Background & Aims
Alagille syndrome is an autosomal-dominant, multisystem disorder caused primarily by mutations in JAG1, resulting in bile duct paucity, cholestasis, cardiac disease, and other features. Liver disease severity in Alagille syndrome is highly variable, however, factors influencing the hepatic phenotype are unknown. We hypothesized that genetic modifiers may contribute to the variable expressivity of this disorder.
Methods
We performed a genome-wide association study in a cohort of Caucasian subjects with known pathogenic JAG1 mutations, comparing patients with mild vs severe liver disease, followed by functional characterization of a candidate locus.
Results
We identified a locus that reached suggestive genome-level significance upstream of the thrombospondin 2 (THBS2) gene. THBS2 codes for a secreted matricellular protein that regulates cell proliferation, apoptosis, and angiogenesis, and has been shown to affect Notch signaling. By using a reporter mouse line, we detected thrombospondin 2 expression in bile ducts and periportal regions of the mouse liver. Examination of Thbs2-null mouse livers showed increased microvessels in the portal regions of adult mice. We also showed that thrombospondin 2 interacts with NOTCH1 and NOTCH2 and can inhibit JAG1–NOTCH2 interactions.
Conclusions
Based on the genome-wide association study results, thrombospondin 2 localization within bile ducts, and demonstration of interactions of thrombospondin 2 with JAG1 and NOTCH2, we propose that changes in thrombospondin 2 expression may further perturb JAG1–NOTCH2 signaling in patients harboring a JAG1 mutation and lead to a more severe liver phenotype. These results implicate THBS2 as a plausible candidate genetic modifier of liver disease severity in Alagille syndrome.
Alagille syndrome (ALGS) is an autosomal-dominant disorder caused by mutations in the Notch pathway ligand
JAG1 in 94% of patients and in 1 of 4 Notch receptors (
NOTCH2) in 1%–2%. ALGS is characterized by bile duct paucity, cholestatic liver disease, congenital heart lesions, eye and skeletal anomalies, and a characteristic facial appearance. However, there is extensive variation in the expressivity of the disease, and variable penetrance of clinical features in ALGS patients has been observed since the earliest description of the syndrome.
1- Alagille D.
- Odievre M.
- Gautier M.
- et al.
Hepatic ductular hypoplasia associated with characteristic facies, vertebral malformations, retarded physical, mental, and sexual development, and cardiac murmur.
Before mutations in
JAG1 were found to be the principal cause of ALGS, examination of inherited cases showed extreme phenotypic variability, even among family members.
2- Dhorne-Pollet S.
- Deleuze J.F.
- Hadchouel M.
- et al.
Segregation analysis of Alagille syndrome.
, 3- Elmslie F.V.
- Vivian A.J.
- Gardiner H.
- et al.
Alagille syndrome: family studies.
, 4- Shulman S.A.
- Hyams J.S.
- Gunta R.
- et al.
Arteriohepatic dysplasia (Alagille syndrome): extreme variability among affected family members.
We suspect that this phenotypic variability, including liver disease severity, is associated with genetic modifiers.
The liver disease seen in ALGS patients is highly variable, ranging from subclinical to severe, and factors influencing the hepatic phenotype are unknown. Unlike the cardiac defects, in which severe forms of cardiac disease can be categorized at initial presentation, liver disease severity cannot be predicted based on the presence of bile duct paucity alone. Early symptoms may resolve and never develop into severe liver disease, however 20%–30% of ALGS patients eventually will require liver transplantation.
5- Emerick K.M.
- Rand E.B.
- Goldmuntz E.
- et al.
Features of Alagille syndrome in 92 patients: frequency and relation to prognosis.
, 6- Hoffenberg E.J.
- Narkewicz M.R.
- Sondheimer J.M.
- et al.
Outcome of syndromic paucity of interlobular bile ducts (Alagille syndrome) with onset of cholestasis in infancy.
, 7- Lykavieris P.
- Hadchouel M.
- Chardot C.
- et al.
Outcome of liver disease in children with Alagille syndrome: a study of 163 patients.
, 8- Quiros-Tejeira R.E.
- Ament M.E.
- Heyman M.B.
- et al.
Variable morbidity in Alagille syndrome: a review of 43 cases.
It also has been observed that liver disease in children younger than 5 years of age is not a stable predictor of long-term need for liver transplantation,
9- Kamath B.M.
- Munoz P.S.
- Bab N.
- et al.
