• No results found

University of Groningen Rare cholestatic childhood diseases van Wessel, Daan

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Rare cholestatic childhood diseases van Wessel, Daan"

Copied!
31
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Rare cholestatic childhood diseases

van Wessel, Daan

DOI:

10.33612/diss.133430251

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

van Wessel, D. (2020). Rare cholestatic childhood diseases: Advances in clinical care. University of

Groningen. https://doi.org/10.33612/diss.133430251

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Introduction and outline of thesis

Chapter 1

(3)

INTRODUCTION AND OUTLINE OF THESIS

Neonatal cholestasis

‘Jaundice’, the yellow discoloration of skin, mucous membranes and bodily fluids,

is observed in up to 80% of new-borns in their first week of life

1,2

. While the cause

is physiological, or breastfeeding-associated in the majority of cases, an underlying

disease may be present. Jaundice and elevation of total serum bilirubin can result

from a decrease in bile flow, or ‘cholestasis’, either by impaired secretion by

hepatocytes, or by obstruction of the intra- or extrahepatic bile ducts. Cholestasis

is defined as a conjugated serum bilirubin level >17 μmol/L if the total bilirubin

is <85.5 μmol/L or >20% of the total bilirubin if the total bilirubin is >85.5 μmol/

L

3

. Apart from its conjugated form (i.e. water soluble after binding to glucuronic

acid within the hepatocyte), bilirubin exists in an unconjugated form (i.e. a waste

product of haemoglobin, bound to albumin in serum until it reaches the hepatocyte).

While unconjugated hyperbilirubinemia resolves in the majority of cases, either by

phototherapy or spontaneously, conjugated hyperbilirubinemia is never physiological

and should always be evaluated for a hepatobiliary disorder.

Over 100 conditions can lead to neonatal cholestasis, including infections, anatomic

obstruction of the biliary system, genetic and metabolic disorders, endocrinopathies,

toxin or drug exposures and cardiovascular abnormalities

3

. Most common are biliary

atresia (BA, 25-40% of cases) and genetic disorders (25% of cases), such as severe

deficiency of the Bile Salt Export Pump (BSEP deficiency) or severe deficiency of the

Familial Intrahepatic Cholestasis protein type 1 (FIC1 deficiency)

4

. While a wide variety

of diseases can cause neonatal cholestasis, most of them are individually rare. Rare

diseases are often diagnosed later than necessary and ideal This can be attributed

to the fact that only one in approximately 350 breastfed neonates being evaluated

for neonatal jaundice within primary care, has an underlying disease responsible for

cholestasis

1

. Due to the relative rarity of such diseases, especially in the primary care

setting, many providers do not regard fractionating serum bilirubin levels as essential

in these particular patients, despite clear guidelines to do so in any jaundiced child at

the age of 3 weeks

4,5

. This prevents them from adequately selecting the neonates in

need of referral to secondary and/or tertiary care, which results in delay and prolonged

exposure to the untreated, underlying disease. An additional disadvantage, from a

more scientific point of view, is that study cohorts in rare disease are generally small

in size. It is often difficult to adequately draw valid conclusions from data derived

from small cohorts due to lack of statistical power. In order to increase the validity

of study data on rare diseases, there has been an increasing incentive over the last

decade to initiate international collaborations to share relevant data of these patients.

(4)

This thesis focusses on improving the clinical care for the rare diseases biliary atresia,

severe BSEP defi ciency and severe FIC1 defi ciency, all responsible for neonatal

cholestasis. A more detailed account of these diseases is given in the following

sections.

Biliary atresia

Biliary atresia (BA) is a rare disease of infancy which is characterized by obliteration of

the intra- and extrahepatic bile ducts due to an unknown cause. The incidence of BA

ranges from approximately 1:5000 live births in Asian countries to 1:20,000 live births

in the Western world

6–12

. Obliteration of the bile ducts results in cholestasis and, if left

untreated, patients die due to end-stage liver disease within two years

13

. BA occurs

in an isolated and syndromal form. There are three anatomical subtypes of isolated

BA, depending on the most proximal location of the biliary obstruction (Figure 1).

Type 3 is regarded the most extensive atresia and accounts for >90% of cases.

Syndromal BA, more specifi cally the ‘Biliary Atresia Splenic Malformation syndrome’

(BASM)

14,15

is observed less frequently. In addition to the phenotype of atretic bile

ducts as in isolated BA, patients with BASM all present with splenic anomalies,

and less often with an absent vena cava (approximately 70% of cases), intestinal

malrotation (approximately 60% of cases) or cardiac anomalies (approximately 45%

of cases, e.g. a ventricular septal defect).

Figure 1. Anatomical subtypes of biliary atresia. 1: left, patent common hepatic ducts. 2: centre, patent

hepatic ducts. 3: right, atresia of all extra-hepatic ducts.

While a signifi cant amount of research has been carried out to understand the

aetiology of BA, the mechanism by which BA originates has still not been conclusively

identifi ed. To date, it is largely accepted that BA is multifactorial in nature. The

most adopted theory is based on a pathogen- or toxin-induced, immune-mediated

destruction of the biliary tree in an individual with genetic susceptibility. This is

supported by evidence of elevated serum levels of Reovirus RNA, Rotavirus DNA,

(5)

CMV DNA and IgM in BA patients, as well as the establishment of BA-associated

single nucleotide polymorphisms (SNPs)

14–38

. A single viral cause has however not

been found and the aforementioned SNPs have only been found in the minority of

BA patients. If the origin of BA would indeed be (partially) attributed to an infectious

insult, likely occurring between conception and the perinatal period, one would

expect differences in the geographical distribution and a seasonal occurrence of BA

corresponding with the variable occurrence of pathogens. To date, some studies

focusing on the clustering of BA in space and time have been carried out, yet

they provide conflicting results

39–41

. Moreover, these studies lack data regarding

environmental factors, such as infectious outbreaks or population density. Studies

describing these factors in relation to the geographical and temporal clustering

of BA are therefore needed, in order to gain further insights in the putative role of

environmental parameters in the pathophysiology of BA.

Some studies have explored the role of the gut microbiome (i.e. the composition of

microorganisms in the gut) in the development of liver disease. A recent study found

that the gut microbiome of BA patients has what is called “decreased richness”

and “structural segregation” compared to healthy individuals

42

, which indicates

that the number of observed taxa was decreased and the types of observed taxa

were different, respectively. In addition, a case report describing two BA infants,

one with good and one with poor outcome, observed differences in the microbiota

of these children, which might suggest that the composition of the gut microbiota

is associated with outcome in BA

43

. Studies in a variety of (established) adult liver

diseases provided comparable results, i.e. that the gut microbiota of patients with

liver disease differs significantly from that of healthy subjects

44–49

. This may be

rather consequence than cause of the liver disease. It might be that changes in

bile composition or the complete absence of bile has a profound impact on the gut

microbiome. Additionally, liver disease and a disruption in the gut microbiome are

associated with increased gut wall permeability, allowing for increased translation

of bacterial products to the liver via the portal vein, potentially provoking constant

hepatocellular damage and/or subclinical inflammation

47,50,51

. In BA there is such a

change in bile metabolism, or even a complete absence of bile. While this interplay

between liver, microbiota and gut is an emerging area of research, there is still very

limited data published on the so-called ‘gut-liver axis’ in BA. Studies which assess

this axis in BA are therefore warranted.

After the obliteration of the biliary tree due to (a) so far unknown event(s), patients

typically present several weeks after birth with jaundice and pale stools. An early

(6)

diagnosis in BA is essential start treatment timely and limit the exposure of the liver

to cholestasis. Yet, this is hindered by the vast differential diagnosis underlying

neonatal cholestasis

4

. Usually BA is diagnosed by a combination of laboratory,

imaging and pathological studies. Ultrasound is used as first line imaging

52

, followed

by liver biopsy. The histological presence of portal fibrosis, ductular reaction or

proliferation, oedema and bile plugs are indicative of BA

53

. If BA is suspected,

clinicians proceed to laparotomy during which the diagnosis is classically based on

intra-operative cholangiography showing discontinuity of the biliary tree between the

liver and the intestine. Upon making the diagnosis, the Kasai portoenterostomy (KPE)

is performed, usually in the same session as the cholangiography was performed

4

. This surgical procedure, first described in 1959 by Morio Kasai

54

, aims to

re-establish bile flow from remaining microscopic intrahepatic ductules towards the gut,

by anastomosing a jejunal ‘Roux-en-y’ conduit to the porta hepatis (Figure 2). The

KPE is deemed therapeutically successful if clearance of jaundice (COJ) is achieved,

which is defined as a total serum bilirubin <20umol/L within six months after KPE. A

surgically well performed procedure does not guarantee COJ, since the restoration of

bile flow is dependent on patent microscopic ductules in the porta hepatis. Patients

without COJ usually require a liver transplantation (LTx) within the first year of life. The

liver graft used to be routinely become available from a deceased donor, but over

the last decade living related LTx is being increasingly performed as an alternative,

to overcome wait-list mortality due to donor organ shortage. In living related LTx,

the partial donor organ is obtained via a surgical procedure on a relative (frequently

one of the parents) or someone else.

