cryptogenic cholestasis in young and adults: atp8b1 ...download.xuebalib.com/1zt6uev62nfa.pdf · lp...

15
ORIGINAL ARTICLE—LIVER, PANCREAS, AND BILIARY TRACT Cryptogenic cholestasis in young and adults: ATP8B1, ABCB11, ABCB4, and TJP2 gene variants analysis by high-throughput sequencing Giovanni Vitale 1 Stefano Gitto 1 Francesco Raimondi 3,4 Alessandro Mattiaccio 2 Vilma Mantovani 2 Ranka Vukotic 1 Antonietta D’Errico 5 Marco Seri 1 Robert B. Russell 3,4 Pietro Andreone 1,6 Received: 25 October 2017 / Accepted: 4 December 2017 Ó Japanese Society of Gastroenterology 2017 Abstract Background Mutations in ATP-transporters ATPB81, ABCB11, and ABCB4 are responsible for progressive familial intrahepatic cholestasis (PFIC) 1, 2 and 3, and recently the gene for tight junction protein-2 (TJP2) has been linked to PFIC4. Aim As these four genes have been poorly studied in young people and adults, we investigated them in this context here. Methods In patients with cryptogenic cholestasis, we analyzed the presence of mutations by high-throughput sequencing. Bioinformatics analyses were performed for mechanistic and functional predictions of their conse- quences on biomolecular interaction interfaces. Results Of 108 patients, 48 whose cause of cholestasis was not established were submitted to molecular analysis. Pathogenic/likely pathogenic mutations were found in ten (21%) probands for 13 mutations: two in ATP8B1, six in ABCB11, two in ABCB4, three in TJP2. We also identified seven variants of uncertain significance: two in ATP8B1, one in ABCB11, two in ABCB4 and two in TJP2. Finally, we identified 11 benign/likely benign variants. Patients with pathogenic/likely pathogenic mutations had higher levels of liver stiffness (measured by FibroScan Ò ) and bile acids, as well as higher rates of cholestatic histological features, compared to the patients without at least likely pathogenic mutations. The multivariate analysis showed that itching was the only independent factor associated with disease-causing mutations (OR 5.801, 95% CI 1.244–27.060, p = 0.025). Conclusions Mutations in the genes responsible for PFIC may be involved in both young and adults with cryptogenic cholestasis in a considerable number of cases, including in heterozygous status. Diagnosis should always be suspected, particularly in the presence of itching. Keywords Progressive familial intrahepatic cholestasis Á Cryptogenic disease Á Pathogenic mutations Á Genetic variants Á Bioinformatics analysis Abbreviations PFIC Progressive familial intrahepatic cholestasis TJP2 Tight junction protein-2 FIC1 Familial intrahepatic cholestasis 1 BSEP Bile salt export pump MDR Multidrug resistance P-glycoprotein 3 Electronic supplementary material The online version of this article (https://doi.org/10.1007/s00535-017-1423-1) contains supple- mentary material, which is available to authorized users. & Pietro Andreone [email protected] 1 Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy 2 Center for Applied Biomedical Research (CRBA), University Hospital, Bologna, Italy 3 CellNetworks, Bioquant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany 4 Bioochemie Zentrum Heidelberg (BZH), Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany 5 Addari Institute of Oncology and Transplant Pathology, Policlinico S. Orsola-Malpighi, University of Bologna, Bologna, Italy 6 Department of Medical and Surgical Sciences and Research Center for the Study of Hepatitis, University of Bologna, Italy, Via Massarenti 9, 40138 Bologna, Italy 123 J Gastroenterol https://doi.org/10.1007/s00535-017-1423-1

Upload: others

Post on 23-Oct-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

  • ORIGINAL ARTICLE—LIVER, PANCREAS, AND BILIARY TRACT

    Cryptogenic cholestasis in young and adults: ATP8B1, ABCB11,ABCB4, and TJP2 gene variants analysis by high-throughputsequencing

    Giovanni Vitale1 • Stefano Gitto1 • Francesco Raimondi3,4 • Alessandro Mattiaccio2 •

    Vilma Mantovani2 • Ranka Vukotic1 • Antonietta D’Errico5 • Marco Seri1 •

    Robert B. Russell3,4 • Pietro Andreone1,6

    Received: 25 October 2017 / Accepted: 4 December 2017

    � Japanese Society of Gastroenterology 2017

    Abstract

    Background Mutations in ATP-transporters ATPB81,

    ABCB11, and ABCB4 are responsible for progressive

    familial intrahepatic cholestasis (PFIC) 1, 2 and 3, and

    recently the gene for tight junction protein-2 (TJP2) has

    been linked to PFIC4.

    Aim As these four genes have been poorly studied in

    young people and adults, we investigated them in this

    context here.

    Methods In patients with cryptogenic cholestasis, we

    analyzed the presence of mutations by high-throughput

    sequencing. Bioinformatics analyses were performed for

    mechanistic and functional predictions of their conse-

    quences on biomolecular interaction interfaces.

    Results Of 108 patients, 48 whose cause of cholestasis was

    not established were submitted to molecular analysis.

    Pathogenic/likely pathogenic mutations were found in ten

    (21%) probands for 13 mutations: two in ATP8B1, six in

    ABCB11, two in ABCB4, three in TJP2. We also identified

    seven variants of uncertain significance: two in ATP8B1,

    one in ABCB11, two in ABCB4 and two in TJP2. Finally,

    we identified 11 benign/likely benign variants. Patients

    with pathogenic/likely pathogenic mutations had higher

    levels of liver stiffness (measured by FibroScan�) and bile

    acids, as well as higher rates of cholestatic histological

    features, compared to the patients without at least likely

    pathogenic mutations. The multivariate analysis showed

    that itching was the only independent factor associated

    with disease-causing mutations (OR 5.801, 95% CI

    1.244–27.060, p = 0.025).

    Conclusions Mutations in the genes responsible for PFIC

    may be involved in both young and adults with cryptogenic

    cholestasis in a considerable number of cases, including in

    heterozygous status. Diagnosis should always be suspected,

    particularly in the presence of itching.

    Keywords Progressive familial intrahepatic cholestasis �Cryptogenic disease � Pathogenic mutations � Geneticvariants � Bioinformatics analysis

    Abbreviations

    PFIC Progressive familial intrahepatic cholestasis

    TJP2 Tight junction protein-2

    FIC1 Familial intrahepatic cholestasis 1

    BSEP Bile salt export pump

    MDR Multidrug resistance P-glycoprotein 3

    Electronic supplementary material The online version of thisarticle (https://doi.org/10.1007/s00535-017-1423-1) contains supple-mentary material, which is available to authorized users.

    & Pietro [email protected]

    1 Department of Medical and Surgical Sciences, University of

    Bologna, Bologna, Italy

    2 Center for Applied Biomedical Research (CRBA), University

    Hospital, Bologna, Italy

    3 CellNetworks, Bioquant, Heidelberg University, Im

    Neuenheimer Feld 267, 69120 Heidelberg, Germany

    4 Bioochemie Zentrum Heidelberg (BZH), Heidelberg

    University, Im Neuenheimer Feld 328, 69120 Heidelberg,

    Germany

    5 Addari Institute of Oncology and Transplant Pathology,

    Policlinico S. Orsola-Malpighi, University of Bologna,

    Bologna, Italy

    6 Department of Medical and Surgical Sciences and Research

    Center for the Study of Hepatitis, University of Bologna,

    Italy, Via Massarenti 9, 40138 Bologna, Italy

    123

    J Gastroenterol

    https://doi.org/10.1007/s00535-017-1423-1

    http://orcid.org/0000-0002-4794-9809https://doi.org/10.1007/s00535-017-1423-1http://crossmark.crossref.org/dialog/?doi=10.1007/s00535-017-1423-1&domain=pdfhttp://crossmark.crossref.org/dialog/?doi=10.1007/s00535-017-1423-1&domain=pdfhttps://doi.org/10.1007/s00535-017-1423-1

  • GGT Gamma-glutamyl-transpeptidase

    AP Alkaline phosphatase

    BRIC Benign intrahepatic cholestasis

    LPAC Low-phospholipid-associated cholelithiasis

    ICP Intrahepatic cholestasis of pregnancy

    DIC Drug-induced cholestasis

    HTS High-throughput sequencing

    NGS Next-generation sequencing

    PSC Primary sclerosing cholangitis

    BA Bile acids

    MAF Minor allele frequency

    SIFT Sorting Intolerant From Tolerant

    HGMD Human Gene Mutation Database

    ACMG American College of Medical Genetics and

    Genomics

    P Pathogenic

    LP Likely pathogenic

    VUS Variants of uncertain significance

    LB Likely benign

    B Benign

    SD Standard deviation

    CI Confidence interval

    SNP Single-nucleotide polymorphism

    ALT Alanine aminotransferase

    OR Odds ratio

    Introduction

    Progressive familial intrahepatic cholestasis (PFIC) is a

    group of autosomal recessive cholestatic diseases that

    affects especially newborns and children, and represents a

    consolidated indication for liver transplantation [1]. These

    disorders are rare and unavoidably progressive to liver

    cirrhosis and portal hypertension. However, their incidence

    is hard to establish because of difficulties in diagnosis [1].