A longitudinal study to identify laboratory predictors of liver disease outcome in Alagille syndrome.
although more recent work has shown that the combinatorial quantification of serum total bilirubin, liver biopsy fibrosis, and the presence of xanthomata is predictive of long-term hepatic disease, offering a prognostic metric for this phenotype.
10- Mouzaki M.
- Bass L.M.
- Sokol R.J.
- et al.
Early life predictive markers of liver disease outcome in an international, multi-center cohort of children with Alagille syndrome.
No environmental factor influencing liver disease severity has been identified to date. Attempts to establish a genotype–phenotype correlation between
JAG1 mutations and the liver phenotype have been unable to substantiate any connection,
11- Crosnier C.
- Driancourt C.
- Raynaud N.
- et al.
Mutations in JAGGED1 gene are predominantly sporadic in Alagille syndrome.
, 12- Krantz I.D.
- Colliton R.P.
- Genin A.
- et al.
Spectrum and frequency of jagged1 (JAG1) mutations in Alagille syndrome patients and their families.
, 13- McElhinney D.B.
- Krantz I.D.
- Bason L.
- et al.
Analysis of cardiovascular phenotype and genotype-phenotype correlation in individuals with a JAG1 mutation and/or Alagille syndrome.
, 14- Spinner N.B.
- Colliton R.P.
- Crosnier C.
- et al.
Jagged1 mutations in Alagille syndrome.
and there presently is no reliable genetic biomarker that is able to explain the high degree of liver disease variability seen in ALGS. We hypothesize that genetic modifying factors contribute to this phenotype, such that some children will progress to end-stage liver disease because of their genetic risk.
We designed a genome-wide association study (GWAS) to identify loci that influence liver disease severity in ALGS patients. The strongest association was found in the genomic region upstream of the gene encoding thrombospondin 2, a matricellular protein known to interact with the Notch signaling pathway.
Materials and Methods
Sample Cohort and Stratification
ALGS patients who were positive for a
JAG1 mutation were enrolled in the study either through the Children’s Hospital of Philadelphia or through the Longitudinal Study of Genetic Causes of Intrahepatic Cholestasis protocol within the Childhood Liver Disease Research Network (ChiLDReN), a National Institute of Diabetes and Digestive and Kidney Diseases/National Institutes of Health–funded network of 16 pediatric academic medical centers across North America. This study was approved by the Institutional Review Boards at each center and informed consent was obtained from parents/guardians or subjects 18 years or older. Data from all patients were reviewed to determine liver disease severity, using a stratification protocol based on a combination of clinical and biochemical findings (
Table 1). At the time of enrollment in this study, there was no reliable predictor of outcome before age 5, therefore stratification was limited to ALGS patients older than 5 years of age.
9- Kamath B.M.
- Munoz P.S.
- Bab N.
- et al.
A longitudinal study to identify laboratory predictors of liver disease outcome in Alagille syndrome.
The 2 cohorts, mild and severe, showed no correlation in
JAG1 mutation type, as has been reported previously (
Supplementary Table 1).
11- Crosnier C.
- Driancourt C.
- Raynaud N.
- et al.
Mutations in JAGGED1 gene are predominantly sporadic in Alagille syndrome.
, 12- Krantz I.D.
- Colliton R.P.
- Genin A.
- et al.
Spectrum and frequency of jagged1 (JAG1) mutations in Alagille syndrome patients and their families.
, 13- McElhinney D.B.
- Krantz I.D.
- Bason L.
- et al.
Analysis of cardiovascular phenotype and genotype-phenotype correlation in individuals with a JAG1 mutation and/or Alagille syndrome.
, 14- Spinner N.B.
- Colliton R.P.
- Crosnier C.
- et al.
Jagged1 mutations in Alagille syndrome.
Table 1Stratification of Liver Disease Severity
Genotyping and Quality Control
There were 234 patients genotyped on the Omni1 (n = 138) and the OmniExpress (n = 96) single-nucleotide polymorphism (SNP) arrays (Illumina, San Diego, CA). Genotype data from both platforms were merged into 1 data set, keeping the 705,132 markers present on both arrays. We followed standard quality control procedures to select samples and SNPs for the association test. SNPs with a minor allele frequency less than 0.05, a missing rate greater than 0.05, or not in Hardy–Weinberg equilibrium (Phwe < .00001) were removed. The remaining 579,677 SNP markers were tested for association. Samples with a missing rate greater than 0.05 also were removed. The X chromosome heterozygosity rate also was used as a quality control metric and did not show any inconsistency between reported sex and genotype data.