(7)

Figure 2. Kasai portoenterostomy as performed in biliary atresia patients. A jejunal conduit is

anasto-mosed to the porta hepatis (‘roux-eny’) in order to re-establish bile fl ow towards the intestine, depending on patent microscopic ductules. Picture taken from 143, illustrated by Holden Groves.

The vast majority (>80%) of BA patients require an LTx during their life time due

to the direct or indirect consequences of end-stage liver disease, even when COJ

had initially been achieved. Therefore, BA is the main indication for paediatric

LTx, accounting for approximately 50% of the transplantations performed during

childhood in the Western world

13,55,56

.

The prognosis of BA has been found to strongly correlate with timely surgical

management by means of KPE, in order to minimize exposure to untreated disease.

It has become generally accepted that the earlier the KPE, the better the outcome.

More specifi cally: a KPE performed before the age of 60 days results in prolonged

native liver survival (NLS, i.e. the time between birth and LTx, death or last follow-up)

as opposed to an age at KPE above 60 days

11,57–61

. Several studies also suggest that

(8)

in the post-KPE prognosis remains controversial. It is believed that steroids might

have an anti-inflammatory effect, either in the liver or locally on the anastomosis,

which both are believed to improve bile flow. Whilst two studies found that

post-operative administration of steroids improved COJ-rates, a profound effect on 1- and

4-year NLS was not established

66,67

. The largest randomized controlled trial to date

by Bezerra et al. did not show a positive effect of steroids on COJ, and NLS did

not significantly improve after a 13-week steroid regimen

68

. Concerningly, an earlier

onset of serious adverse events was observed in the steroid group. In an ancillary

study it was observed that the steroid-use in the specific study was associated with

impaired growth

69

.

The prognosis of BA may also be affected by other factors. For example, preterm

birth has been reported as a risk factor for BA

40,70,71

, while BA might also be

considered a risk factor for preterm birth. To what extent prematurity influences the

prognosis of BA remains largely unknown. The diagnosis of BA in the preterm born

population is even more difficult than in the general population, largely due to the

frequent multifactorial origin of cholestasis in this patient group (e.g. prematurity of

the enterohepatic circulation, sepsis, prolonged total parenteral nutrition

72,73

. These

diagnostic difficulties may result in delayed referral to the treatment centre, to later

treatment and subsequently, to worse outcome. There is a need for establishing

the natural history of BA in preterm infants, in order to be able to optimize care and

awareness for this vulnerable subgroup of BA patients.

While factors predicting the prognosis in the first years after KPE have been studied

extensively, conclusive data regarding factors associated with long-term follow-up

are still scarce. As stated before, approximately half of the patients with BA need an

LTx before the age of two years. It thus seems that reaching the age of two years

with the native liver seems favourable in the prognosis BA. However, little is known

regarding the long-term outcome of BA patients with at least two years of NLS. Also,

factors associated with prolonged survival with native liver after two years of age

have not been established to date. This information, however, can be very helpful

for counselling parents and making treatment decisions.

Severe Bile Salt Export Pump deficiency

Severe deficiency of the Bile Salt Export Pump (BSEP) belongs to the heterogeneous

group of autosomal recessive cholestatic disorders termed ‘Progressive Familial

Intrahepatic Cholestasis’ (PFIC)

74–78

. The most severe form of BSEP deficiency has

been labelled ‘PFIC2’. BSEP belongs to the ATP-binding cassette (ABC) superfamily

(9)

and P-glycoprotein / Multidrug resistance (ABCB/MDR) subfamily of transporters

75,79

. BSEP, among other transporters, is localized at the canalicular membrane of the

liver parenchymal cells (“hepatocytes”) where it facilitates the transport of conjugated

bile acids into the biliary system, against up to 1000-fold concentration gradients

(Figure 3). Severe BSEP deficiency is a rare disease with an estimated incidence of

1:50.000 to 1:100.000 live births

80–82

in the Western world. Higher incidences have

been reported in the Middle East (1:7200)

83

. Sexes seem to be equally affected.

Severe BSEP deficiency results from mutations in the ABCB11 gene which in

humans is localized on chromosome 2 (2q24)

84,85

. Decreased trafficking of BSEP

from the Golgi system towards the canalicular membrane and/or decreased transport

capacity of BSEP

80,84,86–92

, the transport of bile acids from the hepatocyte towards

the canaliculus is impaired. As a result, bile acids accumulate within the hepatocyte

and the systemic circulation and inflict hepatocellular damage, leading to liver fibrosis

and cirrhosis.

Patients typically present within the first months of life with a phenotype

consisting of jaundice, high serum bile acids and transaminases, normal gamma

glutamyltransferase (GGT) levels, malabsorption, fat soluble vitamin deficiencies

and pruritus

81,93

. Some patients present with a relatively milder form of disease,

which is termed ‘Benign Recurrent Intrahepatic Cholestasis type 2’ (BRIC2). This

phenotype is characterized by episodic cholestasis with transient hepatocellular

damage

91,94,95

. BRIC2 might, however, progress to severe BSEP deficiency and

end-stage liver disease at later age, sometimes as late as age 30 years. Liver

histology at presentation in severe BSEP deficiency is characterized by canalicular

cholestasis, hepatocellular necrosis and giant cell transformation

80,96

. Upon

immunohistochemistry, BSEP antibody staining is abnormal or absent in at least

90% of severe cases

80

.

Final diagnosis is preferably made by DNA genotyping techniques, such as whole

exome sequencing. Access to such sequencing facilities might however be limited

in some parts of the world. There, the diagnosis of severe BSEP deficiency still relies

on clinical features and histology, which might lead to delayed diagnosis and even

misdiagnosis. It is therefore important – especially for a rare disease such as severe

BSEP deficiency – to connect various regions of the world through international

networks (e.g. European Reference Networks

97

. Such networks will increase the

exchange of knowledge and facilities, ultimately benefiting patients suffering severe

BSEP deficiency.

(10)

Figure 3. Transporters located at or near the canalicular membrane. The familial intrahepatic

choles-tatis protein type 1 (FIC1) transports aminophospholipids (i.e. phosphatidylcholine, PC) from the outer to the inner leafl et of the canalicular membrane, to maintain an aminophospholipid asymmetry. The bile salt export pump (BSEP) transports bile acids (BAs) from the hepatocyte towards the canaliculus. The func-tionality of these two proteins is impaired in severe FIC1 defi ciency (or progressive familial intrahepatic cholestatis type 1, PFIC1) or severe BSEP defi ciency (or progressive familial intrahepatic cholestasis type 2, PFIC2), respectively.

Ursodeoxycholic acid has been prescribed as treatment for severe BSEP defi ciency,

yet conclusive data regarding its long-term effi cacy have not been established. In

selected cases however, improvements such as de novo or retargeted canalicular

expression of BSEP have been described

81,98

. Some patients may benefi t from

surgery aimed at interrupting the enterohepatic circulation, termed surgical biliary

diversion (SBD). The Partial External Biliary Diversion is most commonly performed

surgical interruption of the enterohepatic circulation, during which an anastomosis

is created between the gallbladder and the skin by means of a short bowel segment

(Figure 4). It aims to divert bile out of the body, thereby decreasing the bile acid pool

which is available for reuptake in the terminal ileum.