    Mutations in four genes have been linked to PFIC. For

    instance, mutations in ATP8B1, which encodes the

    aminophospholipid flippase familial intrahepatic cholesta-

    sis-1 protein (FIC1), are responsible for PFIC1 [2]. Muta-

    tions in ABCB11, encoding a protein that functions as a bile

    salt export pump (BSEP), are responsible for the PFIC2 [3].

    Mutations in ABCB4, which codes for the multidrug

    resistance P-glycoprotein 3 (MDR3), a flippase that medi-

    ates the outflow of phosphatidylcholine into the bile, are

    linked to PFIC3 [4]. Here, levels of gamma-glutamyl-

    transferase (GGT) are always increased [5], in contrast to

    very low values that characterize PFIC1 and PFIC2, in

    which high levels of alkaline phosphatase (AP) are instead

    present. Mutations in a fourth gene, TJP2 (coding for tight

    junction protein-2) were linked to intrahepatic cholestasis

    with low GGT (PFIC4) [6].

    Variants in the same genes cause several other liver

    diseases. Benign intrahepatic cholestasis (BRIC) is linked

    to mutations in ATP8B1 and ABCB11, characterized by

    intermittent cholestasis without progression to cirrhosis.

    Low-phospholipid-associated cholelithiasis (LPAC) is

    related to mutations in ABCB4 with symptomatic intra-

    hepatic lithiasis. Intrahepatic cholestasis of pregnancy

    (ICP) is a reversible pregnancy-specific cholestasis char-

    acterized by pruritus, elevated liver enzymes, and increased

    serum bile acids involving mutations in ATP8B1, ABCB11,

    and ABCB4. Mutations in ABCB11 and ABCB4 are asso-

    ciated with drug-induced cholestasis (DIC), a disorder

    induced by certain drugs. These liver diseases are often

    associated to heterozygous mutations [7, 8].

    Only heterozygous status for ABCB4 mutations has been

    well studied in children with cholestatic liver disease

    [9, 10] while Dröge and colleagues [11] recently revealed a

    high number of different genetic variants by sequencing

    FIC1, BSEP, and MDR3 in a large cohort of patients with

    supposed genetic cholestasis.

    To date, no authors have investigated the four genes

    linked to FIC in young persons and adults, with and

    without progressive forms of liver failure.

    High-throughput sequencing (HTS), including next-

    generation sequencing (NGS), is a technology proposed for

    the molecular diagnosis of PFIC, based on the massive

    parallel sequencing of specific genomic loci, whole exome

    or genomes. Compared to classic Sanger, NGS allows rapid

    sequencing with more information at lower costs

    [6, 12, 13].

    We aimed to develop a targeted NGS panel to investi-

    gate the presence of mutations in ATP8B11, ABCB11,

    ABCB4, and TJP2 in a population with cryptogenic

    cholestasis and related them to the corresponding pheno-

    types and risk factors.

    Materials and methods

    Patients

    From May 2013 to November 2016, all outpatients with

    cholestatic disease aged[ 6 years were enrolled consecu-tively at the Department of Medical and Surgical Sciences,

    a tertiary Italian referral center. We excluded other causes

    of cholestasis as follows; primary biliary cholangitis, pri-

    mary sclerosing cholangitis (PSC), overlap syndromes,

    IgG4-cholangiopathy and obstructive jaundice were

    excluded by demonstration of a normal anatomy of the

    biliary tree and negative specific serological tests. Viral

    hepatitis, alcohol abuse, hemochromatosis, Wilson disease,

    J Gastroenterol

    123

  • alfa1-antitrypsin deficiency were excluded too. Chronic

    idiopathic cholestasis was defined as GGT e/o AP persis-

    tently C 1.5-fold the upper normal values in at least two

    tests or as history of itching combined with elevated serum

    bile acids (BA) concentration ([ 10 lmol/l) for more than6 months. This study was conducted in accordance with

    ethical guidelines of the World Medical Association’s

    Declaration of Helsinki and patients or their legal guar-

    dians provided written informed consent. Subjects affected

    by cryptogenic cholestasis underwent laboratory analysis,

    including BA serum concentration, liver fibrosis evaluation

    by transient elastography (FibroScan�) and, if indicated,

    liver biopsy within 6 months from the execution of the

    genetic tests. Histological bile duct alteration was defined

    as lobular cholestasis (ductal hepatocyte metaplasia, ductal

    proliferation and immunohistochemistry for bile duct

    cytokeratin 7, anti-BSEP, and anti-MDR3 antibodies). An

    expert senior pathologist performed the liver histology.

    Clinical variables considered were history of DIC or

    itching, family history of cholestasis, personal or family

    history of ICP, neonatal jaundice, juvenile cholelithiasis

    (defined as history of gallstones\ 40 years).We compared patients with disease-causing variants

    with the remaining population with idiopathic cholestasis.

    NGS analysis

    We extracted DNA from peripheral blood using the Max-

    well 16 blood DNA purification kit (Promega, Madison,

    WI, USA). Comprehensive molecular analysis of ABCB11,

    ATP8B1, ABCB4, and TJP2 genes was performed by

    multiplex targeted amplicon-based sequencing approach

    using the Ion Torrent technology (Thermo Fisher Scien-

    tific, Waltham, MA, USA). Primers were designed by Ion

    AmpliSeqTM Designer tool to cover the entire coding

    region of the four genes plus 50 bp of intronic flanking

    regions. Target coverage of 98.9% was obtained through

    194 amplicons for 42 kb. Libraries were performed by

    AmpliseqTM Library Kit 2.0 and concentrations were

    evaluated by the Ion Library TaqManTM quantitation kit

    using real-time PCR. Emulsion-PCR was performed by Ion

    PGMTM Hi-Q OT2 kit and the enrichment was realized by

    Ion OneTouchTM ES. NGS was performed by the Ion

    Torrent PGMTM System according to the manufacturer’s

    procedures (Life Technologies, CA, USA). Molecular

    analysis was simultaneously carried out in 12 barcoded

    samples in an Ion 318 Chip v2 for each run.

    The read files obtained from sequencing were mapped to

    the GRCh37/hg19 assembly and the sequence variants

    were identified by Variant Caller and Ion Reporter

    software.

    Allelic variants were reported according to Human

    Genome Variation Society guidelines (http://www.hgvs.

    org/content/guidelines). NGS uncovered regions as well as

    the potential pathogenic variants were confirmed by Sanger

    sequencing using ABI PRISM 3730 Genetic Analyzer

    (Thermo Fisher Scientific, Waltham, MA, USA).

    Prediction of functional consequences of variants

    and classification

    The filtering step of sequence variants was carried out

    according to default criteria for germline mutations:

    insertions or deletions, non-sense variants, splicing-site

    variants, and missense with minor allele frequency

    (MAF) B 0.05 were considered. Three bioinformatics tools

    were used to predict the role of missense variants. Sorting

    Intolerant From Tolerant (SIFT) (http://www.jcvi.org/cms/

    home/), PolyPhen-2 [14] and Mutation Taster [15] software

    can evaluate whether an amino-acid substitution influences

    protein structure and function, according to physical

    modifications and the degree of conservation of protein

    sequence among species. Variants that meet at least one of

    the following criteria were also considered: ever described

    before, reported in Human Gene Mutation Database

    (HGMD), predicted not benign by at least one of the

    bioinformatic tools. According to American College of

    Medical Genetics and Genomics (ACMG) standards [16],

    we classified the filtered variants into five categories:

    pathogenic (P); likely pathogenic (LP); variants of uncer-

    tain significance (VUS); likely benign (LB); and benign

    (B).

    Variants passing the above filtering process were map-

    ped to Uniprot canonical sequences and subjected to

    Mechismo [17] analysis to predict their functional conse-

    quences at biomolecular interaction interfaces. The

    approach matches protein sequence amino-acids to posi-

    tions within structures and identifies sites affecting inter-

    actions with other proteins, DNA/RNA or small molecules.