Individuals of European ancestry accounted for the largest ethnic group in our cohort, and only these samples were used to mitigate biases that may arise from population stratification. European ancestry was inferred by multidimensional scaling with PLINK software.
15- Purcell S.
- Neale B.
- Todd-Brown K.
- et al.
PLINK: a tool set for whole-genome association and population-based linkage analyses.
Multidimensional scaling was performed on our samples and a HapMap data set of 4 populations: Utah residents with northern and western Europe ancestry; Han Chinese in Beijing, China; Japanese in Tokyo, Japan; and Yoruba in Ibadan, Nigeria
16The International HapMap Project.
(
Supplementary Figure 1). From the first 2 multidimensional scaling components, C1 and C2, patients with -0.0525 < C1 < -0.04 and -0.04 < C2 < -0.03 were selected as our cohort with European ancestry based on visual inspection.
To identify identical and related individuals, we estimated identity-by-state by genome-wide pairwise comparisons among all patients. We found 11 samples that were enrolled twice, once in the Children’s Hospital of Philadelphia cohort and once in ChiLDReN. There were 3 pairs of monozygotic twins with concordant liver phenotype, therefore only 1 proband from each pair was included in subsequent analysis. The remaining pairs with π greater than 0.15 (indicating relatedness) were accounted for by our knowledge of their relationships, except for 1 pair. We suspect that in this pair the de-identified ChiLDReN proband was the child of the subject who was enrolled in the Children’s Hospital of Philadelphia cohort. Because of the small number of enrolled families (n = 19), it was not possible to correlate risk allele with severity among relatives.
SNP Association Analysis
After sample quality control including controlling for population stratification and excluding repeated samples, 97 mild and 64 severe liver disease subjects remained for analysis. Because relatives were included in our data set, we used EMMAX, an implementation of the variance component approach that accounts for sample structure,
17- Kang H.M.
- Sul J.H.
- Service S.K.
- et al.
Variance component model to account for sample structure in genome-wide association studies.
to test for association. Suggestive evidence for association was considered if SNPs reached the threshold of 1 × 10
-5, which yields approximately 1 false-positive association per GWAS in populations of European ancestry.
18- Duggal P.
- Gillanders E.M.
- Holmes T.N.
- et al.
Establishing an adjusted p-value threshold to control the family-wide type 1 error in genome wide association studies.
For a finer resolution of the candidate regions, we performed imputation with IMPUTE2, using the recommended settings.
19Genotype imputation for genome-wide association studies.
Reference haplotypes were provided from the 1000 Genomes phase I data set.
20- Abecasis G.R.
- Auton A.
- et al.
1000 Genomes Project Consortium
An integrated map of genetic variation from 1,092 human genomes.
Following these recommendations, SNPs with imputation quality score INFO less than 0.8 and minor allele frequency less than 0.05 were removed from subsequent analysis. The imputed data set was analyzed by SNPTEST using an additive linear model.
19Genotype imputation for genome-wide association studies.
Mouse Lines
Thrombospondin 2 (
Thbs2)-null mice have been described previously.
21- Kyriakides T.R.
- Zhu Y.H.
- Smith L.T.
- et al.
Mice that lack thrombospondin 2 display connective tissue abnormalities that are associated with disordered collagen fibrillogenesis, an increased vascular density, and a bleeding diathesis.
Thbs2–green fluorescent protein (GFP) reporter mice expressing GFP driven by the
Thbs2 promoter were produced by the Gene Expression Nervous System Atlas program using a bacterial artificial chromosome clone spanning 70 kb upstream to 75 kb downstream of the
Thbs2 gene as previously described.
22- Palenski T.L.
- Gurel Z.
- Sorenson C.M.
- et al.
Cyp1B1 expression promotes angiogenesis by suppressing NF-kappaB activity.
Genotyping for all mice was performed by polymerase chain reaction (PCR) analysis using genomic DNA isolated from the tail tip. All procedures involving mice were conducted in accordance with federal guidelines and approved Institutional Animal Care and Use Committee protocols. All animals received humane care according to the criteria outlined in the Guidelines for the Care and Use of Laboratory Animals.
Immunohistochemistry and Immunofluorescence
Standard protocols for immunohistochemistry and immunofluorescence were used on liver tissues from
Thbs2–GFP reporter,
Thbs2-null, and control mice. Further technical information including fixation, antibodies used, and antigen retrieval and blocking can be found in
Supplementary Table 2. Slides were scanned using the Aperio Scan Scope OS at 20× (Leica Microsystems, Buffalo Grove, IL) for immunohistochemistry and were mounted for viewing with the Olympus BX-51 fluorescent microscope (Olympus, Center Valley, PA) for immunofluorescence.
