
11% of the biopsies revealed high scores for inflammation bile ducts;51 its postnatal persistence raises the possibility
and fibrosis, respectively. The r­emaining 70% had mixed that abnormal mesenchymal and cholangiocyte signals
histological features (lower scores for inflammation and halt remodelling of hilar ducts. Although ductal plate

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION  |  3


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

malformation has been described in infants with the Box 1 | Biological categories and pathogenic factors
embryonic form of biliary atresia, a study of the morph­
Abnormal morphogenesis
ology of several portal tracts from each of eight infants
Defective embryogenesis
with the perinatal form of biliary atresia and six with ■■ BASM
BASM identified ductal plate malformation in only 10% ■■ Laterality defects
of the portal tracts, with no predilection to either clinical ■■ Ductal plate malformation
forms of the disease.52 Abnormal prenatal circulation
■■ Vascular abnormalities
Abnormal fetal or prenatal circulation ■■ Arterial hyperplasia or hypertrophy
Genetic factors
Potential defects in prenatal circulation have been impli-
■■ Nonhepatic malformations
cated in disease pathogenesis based on the presence of ■■ SNPs in: CFC1; JAG1; CD14; MIF; ITGB2; ADIPOQ;
anatomical variants of hepatic artery in some patients VEGF; GPC1; and ADD3
with biliary atresia, the findings of arterial hyperplasia or Environmental factors
hypertrophy in liver specimens and on the importance of Toxins
arterial blood flow for integrity of bile ducts.53,54 Although ■■ Biliatresone (from Dysphania plants)
limited data support this mechanism, it is possible that Viruses
arterial changes might be driven by primary growth ■■ Cytomegalovirus
signals, or represent a response to a diseased micro­ ■■ Reovirus
environment that might be rich in growth-promoting ■■ Rotavirus
■■ Human papillomavirus
signals that favour angiogenesis.55,56
■■ Human herpes virus 6
■■ Epstein–Barr virus
Genetic factors
Immune dysregulation
The contribution of genetic defects to the development Abnormal inflammatory response
of biliary atresia is suggested by the association of non- ■■ Activation of dendritic cells
hepatic abnormalities such as polysplenia or asplenia, ■■ T cells
cardio­vascular defects, intestinal malrotation and a spec- ■■ Natural killer cells
trum of laterality defects.57 Experimentally, the inacti­ ■■ Macrophages or Kupffer cells
vation or overexpression of the genes Invs, Hes1, Hnf6 ■■ Expression of IFN‑γ, IL‑2, CD25, TNF, IL‑15, NKG2D,
perforin and granzymes, IL‑8
(also known as Onecut1), Hnf1b, Foxf1, Foxa1 or Foxa2,
Autoimmunity
Sox17, Lgr4 and Pdx1 in mice have been shown to disrupt
■■ HLA-DRA
normal embryogenesis of the extrahepatic biliary system, ■■ Oligoclonal expansion of T cells
causing a spectrum of anatomical malformations of the ■■ Anti-enolase antibodies
gallbladder and extrahepatic bile ducts, including hypo- Susceptibility factors
plasia and agenesis (reviewed elsewhere).58 Gene muta- ■■ Gene SNPs
tions causally linked to biliary atresia in humans, however, ■■ Microchimerism
remain elusive. For example, mutations in INVS were not ■■ TREG cells
■■ α2β1-integrins
identified in children with laterality defects and biliary
Abbreviations: ADD3, adducin 3; BASM, biliary atresia splenic
atresia.59 Promising observations have linked mutations malformation; CFC1, cripto, FRL‑1, cryptic family 1; GPC1,
in CFC1, a different laterality gene,60,61 but they have not glypican 1; HLA-DRA, major histocompatibility complex, class II,
been validated in a large cohort. Studies investigating DR alpha; ITGB2, integrin, beta 2; JAG1, jagged 1; MIF, macrophage
migration inhibitory factor; NKG2D, killer cell lectin-like receptor
gene sequence variants as susceptibility factors for biliary subfamily K, member 1; SNP, single nucleotide polymorphism;
atresia are garnering interest, such as the reports of single TREG cell, regulatory T cell; VEGF, vascular endothelial growth factor.
nucleotide polymorphisms (SNPs) in JAG1, CD14, MIF,
ITGB2, ADIPOQ and VEGF.62–67
A genome-wide association study (GWAS) of 35 the F‑actin binding protein adducin 3.70 This association
patients identified one common heterozygous deletion was replicated in Thai (n = 124) and US (n = 171) patient
of chromosome 2 (2q37.3), which included GPC1 in two cohorts.71,72 Although the biological relevance of this
unrelated patients. GPC1 encodes glypican‑1, a glyco­ SNP is not yet known, the differential hepatic expression
protein located in the apical membrane of cholangio- of ADD3 in affected livers suggest that SNPs in ADD3
cytes mediating intercellular signalling via the Hedgehog might serve as a susceptibility factor or disease modifier.72
pathway. Diseased livers had abnormal localization Further progress in the field will require comprehensive,
of glypican‑1, and morpholino-mediated knockdown of whole-genome mutation surveys in a patient population
gpc1 led to developmental defects in zebrafish bile ducts.68 that is precisely phenotyped and meets stringent criteria
Interestingly, the potential contribution of Hedgehog for adequate statistical analyses, followed by proof-of-
pathway was also implied by results from immunostain- principle experiments to explore the biological relevance
ing and gene expression studies in human liver samples, of SNPs.
which suggested aberrant hepatic Hedgehog signalling in
biliary atresia.69 A different GWAS in a larger cohort of 324 Environmental toxins
Chinese patients identified a high prevalence of the SNP Without clear evidence of disease heritability, there
rs17095355 on 10q24, upstream of ADD3, which encodes remains the possibility that genetic and developmental