SBD has been associated with a decrease in pruritus and might postpone or even

avert liver transplantation (LTx) in selected patients

99–107

. Not all patients respond

to treatment and even after an initial response, many cases progress into end-stage

liver disease within the fi rst years of life. Some patients receive LTx before reaching

this stage due refractory pruritus. Even though BSEP is expressed exclusively

in liver tissue, LTx is not a defi nitive treatment in some cases. Studies report

development of alloantibodies against BSEP in the graft, resulting in a severe BSEP

defi ciency phenotype, even after LTx

108–111

. Alternative treatments for severe BSEP

defi ciency are currently being developed and studied, such as medical interruption

of the enterohepatic circulation by means of apical sodium-dependent bile acid

(11)

transporter (ASBT) inhibitors or organic solute transporter (OST) inhibitors

112–114

, as

well as targeted pharmacotherapy such as chaperone drugs

115–117

. Mainly due to its

rarity, it so far remains unclear to what extend therapy, especially SBD, impacts the

natural history of severe BSEP defi ciency. Perhaps more importantly, it is yet to be

determined which subgroup(s) of patients will likely benefi t from therapy and which

not. Establishing this would allow for improved personalized clinical care for these

patients, as well as better targeting of novel therapeutic strategies. Large multicentre

studies are needed to address such topics.

Figure 4. Schematic overview of a partial external biliary diversion. A jejunal conduit is anastomosed

to the gallbladder and skin in order to divert bile out of the body. Illustrated by Antonia Felzen, with special

(12)

Figure 5. Medical interruption of the enterohepatic circulation by means of inhibition of the apical sodium bile acid transporter (ASBT, blue channel) in the ileum. Inhibition of the ASBT aims to decrease

the amount of bile acids available for reuptake in the liver, thereby limiting the hepatocellular damage.

While it has been established by a variety of studies to what extent mutations

impact BSEP traffi cking or its transport function, it has been diffi cult to establish

associations between the genotype and prognosis; ‘genotype-phenotype

relationships’. Mutations leading to a truncated or otherwise non-functional BSEP

protein, such as insertions, deletions, nonsense and splicing mutations, have been

associated with a severe phenotype in relatively small cohorts

80,92

. In addition,

missense mutations may affect protein traffi cking or processing or lead to a disrupted

protein structure

88,89,118,119

often resulting in a milder phenotype than the complete

abrogation or truncation of protein synthesis. Two examples are common European

missense mutations (c.890A>G; p.E297G and c.1445A>G; p.D482G) which result

in some residual function of BSEP and seem to respond better to some therapies

81,91,99,119

. While the fi ndings above provide some insight in genotype-phenotype

relations, such relations have so far never been established in large, multicentre

studies with genetically defi ned cohorts. This has prevented researchers and

(13)

clinicians to provide proper understanding and characterization of severe BSEP

deficiency, which in turn would allow for improved personalized clinical care.

Although hepatocellular carcinoma (HCC) is rare in young children, research has

demonstrated that patients with severe BSEP deficiency are at a considerable risk

to develop such carcinomas. Up to 15% of patients are diagnosed with HCC, of

which the majority before the age of five years, either clinically diagnosed or for the

first time observed in the explanted organ at the time of transplantation

92

. Based on

anticipated severity, it can be hypothesized that patients with truncating mutations

have a higher chance to develop HCC and possibly at an earlier age as opposed

to patients with milder mutations, e.g. missense mutations. This hypothesis was,

to a certain extent, supported by previously published case series

80,92

, yet an

in-depth analysis of large cohorts of genetically defined patients is lacking. Such data

is essential in targeting those patients in need for early-life screening for HCC and

possibly early referral for liver transplantation.

Severe Familial Intrahepatic Cholestasis protein type 1 deficiency

Severe deficiency of the Familial Intrahepatic Cholestasis protein type 1 (FIC1) results

from mutations in the ATP8B1 gene which in humans is localized on chromosome 18

(18q21). Just as severe BSEP deficiency, it belongs to the heterogeneous group of

autosomal recessive cholestatic disorders termed ‘Progressive Familial Intrahepatic

Cholestasis’

74–78

. The disease was first described as ‘Byler disease’ in the Amish

kindred by Clayton et al

120

. The most severe form of FIC1 deficiency was previously

known as Progressive Familial Intrahepatic Cholestasis type 1 (PFIC1). FIC1 is a

member of ATP-dependent membrane transporters known as phospholipid flippases,

which maintain a phospholipid asymmetry across phospholipid bilayers, such as the

canalicular membrane

84,121

. FIC1 is also expressed in e.g. pancreas, kidney, small

intestine and inner ear. The incidence and prevalence of severe FIC1 deficiency are

estimated to be even lower than the estimate of 1:50.000 to 1:100.000 for severe

BSEP deficiency. So far, a gender predisposition has not been established.

Mutations in ATP8B1 indirectly impair biliary bile salt excretion, although the

exact mechanism by which this occurs remains elusive. It has been suggested

that cholestasis might result from a disturbed phospholipid asymmetry across the

phospholipid bilayer. This asymmetry maintains a protective barrier against the high

bile acid concentration in the canaliculus, and therefore a disruption might enable

bile acids to damage the canalicular membrane, which in turn is believed to inhibit

the functionality of other canalicular membrane transport proteins (e.g. BSEP or the

Multidrug Resistant Protein 3)

121–123

. It has, however, been suggested that the role

(14)

of FIC1 in apical membrane organization may be unrelated to its aminophospholipid

translocase activity

124

.

Due to impaired bile salt excretion, patients with severe FIC1 deficiency typically

present with jaundice and pruritus in early childhood. Due to the expression of

FIC1 in other tissues, patients may also present themselves with concurrent

symptoms such as diarrhoea, pancreatitis, pneumonia, hearing loss, resistance

to parathyroid hormone, kidney stones and, as a result of malabsorption, severe

growth retardation

81,84,93,124–129

. As in severe BSEP deficiency, patients may

initially present with milder, episodic disease which is termed ‘Benign recurrent

intrahepatic cholestasis type 1’ (BRIC1). BRIC1 might evolve into severe FIC1

deficiency at later age

95

. At presentation, patients have elevated serum bile acids,

total serum bilirubin and transaminases, and normal GGT levels. The elevation of

the transaminases may be less pronounced than in patients with BSEP deficiency.

Additionally, liver histology at presentation usually shows intracanalicular cholestasis,

yet no or merely mild portal and lobular fibrosis

81

. Over time, inflammation, bile

duct proliferation and cirrhosis might develop. The final diagnosis is preferably

made by genotyping. As addressed earlier, sequencing techniques are not readily

available in all parts of the world, which hinders adequate diagnosis. International

networks to ensure the exchange of knowledge and resources are therefore needed.

Currently, treatment for the hepatic phenotype of severe FIC1 deficiency consists

of medical symptomatic treatment, surgical biliary diversion (SBD) and LTx.

Severe FIC1 deficiency and even BRIC1 have been largely refractory to medical

treatment; data regarding the symptomatic effect as well as the effect on

long-term outcome are inconclusive

74,81,99,126,130

. SBD procedures, such as the Partial

External Biliary Diversion or Ileal Exclusion, have been successful in decreasing

pruritus and slowing hepatic fibrosis, yet these data are derived from studies with

a proportion of genetically undefined patients

99,105,131–134

. Those patients that do

not (sufficiently) respond to medical treatment or SBD are candidates for LTx.

Since FIC1 is expressed in a variety of tissues, LTx is no definitive treatment for the

disease and complications after LTx have been reported, which will be discussed

below. Despite this, LTx may prolong overall survival. As in severe BSEP deficiency,

there is a – perhaps even larger – paucity of data regarding the symptomatic effect

of medical and surgical treatment strategies, as well as their effect on long-term

outcome. To be able to select those patients that will benefit from either of these

treatments, studies with relatively large, genetically defined cohorts are needed.

Such studies have so far not been published. FIC1 is a rare disease, therefore the

(15)

natural history and genotype-phenotype relationships are not very well established.

Genotype/phenotype associations might actually be difficult to assess in severe

FIC1 deficiency; even within one family with a specific mutation, a wide variability in

phenotype can exists

95,135

. The nature of the mutation might to a certain extent predict

the phenotype. Patients with missense mutations may have milder disease than

patients with truncating mutations

135

. On the other side of the spectrum are patients

harbouring nonsense or frameshift mutations or large deletions, which are expected

to result in a severe phenotype. One variant, p.Ile661Thr, is of European descent

and has been described in mild phenotypes. However, compound heterozygous

patients with this mutation can have severe disease

135

. Interestingly, HCC has never

been described in severe FIC1 deficiency, whilst up to 15% of patients with severe

BSEP deficiency are diagnosed with HCC. Obviously, absence of HCC in severe

FIC1 deficiency patients have precluded and association with severe mutations, in

contrast to BSEP deficiency patients.