Laser Capture Microdissection and Complementary DNA Amplification
Two 1-week-old C57BL/6J mouse livers were snap frozen in optimal cutting temperature compound (Sakura Finetak, USA, Inc, Torrance, CA). Each liver was sectioned at 12-μm thickness under RNAse-free conditions and placed on a polyethylene teraphthalate (PET)-membrane framed slide for laser capture microdissection (ASEE Products, Knoxville, TN). Sections were fixed in a brief dehydration with ethanol and xylene using standard protocols. Portal tracts were identified under 10× objective light microscopy using the Nikon Eclipse TE2000-S microscope (Nikon Instruments, Melville, NY) and approximately 12–20 portal tracts were selected and cut using mmiCellTools v2.3 software equipped with a solid-state UV laser (Molecular Machines and Industries, Eching, Germany). The remaining tissue on each section was extracted (liver lobule) to serve as a control and was considered to be enriched for parenchymal transcripts. Samples were collected using PCR sample tubes for laser capture microdissection (ASEE Products). Extraction buffer was applied to samples and they were placed on dry ice until RNA isolation using the PicoPure RNA isolation kit (Life Technologies, Grand Island, NY).
The Ovation Pico WTA System V2 kit was used to produce approximately 10–20 μg of single primer isothermal amplification complementary DNA (cDNA) from approximately 15–25 ng of total RNA (NuGEN Technologies, San Carlos, CA) following their standard protocol. RNA integrity and purity was confirmed before cDNA synthesis using an Agilent 2100 Bioanalyzer (Agilent Technologies, Inc, Wilmington, DE) and a NanoDrop (Wilmington, DE). Amplified products were purified a second time using Qiagen’s (Germantown, MD) QIAquick PCR purification kit and cDNA yield and purity was assessed again via both a NanoDrop as well as an Agilent DNA 1000 Bioanalyzer.
Gene Expression Analysis
Droplet digital PCR (ddPCR) was performed on a Bio-Rad QX100 ddPCR system (Hercules, CA) using cDNA harvested from laser capture microdissected tissue kindly provided to us by Dr Matthew Ryan (Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia). Droplets containing 60 ng of cDNA from mouse portal tracts were created using TaqMan (Thermo Fisher Scientific, Waltham, MA) mouse
Thbs2 primer and probe set (Mm01279240_m1) with TaqMan primer and probe set for the control gene,
Tbp (Mm00446971_m1). Standard ddPCR methods were used for experimental conditions and subsequent analysis.
23- Tsai E.A.
- Grochowski C.M.
- Falsey A.M.
- et al.
Heterozygous deletion of FOXA2 segregates with disease in a family with heterotaxy, panhypopituitarism, and biliary atresia.
Microvessel, Biliary, and Arterial Counts
Average numbers of microvessels (CD34+) and mature bile ducts (cytokeratin 19 [CK19]+) per portal tract were calculated based on examination of 4 Thbs2-null and 4 control 1-week-old and adult livers using immunofluorescence. Seven to 8 portal tracts were photographed for each sample and portal areas were outlined. Microvessels positively expressing CD34 and mature bile ducts positively expressing CK19 with a visible lumen within the designated portal region were counted and averages were calculated. For arterial counts, immunohistochemistry was performed on paraffin-embedded sections using CK19 and smooth muscle actin antibodies. Four 1-week-old Thbs2-null and control livers and 5 adult Thbs2-null and control livers were examined. The number of arteries were counted across entire liver sections and quantified by determining the average number of arteries per μm2. Results of the microvessel and arterial counts are presented as means and SEMs. Data were analyzed using the unpaired t test with a 2-tailed P value for statistical significance.
Co-immunoprecipitation
The 293A cells plated on 6-well plates were transfected with expression constructs encoding proteins of interest for 48 hours. The immunoprecipitation protocol has been described previously.
24- Meng H.
- Zhang X.
- Hankenson K.D.
- et al.
Thrombospondin 2 potentiates notch3/jagged1 signaling.
Plasmids encoding the ectodomains of NOTCH1 and NOTCH2 fused to a C-terminal V5 epitope tag have been described.
25- Meng H.
- Zhang X.
- Yu G.
- et al.
Biochemical characterization and cellular effects of CADASIL mutants of NOTCH3.