4  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

defects might predispose infants to biliary atresia by trig- Experimental models of biliary injury
gering an abnormal response that targets the bile ducts At least four experimental systems have been used to study
upon exposure to detrimental environmental factors. biliary atresia. The first, serving primarily as an observa-
The potential contribution of environmental toxins tional model of toxin-induced bile duct injury, is repre-
to pathogenesis of disease was suggested by the occur- sented by young lambs and calves in New South Wales,
rence of two biliary atresia-like endemics in lambs Australia, which were reported to have a disease similar
and calves in New South Wales, Australia in 1964 and to biliary atresia.73 The animals have not yet been used
1988.73 In a remarkable effort, an investigative team from in experiment-driven manipulation to study the disease.
Philadelphia, USA, imported two species of Dysphania Second, sea lampreys have been shown to undergo loss
plants from pastures on which the affected animals had of the gallbladder and bile ducts during normal meta­
grazed and screened chemical extracts purified from the morphosis and have been used as a model of ‘programmed
plants using a zebrafish bile excretion assay. They puri- atresia’.89,90 Third, zebrafish have been increasingly used to
fied a phytosterol, which they named biliatresone, which study mutation-induced and toxin-induced defects in the
selectively destroyed extrahepatic bile ducts in zebrafish development of intrahepatic and extrahepatic bile ducts.90,91
larvae in a dose-dependent and time-dependen­t manner. Finally, mice in the first postnatal week have been used
Furthermore, culture of 3D cholangiocyte spheroids as experimental models.92–94 Newborn BALB/c mice
with biliatresone induced lumen occlusion and loss infected with Rhesus rotavirus type A (RRV) constitutes
of cellular polarity.74 Interestingly, a zebrafish mutant a model that recapitulates several features of human
known as ductbend displayed heightened sensitivity to disease. In this model, RRV infection in the first 2 days
biliatresone. Genetic mapping experiments localized the of life reproducibly triggers hepatobiliary injury and
ductbend locus to chromosome 22, in a region that has cholestasis within a week of infection.95,96 Some of the key
conserved synteny to two independent susceptibility loci features shared with the human disease include: a time-
for biliary atresia in humans (10q24.2 and 16p13.3), thus restricted susceptibility of bile duct injury to the early
potentially linking an environmental toxin to a genetic postnatal period; acholic stools; bile duct proliferation and
susceptibility loci.75 portal inflammation; and the type 1 rich inflammatory
response in the liver and bile ducts.25,84,97–101 The model
Viral infection provides opportunities for direct examination of extra­
Microbial agents, especially viruses, have been impli- hepatic bile ducts to investigate different stages of injury
cated in the aetiology of biliary atresia. Since initially and progression to lumenal obstruction, tasks that cannot
suggested by Benjamin Landing in 1974,76 several viruses be performed in humans because of the advanced fibrosis
have been detected directly in injured livers and biliary in biliary remnants at diagnosis. Although, the model is a
remnants or indirectly by the presence of serological unique system to study the role of the immune system in
markers of infection in patients with biliary atresia. The epithelial injury and duct obstruction, it is not suitable to
viruses include cytomegalovirus, human papillomavirus, study how biliary injury results in progressive liver fibrosis
human herpes virus 6, Epstein–Barr virus, reovirus and because of the high mortality before the development of
rotavirus in specific groups of patients. The identification advanced fibrosis or cirrhosis at 2 weeks of age.
of individual viruses in affected tissues has been incon-
sistent in different populations (as reviewed elsewhere77). Data from diseased human livers
One of the factors that might limit the identification of In general, the cellular and molecular profiles of the
viruses includes the variability in methodologies used hepatic microenvironment at the time of diagnosis are
to detect infection. For example cytomegalovirus infec- largely proinflammatory and profibrotic. For example,
tion is detected by measuring serum levels of anti- CD4+ and CD8+ T cells populate affected livers and are
cytomegaloviru­s IgM antibodies, lymphocyte activation associated with the overexpression of activation markers,
and molecular footprinting of an immune response to such as IFN‑γ, IL‑2, the IL‑2 receptor CD25, TNF and the
viral infection.38,40,78,79 Reovirus infection is detected by transferrin receptor CD71.102–106 Cholangiocyte py­knosis
measuring serum levels of anti-reovirus type 3 IgG and and necrosis have been associated with infiltration of
IgM antibodies.80–83 Another potential factor limiting the mononuclear cells in the walls of interlobular bile ducts,
detection of viral infection is the ability of the neonatal duct walls at the porta hepatis and remnants of extra­
immune system to clear the virus. In an experimental hepatic bile ducts.107–109 In vitro studies using T cells show
mouse model of biliary atresia, rotavirus was effectively that patients with biliary atresia have oligoclonal expan-
cleared from the hepatobiliary system by the second sion of CD4+ and CD8+ T cells,110 providing more direct
week of infection.84 evidence that tissue lymphocytes share common biologi-
cal programmes. Despite the limited evidence for disease
Abnormal inflammation and autoimmunity specificity, these technically challenging experiments add
Among all proposed factors linked to the pathogenesis functional relevance of T cells in disease pathology and set
of biliary atresia, the immune system is central, as evi- the stage for future studies investigating their relationship
denced by the infiltration of the liver by inflammatory to molecular epitopes on cholangiocytes and mechanisms
cells, the overexpression of cytokines and/or chemokines of autoimmunity.
at the time of diagnosis and mechanistic experiments in A proinflammatory molecular profile was reported in
disease models.25,85–88 a large-scale gene expression analysis of liver biopsies

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION  |  5


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Dendritic cell NK cell neutrophil chemotaxis.119 Notably, deficiency of Cxcr2,


one of the receptors that recognize Cxcl1, Cxcl2 and Cxcl5
Activation (the murine orthologs of IL‑8) decreases epithelial injury
IL-15 and inflammation of liver and extrahepatic bile ducts.25
Studies of dendritic cells after RRV infection found
plasma­cytoid dendritic cells (pDCs) as the most abundant
Cholangiocyte dendritic cell population in early phases of infection; they
RRV
are targeted by the virus and produce IL‑15.120 The impor-
tance of pDCs is supported by evidence of decreased
Apoptosis epithelial injury and reduced incidence of bile duct
obstruction in mice that are depleted of pDCs after RRV
infection. A similarly milder phenotype was observed by
antibody-mediated inhibition of IL‑15, a potent activa-
tor of natural killer cells,121,122 suggesting that the expres-
Macrophage Neutrophil
sion of pDC-derived cytokines represents an important
­triggering event that targets bile ducts (Figure 1).120
Chemotaxis An even more dramatic effect on the prevention of
Cxcl2 epithelial injury and biliary obstruction was produced
by depletion of natural killer cells.101 In these experi-
Figure 1 | Cellular targets and molecular events after RRV infection. When ments, investigators first found that incubation of hepatic
administered into BALB/c mice in the firstReviews
Nature 2 postnatal days, RRV infects
| Gastroenterology & Hepatology natural killer cells isolated from RRV-infected mice lyzed
cholangiocytes, inducing apoptosis and necrosis. RRV also targets hepatic cholangiocytes in a contact-dependent fashion, with no
DCs, which activate NK cells via Il‑15 and hepatic macrophages, which secrete lysis when the activating Nkg2d receptor (also known
Cxcl2 that attracts neutrophils. Abbreviations: Cxcl2, C-X-C motif chemokine 2; as Klrk1) was blocked on natural killer cells. Consistent
NK, natural killer cell; RRV, Rhesus rotavirus type A. with these in vitro studies, the depletion of natural killer
cells or antibody blockade of Nkg2d immediately after
from infants with biliary atresia.111 The approach identi- birth prevented the development of jaundice in newborn
fied a genetic footprint in which genes involved in the mice infected with RRV.101 Detailed histological analy-
type 1 T helper (TH1) cell response were activated at early sis of extrahepatic bile ducts revealed intact epithelium;
stages of biliary atresia, with simultaneous but transient without duct injury or obstruction, newborn mice grew
suppression of markers of humoral immunity.111 Other into adulthood. Interestingly, this lack of duct injury or
studies reported increased number and activation of obstruction occurred despite the persistence of the virus
Kupffer cells and the expression of the MHC class II in the liver, which suggested that RRV alone is not suffi-
antigen HLA-DRA in the livers of infants with biliary cient to produce biliary obstruction, but requires natural
atresia.104,112,113 The aberrant expression of HLA-DRA by killer cells to breach the continuity of the duct epithelium
cholangiocytes led to the proposal that they might have and initiate a cascade of events that produce the atresia
antigen-presenting properties, but testing this hypothesis phenotype (Figure 2).
in the laboratory found no evidence that virus-infected
cholangiocytes induced lymphocyte proliferation.114 This Mechanisms of duct obstruction
type of experiment lends strong support for the need to The first studies that directly explored disease patho­
complement observations from studies in human tissues genesis at the mechanistic level, focused on the cellular
with mechanistic studies using cell and animal models. and molecular constituents of bile duct obstruction.123 The
studies tested the hypothesis that IFN‑γ is a key cytokine
Mechanisms of epithelial injury in the development of biliary atresia. Using newborn mice
Studies of the cellular localization of RRV and the tem- carry­ing an inactivating mutation in the Ifng gene, infec-
poral dynamics of inflammatory cells in BALB/c mice tion with RRV caused transient cholestasis, but extra­
have provided insight into early mechanisms of epithe- hepatic bile ducts were free of obstruction and newborn
lial injury. Following infection, RRV can be detected mice had improved survival.84 These findings seem to
in cholangiocytes of intrahepatic and extrahepatic bile be unique for IFN‑γ based on other studies that showed
ducts, macrophages and dendritic cells.84,114–117 Infection no improvement in the experimental phenotype if mice
of cholangiocytes is cytopathic and largely dependent on lacked IL‑12 or underwent antibody depletion of TNF.124,125
the expression of α2β1-integrin.116 In culture, cholangio- One of the main sources of IFN‑γ expression is hepatic
cytes infected by RRV express variable levels of cytokines CD4+ T cells, yet the depletion of these cells was not found
and chemokines,114,118 but at levels that are below the to affect the inflammatory obstruction of extrahepatic bile
threshold to induce chemotaxis.119 ducts after RRV infection.100 Instead, the loss of CD8+
Signals that trigger epithelial injury seem to derive from T cells prevented duct obstruction after rotavirus infec-
two other cell types that are targeted by RRV, macrophages tion though enabling the development of cholangitis.100
and dendritic cells. RRV has been detected in the mouse The expression of perforin and granzymes by cytotoxic
hepatic macrophage cell line RAW264.7 resulting in the T cells and natural killer cells seem to be key effector
secretion of Mip2 and/or Cxcl2 and the induction of molecules.126 The similarities between the phenotypes