    We considered low confidence predictions including

    known structures or close (C 30% sequence identity)

    homologs and only very confident, physical protein–pro-

    tein interactions (as defined by Mechismo based on a

    benchmark for the accuracy of perturbed interfaces). In

    case of ATP8B1, no homolog template structure was

    available in Mechismo, so we used a homology model rom

    ModBase [18].

    We annotated mutations and PTMs (phosphorylations

    and acetylations) from Phosphosite [19] of the considered

    genes on corresponding Uniprot canonical sequences using

    lollipop diagrams [20].

    Comparison with an international database

    Allele frequency (AF) of common single-nucleotide poly-

    morphism (SNPs) was matched with data reported in the

    J Gastroenterol

    123

    http://www.hgvs.org/content/guidelineshttp://www.hgvs.org/content/guidelineshttp://www.jcvi.org/cms/home/http://www.jcvi.org/cms/home/

  • international Genome Aggregation Database (gnomAD),

    Cambridge, MA (http://gnomad.broadinstitute.org/;37)

    [allele frequencies accessed October 2017]. We compared

    AF of SNPs in our cohort (with and without P/LP mutation)

    to AFs of European (non-Finnish), East Asian, and

    worldwide population.

    Statistical analysis

    Categorical variables are expressed as number (%), and

    quantitative variables as mean ± standard deviation or as

    median (range). Chi-square or Fisher’s exact test was used

    to compare categorical variables, while for quantitative

    variables the t test or Mann–Whitney’s test (unpaired data)

    or the t test or Wilcoxon’s test (paired data) were used.

    Binary logistic regression was performed for univariate and

    multivariate analyses to identify predictors of causative

    variants (variables were included if p\ 0.1 and removed ifp C 0.05). A p\ 0.05 was considered significant for alltests. The statistical software SPSS version 21.0 (�SPSS

    Inc., Chicago, IL, USA) was used for statistical analyses.

    Results

    Patient characteristics

    We evaluated consecutively 108 cholestatic patients and 48

    fulfilled the established criteria (Supplementary Fig. 1).

    We listed the main laboratory patterns in Table 1. There

    was a slight bias for males (58%) and mean age at time of

    the genetic test was 42 years; the adult population

    (C 18 years) was 93.8%. A history of familial cholestatic

    diseases and DIC was present in 35%, itching in 27%,

    neonatal jaundice in 21%, juvenile cholelithiasis in 17%,

    personal or family history of ICP in 13%. Histologic fea-

    tures of cholestasis were present in 65% of cases (overall

    44 patients agreed to a liver biopsy). Gene analysis

    revealed the presence of P/LP mutations in about one-

    fourth of subjects; clinical features and type of mutations

    found in the 11 patients (ten unrelated probands and one

    affected sister) were reported in Table 2.

    NGS results and variants

    NGS protocol provided an average sequencing depth

    of * 10009, 98.78% of reads having 209 coverage, with99% reads on target and a coverage uniformity of 95.98%.

    Thirty-one variants that satisfied our filtering criteria are

    reported in Supplementary Table 1. Among these, 13 were

    P/LP mutations, five were previously undescribed: three

    were missense, one was a frameshift, and one was a non-

    sense mutation.

    Among P/LP variants, two were in ATP8B1, six in

    ABCB11, two in ABCB4 and three in TJP2. We identified

    seven VUS, two in each of ATP8B1, ABCB4 and TJP2, one

    in ABCB11. Finally, we recorded 11 benign mutations, two

    in each of ATP8B1, ABCB11 and ABCB4, five in TJP2.

    ATP8B1 variants

    Six variants were identified in the coding region of

    ATP8B1, reported below; according to ACMG standards,

    two LP: c.3655G[C (p.D1219H) and c.68C[T (p.P23L);the first was combined with heterozygous LP mutation

    c.1057C[T on TJP2 gene in a 71-year-old woman (case 1).The second is a novel mutation found in a 31-year-old male

    (case 2) who experienced neonatal jaundice and juvenile

    cholelithiasis, with high GGT and bilirubin levels. We

    classified the missense c.134A[C (p.N45T) and c.607A[G(p.K203E) as VUS and each was previously described as

    risk allele in ICP [21]. We found these variants in com-

    pound heterozygous in a 36-year-old female who showed a

    Table 1 Baseline features of the entire study population

    N 48

    Male N (%) 28 (58.3)

    Adult population C 18 years N (%) 45 (93.8)

    Age (years, mean ± SD)

    At time of genetic test 42 ± 15

    At presentation 32 ± 15

    Risk factor for cholestasis N (%)

    DIC history 17 (35.4)

    Neonatal jaundice 10 (20.8)

    Itching history 13 (27.1)

    ICP history 6 (12.5)

    Juvenile cholelithiasis 8 (16.7)

    Familiarity 17 (35.4)

    Laboratory (median, range)

    GGT (UI/l) 139 (5–597)

    FA (UI/l) 283 (122–1012)

    ALT (UI/l) 45 (9–387)

    Bilirubin (mg/dl) 0.7 (0.4–11.2)

    Bile acids (lmol/l) 11 (2.3–403)

    Cholesterol (mg/dl) 211 (87–328)

    Albumin (g/dl) 4.3 (2.9–5.2) 4.3 (2.9–5.2)

    Platelets (n 9 103/ll) 229 (61–417)

    FibroScan (kPa) (median, range) 5.3 (3.1–35.8)

    Histologic features of cholestasis N (%) 31 (64.6)

    N number, SD standard deviation, DIC drug-induced cholestasis, ICP

    intrahepatic cholestasis of pregnancy, GGT gamma-glutamyltrans-

    ferase, AP alkaline phosphatase, ALT alanine transaminases, kPa

    kiloPascal

    J Gastroenterol

    123

    http://gnomad.broadinstitute.org/%3b37

  • Table 2 Baseline features of the population with likely pathogenic (LP) and pathogenic (P) mutations

    ID Sex LP and P mutations Additional variants Age

    (years)

    GGT

    (U(L)

    PA

    (U/l)

    Bilirubin

    (mg/dl)

    FibroScan

    (kPa)

    Notes

    1_17421 F ATP8B1:

    p.[D1219H]; [=]

    TJP2:

    p.[R322W]; [=]

    ATP8B1:

    p.[R952Q]; [=]

    ABCB11:

    p.[V444A(;)M677V]

    TJP2:

    p.[M668I]; [=]

    71 121 394 1.2 5.5 No affected relatives

    Segregation not

    assessed

    2_15710 M ATP8B1:

    p.[P23L]; [=]

    ABCB11:

    p.[V444A(;)M677V]

    31 325 138 3.3 7.4 Familiarity

    Juvenile

    cholelithiasis

    Neonatal jaundice

    The mutation was

    inherited from the

    mother and

    segregates in both

    brothers and in a

    sister

    3_16545 F ABCB11:

    p.[Y93S(;)V597L(;)R1128]

    ABCB11:

    p.[V444A]; [=]

    ABCB4:

    p.[I237=]; [=]

    29 5 466 1.2 12 DIC

    Neonatal jaundice

    Itching

    Juvenile

    cholelithiasis

    ICP

    Segregation not

    assessed

    4_15502 F ABCB11:

    p.[A523G]; [A523G]

    ABCB11:

    p.[V444A];

    [V444A]

    ATP8B1:

    p.[R952Q]; [=]

    ABCB4:

    p.[I237=]; [=]

    20 15 672 11.2 20.9 Familiarity

    DIC

    Neonatal jaundice

    itching

    The mutations are

    inherited from

    healthy parents

    4a_15505 F ABCB11:

    p.[A523G]; [A523G]

    ABCB11:

    p.[V444A];

    [V444A]

    ABCB4:

    p.[I237=]; [=]

    7 13 1012 3.5 10.1 Familiarity

    DIC

    Neonatal jaundice

    itching

    Affected sister of

    case 4

    The mutations

    segregate in both

    affected sisters

    5_15507 M ABCB11:

    p.[E135K]; [S1100Qfs*38]

    ABCB11:

    p.[V444A];

    [V444A]

    16 8 467 2.2 11.5 Neonatal jaundice

    Itching

    The mutations are

    inherited from

    healthy parents

    6_17248 F ABCB4:

    p.[K672*]; [=]

    ABCB11:

    p.[V444A];

    [V444A]

    ABCB4:

    p.[I237=]; [=] TJP2:

    p.[T1124=]; [=]

    39 363 562 0.4 7.7 Familiarity

    DIC

    Itching

    The mutation was

    inherited from the

    affected father

    J Gastroenterol

    123

  • cholestatic disease characterized by normal GGT and high

    BA levels, neonatal jaundice, juvenile cholelithiasis com-

    plicated by LPAC, and a history of ICP.