Retrovirus Production and Transduction
Phoenix A (amphotropic) cells were plated at a density of 4 × 105 cells/well on a 6-well plate. On the following day, culture media was replaced with fresh media (Dulbecco’s modified Eagle medium, high glucose, 10% fetal bovine serum, pen-strep, L-glutamine). Chloroquine dihydrochloride (S764663; Sigma, St. Louis, MO) was added to Phoenix cells 5 minutes before transfection and 2 μg of plasmid DNA was transfected into cells using the Profection kit (E1200; Promega, Madison, WI). The media was changed 10 hours after transfection. Retroviral supernatant was collected the following day and 2 mL was added to GP+E86 cells, plated at 4 × 104 cells/well on a 6-well dish the previous day, after filtration (0.45 μm) and supplemented with protamine sulfate (5 μg/mL). GP+E86 cells were transduced again on the following day. Viral supernatant from GP+E86 cells was collected on the following 2 days and used to transduce C3H10T1/2 cells.
Binding Assays
Recombinant proteins from R&D Systems (Minneapolis, MN) in this study include the following: recombinant human NOTCH2-Fc and NOTCH3-Fc fusions (containing the first 11 epidermal growth factor-like repeats), rat JAG1-Fc, human full-length thrombospondin 2, and control human IgG1 Fc. For protein labeling, 5 μg of protein was incubated with Alexa700-succinimide (10 μg) in phosphate-buffered saline at room temperature for 1 hour. Unincorporated label was removed using a size exclusion column equilibrated in Tris buffer (25-kilodalton cut-off level; Bio-Rad). Labeling of proteins was verified by sodium dodecyl sulfate–polyacrylamide gel electrophoresis under reducing conditions, followed by visualization of label using a LI-COR Odyssey flatbed infrared detector (LI-COR Biosciences, Lincoln, NE). All proteins were predominantly 1 band of the expected molecular weight at an apparent purity of at least 90%. Target proteins were coated on 96-well, flat-bottom enzyme-linked immunosorbent assay plates at 5 μg/mL in Tris-buffered saline (50 mmol/L Tris, 150 mmol/L NaCl) with 2 mmol/L CaCl
2 overnight at 4°C and then blocked with 1% bovine serum albumin (BSA) in Tris-buffered saline with 2 mmol/L CaCl
2 for 1 hour at room temperature. The wells then were incubated with Alexa700-succinimide–labeled proteins in Tris-buffered saline plus 2 mmol/L CaCl
2 and 0.05% Tween 20 at 4°C overnight. In some experiments, unlabeled thrombospondin 2 was added to the binding reaction to assess the effect on binary interactions. Bound, labeled proteins were quantified with a LI-COR Odyssey flatbed infrared detector (LI-COR Biosciences). Ligand binding studies of proteins immobilized on plastic have been described previously.
24- Meng H.
- Zhang X.
- Hankenson K.D.
- et al.
Thrombospondin 2 potentiates notch3/jagged1 signaling.
As in the previous study, control studies of the labeled proteins used here showed only trivial amounts of labeling of Fc and BSA control-coated plates.
Generation of Thrombospondin 2 Type I Repeat Deletion Constructs
A retroviral construct missing all 3 type I repeat domains (amino acids 379–549, THBS2-Del3) was generated by PCR amplification of a 1169-base pair fragment encoding amino acids 1–378 (fragment 1) and a 1925-base pair fragment encoding amino acids 550–1172 (fragment 2), followed by cloning into the pCRII-TOPO vector (Invitrogen, Carlsbad, CA). SfoI restriction sites were added to the 3’ primer of fragment 1 and the 5’ primer of fragment 2 to allow for ligation. Fragments 1 and 2 were released from pCRII-TOPO using BamHI/SfoI and SfoI/XbaI, respectively, and ligated into the pBluescript cloning vector. The full-length THBS2 and THBS2-Del3 fragment were released from pBluescript using EcoRI and cloned into the pRET retroviral vector.
Cell Culture
Stromal cells were harvested from femurs and tibias of
Thbs2-null and control mice and cultured as previously described.
26- Hankenson K.D.
- Bornstein P.
The secreted protein thrombospondin 2 is an autocrine inhibitor of marrow stromal cell proliferation.
At 80% confluence, cells were trypsinized and plated on 12-well tissue culture plates at a density of 1 × 10
5 cells per well. Notch signaling was activated by precoating tissue culture plates with 10 μg/mL of antibody against the Fc portion of human IgG (Jackson ImmonoResearch, West Grove, PA) for 1 hour and then incubating with the indicated concentration of recombinant rat JAG1/human Fc IgG chimeric protein (R&D Systems) for 2 hours as previously described.