6  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

IL-33
Fibrosis Additional experiments revealed that IL‑33 promotes
IL-13 cholangiocyte proliferation by inducing type 2 innate
TGF-β
Liver

Osteopontin Cirrhosis lymphoid cells (ILC2s) to release IL‑13, thus identifying


Bile acids? a new paracrine system in which an IL‑33–ILC2s–IL‑13
Hepatic
stellate cell axis works in concert to regulate proliferation in neigh-
ILC2
bouring bile duct epithelial cells.128 Interestingly, in the
liver, IL‑33 activates hepatic stellate cells and promotes
Bile duct

RRV IFN-γ IL-33 hepatic fibrosis, raising the possibility that this para­
crine system could be a contributor to the progression
Extrahepatic
and intrahepatic
of f­ibrosis characteristic of biliary atresia (Figure 3).129
Bile duct
Inflammatory obstruction fibrosis
Epithelium Atresia
response Susceptibility factors
breach
IL-8
EHBD

IL-15 The biological basis for the time-restricted onset of


Nkg2d biliary atresia is not fully understood. One possibility is
DC a genetic predisposition caused by SNPs in CFC1 and
NK cell CD8+
T cell other genes (see earlier); however, the lack of congenital
Macrophage malformations in the majority of patients suggests that
Figure 2 | Biological processes andNature
effectors of injury
Reviews in biliary atresia.&The
| Gastroenterology release
Hepatology other biological processes might be susceptibility factors.
of IL-15 by DCs after RRV infection or toxin activates NK cells and CD8+ T cells. Among such processes is an inflammatory response trig-
Targeting of cholangiocytes by infection or toxin activates inflammatory cells and gered by the presence of rare maternal cells in the liver
the release of IL-15, perforin and granzymes, which injures the epithelium. IFN-γ of affected infants (maternal chimerism).130,131 Evidence
expression is linked to an amplification of epithelial injury and duct obstruction by for this reaction to maternal cells is largely circumstantial
prominent infiltration of lymphocytes and myeloid cells. The release of IL-33 and at this time. Another susceptibility factor is an imbal-
IL-13 by ILC2 promotes epithelial repair in bile ducts, and is linked to fibrosis in
anced response by the immune system to an exogenous
the liver (and perhaps of extrahepatic ducts). Abbreviations: DC, dendritic cell;
EHBD, extrahepatic bile duct; ILC2, innate lymphocyte cell type 2; M, macrophage;
insult at a vulnerable time of postnatal development.
N, neutrophil; NK, natural killer cell; RRV, Rhesus rotavirus type A. The mouse model of biliary atresia has a unique sus-
ceptibility to disease in early postnatal life. Specific tar-
geting of cholangiocytes by RRV has been linked to the
produced by the loss of CD8+ T cells or Ifnγ expression expression of α2β1-integrin, which influenced viral attach-
suggest that both factors work in parallel to promote duct ment and biliary injury.116 Another important suscepti-
obstruction (Figure 2).84,100 Although these data pointed bility factor are regulatory T (TREG) cells, which have an
to key roles of the TH1 cell response in pathogenesis of important immunomodulatory function; their absence
experimental biliary atresia, the administration of RRV leads to an array of autoimmune phenotypes. In the
into newborn mice engineered to lack a TH1 cell response RRV mouse model, the number of TREG cells in the first
by the inactivation of Stat1 gene unexpectedly resulted in 3 days of life is very low, even after RRV challenge,132,133
the full obstructive phenotype, in addition to focal cystic and their abundance correlates inversely with the degree
dilatations of extrahepatic bile ducts.33 In Stat1-knockout of liver and biliary injury induced by RRV infection.133,134
mice, RRV induced a prominent TH2 lymphoid and Mechanistically, TREG cells regulate the activation of hepatic
type 2 myeloid responses, with IL‑13 responsible for the T cells by CD86-dependent co-stimulation with dendritic
o­bstructive phenotype. cells.134 How these TREG cells modulate s­usceptibility of
biliary injury in humans remains poorly defined.
Type 2 immunity and epithelial integrity
The unexpected obstruction of bile ducts in mice over- A working model of disease
expressing type 2 cytokines suggested that the presence Previous models of pathogenesis broadly linked disease
of an epithelial injury and an inflammatory response are phenotype to the interplay between environmental
sufficient to occlude the duct lumen and disrupt bile flow, factors and developmentally regulated tissue responses
even when the epithelial injury is functionally diverse that target the biliary system and disrupt bile flow.
such as in type 1 and 2 immune responses. Notably, these Contemporary studies that more precisely characterize
responses have also been documented in serum (at the the clinical features, quantify the cellular and molecular
protein level) and liver samples (as gene expression) at composition of affected tissues and explore mechanisms
the time of diagnosis.33 The cohort of patients could have of bile duct injury enable a revised proposal that includes
been divided into subgroups that share similar molec- a more-refined model of disease.
ular profiles,33 in a fashion akin to the staging of liver An inclusive model of disease begins with the expo-
disease by gene expression signatures.26 The molecular sure to environmental toxins or viruses (Figure 2).
profiles might reflect a transition from a proinflamma- Experimentally, biliatresone targets cholangiocytes,
tory type 1 immune response to a fibrosis-related type 2 while rotavirus infects cholangiocytes, macrophages
immune response.127 IL‑33, one of the type 2 cytokines and dendritic cells. Disease susceptibility probably
with alarmin properties, exerts a potent proliferative involves sequence variants in candidate genes (for
response in cholangiocytes of extrahepatic bile ducts example, ADD3), functional immaturity of TREG cells
and promotes epithelial integrity after RRV infection.128 and other developmental factors. Gene variants might