    We found two B/LB variants: c.913T[A (p.F305I) andc.2855G[A (p.R952Q). The SNP p.R952Q was found infive cases and in two patients was combined with LP

    mutations, one on ABCB11 and one on TJP2, respectively.

    AF of p.R952Q was significantly higher in comparison

    with AF of East Asian population (10.4 vs. 0.03%:

    p\ 0.0001) and comparable with European and worldwidedata (Table 3).

    Analysis of variants in the context of sequence and

    structural annotations on structures or homology models

    revealed that two of them, i.e., p.K203E and p.F305I are

    found in spatial proximity within the ATPase catalytic

    domain (Fig. 1a), suggesting potential converging effect on

    the enzymatic activity, while the p.R952Q is found at the

    transmembrane (i.e., phospholipid translocating) domain.

    The LP variants, p.P23L and p.D1219H, reside at the N-

    and C-terminals of the protein, outside the structured por-

    tion (Fig. 1b).

    ABCB11 variations

    Nine variants were identified in the coding region of

    ABCB11 and six resulted in P/LP mutations: three causa-

    tive mutations, c.278A[C (p.Y93S), c.1789G[C

    (p.V597L) and c.3382C[T (R1128C) were combined in ayoung female (29 years) presenting with a history of

    neonatal jaundice, DIC, ICP, juvenile cholelithiasis, itch-

    ing, normal GGT levels (case 3). Two sisters (20 and

    7 years) were carriers of the LP homozygous mutation

    c.1568C[G (p.A523G): both had history of neonataljaundice, itching and DIC, (cases 4–4a). Last two P/LP

    mutations, c.3297delC (p.S1100Qfs*38) and c.403G[A(p.E135K), were responsible for a double heterozygosis in

    a boy with history of itching and neonatal jaundice, listed

    for liver transplantation (case 5); c.3297delC was recently

    described [22] while c.403G[A mutation was linked byAnzivino et al. to ICP conditions [23]. The VUS

    c.1268A[G (p.H423R) was found in a 43-year-old womanwho presented only high GGT without fibrosis.

    Benign mutations detected were among those previously

    described [7, 11, 24]: c.2029A[G (p.M677V), detected innine patients, and c.1331T[C (p.V444A), found in 40cases (83.3% of population studied). Of note, SNP

    c.1331T[C was reported associated to DIC and ICP inprevious reports [7, 24] and recently to cholestatic phe-

    notype in patients without disease-causing mutations in the

    respective gene [11]. AF of p.V444A was significantly

    more frequent in our cohort of patients without P/LP

    mutations in comparison with European and worldwide

    populations (81 vs. 60%: p = 0.008; 81 vs. 57%:

    p = 0.002). We also observed a higher AF of p.M677V in

    Table 2 continued

    ID Sex LP and P mutations Additional variants Age

    (years)

    GGT

    (U(L)

    PA

    (U/l)

    Bilirubin

    (mg/dl)

    FibroScan

    (kPa)

    Notes

    7_15873 M ABCB4:

    p.[A364V]; [=]

    ABCB4:

    p.[I237=]; [=] TJP2

    gene:

    p.[Q128K(;)M668I]

    57 379 606 0.4 35.8 DIC

    HCC

    No affected relatives.

    Segregation not

    assessed

    8_17362 F TJP2:

    p.[T62M]; [=]

    ABCB11:

    p.[V444A(;)M677V]

    ABCB4:

    p.[I237=]; [I237=]

    51 84 205 0.6 4.4 DIC

    ICP

    No affected relatives.

    Segregation not

    assessed

    9_17374 M TJP2:

    p.[T62M]; [=]

    ABCB11:

    p.[V444A(;)M677V]

    50 130 175 0.6 3.3 No affected relatives.

    Segregation not

    assessed

    10_16643 M TJP2:

    p.[I875T]; [=]

    ABCB11:

    p.[V444A];

    [V444A]

    ABCB4:

    p.[T775M]; [=]

    TJP2:

    p.[R12H]; [R12H]

    37 217 399 0.6 8.1 Familiarity

    Both mutations

    segregate in an

    affected brother

    with history of high

    GGT

    ID 4 and 4a are affected sibling. Sequence variants are reported according to HGVS recommendations (http://varnomen.hgvs.org)

    J Gastroenterol

    123

    http://varnomen.hgvs.org

  • our cohort with and without P/LP mutations respect to all

    other populations (13.5 vs. 1.76% European, 0% East

    Asian, 2.7% worldwide: all p\ 0.0001) (Table 3).All nine reported mutations for ABCB11 were mapped

    to homolog structures through Mechismo (Fig. 1a). P/LP

    variants were found to be located either at ATP-binding

    domain, like p.A523G, p.V597L, p.S1100fs and p.R1128C,

    or at transmembrane domain, like p.E135K or p.Y93S,

    which are also found in spatial proximity (Fig. 1b).

    Moreover, the variant p.H423R was found at the interface

    with the ATP-binding pocket (Supplementary Table 2).

    This overall suggests that deleterious mutations on

    ABCB11 might similarly affect the ATP-binding cassette

    (ABC) transporter’s functionality by impairing either ABC

    catalytic activity, which couples ATP-hydrolysis to

    transport, or by perturbing transmembrane region, which

    mediates ligands transport to the extracellular side [25].

    ABCB4 variations

    We identified six variants in the coding region of ABCB4

    and two were P/LP mutations. Disease-causing variants

    were: c.2014A[T (p.Lys672*), previously undescribed,was found in a 39-year-old woman with familiarity for

    cholestatic diseases (mutation was inherited from the

    affected father with high GGT), history of DIC and itching

    (case 6) while c.1091C[T (p.A364V), described byDegiorgio et al. [26], was present in a 57-year-old cirrhotic

    patient with DIC and hepatocellular carcinoma (case 7).

    Two VUS, p.L73V (c.217C[G) and c.2324C[T

    Table 3 Allele frequencies of detected SNPs

    Gene Mutation Protein dbSNPs AF full

    cohort

    (a) (%)

    AF Pts w/o

    P/LP

    mutations

    (b) (%)

    AF

    EU

    (1)

    (%)

    AF East

    Asia (2)

    (%)

    AF

    world

    (3) (%)

    p values f

    ATP8B1 c.2855G[A p.R952Q rs12968116 10.4 10.8 12 0.03 8.3 a-1 = 0.9151 b-1 = 0.8297a-2 < 0.0001 b-2 < 0.0001

    a-3 = 0.5888 b-3 = 0.5744

    ABCB11 c.1331T[C p.V444A rs2287622 83 81 60 73 57 a-1 < 0.0001 b-1 = 0.008a-2 = 0.1329 b-2 = 0.3318

    a-3 =

  • E135K Y93S

    A523GH423RV444A V597L

    M677V R1128C

    S1100Qfs

    Pholip_ATPase

    ATP8B1 ABCB4ABCB11

    cytosol

    extracellular

    ABC_tran

    PDZ

    PDZ

    PDZ

    SH3_2Guanylate_kin

    TJP2

    ATP8B1P23

    LN45

    TK20

    3EF30

    5IR95

    2Q D1219H

    PhoLip_ATPase E1-E2 ATPase Cation_ATPase Phospholipid-translocating P-type ATPase C-terminal

    ABCB11Y93

    SE13

    5KH42

    3R V444A

    A523G

    V597L

    M677V

    S1100Q

    fs*38R11

    28C

    ABC_membrane ABC_tran ABC_membrane ABC_tran

    0 93 135 371 423 523 586 677 755 858 1029 1100 12141247 1321

    ABCB4L73

    V A36

    4V R652G L67

    2*

    T775M

    ABC_membrane ABC_tran ABC_membrane ABC_tran

    0 73 121 237 364 424 524 561 624 672 711 775 985 1052 1210 1286

    TJP2

    V3LR12H

    T62M Q12

    8KR32

    2W

    M668I

    I875T

    PDZ PDZ PDZ SH3_2 Guanylate_kin

    3 33 62 91 319 668 875 1190

    2338 66 145 172 203 305 413 440 476 532 632 658667 779789 902924952 992 11731203 12170

    R4E

    (L)B or VUS(L)PPhosphorylation siteAcetylation site

    R952Q

    K203E

    F305IE1_E2_ATPase

    (P23L)(N45T)

    (D1219H)

    ABC_membraneL73V

    A364V

    R652G

    T775M

    (K672*)

    T62M

    Q128K

    R322W M668I

    I875T

    A256V

    (V3L)(R4E)(R12H)

    (A256V)

    (a)

    (b)

    J Gastroenterol

    123

  • (p.T775M), were identified, previously associated to dif-

    ferent PFIC3 phenotypes [10, 26].