27- Zhu F.
- Sweetwyne M.T.
- Hankenson K.D.
PKCdelta is required for Jagged-1 induction of human mesenchymal stem cell osteogenic differentiation.
For Notch reporter activity experiments, Fc-JAG1 or TNF-related apoptosis-inducing ligand (control) conditioned media
28- Buas M.F.
- Kabak S.
- Kadesch T.
Inhibition of myogenesis by Notch: evidence for multiple pathways.
were used to coat plates instead of the recombinant Fc-JAG1 protein.
RNA Extraction and Quantitative Reverse-Transcription PCR
RNA was extracted using the RNeasy RNA extraction kit (Qiagen, Venlo, The Netherlands) according to the manufacturer’s instructions and 1 μg of total RNA was reverse-transcribed. Quantitative real-time PCR reactions using SybrGreen I were performed on an ABI 7500 Fast Real-Time PCR System (Life Technologies, Carlsbad, CA). Relative changes in gene expression were determined by the 2ΔΔCT method using β-actin as an endogenous control.
Transient Transfection and CSL-Luciferase Assay
C3H10T1/2 cells were plated on 12-well plates coated with either Fc-Trail or Fc-JAG1 recombinant protein. After 36 hours, cells were co-transfected with 0.5 μg of CSL-luciferase, a synthetic DNA construct in which 4 tandem repeats of CBF-1, Suppressor of Hairless, Lag-1 binding elements drive expression of firefly luciferase, and 100 ng of pRL-TK, a constitutively active TK promoter driving expression of renilla luciferase activity, using Lipofectamine and Plus reagent (Invitrogen). Cells were harvested 48 hours after transfection and firefly and renilla luciferase activities were measured using the Dual Luciferase assay kit (Promega).
Immunoblotting
Protein was extracted from cultured human fibroblast samples using RIPA buffer (Sigma-Aldrich, St. Louis, MO) supplemented with proteinase inhibitors (Thermo Fisher Scientific). All Western blots were run using NuPAGE Tris-acetate pre-cast gels (Thermo Fisher Scientific). Membranes were blocked with 5% milk and 2% BSA and incubated for either 1 (β-actin, 1:5000) or 2 (thrombospondin 2, 1:50) nights with primary antibody. Membranes were incubated in horseradish-peroxidase–conjugated secondary antibody for 1 hour at room temperature and subsequently were developed using Pierce ECL Western Blotting Substrate (Thermo Fisher Scientific). Antibodies used included thrombospondin 2 (BD Biosciences, San Jose, CA) and β-actin (Millipore, Billerica, MA). Thrombospondin 2 expression was quantified and normalized to β-actin using ImageJ software (National Institutes of Health, Bethesda, MD).
Discussion
Our study has identified
THBS2 as a compelling candidate modifier gene of liver disease severity in ALGS. Thrombospondin 2 is a matricellular protein that previously was shown to potentiate JAG1–NOTCH3 interactions—a function that is specific to thrombospondin 2 and not other thrombospondin family members.
24- Meng H.
- Zhang X.
- Hankenson K.D.
- et al.
Thrombospondin 2 potentiates notch3/jagged1 signaling.
Although thrombospondin 2 has been studied in other tissues and found to be associated with anti-angiogenic effects during wound healing,
36- Kyriakides T.R.
- Maclauchlan S.
The role of thrombospondins in wound healing, ischemia, and the foreign body reaction.
its role in the liver has not been studied to date. Given the importance of both the
JAG1 gene and Notch signaling in the etiology of ALGS, we pursued additional functional studies after identifying this gene through our GWAS. We further show that thrombospondin 2 interacts with NOTCH1 and NOTCH2, and that increased concentration of thrombospondin 2 results in reduced binding of the JAG1–NOTCH2 ligand-receptor pair. We also show that thrombospondin 2 is expressed specifically in the biliary epithelium and portal tracts of mouse liver (
Figure 2). Together these data are highly suggestive of a role for thrombospondin 2 in the liver phenotype of ALGS.