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION  |  7


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Cholangiocyte contribute to early epithelial injury or to the ongoing


in­trahepatic biliary disease after portoenterostomy.135
EHBD Mitosis Less well known are the factors responsible for progres-
sive biliary fibrosis, but emerging data point to the exist-
IL-33 IL-13
ence of a more fibrogenic type 2 immune response driven
ILC2
by ILC2s, hepatic stellate cells and the expression of IL‑33,
IL‑13, TGF‑β1, osteopontin and related extra­cellular
Activation matrix molecules.136–138 Other factors that could con-
Liver ? tribute to this phenotype include the toxicity of retained
Quiescent Activated
hepatic hepatic bile acids and other components of the inflammatory
stellate cell stellate cell response, such as IL‑17 and related cytokines; however,
Figure 3 | Epithelial and fibrogenic response induced by IL‑33. The presence these factors remained largely unexplored (Figure 2).
Nature Reviews | Gastroenterology & Hepatology
of IL‑33 activates type 2 innate lymphoid cells (ILC2) to release IL‑13, which
induces cholangiocyte proliferation in the neighbouring epithelium of extrahepatic Conclusions
bile ducts. In the liver, the IL‑33–ILC2–IL‑13 circuit activates the profibrogenic Research on biliary atresia has entered a new phase
phenotype of hepatic stellate cells. Abbreviation: EHBD, extrahepatic bile duct.
of discoveries, fuelled largely by careful phenotyping of
patients and by mechanistic experiments using model
Immune suppression systems. Moving forward, investigation will be facilitated
by the access to large cohorts via multicentre studies,
such as the European consortium of Biliary Atresia
and Related Diseases139 or the Childhood Liver Disease
Inflammation Research Network (ChiLDReN).140
The systematic use of complementary approaches to
detect biomarkers of infection will aid the search for a
viral aetiology of biliary atresia. Testing for cytomegalo­
virus in multicentre cohorts might be particularly
rele­vant based on its preliminary relationship to poor
Fibrosis outcome after hepatoportoenterostomy. As for environ-
mental toxins, the field awaits greater knowledge on the
cytotoxic or cytopathic effects of biliatresone, its effects
on the immune system and whether it is detected in
tissues from affected patients.
Antifibrotics
Using larger cohorts in GWAS or exome sequencing
Figure 4 | Designing clinical
Nature Reviews trials based on biological
| Gastroenterology & Hepatology experiments will increase the power of gene association
stages of liver disease. Histopathology and molecular studies in biliary atresia. On the basis of the low inci-
methods to stage the liver disease might identify dence of the disease, the inclusion of trios (the patients
predominantly inflammation (red), fibrosis (blue) and mixed
and parents) in sequencing experiments might narrow
(grey) subgroups of patients at diagnosis. Matching of the
disease stages with specific therapies in future trials could the number of sequence variants and aid discoveries on
yield improved efficacy and prevent unnecessary exposure genetic susceptibility.
of patients to drug toxicity. The combination of human and mouse studies has
identified several inflammatory pathways that can serve
as therapeutic targets. However, blocking individual
be particularly important for pathogenesis in children pathways requires preclinical testing in a model that
with BASM and/or laterality defects. more closely recapitulates the progressive liver disease
Injured or infected cholangiocytes and cells of the innate after hepatoportoenterostomy. One area of particular
immune system produce chemoattractants to myeloid interest might be the IL‑33–ILC2–IL‑13 circuit based
cells (for example, Cxcl2 in mice or IL‑8 in humans) and on its role in promoting experimental fibrosis. Future
engage natural killer cells to amplify cholangiocyte injury studies will investigate whether the disruption of this
and break the continuity of the biliary epithelium. Key circuit suppresses the inflammation-induced fibrosis
effector molecules in early phases of epithelial injury in and lowers the rate of progression to cirrhosis.
mice include Il‑15, Nkg2d, Rae1, and Cxcl2 (or IL‑8 Prospective validation studies, perhaps using ‘disease
in humans). What follows is a prominent response of the modelling’ strategies, are important to better define the
adaptive immune system, with the activation of CD8+ clinical relevance of different phenotypic forms, treat-
T cells and expression of IFN‑γ, perforin and granzymes, ment responses and progression of liver disease. The
which targets the epithelium; together with other inflam- same is true for staging of liver disease at diagnosis and
matory cells, a luminal plug is formed that stops bile flow. the identification of cytomegalovirus-positive patient
Each one of these effectors of injury in experimental cohorts. These studies will enable patient-based investi­
atresia has been validated in patients’ livers at the time gations that more consistently implicate pathogenic
of diagnosis. The production of autoantibodies by B cells mechanisms with clinical phenotypes and speed up the
targeting enolase and other cholangiocyte epitopes might discovery of disease biomarkers.

8  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

If the different clinical phenotypes and stages of liver positive for anti-cytomegalovirus IgM antibodies)
disease are validated, they will form a strong rationale showed improved short-term bile drainage.143 This type
for personalized clinical trials. Despite the predomi- of open-label trial and the emerging evidence discussed
nant inflammatory signature uncovered in clinical and here, point to the possibility of future trials that match
preclinical studies, the use of corticosteroids in patients therapy with a predominant biological disease process,
with biliary atresia has not been shown to reliably improve thus enabling the inclusion of patients that will benefit
biliary drainage after hepatoportoentero­stomy.141 In the from targeted therapies and minimizing unwarranted
prospective, placebo-controlled, double blind START exposure to drug-induced toxicity (Figure 4).
trial (steroid in biliary atresia randomized trial, con-
ducted by ChiLDReN),142 high-dose cortico­steroids after
hepatoportoenterostomy did not significantly increase Review criteria
the proportion of patients achieving biliary drainage This Review presents a perspective of how recent
when compared to placebo (58.6% versus 48.6%, respec- patient-based and laboratory-based studies advance the
tively; P = 0.43), or transplant-free survival (58.7% versus understanding of pathogenic mechanisms of biliary atresia.
59.4%, respectively; P = 0.99).142 Interestingly, an open- To select the studies, we performed a MEDLINE search
label corticosteroid trial that included patients younger combining the medical subject headings of “biliary atresia”
than 70 days at the time of hepatoportoenterostomy and “children,” published in the English literature as
original articles up to 30 September 2014.
and excluding specific phenotypes (BASM and patients