The prognosis of severe FIC1 deficiency after LTx is unpredictable and might be even

worse than before transplantation, especially in terms of quality of life. In several

patients with severe FIC1 deficiency, diarrhoea might worsen after LTx and fluid

resuscitation might be needed

132,136

. Bile chelators or clonidine may milden diarrhoea

in selected patients

137

. Some patients develop post-LTx pancreatitis. Additionally,

development of steatohepatitis is a worrisome complication; it might necessitate

re-LTx. Performing a total biliary diversion (sometimes performed already at the

transplant itself) may reduce liver graft steatosis

99,136,138–142

.

So far, there has been no detailed investigation of the natural history or prognosis

in FIC1 deficiency, especially not in large samples of genetically defined patients.

Healthcare is increasingly moving towards patient tailored approaches, yet such

approaches are so far not been possible to patients with severe FIC1 deficiency (or

severe BSEP deficiency), because of lack of relevant natural history data. To provide

for this need, large study cohorts of severe FIC1 patients are needed to properly

address genotype/phenotype associations, in order to gain detailed insight in the

natural history and prognosis of this disease.

(16)

Outline

The aim of this study was to improve the clinical care for patients with the rare,

cholestatic childhood diseases biliary atresia (BA), severe BSEP deficiency

and severe FIC1 deficiency. Patients with BA undergo Kasai portoenterostomy,

preferably in the first weeks of life, to re-establish bile flow towards the intestine.

Unfortunately, the majority of patients require liver transplantation (LTx) before

the age of two. LTx after the age of two is relatively uncommon in BA and one

might therefore hypothesize that reaching the age of two with a native liver seems

rather favourable. While prognostic factors within the first two years have been

studied extensively, neither the native liver survival (NLS) beyond the age of two

years, nor the factors associated with it have however been characterized in detail.

The objective in

Chapter 2 was therefore to determine the prognosis of patients

with BA after 2 years of NLS. In addition, we aimed to identify early-life factors

which impacted continued NLS after two years of age. Another factor which might

impact the short- and long-term prognosis of BA is preterm birth. While it is known

that preterm birth is a risk factor for BA (and possibly vice versa), to what extent

prematurity affects the disease course of BA remains largely unknown, especially

in Western countries. In

Chapter 3 we characterized the course of BA in preterm

infants in the Netherlands in terms of clearance of jaundice, NLS and mortality. While

the disease course of BA has been relatively well established, its origin remains

elusive despite extensive scientific effort. A generally accepted theory is that BA

is multifactorial in nature. It is believed that an environmental factor (e.g. viruses,

bacteria or toxins) triggers an exaggerated immune response in an individual with

increased susceptibility for these triggers or their associated pathways. If BA would

indeed be triggered by pathogen, differences in the geographical distribution and

seasonal occurrence should to some extent correspond with abundance of these

pathogens. To provide further support for a pathophysiological role of environmental

factors in BA, we performed an epidemiological study in

Chapter 4. In this study, we

assessed if temporal and geographical clustering of BA existed in the Netherlands,

and if this corresponded with the number of nationwide confirmed infections and

population density. It is increasingly recognized that the gut microbiota is associated

with health and disease. More specifically, the presence and prognosis of several

(adult) liver disease has already been associated with the composition of the gut

microbiota. Data regarding the microbiota and BA are scarce. To establish if in a

BA specific microbiota is present and if the prognosis of BA is associated with

the microbiota in the gut, we performed a prospective cohort study in

Chapter

5. We determined the gut microbiota by means of 16S rRNA sequencing in BA

infants longitudinally, up to six months after Kasai portoenterostomy. Moreover,

(17)

we compared the composition of the gut microbiota from that of BA patients to

that of infants undergoing inguinal hernia repair. Severe deficiency of the bile salt

export pump (BSEP) is a rare disease and results from mutations in the ABCB11

gene. Due to its rarity, it has so far been impossible to assess the natural history,

genotype-phenotype associations and the effect of surgical interruption of the

enterohepatic circulation on long-term outcome in large, genetically well-defined

cohorts. In

Chapter 6 we aimed to provide these data and for that we used a

global, multicentre cohort of genetically defined severe BSEP deficiency patients.

Severe BSEP deficiency is mostly recessive in nature, hence it results from two

mutations in ABCB11. Two common European mutations (i.e. c.1445A>G; p.D482G

and c.890A>G; p.E297G) have been associated with residual BSEP functionality and

patients harbouring at least one of these mutations seem to present relatively mild

phenotype. To what extent the mutation other than p.D482G or p.E297G impacts

the phenotype has not been described before. In

Chapter 7 we set out to scrutinize

patients with severe BSEP deficiency that harbour at least one p.D482G or p.E297G

mutation. By subcategorizing patients based on the severity on the second allele

(i.e. the mutation other than p.D482G or p.E297G), we aimed to find

genotype-phenotype associations to be able to address if the combined residual function

of the two mutations in ABCB11 is associated with the severe BSEP deficiency

phenotype. Severe deficiency of the familial intrahepatic cholestasis protein type

1 (FIC1) results from mutations in the ATP8B1 gene. Severe deficiency of FIC1 is a

rare disease, likely even rarer than severe BSEP deficiency. Therefore, the natural

course of disease has not been established in a proper manner. Moreover,

genotype-phenotype associations have not been addressed. Lastly, the effect of interruption of

the enterohepatic circulation on long-term outcome remains unknown. In

Chapter

8 we performed a global, multicentre cohort study to provide these insights.

In

Chapter 9 we discuss the outcomes and clinical implications of the studies

reported in this thesis and future directions for research in neonatal cholestasis as

a whole, and biliary atresia, severe BSEP deficiency and severe FIC1 deficiency

separately.

(18)

REFERENCES

1. Kelly DA, Stanton A. Jaundice in babies: implications for community screening for biliary atresia. BMJ (Clinical research ed). 1995 May 6;310(6988):1172–3.

2. Bhutani VK, Stark AR, Lazzeroni LC, Poland R, Gourley GR, Kazmierczak S, et al. Predischarge screening for severe neonatal hyperbilirubinemia identifies infants who need phototherapy. The Journal of pediatrics. 2013 Mar;162(3):477-482.e1.

3. Feldman AG, Sokol RJ. Neonatal cholestasis: emerging molecular diagnostics and potential novel therapeutics. Nature reviewsGastroenterology & hepatology. 2019 Jun;16(6):346–60.

4. Fawaz R, Baumann U, Ekong U, Fischler B, Hadzic N, Mack CL, et al. Guideline for the Evaluation of Cholestatic Jaundice in Infants: Joint Recommendations of the North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition and the European Society for Pediatric Gastroenterology, Hepatology, and Nutriti. Journal of pediatric gastroenterology and nutrition. 2017 Jan;64(1):154–68.

5. Dijk PH, de Vries TW, de Beer JJ, Association DP. Guideline “Prevention, diagnosis and treatment of hyperbilirubinemia in the neonate with a gestational age of 35 or more weeks.” Nederlands tijdschrift voor geneeskunde. 2009;153:A93.

6. McKiernan PJ, Baker AJ, Kelly DA. The frequency and outcome of biliary atresia in the UK and Ireland. Lancet (London, England). 2000 Jan 1;355(9197):25–9.

7. Lin YC, Chang MH, Liao SF, Wu JF, Ni YH, Tiao MM, et al. Decreasing rate of biliary atresia in Taiwan: a survey, 2004-2009. Pediatrics. 2011 Sep;128(3):530.

8. Hopkins PC, Yazigi N, Nylund CM. Incidence of Biliary Atresia and Timing of Hepatoportoenterostomy in the United States. The Journal of pediatrics. 2017 Aug;187:253–7.

9. Yoon PW, Bresee JS, Olney RS, James LM, Khoury MJ. Epidemiology of biliary atresia: a population-based study. Pediatrics. 1997 Mar;99(3):376–82.

10. Fanna M, Masson G, Capito C, Girard M, Guerin F, Hermeziu B, et al. Management of Biliary Atresia in France 1986-2015: Long Term Results. Journal of pediatric gastroenterology and nutrition. 2019 Jul 22;

11. de Vries W, de Langen ZJ, Groen H, Scheenstra R, Peeters PM, Hulscher JB, et al. Biliary atresia in the Netherlands: outcome of patients diagnosed between 1987 and 2008. The Journal of pediatrics. 2012 Apr;160(4):638-644.e2.