Through the GWAS, we identified a SNP, rs7382539, located 14 kb upstream of
THBS2 on 6q27, that is associated with liver disease severity in ALGS patients with a JAG1 mutation. Although rs7382539 did not pass the cut-off for genome-wide significance, it did reach a suggestive level of significance for European populations. We identified 2 additional SNPs, rs4078087 and rs7451470, in strong linkage disequilibrium with each other and with rs7382539 (r
2 = 1) and highly associated with liver disease severity through imputation (
Figure 1). These 3 SNPs are located in regions with histone marks that are indicative of enhancer activity, including acetylated histone H3 lysine 27 and monomethylated histone H3 lysine 4.
30- Creyghton M.P.
- Cheng A.W.
- Welstead G.G.
- et al.
Histone H3K27ac separates active from poised enhancers and predicts developmental state.
This suggests that although the causal marker may not be in the coding sequence of
THBS2, it still potentially can play a role in regulating this gene.
By using a reporter mouse, we identified a pattern of expression for thrombospondin 2 in the portal tracts and biliary epithelium of the adult liver (
Figure 2) and showed that this pattern extends into stromal cells and is seen rarely in periportal hepatocytes in young mice (1 week old) (
Figure 3). Studies aiming to identify expression patterns of thrombospondin 2 during murine embryogenesis found high levels of expression in the heart, vessels, and mesenchyme.
37- Iruela-Arispe M.L.
- Liska D.J.
- Sage E.H.
- et al.
Differential expression of thrombospondin 1, 2, and 3 during murine development.
In addition, a role for thrombospondin 2 in angiogenesis has been well described for multiple tissue types.
21- Kyriakides T.R.
- Zhu Y.H.
- Smith L.T.
- et al.
Mice that lack thrombospondin 2 display connective tissue abnormalities that are associated with disordered collagen fibrillogenesis, an increased vascular density, and a bleeding diathesis.
, 31- Fears C.Y.
- Grammer J.R.
- Stewart Jr., J.E.
- et al.
Low-density lipoprotein receptor-related protein contributes to the antiangiogenic activity of thrombospondin-2 in a murine glioma model.
, 32- Volpert O.V.
- Tolsma S.S.
- Pellerin S.
- et al.
Inhibition of angiogenesis by thrombospondin-2.
, 33Matrix metalloproteinase-9 deficiency leads to prolonged foreign body response in the brain associated with increased IL-1beta levels and leakage of the blood-brain barrier.
, 34- Reinecke H.
- Robey T.E.
- Mignone J.L.
- et al.
Lack of thrombospondin-2 reduces fibrosis and increases vascularity around cardiac cell grafts.
, 35- Taylor D.K.
- Meganck J.A.
- Terkhorn S.
- et al.
Thrombospondin-2 influences the proportion of cartilage and bone during fracture healing.
A recent report identified a thinning of periportal vascular smooth muscle cells in mice haploinsufficient for
Jag1, suggesting that an angiogenic effect associated with Notch signaling also is seen in the liver.
38- Thakurdas S.M.
- Lopez M.F.
- Kakuda S.
- et al.
Jagged1 heterozygosity in mice results in a congenital cholangiopathy which is reversed by concomitant deletion of one copy of Poglut1 (Rumi).
We investigated a role for thrombospondin 2 in liver vasculature and found that adult mice lacking thrombospondin 2 had increased numbers of microvessels per portal tract whereas 1-week-old mice, but not adult mice, showed an increase in the number of arteries per portal tract (
Tables 2 and
3). This finding is in keeping with the results of Krady et al,
39- Krady M.M.
- Zeng J.
- Yu J.
- et al.
Thrombospondin-2 modulates extracellular matrix remodeling during physiological angiogenesis.
who found an increase in baseline arteriogenesis in
Thbs2-null animals in an induced-ischemia model of the hindlimb. Our finding within the 1-week-old livers raises the possibility that the rate at which arteries develop may be increased in the
Thbs2-null livers, but that once maturity is reached, the number of arteries either normalizes or is reduced slightly. Our results both substantiate our expression data and suggest that thrombospondin 2 may have combined roles in biliary development and liver vasculature. In addition, they suggest that it may be interesting to investigate thrombospondin 2 as a modifier of the heart and vascular phenotypes in ALGS. Indeed, recent studies have found an association of
THBS2 with a variety of heart conditions, including coronary artery disease,
40- Zhang X.J.
- Wei C.Y.
- Li W.B.
- et al.
Association between single nucleotide polymorphisms in thrombospondins genes and coronary artery disease: a meta-analysis.
, 41- Yamada Y.
- Matsui K.
- Takeuchi I.
- et al.
Association of genetic variants with coronary artery disease and ischemic stroke in a longitudinal population-based genetic epidemiological study.
thoracic aortic dissection,
42- Wang H.Q.