1. Dehghani, S. M., Efazati, N., Shahramian, I., 15. Fallon, S. C., Chang, S., Finegold, M. J., 29. Davenport, M. et al. The biliary atresia splenic
Haghighat, M. & Imanieh, M. H. Evaluation of Karpen, S. J. & Brandt, M. L. Discordant malformation syndrome: a 28-year single-center
cholestasis in Iranian infants less than three presentation of biliary atresia in premature retrospective study. J. Pediatr. 149, 393–400
months of age. Gastroenterol. Hepatol. Bed monozygotic twins. J. Pediatr. Gastroenterol. Nutr. (2006).
Bench 8, 42–48 (2015). 57, e22–e23 (2013). 30. Schwarz, K. B. et al. Extrahepatic anomalies
2. Hoerning, A. et al. Diversity of disorders 16. Hyams, J. S., Glaser, J. H., Leichtner, A. M. in infants with biliary atresia: results of a large
causing neonatal cholestasis—the experience & Morecki, R. Discordance for biliary atresia in prospective North American multicenter study.
of a tertiary pediatric center in Germany. two sets of monozygotic twins. J. Pediatr. 107, Hepatology 58, 1724–1731 (2013).
Front. Pediatr. 2, 65 (2014). 420–422 (1985). 31. Muise, A. M. et al. Biliary atresia with
3. Lee, W. S., Chai, P. F., Boey, C. M. & Looi, L. M. 17. Lachaux, A. et al. Familial extrahepatic biliary choledochal cyst: implications for classification.
Aetiology and outcome of neonatal cholestasis atresia. J. Pediatr. Gastroenterol. Nutr. 7, 280–283 Clin. Gastroenterol. Hepatol. 4, 1411–1414
in Malaysia. Singapore Med. J. 51, 434–439 (1988). (2006).
(2010). 18. Smith, B. M., Laberge, J. M., Schreiber, R., 32. Davenport, M., Caponcelli, E., Livesey, E.,
4. Stormon, M. O., Dorney, S. F., Kamath, K. R., Weber, A. M. & Blanchard, H. Familial biliary Hadzic, N. & Howard, E. Surgical outcome in
O’Loughlin, E. V. & Gaskin, K. J. The changing atresia in three siblings including twins. biliary atresia: etiology affects the influence of
pattern of diagnosis of infantile cholestasis. J. Pediatr. Surg. 26, 1331–1333 (1991). age at surgery. Ann. Surg. 247, 694–698 (2008).
J. Paediatr. Child Health 37, 47–50 (2001). 19. Strickland, A. D., Shannon, K. & Coln, C. D. 33. Li, J. et al. Th2 signals induce epithelial injury
5. Schreiber, R. A. & Kleinman, R. E. Biliary atresia. Biliary atresia in two sets of twins. J. Pediatr. in mice and are compatible with the biliary
J. Pediatr. Gastroenterol. Nutr. 35 (Suppl. 1), 107, 418–420 (1985). atresia phenotype. J. Clin. Invest. 121, 4245–4256
S11–S16 (2002). 20. Kasai, M. & Suzuki, S. A new operation for (2011).
6. Russo, P. et al. Design and validation of the “non‑correctable” biliary atresia, hepatic 34. Xu, Y. et al. The perinatal infection of
biliary atresia research consortium histologic portoenterostomy. Shujutsu 13, 733–739 cytomegalovirus is an important etiology for
assessment system for cholestasis in infancy. (1959). biliary atresia in China. Clin. Pediatr. (Phila) 51,
Clin. Gastroenterol. Hepatol. 9, 357–362 (2011). 21. Chardot, C. et al. Improving outcomes of biliary 109–113 (2012).
7. Chardot, C. et al. Epidemiology of biliary atresia: French national series 1986–2009. 35. Shen, C., Zheng, S., Wang, W. & Xiao, X. M.
atresia in France: a national study 1986–1996. J. Hepatol. 58, 1209–1217 (2013). Relationship between prognosis of biliary atresia
J. Hepatol. 31, 1006–1013 (1999). 22. Schreiber, R. A. et al. Biliary atresia: the and infection of cytomegalovirus. World J. Pediatr.
8. Houwen, R. H. et al. Time-space distribution of Canadian experience. J. Pediatr. 151, 659–665 4, 123–126 (2008).
extrahepatic biliary atresia in the Netherlands (2007). 36. Davenport, M. & Grieve, A. Maximizing Kasai
and West Germany. Surg. Inf. Child 43, 68–71 23. Superina, R. et al. The anatomic pattern portoenterostomy in the treatment of biliary
(1988). of biliary atresia identified at time of Kasai atresia: medical and surgical options.
9. Hsiao, C. H. et al. Universal screening for biliary hepatoportoenterostomy and early South African Med. J. 102, 865–867 (2012).
atresia using an infant stool color card in Taiwan. postoperative clearance of jaundice are 37. Davenport, M. Biliary atresia: clinical aspects.
Hepatology 47, 1233–1240 (2008). significant predictors of transplant-free Semin. Pediatr. Surg. 21, 175–184 (2012).
10. McKiernan, P. J., Baker, A. J. & Kelly, D. A. survival. Ann. Surg. 254, 577–585 (2011). 38. Fischler, B., Svensson, J. F. & Nemeth, A. Early
The frequency and outcome of biliary atresia in 24. Ohi, R. Biliary atresia: a surgical perspective. cytomegalovirus infection and the long-term
the UK and Ireland. Lancet 355, 25–29 (2000). Clin. Liver Dis. 4, 779–804 (2000). outcome of biliary atresia. Acta Paediatr. 98,
11. Yoon, P. W., Bresee, J. S., Olney, R. S., 25. Bessho, K. & Bezerra, J. A. Biliary atresia: 1600–1602 (2009).
James, L. M. & Khoury, M. J. Epidemiology will blocking inflammation tame the disease? 39. Schukfeh, N., Al-Gamrah, A., Petersen, C. &
of biliary atresia: a population-based study. Annu. Rev. Med. 62, 171–185 (2011). Kuebler, J. F. Detection of hepatotropic viruses
Pediatrics 99, 376–382 (1997). 26. Moyer, K. et al. Staging of biliary atresia at has no impact on the prognosis after Kasai
12. Strickland, A. D. & Shannon, K. Studies in diagnosis by molecular profiling of the liver. procedure. J. Pediatr. Surg. 47, 1828–1832
the etiology of extrahepatic biliary atresia: Genome Med. 2, 33 (2010). (2012).
time‑space clustering. J. Pediatr. 100, 749–753 27. Harpavat, S., Finegold, M. J. & Karpen, S. J. 40. Brindley, S. M. et al. Cytomegalovirus-specific
(1982). Patients with biliary atresia have elevated T-cell reactivity in biliary atresia at the time
13. Wada, H. et al. Insignificant seasonal and direct/conjugated bilirubin levels shortly after of diagnosis is associated with deficits in
geographical variation in incidence of biliary birth. Pediatrics 128, e1428–e1433 (2011). regulatory T cells. Hepatology 55, 1130–1138
atresia in Japan: a regional survey of over 20 28. Davenport, M., Savage, M., Mowat, A. P. (2012).
years. J. Pediatr. Surg. 42, 2090–2092 (2007). & Howard, E. R. Biliary atresia splenic 41. Sokol, R. J. et al. Screening and outcomes in
14. Shneider, B. L. et al. A multicenter study of the malformation syndrome: an etiologic and biliary atresia: summary of a national institutes
outcome of biliary atresia in the United States, prognostic subgroup. Surgery 113, 662–668 of health workshop. Hepatology 46, 566–581
1997 to 2000. J. Pediatr. 148, 467–474 (2006). (1993). (2007).