    The missense variant p.T775M was detected together

    with LP c.2717T[C (p.I906T) in exon 18 of TJP2 in a37-year-old male presenting a familiarity for cholestasis

    (case 3), while the variant p.L73V was detected in a

    23-year-old female with high GGT levels, without evi-

    dence of significant liver fibrosis at FibroScan�. The

    known LB c.711A[T (p.I237=) [11, 27] and c.1954A[G(p.R652G) [11, 28] mutations were found in 16 and four

    cases, respectively. The SNP p.I237= was associated in

    previous reports with elevated GGT-cholestasis, ICP, and

    gallstones [11].

    AF of p.I237= was significantly more common in our

    cohort of patients with disease-causing mutations than in

    European and worldwide population (33 vs. 18%:

    p = 0.004; 33 vs. 21%: p = 0.033; Table 3).

    AF of p.R652G was similar between our cohort and both

    European and worldwide population while it appeared to

    be more common in East Asian subgroup (10.8 vs. 27%;

    p = 0.00106).

    Similarly to ABCB11, ABCB4 non-synonymous muta-

    tions were mapped to homolog structures with exception of

    the stop-gain p.L672*. Mutations were essentially located

    in transmembrane region (Fig. 1a), suggesting a perturba-

    tion of ligand transport mechanisms. The stop-gain

    p.L672*, located in the middle of the gene (Fig. 1b), would

    result in a halved transcript and protein sequence, lacking

    one ATP-binding cassette and one transmembrane domain,

    further stressing its high severity.

    TJP2 variations

    Variants identified in the coding region of TJP2 were ten:

    three LP mutations, two VUS, and five B/lB variants.

    The LP mutations were: c.278C[T (p.T62M) in twocases (cases 8 and 9), one who experienced DIC and ICP,

    c.1057C[T (p.R322W), combined to LP heterozygousp.D1219H on ATP8B1 in a 71-year-old man presenting

    only high GGT (case 1), and c.2717T[C (p.I875T) asso-ciated with VUS p.T775M in ABCB4 in one patient with

    familiarity for cholestatic diseases (case 10).

    Differently from what has been reported in a previous

    report [6], all three patients presented high GGT. Two VUS

    were c.43G[C (p.V3L) and c.46A[G (p.R4E) while B/LBmutations were c.71G[A (p.R12H), c.475C[A (p.Q128K),c.860C[T (p.A256V), c.2097G[A (p.M668I), andc.3465G[AT (p.T1124=).

    VUS p.R4E was present in a 31-year-old female asso-

    ciated to benign C1954A[G and C711A[T mutations inABCB4: the patient had a history of ICP, juvenile

    cholelithiasis and recurrent DIC to different drugs used for

    multiple sclerosis. Finally, a 31-year-old man with high

    GGT and BA levels presented another VUS p.V3L.

    SNP p.M668I was significantly more frequent in our

    cohort of patients with disease-causing mutation compared

    to European, East Asian, and worldwide population (14.6

    vs. 6.3%: p = 0.0385, 0.03%: p\ 0.001, and 5.3%:p = 0.0043) (Table 3).

    Most deleterious TJP2 mutations were found at the level

    of PDZ, SH3, and guanylate kinase domains (Fig. 1a, b).

    This suggests a potential perturbation of TJP2’s function,

    i.e., organizing tight and adherent junctions by binding to

    the cytoplasmic C termini of junctional transmembrane

    proteins and linking them to the actin cytoskeleton [29].

    Indeed, one variant (p.R322W) is predicted to affect the

    homo- and hetero-dimerization interface with TJP1

    (Fig. 2).

    Summary mutation profile for FIC patients

    Excluding the common SNP V444A on ABCB11, (present

    in 83.3% of cases) and I237= on ABCB4 (33%), 26/48

    patients (58.3%), have at least one mutation in one or more

    genes: eight patients had only one mutated allele while 18

    patients had two or more mutated alleles. ABCB11 and

    TJP2 resulted most affected genes and they had mutual

    exclusive but non-significant tendency. In particular,

    ABCB11/M677V, TJP2/M668I, TJP2/Q128K, and TJP2/

    R12H had a tendency to affect patients in a mutual

    exclusive way between each other (but in combination,

    within the same patient, with various mutations). Figure 3

    summarizes mutations discovered contemporary, showing

    allele variants in the four genes, according to pathogenicity

    prediction (P/LP vs. B, LB, and VUS mutations), zygosity,

    and the clinical data (age, FibroScan�, GGT, neonatal

    jaundice, itching history, bile acids and cholestasis at

    histology).

    It is possible that patients with multiple alleles mutated

    have more severe phenotypes by a synergistic effect at

    different bile transporters’ sites as predicted by Mechismo.

    However, patients having C 2 non-synonymous mutations

    (with at least one predicted P/LP), tend to have higher liver

    stiffness comparing to patients with\ 2 non-synonymousmutations (kPa, 7.9 [3.3–35.8] vs. 4.9 [3.1–29.9],

    bFig. 1 a Protein sequence annotations of FIC-selected non-synony-mous mutations displayed as pink and red lollipops to indicate

    respectively B/LB or VUS and P/LP predicted consequences. Circle

    diameters are proportional to the number of patients affected. Blue

    and green lollipops indicate phosphorylation and acetylation sites.

    Protein sequence regions corresponding to conserved domains are

    highlighted by colored boxes and are indicated by their respective

    Pfam names; b non-synonymous mutation annotations on available3D structures as assessed through Mechismo, are displayed following

    the same coloring scheme as in a. Labels in parenthesis indicatemutations with no structural information

    J Gastroenterol

    123

  • p = 0.028), indicating a potential role of these variants as

    disease modifiers.

    Subanalysis of patients with P and LP mutations vs.

    remaining cholestatic population

    Significant differences between subjects with and without

    P/LP mutations were not observed in sex, age at symptoms

    presentation, and the following risk factors for PFIC: DIC

    history, family or personal history of ICP, juvenile

    cholelithiasis and family history for cholestatic diseases

    (Table 4). However, at least one of these risk factors was

    present in 9/11 patients with P/LP mutations (Table 2).

    Patients with P/LP mutations had more frequently neonatal

    jaundice (45.5 vs. 13.5%, p = 0.036) and itching (54.5 vs.

    18.9%, p = 0.029).

    Regarding laboratory tests, significant differences were

    observed between the two subgroups only in terms of BA

    concentration (23.8 [4.4–403] vs. 8.8 [2.3–114],

    p = 0.003).

    Liver stiffness was greater in subjects with P/LP muta-

    tions in comparison with the others (8.1 [3.3–35.8] vs. 4.8

    [3.1–29.9], p = 0.009). All subjects of the first group

    showed histological features of intrahepatic cholestasis

    while 72% of patients without P/LP mutations presented it

    (p = 0.018). The univariate analysis for predictors of P/LP

    mutations indicated that AP (odds ratio [OR] 0.995, 95%

    CI 0.990–0.999, p = 0.019), liver stiffness (OR 0.921,

    95% CI 0.841–1.010, p = 0.081), itching (OR 5.143, 95%

    CI 1.214–21.795, p = 0.026), neonatal jaundice (OR

    5.330, 95% CI (1.172–24.277, p = 0.030) showed a

    p\ 0.1 (Supplementary Table 3). Multivariate analysis(Supplementary Table 3) showed that only itching was an

    independent predictor of P/LP mutations in patients with

    cryptogenic cholestasis (OR 5.801, 95% CI 1.244–27.060,

    p = 0.025).

    Fig. 2 a Prediction of the functional consequences of TJP2’sp.R322W variant through Mechismo. Green and orange arrows,

    respectively, indicate an enabling and mixed (i.e., both enabling and

    disabling) effect of the p.R322W mutation towards interactors.

    b Structural details of the TJP2 dimerization interface predicted to beaffected by the p.R322W mutation

    *(L)P(L)B or VUSHM

    Age at testFibroscan

    High GGT Yes No

    7 71

    3.1 35.8

    Patient ID

    NA NA NA

    Bile acidsNeonatal jaundice

    Itching

    Cholestasis at histology

    2.3 403

    Yes No

    Yes No

    Yes No

    Fig. 3 Bi-dimensional representation showing genes affected by non-synonymous mutations (row) in each patient (column). Mutated genes

    are sorted according to their mutation rate and each mutation is

    colored according to pathogenicity prediction (P/LP in red, B/LB and

    VUS in pink). Homozygous mutations are indicated by an asterisk.