- Jian T.
- Wang F.
- et al.
Impact of thrombospondin-2 gene variations on the risk of thoracic aortic dissection in a Chinese Han population.
and heart failure with preserved ejection fraction.
43- Kimura Y.
- Izumiya Y.
- Hanatani S.
- et al.
High serum levels of thrombospondin-2 correlate with poor prognosis of patients with heart failure with preserved ejection fraction.
Biologically,
THBS2 represents an intriguing putative modifier of ALGS liver disease severity because it previously has been shown in binding assays that thrombospondin 2 is capable of physically binding to NOTCH3 and JAG1.
24- Meng H.
- Zhang X.
- Hankenson K.D.
- et al.
Thrombospondin 2 potentiates notch3/jagged1 signaling.
Here, we extended these observations and show that thrombospondin 2 also interacts with NOTCH2 (
Figure 5) and inhibits the expression of downstream Notch target genes in the presence of JAG1 (
Figure 6). Moreover, we determined that a specific
THBS2 regulatory domain, the type I repeat, is required for this function (
Figure 7). Type I repeats are known to be required for binding to CD36, a glycoprotein expressed on platelets, and for the antiangiogenic function of thrombospondin 2.
44- Dawson D.W.
- Pearce S.F.
- Zhong R.
- et al.
CD36 mediates the In vitro inhibitory effects of thrombospondin-1 on endothelial cells.
Given the increased numbers of microvessels and arteries in
Thbs2-null adult and 1-week-old mice, respectively, these data suggest that thrombospondin 2 may function via Notch signaling in the liver vasculature such that enhanced thrombospondin 2 levels could decrease Notch signaling and impair angiogenesis during liver development, thereby modifying the liver phenotype in ALGS. The precise molecular mechanism of how thrombospondin 2 blocks NOTCH2 function remains to be determined. Further studies will be needed to distinguish between competitive inhibition of JAG1–NOTCH2 inhibition by thrombospondin 2 vs the allosteric effect of thrombospondin 2 on one of the binding partners.
We acknowledge that our study had several limitations. First, because of the rarity of the syndrome, our patient cohort was small and the marker identified in the GWAS at 6q27 did not reach genome-wide statistical significance. However, given the known interaction of thrombospondin 2 with JAG1/NOTCH3, we were interested in further pursuing functional studies in the liver.
24- Meng H.
- Zhang X.
- Hankenson K.D.
- et al.
Thrombospondin 2 potentiates notch3/jagged1 signaling.
Second, we were unable to show a significant correlation between SNP genotype and
THBS2 expression level. It is possible that the GWAS-identified SNP, located upstream of the
THBS2 locus in a region with histone acetylation signatures, functions to regulate
THBS2 expression in a tissue-specific manner. Investigation of this possibility is outside the scope of the current study, but might include chromosome conformation capture, more detailed analysis of patient liver samples, and studies in animal models. Despite these limitations, we believe that the expression data and functional studies provide compelling evidence that thrombospondin 2 could modulate JAG1–NOTCH2 interactions, thereby modifying liver disease severity in ALGS.
Ultimately, these data provide genetic, expression, and functional evidence to support a role for
THBS2 as a modifier of liver disease severity in ALGS. These findings could have significant clinical applications when the current treatment course of liver disease is unpredictable,
9- Kamath B.M.
- Munoz P.S.
- Bab N.
- et al.
A longitudinal study to identify laboratory predictors of liver disease outcome in Alagille syndrome.
and the identification of a genetic modifier may help in identifying patient populations that will require liver transplantation at an earlier age.
Article info
Publication history
Published online: May 27, 2016
Accepted:
May 17,
2016
Received:
March 10,
2016
Footnotes
Conflicts of interest The authors disclose no conflicts.
Funding This work was supported in part by the Childhood Liver Disease Research Network grant U01-DK062481 (K.M.L.); National Institutes of Health grants R01-DK081702-05 (N.B.S.) and R01-AR049682 (K.D.H.); National Institutes of Health training grant T32-HG000046 (E.A.T.); and VA Merit Award BX000375 (M.M.W.). The Childhood Liver Disease Research Network Data Coordinating Center is supported by U01 DK062456 from the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK). This work also was supported by the Fred and Suzanne Biesecker Liver Center at the Children’s Hospital of Philadelphia.
Copyright
© 2016 The Authors. Published by Elsevier Inc. on behalf of the AGA Institute.