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION  |  9


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

42. Volpert, D. et al. Outcome of early hepatic right axis development and biliary atresia. 78. Jevon, G. P. & Dimmick, J. E. Biliary atresia
portoenterostomy for biliary atresia. J. Pediatr. Human Genetics 110, 157–165 (2002). and cytomegalovirus infection: a DNA study.
Gastroenterol. Nutr. 32, 265–269 (2001). 60. Davit-Spraul, A., Baussan, C., Hermeziu, B., Pediatr. Dev. Pathol. 2, 11–14 (1999).
43. Azarow, K. S., Phillips, M. J., Sandler, A. D., Bernard, O. & Jacquemin, E. CFC1 gene 79. Al-Masri, A. N. et al. Expression of the interferon-
Hagerstrand, I. & Superina, R. A. Biliary atresia: involvement in biliary atresia with polysplenia induced Mx proteins in biliary atresia. J. Pediatr.
should all patients undergo a portoenterostomy? syndrome. J. Pediatr. Gastroenterol. Nutr. 46, Surg. 41, 1139–1143 (2006).
J. Pediatr. Surg. 32, 168–172 (1997). 111–112 (2008). 80. Glaser, J. H., Balistreri, W. F. & Morecki, R.
44. Pape, L., Olsson, K., Petersen, C., von Wasilewski, R. 61. Jacquemin, E., Cresteil, D., Raynaud, N. & Role of reovirus type 3 in persistent infantile
& Melter, M. Prognostic value of computerized Hadchouel, M. CFC1 gene mutation and biliary cholestasis. J. Pediatr. 105, 912–915 (1984).
quantification of liver fibrosis in children with atresia with polysplenia syndrome. J. Pediatr. 81. Morecki, R., Glaser, J. H., Johnson, A. B.
biliary atresia. Liver Transpl. 15, 876–882 Gastroenterol. Hepatol. Nutr. 34, 326–327 & Kress, Y. Detection of reovirus type 3 in the
(2009). (2002). porta hepatis of an infant with extrahepatic biliary
45. Weerasooriya, V. S., White, F. V. & Shepherd, R. W. 62. Arikan, C. et al. Polymorphisms of the ICAM-1 gene atresia: ultrastructural and immunocytochemical
Hepatic fibrosis and survival in biliary atresia. are associated with biliary atresia. Dig. Dis. Sci. study. Hepatology 4, 1137–1142 (1984).
J. Pediatr. 144, 123–125 (2004). 53, 2000–2004 (2008). 82. Richardson, S. C., Bishop, R. F. & Smith, A. L.
46. Santos, J. L. et al. The extent of biliary proliferation 63. Shih, H. H. et al. Promoter polymorphism of the Reovirus serotype 3 infection in infants with
in liver biopsies from patients with biliary atresia CD14 endotoxin receptor gene is associated extrahepatic biliary atresia or neonatal hepatitis.
at portoenterostomy is associated with the with biliary atresia and idiopathic neonatal J. Gastroenterol. Hepatol. 9, 264–268 (1994).
postoperative prognosis. J. Pediatr. Surg. 44, cholestasis. Pediatrics 116, 437–441 (2005). 83. Tyler, K. L. et al. Detection of reovirus RNA in
695–701 (2009). 64. Kohsaka, T. et al. The significance of human hepatobiliary tissues from patients with
47. Balistreri, W. F. et al. Biliary atresia: current jagged 1 mutations detected in severe cases extrahepatic biliary atresia and choledochal
concepts and research directions. Summary of a of extrahepatic biliary atresia. Hepatology 36, cysts. Hepatology 27, 1475–1482 (1998).
symposium. Hepatology 23, 1682–1692 (1996). 904–912 (2002). 84. Shivakumar, P. et al. Obstruction of extrahepatic
48. Bezerra, J. A. The next challenge in pediatric 65. Lee, H. C. et al. Genetic variation in the vascular bile ducts by lymphocytes is regulated by IFN-γ
cholestasis: deciphering the pathogenesis of endothelial growth factor gene is associated in experimental biliary atresia. J. Clin. Invest. 114,
biliary atresia. J. Pediatr. Gastroenterol. Nutr. 43 with biliary atresia. J. Clin. Gastroenterol. 44, 322–329 (2004).
(Suppl. 1), S23–29 (2006). 135–139 (2010). 85. Feldman, A. G. & Mack, C. L. Biliary atresia:
49. Desmet, V. J. Congenital diseases of 66. Udomsinprasert, W. et al. +276 G/T single cellular dynamics and immune dysregulation.
intrahepatic bile ducts: variations on the theme nucleotide polymorphism of the adiponectin Semin. Pediatr. Surg. 21, 192–200 (2012).
“ductal plate malformation”. Hepatology 16, gene is associated with the susceptibility to 86. Hartley, J. L., Davenport, M. & Kelly, D. A.
1069–1083 (1992). biliary atresia. World J. Pediatr. 8, 328–334 Biliary atresia. Lancet 374, 1704–1713 (2009).
50. Tan, C. E., Davenport, M., Driver, M. (2012). 87. Mack, C. L., Feldman, A. G. & Sokol, R. J.
& Howard, E. R. Does the morphology of the 67. Zhao, R. et al. Polymorphism of ITGB2 gene Clues to the etiology of bile duct injury in biliary
extrahepatic biliary remnants in biliary atresia 3’-UTR+145C/A is associated with biliary atresia. Semin. Liver. Dis. 32, 307–316 (2012).
influence survival? A review of 205 cases. atresia. Digestion 88, 65–71 (2013). 88. Petersen, C. & Davenport, M. Aetiology of biliary
J. Pediatr. Surg. 29, 1459–1464 (1994). 68. Cui, S. et al. Evidence from human and zebrafish atresia: what is actually known? Orphanet. J.
51. Takashima, Y., Terada, M., Kawabata, M. that GPC1 is a biliary atresia susceptibility gene. Rare. Dis. 8, 128 (2013).
& Suzuki, A. Dynamic three-dimensional Gastroenterology 144, 1107–1115 (2013). 89. Cai, S. Y. et al. Adult sea lamprey tolerates biliary