12. Wada H, Muraji T, Yokoi A, Okamoto T, Sato S, Takamizawa S, et al. Insignificant seasonal and geographical variation in incidence of biliary atresia in Japan: a regional survey of over 20 years. Journal of pediatric surgery. 2007 Dec;42(12):2090–2.

13. Hartley JL, Davenport M, Kelly DA. Biliary atresia. Lancet (London, England). 2009 Nov 14;374(9702):1704–13.

14. Davenport M, Savage M, Mowat AP, Howard ER. Biliary atresia splenic malformation syndrome: an etiologic and prognostic subgroup. Surgery. 1993 Jun;113(6):662–8. 15. Davenport M, Tizzard SA, Underhill J, Mieli-Vergani G, Portmann B, Hadzic N. The biliary

atresia splenic malformation syndrome: a 28-year single-center retrospective study. The Journal of pediatrics. 2006 Sep;149(3):393–400.

(19)

16. Oetzmann von Sochaczewski C, Pintelon I, Brouns I, Dreier A, Klemann C, Timmermans JP, et al. Rotavirus particles in the extrahepatic bile duct in experimental biliary atresia. Journal of pediatric surgery. 2014 Apr;49(4):520–4.

17. Waisbourd-Zinman O, Koh H, Tsai S, Lavrut PM, Dang C, Zhao X, et al. The toxin biliatresone causes mouse extrahepatic cholangiocyte damage and fibrosis through decreased glutathione and SOX17. Hepatology (Baltimore, Md). 2016 Sep;64(3):880–93. 18. Ramachandran P, Balamurali D, Peter JJ, Kumar MM, Safwan M, Vij M, et al.

RNA-seq reveals outcome-specific gene expression of MMP7 and PCK1 in biliary atresia. Molecular biology reports. 2019 Jul 24;

19. Lorent K, Gong W, Koo KA, Waisbourd-Zinman O, Karjoo S, Zhao X, et al. Identification of a plant isoflavonoid that causes biliary atresia. Science translational medicine. 2015 May 6;7(286):286ra67.

20. Fratta LX, Hoss GR, Longo L, Uribe-Cruz C, da Silveira TR, Vieira SM, et al. Hypoxic-ischemic gene expression profile in the isolated variant of biliary atresia. Journal of hepato-biliary-pancreatic sciences. 2015 Dec;22(12):846–54.

21. dos Santos JL, da Silveira TR, da Silva VD, Cerski CT, Wagner MB. Medial thickening of hepatic artery branches in biliary atresia. A morphometric study. Journal of pediatric surgery. 2005 Apr;40(4):637–42.

22. Hill R, Quaglia A, Hussain M, Hadzic N, Mieli-Vergani G, Vergani D, et al. Th-17 cells infiltrate the liver in human biliary atresia and are related to surgical outcome. Journal of pediatric surgery. 2015 Aug;50(8):1297–303.

23. Karjoo S, Hand NJ, Loarca L, Russo PA, Friedman JR, Wells RG. Extrahepatic cholangiocyte cilia are abnormal in biliary atresia. Journal of pediatric gastroenterology and nutrition. 2013 Jul;57(1):96–101.

24. Klemann C, Schroder A, Dreier A, Mohn N, Dippel S, Winterberg T, et al. Interleukin 17, Produced by gammadelta T Cells, Contributes to Hepatic Inflammation in a Mouse Model of Biliary Atresia and Is Increased in Livers of Patients. Gastroenterology. 2016 Jan;150(1):229-241.e5.

25. Leonhardt J, Kuebler JF, Turowski C, Tschernig T, Geffers R, Petersen C. Susceptibility to experimental biliary atresia linked to different hepatic gene expression profiles in two mouse strains. Hepatology research : the official journal of the Japan Society of Hepatology. 2010 Feb;40(2):196–203.

26. Girard M, Panasyuk G. Genetics in biliary atresia. Current opinion in gastroenterology. 2019 Mar;35(2):73–81.

27. Jafri M, Donnelly B, Allen S, Bondoc A, McNeal M, Rennert PD, et al. Cholangiocyte expression of alpha2beta1-integrin confers susceptibility to rotavirus-induced experimental biliary atresia. American journal of physiologyGastrointestinal and liver physiology. 2008 Jul;295(1):G16–26.

28. Mysore KR, Shneider BL, Harpavat S. Biliary Atresia as a Disease Starting In Utero: Implications for Treatment, Diagnosis, and Pathogenesis. Journal of pediatric gastroenterology and nutrition. 2019 Jul 22;

29. Saxena V, Shivakumar P, Sabla G, Mourya R, Chougnet C, Bezerra JA. Dendritic cells regulate natural killer cell activation and epithelial injury in experimental biliary atresia. Science translational medicine. 2011 Sep 28;3(102):102ra94.

30. Tucker RM, Feldman AG, Fenner EK, Mack CL. Regulatory T cells inhibit Th1 cell-mediated bile duct injury in murine biliary atresia. Journal of hepatology. 2013 Oct;59(4):790–6.

(20)

31. Lages CS, Simmons J, Chougnet CA, Miethke AG. Regulatory T cells control the CD8 adaptive immune response at the time of ductal obstruction in experimental biliary atresia. Hepatology (Baltimore, Md). 2012 Jul;56(1):219–27.

32. Lu BR, Brindley SM, Tucker RM, Lambert CL, Mack CL. Alpha-Enolase Autoantibodies Cross-Reactive to Viral Proteins in a Mouse Model of Biliary Atresia. Gastroenterology. 2010 Nov;139(5):1753–61.

33. Feldman AG, Tucker RM, Fenner EK, Pelanda R, Mack CL. B cell deficient mice are protected from biliary obstruction in the rotavirus-induced mouse model of biliary atresia. PloS one. 2013 Aug 21;8(8):e73644.

34. Cheng G, Tang CS, Wong EH, Cheng WW, So MT, Miao X, et al. Common genetic variants regulating ADD3 gene expression alter biliary atresia risk. Journal of hepatology. 2013 Dec;59(6):1285–91.

35. Cui S, Leyva-Vega M, Tsai EA, EauClaire SF, Glessner JT, Hakonarson H, et al. Evidence from human and zebrafish that GPC1 is a biliary atresia susceptibility gene. Gastroenterology. 2013 May;144(5):1107-1115.e3.

36. Davit-Spraul A, Baussan C, Hermeziu B, Bernard O, Jacquemin E. CFC1 gene involvement in biliary atresia with polysplenia syndrome. Journal of pediatric gastroenterology and nutrition. 2008 Jan;46(1):111–2.

37. Mack CL. What Causes Biliary Atresia? Unique Aspects of the Neonatal Immune System Provide Clues to Disease Pathogenesis. Cellular and molecular gastroenterology and hepatology. 2015 May 1;1(3):267–74.

38. Bezerra JA. Potential etiologies of biliary atresia. Pediatric transplantation. 2005 Oct;9(5):646–51.

39. Strickland AD, Shannon K. Studies in the etiology of extrahepatic biliary atresia: Time-space clustering. The Journal of Pediatrics. 1982;

40. Fischler B, Haglund B, Hjern A. A population-based study on the incidence and possible pre- and perinatal etiologic risk factors of biliary atresia. The Journal of pediatrics. 2002 Aug;141(2):217–22.

41. Chardot C, Carton M, Spire-Bendelac N, le Pommelet C, Golmard JL, Auvert B. Epidemiology of biliary atresia in France: a national study 1986-96. Journal of hepatology. 1999 Dec;31(6):1006–13.

42. Wang J, Qian T, Jiang J, Yang Y, Shen Z, Huang Y, et al. Gut microbial profile in biliary atresia: a case-control study. Journal of gastroenterology and hepatology. 2019 Jul 4; 43. Elaine Chen Y-F, Lai M-W, Tsai C-N, Lai J-Y, Yang Y-C, Chen S-Y. Gut Microbiota

Composition and Copy Number Variation in Biliary Atresia Infants with Different Outcomes after Kasai Operation. Pediatrics & Neonatology. 2020;

44. Sabino J, Vieira-Silva S, Machiels K, Joossens M, Falony G, Ballet V, et al. Primary sclerosing cholangitis is characterised by intestinal dysbiosis independent from IBD. Gut. 2016 Oct;65(10):1681–9.