    For each patient, the following clinical data are also displayed: age of

    patient, FibroScan� values, GGT levels, neonatal jaundice, itching

    history, bile acids and cholestasis at histology

    J Gastroenterol

    123

  • Discussion

    ABCB11 and ABCB4 are ATP-binding cassette proteins

    and members of MDR/TAP subfamily. They are membrane

    proteins, with a long intracellular domain, which makes

    them partly similar to ATP8B1, though it shares no close

    homology with either of them. These three genes are pre-

    dicted, but equivalents in non-human species, to interact

    both physically and functionally [30]. Specifically,

    ATP8B1 homolog in Drosophila MRP has been seen to

    interact with ABCB11/B4 homolog CG31729 and yeast

    equivalents YOR1 and DNF3 are genetic interactors. The

    majority of mutations described in these three genes lie in

    or near the intracellular domains in these three proteins.

    TJP2, while lacking trans-membrane domains is never-

    theless a peripheral membrane protein attached (when not

    in the nucleus) via lipid head groups to the cytoplasmic

    side of the membrane. Functionally, the formation of tight-

    junctions is thought to be related to separation of bile from

    plasma (which in itself is intricately linked to their trans-

    port) [31]. It is possible that some or all of these proteins

    form a complex at some point during the production or

    transport of liver metabolites.

    PFIC are considered pediatric diseases related to liver

    failure. Mutations in ATP8B1, ABCB11, ABCB4 and TJP2

    have been associated to a plethora of cholestatic disorders;

    hepatocellular carcinoma and cholangiocarcinoma (to

    ABCB11 and TJP2), ICP (linked to ATP8B1, ABCB11 and

    ABCB4), LPAC (to ABCB4), DIC (to ABCB11 and ABCB4)

    and BRIC (to ATP8B1 and ABCB11), are related and may

    coexist in the same patient. Furthermore, all four genes may

    be responsible for progressive forms of cholestasis, tradi-

    tionally considered exclusive of childhood [5, 6, 32, 33].

    Of interest, we found a mean age of 37 years in subjects

    with disease-causing variants to confirm that P/LP muta-

    tions are not present only in children. Few studies linked

    mutations in PFIC genes with non-progressive diseases,

    especially in heterozygous subjects. In our cohort of

    patients with at least one disease-causing mutation, only

    four patients were homozygous or compound heterozygous

    for disease-causing variants in the same gene.

    Colombo et al. [10] described pathogenic mutations in

    ABCB4 in about a quarter of asymptomatic children where

    cholestatic disease was incidentally discovered via liver

    enzyme abnormalities and, not surprisingly, some of these

    patients carried a single heterozygous mutation. Gordo-

    Table 4 Main features of patients according to presence of P/LP mutations

    Pts with P/LP mutations N = 11 Pts without P/LP mutations N = 37 p value

    Male N/Tot N (%) 5/11 (45.5) 23/37 (62.2) 0.260

    Age: (years, mean ± SD)

    At time of genetic test 37 ± 19 44 ± 14 0.244

    At symptoms presentation 27 ± 12 33 ± 15 0.308

    Risk factor of cholestasis N/Tot N (%)

    DIC history 6/11 (54.4) 11/37 (29.7) 0.126

    Neonatal jaundice 5/11 (45.5) 5/37 (13.5) 0.036

    Itching history 6/11 (54.5) 7/37 (18.9) 0.029

    ICP history 2/11 (18.2) 4/37 (10.8) 0.420

    Juvenile cholelithiasis 3/11 (27.3) 5/37 (13.5) 0.259

    Family history 5/11 (45.5) 12/37 (32.4) 0.327

    Laboratory, median (range)

    GammaGT (UI/l) 121 (5–379) 148 (18–597) 0.425

    AP (UI/l) 467 (138–1012) 280 (122–610) 0.061

    ALT (UI/l) 41 (9–387) 46 (9–358) 0.608

    Bilirubin (mg/dl) 1.2 (0.4–11.2) 0.7 (0.4–6.2) 0.484

    Bile acids (lmol/l) 23.8 (4.4–403) 8.8 (2.3–114) 0.034

    Cholesterol (mg/dl) 182 (132–303) 215 (87–328) 0.484

    Albumin (g/dl) 4.3 (2.9–5.2) 4 (3.6–4.7) 4.3 (2.9–5.2) 0.126

    Platelets (103/lL) 223 (128–412) 233 (61–417) 0.873

    FibroScan (kPa), median (range) 8.1 (3.3–35.8) 4.8 (3.1–29.9) 0.009

    Histologic features of cholestasis N/Tot N (%) 10/10 (100) 21/34 (61.8) 0.018

    N number, Tot N total number, DIC drug-induced cholestasis, SD standard deviation, P/LP mutations pathogenic and likely mutations, ICP

    intrahepatic cholestasis of pregnancy, AP alkaline phosphatase, ALT alanine transaminases, kPa kiloPascal

    J Gastroenterol

    123

  • Gilart et al. [9] reported similar results in another cohort of

    pediatric patients in whom defects in a single allele of

    ABCB4 were identified in 9/67 subjects. Some authors

    suggested that cryptogenic cholestasis in adults should be

    added to the spectrum of conditions associated with

    ABCB4 mutations [34–36]. Furthermore, adults with PSC

    had early onset of disease if MDR3 deficiency was found

    [34].

    In a recent study analyzing 427 cholestatic patients, 149

    subjects presented at least one disease-causing variant on

    ATP8B1, ABCB11 and ABCB4. Surprisingly, 25 patients

    with only one heterozygous variant experienced symptoms

    before first year of life, suggesting the presence of muta-

    tions in other genes, epigenetic changes or environmental

    factors responsible of cholestatic phenotype severity [11].

    However, adult population data lack clinical significance

    of heterozygosity in ABCB11, ATP8B1, and TJP2, though

    the first two genes have been correlated to BRIC when only

    one heterozygous variant was present. In some instances,

    these diseases may evolve towards progressive forms (e.g.,

    PFIC), thus representing a clinical continuum [22, 37].

    Our study represents the first report of simultaneous

    sequencing of ATP8B1, ABCB11, ABCB4 and TJP2 in

    patients with cryptogenic cholestasis.

    In our cohort, 21% of patients had at least one P/LP

    mutation and 17% with a disease-causing mutation pre-

    sented a liver disorder in adulthood ([ 18 years), sug-gesting once again that variants in PFIC genes are not

    exclusive to cholestatic diseases in childhood.

    Patients with P/LP mutations had higher liver stiffness

    and BA levels than patients without disease-causing vari-

    ants and all exhibited histological evidence of lobular

    cholestasis, confirming a more aggressive phenotype in

    subjects with causative mutations.

    Many patients had multiple variants in these four genes.

    It is thus tempting to hypothesize a synergistic effect in

    determining different cholestasis phenotypes and/or speci-

    fic interactions with environmental factors, in particular

    certain drugs. A reduction of bile flow represents a

    pathophysiological state and can be critical for metabolism

    of many drugs. This is supported by MDR3 and BSEP

    deficiencies being known to be associated to different

    levels of DIC [7, 38]. Examples are SNPs V444A/M677V

    in ABCB11 and I237= in ABCB4. We found V444A in

    83.3% of patients (38% in homozygous state): this benign

    variant was reported with AF in the general population of

    56.9 and of 65.9% in cholestatic patients with no BSEP

    disease-causing mutations [11]. It is associated with DIC,

    ICP and reduced expression of BSEP levels in previous

    studies [24, 39–41]. Specifically in DIC, patients carrying

    V444A were at increased risk of drug-induced liver injury,

    when taking drugs containing a carbocyclic system with

    aromatic rings [42].

    SNP M677V occurred in 13.5% of our cohort of cho-

    lestatic patients without P/LP mutations, more significantly

    frequent than in the other zones (AF in word 2.73%).

    In our cohort, SNP p.I237= has an allele frequency of

    25%, while Dröge et al. [11] reported 15.4% in an assumed

    genetic cholestatic population. Our AFs were significantly

    increased respect to AFs reported in worldwide and

    European population. This ABCB4 polymorphism increa-

    ses the risk of development of cholestatic disease, ICP and

    LPAC in previous reports [11, 43].

    Common SNPs V444A/M677V/I237= were detected in

    a very high number of cases, with or without disease-

    causing mutations, suggesting that these common variants

    might contribute to cholestasis development or worsening.