morphogenesis of intrahepatic bile ducts 69. Omenetti, A. et al. Hedgehog activity, epithelial- atresia by altering bile salt composition and
in mouse liver development. Hepatology 61, mesenchymal transitions, and biliary renal excretion. Hepatology 57, 2418–2426
1003–1011 (2014). dysmorphogenesis in biliary atresia. Hepatology (2013).
52. Pacheco, M. C., Campbell, K. M. & Bove, K. E. 53, 1246–1258 (2011). 90. Yeoh, E. J. et al. Classification, subtype
Ductal plate malformation-like arrays in 70. Garcia-Barcelo, M. M. et al. Genome-wide discovery, and prediction of outcome in
early explants after a Kasai procedure are association study identifies a susceptibility pediatric acute lymphoblastic leukemia by gene
independent of splenic malformation complex locus for biliary atresia on 10q24.2. Hum. Mol. expression profiling. [comment]. Cancer Cell 1,
(heterotaxy). Pediatr. Dev. Pathol. 12, 355–360 Genet. 19, 2917–2925 (2010). 133–143 (2002).
(2009). 71. Kaewkiattiyot, S., Honsawek, S., Vejchapipat, P., 91. Cui, H. et al. Loss of IGF2 imprinting: a potential
53. dos Santos, J. L., da Silveira, T. R., da Silva, V. D., Chongsrisawat, V. & Poovorawan, Y. marker of colorectal cancer risk. Science 299,
Cerski, C. T. & Wagner, M. B. Medial thickening Association of X-prolyl aminopeptidase 1 1753–1755 (2003).
of hepatic artery branches in biliary atresia. rs17095355 polymorphism with biliary atresia 92. Bangaru, B., Morecki, R., Glaser, J. H.,
A morphometric study. J. Pediatr. Surg. 40, 637–642 in Thai children. Hepatol. Res. 41, 1249–1252 Gartner, L. M. & Horwitz, M. S. Comparative
(2005). (2011). studies of biliary atresia in the human newborn
54. Ho, C. W. et al. The pathogenesis of biliary 72. Tsai, E. A. et al. Replication of a GWAS signal and reovirus-induced cholangitis in weanling
atresia: a morphological study of the in a Caucasian population implicates ADD3 in mice. Lab. Invest. 43, 456–462 (1980).
hepatobiliary system and the hepatic artery. susceptibility to biliary atresia. Hum. Genet. 133, 93. Szavay, P. O., Leonhardt, J., Czech-Schmidt, G.
J. Pediatr. Gastroenterol. Nutr. 16, 53–60 (1993). 235–243 (2014). & Petersen, C. The role of reovirus type 3
55. de Souza, A. F. et al. Angiopoietin 1 and 73. Harper, P., Plant, J. W. & Unger, D. B. Congenital infection in an established murine model for
angiopoietin 2 are associated with medial biliary atresia and jaundice in lambs and calves. biliary atresia. Eur. J. Pediatr. Surg. 12, 248–250
thickening of hepatic arterial branches in biliary Aust. Vet. J. 67, 18–22 (1990). (2002).
atresia. Pediatr. Res. 75, 22–28 (2014). 74. Waisbourd-Zinman, O. et al. A novel toxin 94. Wilson, G. A., Morrison, L. A. & Fields, B. N.
56. Edom, P. T., Meurer, L., da Silveira, T. R., responsible for outbreaks of biliary atresia in Association of the reovirus S1 gene with
Matte, U. & dos Santos, J. L. Immunolocalization livestock causes lumen obstruction in a serotype 3-induced biliary atresia in mice.
of VEGF A and its receptors, VEGFR1 and cholangiocyte spheroid model [abstract 104]. J. Virol. 68, 6458–6465 (1994).
VEGFR2, in the liver from patients with biliary Hepatology 60 (Suppl.1), 249A (2014). 95. Petersen, C. et al. New aspects in a murine
atresia. Appl. Immunohist. Mol. Morphol. 19, 75. Zhao, X. et al. The zebrafish duct bend mutation model for extrahepatic biliary atresia. J. Pediatr.
360–368 (2011). is a sensitizer to toxin-induced biliary atresia Surg. 32, 1190–1195 (1997).
57. Carmi, R., Magee, C. A., Neill, C. A. & Karrer, F. M. and a potential homologue of a human biliary 96. Riepenhoff-Talty, M. et al. Group A rotaviruses
Extrahepatic biliary atresia and associated atresia modifier [abstract 156]. Hepatology 60 produce extrahepatic biliary obstruction in
anomalies: etiologic heterogeneity suggested by (Suppl. 1), 274A (2014). orally inoculated newborn mice. Pediatr. Res.
distinctive patterns of associations. Am. J. Med. 76. Landing, B. H. Considerations of the 33, 394–399 (1993).
Genet. 45, 683–693 (1993). pathogenesis of neonatal hepatitis, biliary 97. Carvalho, E. et al. Analysis of the Biliary
58. Yokoyama, T. et al. Reversal of left-right atresia and choledochal cyst—the concept Transcriptome in Experimental Biliary Atresia.
asymmetry: a situs inversus mutation. Science of infantile obstructive cholangiopathy. Gastroenterology 129, 713–717 (2005).
260, 679–682 (1993). Prog. Pediatr. Surg. 6, 113–139 (1974). 98. Leonhardt, J. et al. Gene expression profile of
59. Schon, P. et al. Identification, genomic 77. Mack, C. L. The pathogenesis of biliary atresia: the infective murine model for biliary atresia.
organization, chromosomal mapping and evidence for a virus-induced autoimmune Pediatr. Surg. Int. 22, 84–89 (2006).
mutation analysis of the human INV gene, disease. Semin. Liver. Dis. 27, 233–242 99. Mack, C. L., Tucker, R. M., Sokol, R. J.
the ortholog of a murine gene implicated in left- (2007). & Kotzin, B. L. Armed CD4+ TH1 effector cells