45. Boursier J, Mueller O, Barret M, Machado M, Fizanne L, Araujo-Perez F, et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology (Baltimore, Md). 2016 Mar;63(3):764–75.

46. Quigley EM. Primary Biliary Cirrhosis and the Microbiome. Seminars in liver disease. 2016 Sep;36(4):349–53.

(21)

47. Kakiyama G, Pandak WM, Gillevet PM, Hylemon PB, Heuman DM, Daita K, et al. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. Journal of hepatology. 2013 May;58(5):949–55.

48. Hartstra A v, Bouter KE, Backhed F, Nieuwdorp M. Insights into the role of the microbiome in obesity and type 2 diabetes. Diabetes care. 2015 Jan;38(1):159–65.

49. Brandi G, de Lorenzo S, Candela M, Pantaleo MA, Bellentani S, Tovoli F, et al. Microbiota, NASH, HCC and the potential role of probiotics. Carcinogenesis. 2017 Mar 1;38(3):231– 40.

50. Stenman LK, Holma R, Eggert A, Korpela R. A novel mechanism for gut barrier dysfunction by dietary fat: epithelial disruption by hydrophobic bile acids. American journal of physiologyGastrointestinal and liver physiology. 2013 Feb 1;304(3):227. 51. Pereira-Fantini PM, Lapthorne S, Joyce SA, Dellios NL, Wilson G, Fouhy F, et al. Altered

FXR signalling is associated with bile acid dysmetabolism in short bowel syndrome-associated liver disease. Journal of hepatology. 2014 Nov;61(5):1115–25.

52. Farrant P, Meire HB, Mieli-Vergani G. Improved diagnosis of extraheptic biliary atresia by high frequency ultrasound of the gall bladder. The British journal of radiology. 2001 Oct;74(886):952–4.

53. Russo P, Magee JC, Boitnott J, Bove KE, Raghunathan T, Finegold M, et al. Design and validation of the biliary atresia research consortium histologic assessment system for cholestasis in infancy. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2011 Apr;9(4):357-362. e2.

54. Kasai M, Suzuki M. A new operation for non-correctable biliary atresia: hepatic portoenterostomy.. 1959;(13):733–9.

55. Kim SR, Saito Y, Itoda M, Maekawa K, Kawamoto M, Kamatani N, et al. Genetic variations of the ABC transporter gene ABCB11 encoding the human bile salt export pump (BSEP) in a Japanese population. Drug metabolism and pharmacokinetics. 2009;24(3):277–81. 56. Scheenstra R, Peeters PM, Verkade HJ, Gouw AS. Graft fibrosis after pediatric liver transplantation: ten years of follow-up. Hepatology (Baltimore, Md). 2009 Mar;49(3):880– 6.

57. Schreiber RA, Barker CC, Roberts EA, Martin SR, Alvarez F, Smith L, et al. Biliary atresia: the Canadian experience. The Journal of pediatrics. 2007 Dec;151(6):659–65, 665.e1. 58. Hsiao CH, Chang MH, Chen HL, Lee HC, Wu TC, Lin CC, et al. Universal screening for

biliary atresia using an infant stool color card in Taiwan. Hepatology (Baltimore, Md). 2008 Apr;47(4):1233–40.

59. Serinet MO, Wildhaber BE, Broue P, Lachaux A, Sarles J, Jacquemin E, et al. Impact of age at Kasai operation on its results in late childhood and adolescence: a rational basis for biliary atresia screening. Pediatrics. 2009 May;123(5):1280–6.

60. Chardot C, Buet C, Serinet MO, Golmard JL, Lachaux A, Roquelaure B, et al. Improving outcomes of biliary atresia: French national series 1986-2009. Journal of hepatology. 2013 Jun;58(6):1209–17.

61. de Vries W, Homan-Van der Veen J, Hulscher JB, Hoekstra-Weebers JE, Houwen RH, Verkade HJ, et al. Twenty-year transplant-free survival rate among patients with biliary atresia. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2011 Dec;9(12):1086–91.

(22)

62. Davenport M, Ong E, Sharif K, Alizai N, McClean P, Hadzic N, et al. Biliary atresia in England and Wales: results of centralization and new benchmark. Journal of pediatric surgery. 2011 Sep;46(9):1689–94.

63. Lampela H, Ritvanen A, Kosola S, Koivusalo A, Rintala R, Jalanko H, et al. National centralization of biliary atresia care to an assigned multidisciplinary team provides high-quality outcomes. Scandinavian Journal of Gastroenterology. 2012 Jan;47(1):99–107. 64. Davenport M, de Ville de Goyet J, Stringer MD, Mieli-Vergani G, Kelly DA, McClean P, et

al. Seamless management of biliary atresia in England and Wales (1999-2002). Lancet (London, England). 2004 Apr 24;363(9418):1354–7.

65. Leonhardt J, Kuebler JF, Leute PJ, Turowski C, Becker T, Pfister ED, et al. Biliary atresia: lessons learned from the voluntary German registry. European journal of pediatric surgery : official journal of Austrian Association of Pediatric Surgery .[et al] = Zeitschrift fur Kinderchirurgie. 2011 Mar;21(2):82–7.

66. Davenport M, Parsons C, Tizzard S, Hadzic N. Steroids in biliary atresia: single surgeon, single centre, prospective study. Journal of hepatology. 2013 Nov;59(5):1054–8. 67. Davenport M, Stringer MD, Tizzard SA, McClean P, Mieli-Vergani G, Hadzic N.

Randomized, double-blind, placebo-controlled trial of corticosteroids after Kasai portoenterostomy for biliary atresia. Hepatology (Baltimore, Md). 2007 Dec;46(6):1821–7. 68. Bezerra JA, Spino C, Magee JC, Shneider BL, Rosenthal P, Wang KS, et al. Use of corticosteroids after hepatoportoenterostomy for bile drainage in infants with biliary atresia: the START randomized clinical trial. Jama. 2014 May 7;311(17):1750–9. 69. Alonso EM, Ye W, Hawthorne K, Venkat V, Loomes KM, Mack CL, et al. Impact of Steroid

Therapy on Early Growth in Infants with Biliary Atresia: The Multicenter Steroids in Biliary Atresia Randomized Trial. The Journal of pediatrics. 2018 Nov;202:179-185.e4. 70. Chiu CY, Chen PH, Chan CF, Chang MH, Wu TC, Group TISCCS. Biliary atresia in preterm

infants in Taiwan: a nationwide survey. The Journal of pediatrics. 2013 Jul;163(1):100-3. e1.

71. Chen HW, Hsu WM, Chang MH, Chen CY, Chou HC, Tsao PN, et al. Embryonic biliary atresia in a very-low-birth-weight premature infant. Journal of the Formosan Medical Association = Taiwan yi zhi. 2007 Jan;106(1):78–81.

72. Fallon SC, Chang S, Finegold MJ, Karpen SJ, Brandt ML. Discordant presentation of biliary atresia in premature monozygotic twins. Journal of pediatric gastroenterology and nutrition. 2013 Oct;57(4):22.

73. Mourier O, Franchi-Abella S, Ackermann O, Branchereau S, Gonzales E, Bernard O, et al. Delayed postnatal presentation of biliary atresia in 2 premature neonates. Journal of pediatric gastroenterology and nutrition. 2011 Apr;52(4):489–91.

74. Bull LN, Thompson RJ. Progressive Familial Intrahepatic Cholestasis. Clinics in liver disease. 2018 Nov;22(4):657–69.

75. Thompson R, Strautnieks S. BSEP: function and role in progressive familial intrahepatic cholestasis. Seminars in liver disease. 2001 Nov;21(4):545–50.

76. Knisely AS. Progressive familial intrahepatic cholestasis: a personal perspective. Pediatric and developmental pathology : the official journal of the Society for Pediatric Pathology and the Paediatric Pathology Society. 2000;3(2):113–25.

77. Pauli-Magnus C, Stieger B, Meier Y, Kullak-Ublick GA, Meier PJ. Enterohepatic transport of bile salts and genetics of cholestasis. Journal of hepatology. 2005 Aug;43(2):342–57.

(23)

78. Elferink RO, Groen AK. Genetic defects in hepatobiliary transport. Biochimica et biophysica acta. 2002 Mar 16;1586(2):129–45.

79. Stieger B, Meier Y, Meier PJ. The bile salt export pump. Pflugers Archiv : European journal of physiology. 2007 Feb;453(5):611–20.