    The integration of standard variant prediction and clas-

    sification tools with more advanced bioinformatic analyses,

    taking into account biological pathways and/or biomolec-

    ular structures, is becoming increasingly used to get a

    deeper understanding on the role of genetic variation from

    large NGS datasets, as we previously showed for genetic

    diseases [44] and cancer [45]. This is also witnessed by

    recent work by Dröge et al. [11] for a larger panel of

    cholestatic liver disease patients. For example, we showed

    that the common SNP p.V444A of ABCB11 is found at the

    catalytic domain of the ABC transporter (Fig. 1) in close

    proximity but not in direct physical contact with the ATP-

    binding cassette. We thus speculate that while it is unlikely

    that this variant directly perturbs the enzymatic activity of

    the transporter, it might still induce subtle structural rear-

    rangements on the catalytic domain leading to a slightly

    perturbed, but still functional enzyme. Such a genotype

    would give rise to a disease phenotype when in co-presence

    with other genetic or environmental factors.

    Disease-causing mutations in TJP2 are also well estab-

    lished. Sambrotta et al. [6] associated them with progres-

    sive forms of normal GGT cholestasis in pediatric patients,

    while our four patients had heterozygous status, different

    ages of onset, and high GGT levels. Our results highlight

    the variability of clinical presentation in patients having

    mutations in ATP8B1, ABCB11, ABCB4, and TJP2, espe-

    cially when only one allele was involved. Our structural

    analysis, while corroborating pathogenicity predictions by

    standard bioinformatic tools, also provided novel mecha-

    nistic insights on the functional consequences of these

    variants. For example, the TJP2 p.R322W mutation, pre-

    dicted to be pathogenic, was shown by Mechismo to affect

    interfaces mediating homo- and hetero-dimers (Fig. 2),

    thus suggesting to likely impact TJP2’s physiological

    function.

    Similarly to other studies, we found it difficult to assess

    the causative role of many missense variants. We classified

    the variants detected in our cohort according to ACMG

    criteria, and some missense resulted as VUS. The patients

    J Gastroenterol

    123

  • showing only VUS were not included in our analyses of

    mutated patients (Table 4 and Supplementary Table 1) but

    some of these variants might be re-classified as likely

    pathogenic when related to the clinical phenotype.

    However, in our cohort of patients, the etiology of the

    cholestatic disease remains elusive, with only 21%

    attributable to mutations in the known causative genes.

    Mutational combinations at different loci as well as the

    several VUS detected in one or more FIC-genes and their

    potential synergistic effect could also contribute to the

    disease onset.

    The main limit of our study is that we have considered

    four genes responsible for intrahepatic cholestasis, which

    does not exclude that other genes could potentially be

    associated with this condition. Other studies proposed

    multi-gene panels able to facilitate genetic diagnosis, but

    only in children with intrahepatic cholestasis [13, 46, 47].

    For instance, the recent discovery of new genes

    responsible of FIC: myosin-5B gene, linked to microvillus

    inclusion disease and NR1H4 gene, encoding farnesoid

    X-receptor, a bile acid-activated nuclear hormone receptor

    that regulates bile acid metabolism and suppression of bile

    acid production. Both genes are able to lead to progressive

    cholestatic liver disease [48–50].

    Nevertheless, the highest rates of BA levels, liver

    fibrosis, itching, and bile histologic alterations detected in

    the patients with P/lP mutations make it possible to classify

    the pathogenicity of variants in accordance with the

    ACMG standards [16].

    In conclusion, the remarkable number of cases with

    causative mutations found in adults with cryptogenic

    cholestasis confirms the usefulness of mutational screening

    in ATP8B1, ABCB11, ABCB4, and TJP2, especially in

    those cases with itching history, regardless of age of

    cholestasis onset. Remains to be evaluated the synergistic

    role of mutations at different loci and of VUS in these four

    genes. Further observational studies are needed to under-

    stand the long-term clinical significance of mutations in

    genes responsible for FIC. Finally, the high proportion of

    unsolved cases suggests novel genetic etiologies that

    remain to be elucidated.

    Author contributions GV and PA designed the study and collecteddata. AM, VM, and MS performed the DNA sequencing and applied

    prediction tools; AD supervised the histological evaluations, FR and

    RBR performed protein modeling by Mechismo; SG, AM, VM, GV,

    RV, and PA analyzed the patients’ data. GV wrote the manuscript; all

    authors critically revised the manuscript.

    Compliance with ethical standards

    Conflict of interest The authors declare that they have no conflict ofinterest.

    Financial support No grants and other financial support werereceived.

    References

    1. Hori T, Nguyen JH, Uemoto S. Progressive familial intrahepatic

    cholestasis. Hepatobiliary Pancreat Dis Int. 2010;9:570–8.

    2. Paulusma CC, Elferink RP, Jansen PL. Progressive familial

    intrahepatic cholestasis type 1. Semin Liver Dis.

    2010;30:117–24.

    3. Lam P, Soroka CJ, Boyer JL. The bile salt export pump: clinical

    and experimental aspects of genetic and acquired cholestatic liver

    disease. Semin Liver Dis. 2010;30:125–33.

    4. Smit JJ, Schinkel AH, Oude Elferink RP, et al. Homozygous

    disruption of the murine mdr2 P-glycoprotein gene leads to a

    complete absence of phospholipid from bile and to liver disease.

    Cell. 1993;75:451–62.

    5. Davit-Spraul A, Gonzales E, Baussan C, et al. The spectrum of

    liver diseases related to ABCB4 gene mutations: pathophysiology

    and clinical aspects. Semin Liver Dis. 2010;30:134–46.

    6. Sambrotta M, Strautnieks S, Papouli E, et al. Mutations in TJP2

    cause progressive cholestatic liver disease. Nat Genet.

    2014;46:326–8.

    7. Pauli-Magnus C, Meier PJ, Stieger B. Genetic determinants of

    drug-induced cholestasis and intrahepatic cholestasis of preg-

    nancy. Semin Liver Dis. 2010;30:147–59.

    8. Poupon R, Rosmorduc O, Boëlle PY, et al. Genotype-phenotype

    relationships in the low-phospholipid-associated cholelithiasis

    syndrome: a study of 156 consecutive patients. Hepatology.

    2013;58:1105–10.

    9. Gordo-Gilart R, Hierro L, Andueza S, et al. Heterozygous

    ABCB4 mutations in children with cholestatic liver disease. Liver

    Int. 2016;36:258–67.

    10. Colombo C, Vajro P, Degiorgio D, et al. SIGENP Study Group

    for Genetic Cholestasis. Clinical features and genotype-pheno-

    type correlations in children with progressive familial intrahep-

    atic cholestasis type 3 related to ABCB4 mutations. J Pediatr

    Gastroenterol Nutr. 2011;52:73–83.

    11. Dröge C, Bonus M, Baumann U, et al. Sequencing of FIC1, BSEP

    and MDR3 in a large cohort of patients with cholestasis revealed

    a high number of different genetic variants. J Hepatol.

    2017;S0168–8278:32147–55.

    12. Xuan J, Yu Y, Qing T, et al. Next-generation sequencing in the

    clinic: promises and challenges. Cancer Lett. 2013;340:284–95.

    13. Herbst SM, Schirmer S, Posovszky C, et al. Taking the next step

    forward—diagnosing inherited infantile cholestatic disorders

    with next generation sequencing. Mol Cell Probes.

    2015;29:291–8.

    14. Adzhubei IA, Schmidt S, Peshkin L, et al. A method and server

    for predicting damaging missense mutations. Nat Methods.

    2010;7:248–9.

    15. Schwarz JM, Cooper DN, Schuelke M, et al. MutationTaster2:

    mutation prediction for the deep-sequencing age. Nat Methods.

    2014;11:361–2.

    16. Richards S, Aziz N, Bale S, et al. ACMG Laboratory Quality

    Assurance Committee. Standards and guidelines for the inter-

    pretation of sequence variants: a joint consensus recommendation

    of the American College of Medical Genetics and Genomics and

    the Association for Molecular Pathology. Genet Med.

    2015;17:405–24.

    17. Betts MJ, Lu Q, Jiang Y, et al. Mechismo: predicting the

    mechanistic impact of mutations and modifications on molecular

    interactions. Nucleic Acids Res. 2015;43:e10.

    J Gastroenterol

    123

  • 18. Pieper U, Webb BM, Dong GQ, et al. ModBase, a database of

    annotated comparative protein structure models and associated

    resources. Nucleic Acids Res. 2014;42(Database issue):D336–46.