10  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

and activated macrophages participate in bile 116. Jafri, M. et al. Cholangiocyte expression of quantification and phenotypes of maternal
duct injury in murine biliary atresia. Clin. Immunol. α2β1‑integrin confers susceptibility to rotavirus- cells in the liver. Pediatrics 121, 517–521
115, 200–209 (2005). induced experimental biliary atresia. Am. J. (2008).
100. Shivakumar, P. et al. Effector role of neonatal Physiol. Gastrointest. Liver Physiol. 295, G16–G26 132. Miethke, A. G. et al. Post-natal paucity of
hepatic CD8+ lymphocytes in epithelial injury (2008). regulatory T cells and control of NK cell
and autoimmunity in experimental biliary atresia. 117. Erickson, N. et al. Temporal-spatial activation activation in experimental biliary atresia.
Gastroenterology 133, 268–277 (2007). of apoptosis and epithelial injury in murine J. Hepatol. 52, 718–726 (2010).
101. Shivakumar, P., Sabla, G. E., Whitington, P., experimental biliary atresia. Hepatology 47, 133. Tucker, R. M., Feldman, A. G., Fenner, E. K.
Chougnet, C. A. & Bezerra, J. A. Neonatal NK 1567–1577 (2008). & Mack, C. L. Regulatory T cells inhibit TH1
cells target the mouse duct epithelium via 118. Jafri, M., Donnelly, B., Bondoc, A., Allen, S. & cell‑mediated bile duct injury in murine biliary
Nkg2d and drive tissue-specific injury in Tiao, G. Cholangiocyte secretion of chemokines atresia. J. Hepatol. 59, 790–796 (2013).
experimental biliary atresia. J. Clin. Invest. 119, in experimental biliary atresia. J. Pediatr. Surg. 134. Lages, C. S., Simmons, J., Chougnet, C. A.
2281–2290 (2009). 44, 500–507 (2009). & Miethke, A. G. Regulatory T cells control
102. Ahmed, A. F. et al. CD8+ T cells infiltrating into 119. Mohanty, S. K., Ivantes, C. A., Mourya, R., the CD8 adaptive immune response at the
bile ducts in biliary atresia do not appear to Pacheco, C. & Bezerra, J. A. Macrophages are time of ductal obstruction in experimental biliary
function as cytotoxic T cells: a clinicopathological targeted by rotavirus in experimental biliary atresia. Hepatology 56, 219–227 (2012).
analysis. J. Pathol. 193, 383–389 (2001). atresia and induce neutrophil chemotaxis by 135. Lu, B. R., Brindley, S. M., Tucker, R. M.,
103. Broome, U., Nemeth, A., Hultcrantz, R. mip2/cxcl2. Pediatr. Res. 67, 345–351 (2010). Lambert, C. L. & Mack, C. L. alpha-enolase
& Scheynius, A. Different expression of HLA-DR 120. Saxena, V. et al. Dendritic cells regulate natural autoantibodies cross-reactive to viral proteins in a
and ICAM-1 in livers from patients with biliary killer cell activation and epithelial injury in mouse model of biliary atresia. Gastroenterology
atresia and Byler’s disease. J. Hepatol. 26, experimental biliary atresia. Sci. Transl. Med. 3, 139, 1753–1761 (2010).
857–862 (1997). 102ra194 (2011). 136. Chiaramonte, M. G., Donaldson, D. D.,
104. Davenport, M. et al. Immunohistochemistry of 121. Kuwajima, S. et al. Interleukin 15-dependent Cheever, A. W. & Wynn, T. A. An IL-13 inhibitor
the liver and biliary tree in extrahepatic biliary crosstalk between conventional and blocks the development of hepatic fibrosis
atresia. J. Pediatr. Surg. 36, 1017–1025 (2001). plasmacytoid dendritic cells is essential for during a T-helper type 2-dominated
105. Dillon, P. W., Belchis, D., Minnick, K. & Tracy, T. CpG‑induced immune activation. Nat. Immunol. inflammatory response. J. Clin. Invest. 104,
Differential expression of the major 7, 740–746 (2006). 777–785 (1999).
histocompatibility antigens and ICAM-1 on bile 122. Sun, C. M., Fiette, L., Tanguy, M., Leclerc, C. 137. McHedlidze, T. et al. Interleukin 33 dependent
duct epithelial cells in biliary atresia. Tohoku J. & Lo-Man, R. Ontogeny and innate properties innate lymphoid cells mediate hepatic fibrosis.
Exp. Med. 181, 33–40 (1997). of neonatal dendritic cells. Blood 102, 585–591 Immunity 39, 357–371 (2013).
106. Mack, C. L. et al. Biliary atresia is associated (2003). 138. Syn, W. K. et al. Osteopontin is induced by
with CD4+ TH1 cell-mediated portal tract 123. Shivakumar, P. & Bezerra, J. A. Biliary atresia hedgehog pathway activation and promotes
inflammation. Pediatr. Res. 56, 79–87 (2004). and Th1 function: linking lymphocytes and bile fibrosis progression in nonalcoholic
107. Bill, A. H., Haas, J. E. & Foster, G. L. Biliary ducts: commentary on the article by Mack. et al. steatohepatitis. Hepatology 53, 106–115
atresia: histopathologic observations and on page 79. Pediatr. Res. 56, 9–10 (2004). (2011).
reflections upon its natural history. J. Pediatr. 124. Mohanty, S. K., Shivakumar, P., Sabla, G. 139. European consortium of Biliary Atresia and
Surg. 12, 977–982 (1977). & Bezerra, J. A. Loss of interleukin-12 modifies Related Diseases [online], http://www.bard-
108. Gosseye, S., Otte, J. B., De Meyer, R. & the pro-inflammatory response but does not online.com.
Maldague, P. A histological study of extrahepatic prevent duct obstruction in experimental biliary 140. Childhood Liver Disease Research Network
biliary atresia. Acta Paediatr. Belg. 30, 85–90 atresia. BMC Gastroenterol. 6, 14 (2006). [online], http://www.childrennetwork.org (2015).
(1977). 125. Tucker, R. M., Hendrickson, R. J., Mukaida, N., 141. Sarkhy, A., Schreiber, R. A., Milner, R. A.
109. Ohya, T., Fujimoto, T., Shimomura, H. & Miyano, T. Gill, R. G. & Mack, C. L. Progressive biliary & Barker, C. C. Does adjuvant steroid therapy
Degeneration of intrahepatic bile duct with destruction is independent of a functional post-Kasai portoenterostomy improve outcome
lymphocyte infiltration into biliary epithelial cells tumor necrosis factor-alpha pathway in a of biliary atresia? Systematic review and meta-
in biliary atresia. J. Pediatr. Surg. 30, 515–518 rhesus rotavirus-induced murine model of analysis. Can. J. Gastroenterol. 25, 440–444
(1995). biliary atresia. Viral Immunol. 20, 34–43 (2007). (2011).
110. Mack, C. L. et al. Oligoclonal expansions 126. Shivakumar, P., Mourya, R. & Bezerra, J. A. 142. Bezerra, J. A. et al. Use of corticosteroids after
of CD4+ and CD8+ T-cells in the target organ of Perforin and granzymes work in synergy to hepatoportoenterostomy for bile drainage in
patients with biliary atresia. Gastroenterology mediate cholangiocyte injury in experimental infants with biliary atresia: the START randomized
133, 278–287 (2007). biliary atresia. J. Hepatol. 60, 370–376 (2014). clinical trial. JAMA 311, 1750–1759 (2014).
111. Bezerra, J. A. et al. Genetic induction of 127. Pellicoro, A., Ramachandran, P., Iredale, J. P. 143. Davenport, M., Parsons, C., Tizzard, S.
proinflammatory immunity in children with biliary & Fallowfield, J. A. Liver fibrosis and repair: & Hadzic, N. Steroids in biliary atresia:
atresia. Lancet 360, 1563–1659 (2002). immune regulation of wound healing in a solid single surgeon, single centre, prospective study.
112. Tracy, T. F. Jr, Dillon, P., Fox, E. S., Minnick, K. organ. Nat. Rev. Immunol. 14, 181–194 (2014). J. Hepatol. 59, 1054–1058 (2013).
& Vogler, C. The inflammatory response in 128. Li, J. et al. Biliary repair and carcinogenesis
pediatric biliary disease: macrophage phenotype are mediated by IL 33 dependent cholangiocyte Acknowledgements
and distribution. J. Pediatr. Surg. 31, 121–125; proliferation. J. Clin. Invest. 124, 3241–3251 The authors are supported by the NIH grants
discussion 125–126 (1996). (2014). DK64008 and DK83781 to J.A.B. and DK95001
113. Urushihara, N. et al. Elevation of serum 129. Marvie, P. et al. Interleukin-33 overexpression to A.M. J.A.B. is the Cincinnati Principal Investigator
interleukin-18 levels and activation of is associated with liver fibrosis in mice and of the NIDDK-funded Childhood Liver Disease and
Kupffer cells in biliary atresia. J. Pediatr. Surg. humans. J. Cell. Mol. Med. 14, 1726–1739 Research Network (NIH grant DK62497).
35, 446–449 (2000). (2010).
114. Barnes, B. H. et al. Cholangiocytes as immune 130. Hayashida, M. et al. The evidence of maternal Author contributions
modulators in rotavirus-induced murine biliary microchimerism in biliary atresia using J.A.B. and A.A. contributed equally to researching
atresia. Liver Int. 29, 1253–1261 (2009). fluorescent in situ hybridization. J. Pediatr. Surg. data for the manuscript. All authors contributed
115. Allen, S. R. et al. Effect of rotavirus strain on 42, 2097–2101 (2007). equally to the discussion of content, writing
the murine model of biliary atresia. J. Virol. 81, 131. Muraji, T. et al. Maternal microchimerism in and reviewing and editing the manuscript
1671–1679 (2007). underlying pathogenesis of biliary atresia: before submission.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION  |  11


© 2015 Macmillan Publishers Limited. All rights reserved

Você também pode gostar