80. Strautnieks SS, Byrne JA, Pawlikowska L, Cebecauerova D, Rayner A, Dutton L, et al. Severe bile salt export pump deficiency: 82 different ABCB11 mutations in 109 families. Gastroenterology. 2008 Apr;134(4):1203–14.

81. Davit-Spraul A, Fabre M, Branchereau S, Baussan C, Gonzales E, Stieger B, et al. ATP8B1 and ABCB11 analysis in 62 children with normal gamma-glutamyl transferase progressive familial intrahepatic cholestasis (PFIC): phenotypic differences between PFIC1 and PFIC2 and natural history. Hepatology (Baltimore, Md). 2010 May;51(5):1645–55.

82. Kamath BM, Chen Z, Romero R, Fredericks EM, Alonso EM, Arnon R, et al. Quality of Life and Its Determinants in a Multicenter Cohort of Children with Alagille Syndrome. The Journal of pediatrics. 2015 Aug;167(2):390-6.e3.

83. Shagrani M, Burkholder J, Broering D, Abouelhoda M, Faquih T, El-Kalioby M, et al. Genetic profiling of children with advanced cholestatic liver disease. Clinical genetics. 2017 Jul;92(1):52–61.

84. Strautnieks SS, Bull LN, Knisely AS, Kocoshis SA, Dahl N, Arnell H, et al. A gene encoding a liver-specific ABC transporter is mutated in progressive familial intrahepatic cholestasis. Nature genetics. 1998 Nov;20(3):233–8.

85. Jansen PL, Strautnieks SS, Jacquemin E, Hadchouel M, Sokal EM, Hooiveld GJ, et al. Hepatocanalicular bile salt export pump deficiency in patients with progressive familial intrahepatic cholestasis. Gastroenterology. 1999 Dec;117(6):1370–9.

86. Wang L, Dong H, Soroka CJ, Wei N, Boyer JL, Hochstrasser M. Degradation of the bile salt export pump at endoplasmic reticulum in progressive familial intrahepatic cholestasis type II. Hepatology (Baltimore, Md). 2008 Nov;48(5):1558–69.

87. Wang L, Soroka CJ, Boyer JL. The role of bile salt export pump mutations in progressive familial intrahepatic cholestasis type II. The Journal of clinical investigation. 2002 Oct;110(7):965–72.

88. Lam P, Pearson CL, Soroka CJ, Xu S, Mennone A, Boyer JL. Levels of plasma membrane expression in progressive and benign mutations of the bile salt export pump (Bsep/ Abcb11) correlate with severity of cholestatic diseases. American journal of physiologyCell physiology. 2007 Nov;293(5):1709.

89. Hayashi H, Sugiyama Y. 4-Phenylbutyrate Enhances the Cell Surface Expression and the Transport Capacity of Wild-Type and Mutated Bile Salt Export Pumps. Hepatology (Baltimore, Md). 2007 Jun;45(6):1506–16.

90. Byrne JA, Strautnieks SS, Ihrke G, Pagani F, Knisely AS, Linton KJ, et al. Missense mutations and single nucleotide polymorphisms in ABCB11 impair bile salt export pump processing and function or disrupt pre-messenger RNA splicing. Hepatology (Baltimore, Md). 2009 Feb;49(2):553–67.

91. Droge C, Bonus M, Baumann U, Klindt C, Lainka E, Kathemann S, et al. Sequencing of FIC1, BSEP and MDR3 in a large cohort of patients with cholestasis revealed a high number of different genetic variants. Journal of hepatology. 2017 Dec;67(6):1253–64. 92. Knisely AS, Strautnieks SS, Meier Y, Stieger B, Byrne JA, Portmann BC, et al.

Hepatocellular carcinoma in ten children under five years of age with bile salt export pump deficiency. Hepatology (Baltimore, Md). 2006 Aug;44(2):478–86.

(24)

93. Pawlikowska L, Strautnieks S, Jankowska I, Czubkowski P, Emerick K, Antoniou A, et al. Differences in presentation and progression between severe FIC1 and BSEP deficiencies. Journal of hepatology. 2010 Jul;53(1):170–8.

94. van Mil SW, van der Woerd WL, van der Brugge G, Sturm E, Jansen PL, Bull LN, et al. Benign recurrent intrahepatic cholestasis type 2 is caused by mutations in ABCB11. Gastroenterology. 2004 Aug;127(2):379–84.

95. van Ooteghem NA, Klomp LW, van Berge-Henegouwen GP, Houwen RH. Benign recurrent intrahepatic cholestasis progressing to progressive familial intrahepatic cholestasis: low GGT cholestasis is a clinical continuum. Journal of hepatology. 2002 Mar;36(3):439–43. 96. Davit-Spraul A, Gonzales E, Baussan C, Jacquemin E. Progressive familial intrahepatic

cholestasis. Orphanet journal of rare diseases. 2009 Jan 8;4:1.

97. Heon-Klin V. European Reference networks for rare diseases: what is the conceptual framework? Orphanet journal of rare diseases. 2017 Aug 7;12(1):133–7.

98. Varma S, Revencu N, Stephenne X, Scheers I, Smets F, Beleza-Meireles A, et al. Retargeting of bile salt export pump and favorable outcome in children with progressive familial intrahepatic cholestasis type 2. Hepatology (Baltimore, Md). 2015 Jul;62(1):198– 206.

99. Bull LN, Pawlikowska L, Strautnieks S, Jankowska I, Czubkowski P, Dodge JL, et al. Outcomes of surgical management of familial intrahepatic cholestasis 1 and bile salt export protein deficiencies. Hepatology communications. 2018 Mar 30;2(5):515–28. 100. Ellinger P, Stindt J, Droge C, Sattler K, Stross C, Kluge S, et al. Partial external biliary

diversion in bile salt export pump deficiency: Association between outcome and mutation. World journal of gastroenterology. 2017 Aug 7;23(29):5295–303.

101. Lemoine C, Bhardwaj T, Bass LM, Superina RA. Outcomes following partial external biliary diversion in patients with progressive familial intrahepatic cholestasis. Journal of pediatric surgery. 2017 Feb;52(2):268–72.

102. Yang H, Porte RJ, Verkade HJ, de Langen ZJ, Hulscher JB. Partial external biliary diversion in children with progressive familial intrahepatic cholestasis and Alagille disease. Journal of pediatric gastroenterology and nutrition. 2009 Aug;49(2):216–21.

103. Ng VL, Ryckman FC, Porta G, Miura IK, de Carvalho E, Servidoni MF, et al. Long-term outcome after partial external biliary diversion for intractable pruritus in patients with intrahepatic cholestasis. Journal of pediatric gastroenterology and nutrition. 2000 Feb;30(2):152–6.

104. Arnell H, Bergdahl S, Papadogiannakis N, Nemeth A, Fischler B. Preoperative observations and short-term outcome after partial external biliary diversion in 13 patients with progressive familial intrahepatic cholestasis. Journal of pediatric surgery. 2008 Jul;43(7):1312–20.

105. Kurbegov AC, Setchell KD, Haas JE, Mierau GW, Narkewicz M, Bancroft JD, et al. Biliary diversion for progressive familial intrahepatic cholestasis: improved liver morphology and bile acid profile. Gastroenterology. 2003 Oct;125(4):1227–34.

106. Melter M, Rodeck B, Kardorff R, Hoyer PF, Petersen C, Ballauff A, et al. Progressive familial intrahepatic cholestasis: partial biliary diversion normalizes serum lipids and improves growth in noncirrhotic patients. The American Journal of Gastroenterology. 2000 Dec;95(12):3522–8.

(25)

107. Emond JC, Whitington PF. Selective surgical management of progressive familial intrahepatic cholestasis (Byler’s disease). Journal of pediatric surgery. 1995 Dec;30(12):1635–41.

108. Stindt J, Kluge S, Droge C, Keitel V, Stross C, Baumann U, et al. Bile salt export pump-reactive antibodies form a polyclonal, multi-inhibitory response in antibody-induced bile salt export pump deficiency. Hepatology (Baltimore, Md). 2016 Feb;63(2):524–37. 109. Kubitz R, Droge C, Kluge S, Stross C, Walter N, Keitel V, et al. Autoimmune BSEP

disease: disease recurrence after liver transplantation for progressive familial intrahepatic cholestasis. Clinical reviews in allergy & immunology. 2015 Jun;48(2–3):273–84. 110. Maggiore G, Gonzales E, Sciveres M, Redon MJ, Grosse B, Stieger B, et al. Relapsing

features of bile salt export pump deficiency after liver transplantation in two patients with progressive familial intrahepatic cholestasis type 2. Journal of hepatology. 2010 Nov;53(5):981–6.