    19. Hornbeck PV, Chabra I, Kornhauser JM, et al. PhosphoSite: a

    bioinformatics resource dedicated to physiological protein phos-

    phorylation. Proteomics. 2004;4:1551–61.

    20. Jay JJ, Brouwer C. Lollipops in the CLINIC: information dense

    mutation plots for precision medicine. PLoS ONE.

    2016;11:e0160519.

    21. Painter JN, Savander M, Ropponen A, et al. Sequence variation in

    the ATP8B1 gene and intrahepatic cholestasis of pregnancy. Eur

    J Hum Genet. 2005;13:435–9.

    22. Vitale G, Pirillo M, Mantovani V, et al. Bile salt export pump

    deficiency disease: two novel, late onset, ABCB11 mutations

    identified by next generation sequencing. Ann Hepatol.

    2016;15:795–800.

    23. Anzivino C, Odoardi MR, Meschiari E, et al. ABCB4 and

    ABCB11 mutations in intrahepatic cholestasis of pregnancy in an

    Italian population. Dig Liver Dis. 2013;45:226–32.

    24. Lang C, Meier Y, Stieger B, et al. Mutations and polymorphisms

    in the bile salt export pump and the multidrug resistance protein 3

    associated with drug-induced liver injury. Pharmacogenet

    Genomics. 2007;17:47–60.

    25. Jin MS, Oldham ML, Zhang Q, et al. Crystal structure of the

    multidrug transporter P-glycoprotein from Caenorhabditis ele-

    gans. Nature. 2012;490:566–9.

    26. Degiorgio D, Colombo C, Seia M, et al. Molecular characteri-

    zation and structural implications of 25 new ABCB4 mutations in

    progressive familial intrahepatic cholestasis type 3 (PFIC3). Eur J

    Hum Genet. 2007;15:1230–8.

    27. Müllenbach R, Linton KJ, Wiltshire S, et al. ABCB4 gene

    sequence variation in women with intrahepatic cholestasis of

    pregnancy. J Med Genet. 2003;40:e70.

    28. Liu C, Aronow BJ, Jegga AG, et al. Novel resequencing chip

    customized to diagnose mutations in patients with inherited

    syndromes of intrahepatic cholestasis. Gastroenterology.

    2007;132:119–26.

    29. Fanning AS, Anderson JM. Zonula occludens-1 and -2 are

    cytosolic scaffolds that regulate the assembly of cellular junc-

    tions. Ann NY Acad Sci. 2009;1165:113–20.

    30. Szklarczyk D, Franceschini A, Wyder S, et al. STRING v10:

    protein–protein interaction networks, integrated over the tree of

    life. Nucleic Acids Res. 2015;43:D447–52.

    31. van Mil SW, Houwen RH, Klomp LW. Genetics of familial

    intrahepatic cholestasis syndromes. J Med Genet.

    2005;42:449–63.

    32. Alissa FT, Jaffe R, Shneider BL. Update on progressive familial

    intrahepatic cholestasis. J Pediatr Gastroenterol Nutr.

    2008;46:241–52.

    33. Zhou S, Hertel PM, Finegold MJ, et al. Hepatocellular carcinoma

    associated with tight-junction protein 2 deficiency. Hepatology.

    2015;62:1914–6.

    34. Degiorgio D, Crosignani A, Colombo C, et al. ABCB4 mutations

    in adult patients with cholestatic liver disease: impact and phe-

    notypic expression. J Gastroenterol. 2016;51:271–80.

    35. Gotthardt D, Runz H, Keitel V, et al. A mutation in the

    canalicular phospholipid transporter gene, ABCB4, is associated

    with cholestasis, ductopenia, and cirrhosis in adults. Hepatology.

    2008;48:1157–66.

    36. Ziol M, Barbu V, Rosmorduc O, et al. ABCB4 heterozygous gene

    mutations associated with fibrosing cholestatic liver disease in

    adults. Gastroenterology. 2008;135:131–41.

    37. Van Ooteghem NA, Klomp LW, Van Berge-Henegouwen GP,

    et al. Benign recurrent intrahepatic cholestasis progressing to

    progressive familial intrahepatic cholestasis: low GGT cholesta-

    sis is a clinical continuum. J Hepatol. 2002;36:439–43.

    38. Padda MS, Sanchez M, Akhtar AJ, et al. Drug-induced

    cholestasis. Hepatology. 2011;53:1377–87.

    39. Meier Y, Zodan T, Lang C, et al. Increased susceptibility for

    Intrahepatic cholestasis of pregnancy and contraceptive-induced

    cholestasis in carriers of the 1331T[C polymorphism in the bilesalt export pump. World J Gastroenterol. 2008;14:38–45.

    40. Pauli-Magnus C, Lang T, Meier Y, et al. Sequence analysis of

    bile salt export pump (ABCB11) and multidrug resistance p-

    glycoprotein 3 (ABCB4, MDR3) in patients with intrahepatic

    cholestasis of pregnancy. Pharmacogenetics. 2004;14:91–102.

    41. Dixon PH, van Mil SW, Chambers J, et al. Contribution of variant

    alleles of ABCB11 to susceptibility to intrahepatic cholestasis of

    pregnancy. Gut. 2009;58:537–44.

    42. Ulzurrun E, Stephens C, Crespo E, et al. Role of chemical

    structures and the 1331T[C bile salt export pump polymorphismin idiosyncratic drug-induced liver injury. Liver Int.

    2013;33:1378–85.

    43. Gudbjartsson DF, Helgason H, Gudjonsson SA, et al. Large-scale

    whole-genome sequencing of the Icelandic population. Nat

    Genet. 2015;47:435–44.

    44. Boldt K, van Reeuwijk J, Lu Q, et al. UK10K Rare Diseases

    Group. An organelle-specific protein landscape identifies novel

    diseases and molecular mechanisms. Nat Commun.

    2016;7:11491. https://doi.org/10.1038/ncomms11491.

    45. Raimondi F, Singh G, Betts MJ, et al. Insights into cancer

    severity from biomolecular interaction mechanisms. Sci Rep.

    2016;6:34490. https://doi.org/10.1038/srep34490.

    46. Wang NL, Lu YL, Zhang P, et al. Specially designed multi-gene

    panel facilitates genetic diagnosis in children with intrahepatic

    cholestasis: simultaneous test of known large insertions/deletions.

    PLoS ONE. 2016;11:e0164058.

    47. Togawa T, Sugiura T, Ito K, et al. Molecular genetic dissection

    and neonatal/infantile intrahepatic cholestasis using targeted

    next-generation sequencing. J Pediatr. 2016;171:171–4.

    48. Gomez-Ospina N, Potter CJ, Xiao R, et al. Mutations in the

    nuclear bile acid receptor FXR cause progressive familial intra-

    hepatic cholestasis. Nat Commun. 2016;18(7):10713.

    49. Qiu YL, Gong JY, Feng JY, et al. Defects in myosin VB are

    associated with a spectrum of previously undiagnosed low c-glutamyltransferase cholestasis. Hepatology. 2017;65:1655–69.

    50. Gonzales E, Taylor SA, Davit-Spraul A, et al. MYO5B mutations

    cause cholestasis with normal serum gamma-glutamyl transferase

    activity in children without microvillous inclusion disease.

    Hepatology. 2017;65:164–73.

    J Gastroenterol

    123

    https://doi.org/10.1038/ncomms11491https://doi.org/10.1038/srep34490

  • 本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

    学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

    提供一站式文献检索和下载服务”的24 小时在线不限IP

    图书馆。

    图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

    图书馆导航:

    图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具

    http://www.xuebalib.com/cloud/http://www.xuebalib.com/http://www.xuebalib.com/cloud/http://www.xuebalib.com/http://www.xuebalib.com/vip.htmlhttp://www.xuebalib.com/db.phphttp://www.xuebalib.com/zixun/2014-08-15/44.htmlhttp://www.xuebalib.com/

    Cryptogenic cholestasis in young and adults: ATP8B1, ABCB11, ABCB4, and TJP2 gene variants analysis by high-throughput sequencingAbstractBackgroundAimMethodsResultsConclusions

    IntroductionMaterials and methodsPatientsNGS analysisPrediction of functional consequences of variants and classificationComparison with an international databaseStatistical analysis

    ResultsPatient characteristicsNGS results and variantsATP8B1 variantsABCB11 variationsABCB4 variationsTJP2 variationsSummary mutation profile for FIC patientsSubanalysis of patients with P and LP mutations vs. remaining cholestatic population

    DiscussionAuthor contributionsReferences

    学霸图书馆link:学霸图书馆