111. Jara P, Hierro L, Martinez-Fernandez P, Alvarez-Doforno R, Yanez F, Diaz MC, et al. Recurrence of bile salt export pump deficiency after liver transplantation. The New England journal of medicine. 2009 Oct 1;361(14):1359–67.

112. Baghdasaryan A, Fuchs CD, Osterreicher CH, Lemberger UJ, Halilbasic E, Pahlman I, et al. Inhibition of intestinal bile acid absorption improves cholestatic liver and bile duct injury in a mouse model of sclerosing cholangitis. Journal of hepatology. 2016 Mar;64(3):674–81.

113. Miethke AG, Zhang W, Simmons J, Taylor AE, Shi T, Shanmukhappa SK, et al. Pharmacological inhibition of apical sodium-dependent bile acid transporter changes bile composition and blocks progression of sclerosing cholangitis in multidrug resistance 2 knockout mice. Hepatology (Baltimore, Md). 2016 Feb;63(2):512–23.

114. Soroka CJ, Mennone A, Hagey LR, Ballatori N, Boyer JL. Mouse organic solute transporter alpha deficiency enhances renal excretion of bile acids and attenuates cholestasis. Hepatology (Baltimore, Md). 2010 Jan;51(1):181–90.

115. Gonzales E, Grosse B, Cassio D, Davit-Spraul A, Fabre M, Jacquemin E. Successful mutation-specific chaperone therapy with 4-phenylbutyrate in a child with progressive familial intrahepatic cholestasis type 2. Journal of hepatology. 2012 Sep;57(3):695–8. 116. Gonzales E, Grosse B, Schuller B, Davit-Spraul A, Conti F, Guettier C, et al. Targeted

pharmacotherapy in progressive familial intrahepatic cholestasis type 2: Evidence for improvement of cholestasis with 4-phenylbutyrate. Hepatology (Baltimore, Md). 2015 Aug;62(2):558–66.

117. Gonzales E, Jacquemin E. Mutation specific drug therapy for progressive familial or benign recurrent intrahepatic cholestasis: a new tool in a near future? Journal of hepatology. 2010 Aug;53(2):385–7.

118. Hayashi H, Takada T, Suzuki H, Akita H, Sugiyama Y. Two common PFIC2 mutations are associated with the impaired membrane trafficking of BSEP/ABCB11. Hepatology (Baltimore, Md). 2005 Apr;41(4):916–24.

119. Kagawa T, Watanabe N, Mochizuki K, Numari A, Ikeno Y, Itoh J, et al. Phenotypic differences in PFIC2 and BRIC2 correlate with protein stability of mutant Bsep and impaired taurocholate secretion in MDCK II cells. American journal of physiologyGastrointestinal and liver physiology. 2008 Jan;294(1):58.

120. Clayton RJ, Iber FL, Ruebner BH, McKusick VA. Byler disease. Fatal familial intrahepatic cholestasis in an Amish kindred. American Journal of Diseases of Children (1960). 1969 Jan;117(1):112–24.

(26)

121. Andersen JP, Vestergaard AL, Mikkelsen SA, Mogensen LS, Chalat M, Molday RS. P4-ATPases as Phospholipid Flippases-Structure, Function, and Enigmas. Frontiers in physiology. 2016 Jul 8;7:275.

122. Paulusma CC, de Waart DR, Kunne C, Mok KS, Elferink RP. Activity of the bile salt export pump (ABCB11) is critically dependent on canalicular membrane cholesterol content. The Journal of biological chemistry. 2009 Apr 10;284(15):9947–54.

123. Folmer DE, van der Mark VA, Ho-Mok KS, Oude Elferink RP, Paulusma CC. Differential effects of progressive familial intrahepatic cholestasis type 1 and benign recurrent intrahepatic cholestasis type 1 mutations on canalicular localization of ATP8B1. Hepatology (Baltimore, Md). 2009 Nov;50(5):1597–605.

124. Verhulst PM, van der Velden LM, Oorschot V, van Faassen EE, Klumperman J, Houwen RH, et al. A flippase-independent function of ATP8B1, the protein affected in familial intrahepatic cholestasis type 1, is required for apical protein expression and microvillus formation in polarized epithelial cells. Hepatology (Baltimore, Md). 2010 Jun;51(6):2049– 60.

125. Ray NB, Durairaj L, Chen BB, McVerry BJ, Ryan AJ, Donahoe M, et al. Dynamic regulation of cardiolipin by the lipid pump Atp8b1 determines the severity of lung injury in experimental pneumonia. Nature medicine. 2010 Oct;16(10):1120–7.

126. Folvik G, Hilde O, Helge GO. Benign recurrent intrahepatic cholestasis: review and long-term follow-up of five cases. Scandinavian Journal of Gastroenterology. 2012 Apr;47(4):482–8.

127. Tygstrup N, Steig BA, Juijn JA, Bull LN, Houwen RH. Recurrent familial intrahepatic cholestasis in the Faeroe Islands. Phenotypic heterogeneity but genetic homogeneity. Hepatology (Baltimore, Md). 1999 Feb;29(2):506–8.

128. Stapelbroek JM, Peters TA, van Beurden DH, Curfs JH, Joosten A, Beynon AJ, et al.

ATP8B1 is essential for maintaining normal hearing. Proceedings of the National Academy

of Sciences of the United States of America. 2009 Jun 16;106(24):9709–14.

129. Nagasaka H, Yorifuji T, Kosugiyama K, Egawa H, Kawai M, Murayama K, et al. Resistance to parathyroid hormone in two patients with familial intrahepatic cholestasis: possible involvement of the ATP8B1 gene in calcium regulation via parathyroid hormone. Journal of pediatric gastroenterology and nutrition. 2004 Oct;39(4):404–9.

130. Mizuochi T, Kimura A, Tanaka A, Muto A, Nittono H, Seki Y, et al. Characterization of urinary bile acids in a pediatric BRIC-1 patient: effect of rifampicin treatment. Clinica chimica acta; international journal of clinical chemistry. 2012 Aug 16;413(15–16):1301–4. 131. Bustorff-Silva J, Sbraggia Neto L, Olimpio H, de Alcantara R v, Matsushima E, de

Tommaso AM, et al. Partial internal biliary diversion through a cholecystojejunocolonic anastomosis--a novel surgical approach for patients with progressive familial intrahepatic cholestasis: a preliminary report. Journal of pediatric surgery. 2007 Aug;42(8):1337–40. 132. Kalicinski PJ, Ismail H, Jankowska I, Kaminski A, Pawlowska J, Drewniak T, et al. Surgical

treatment of progressive familial intrahepatic cholestasis: comparison of partial external biliary diversion and ileal bypass. European journal of pediatric surgery : official journal of Austrian Association of Pediatric Surgery .[et al] = Zeitschrift fur Kinderchirurgie. 2003 Oct;13(5):307–11.

133. Clifton MS, Romero R, Ricketts RR. Button cholecystostomy for management of progressive familial intrahepatic cholestasis syndromes. Journal of pediatric surgery. 2011 Feb;46(2):304–7.

Referenties

GERELATEERDE DOCUMENTEN

Chapter 2 Prognosis of biliary atresia after 2-year survival with native liver: a nationwide cohort

From the Netherlands Study group on Biliary Atresia Registry (NeSBAR) database we included all patients with confirmed BA who reached the age of two years with their native liver..

Also, preterm infants with BA are treated relatively late in life and have poor post-operative outcome, regarding clearance of jaundice, need for LTx, and (transplant-free)

Proportion of BSEP deficiency patients alive with native liver over time in patients with homozygous (dotted line) or compound heterozygous (solid line) E297G

There is a complete absence of a systematic, dynamic and effective progress monitoring assessment system addressing the early literacy skills of foundation phase

H3: Voor zowel supermarkten als banken zorgen facebookberichten waarin gecommuniceerd wordt over een beloning voor meer engagement (liken, reageren en delen) dan berichten waarin

This dissertation focuses upon the detailed site investigation and site characterisation works that were undertaken by the author between 2014 and 2015 in order to inform and develop

The emergence of a voluminous legal corpus concerning the organisation of a transition from fossil and fissile fuels to renewable energy sources in France has led the author to ask