Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 304
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 111(8): 1656-1672, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39043182

ABSTRACT

Pathogenic variants in the JAG1 gene are a primary cause of the multi-system disorder Alagille syndrome. Although variant detection rates are high for this disease, there is uncertainty associated with the classification of missense variants that leads to reduced diagnostic yield. Consequently, up to 85% of reported JAG1 missense variants have uncertain or conflicting classifications. We generated a library of 2,832 JAG1 nucleotide variants within exons 1-7, a region with a high number of reported missense variants, and designed a high-throughput assay to measure JAG1 membrane expression, a requirement for normal function. After calibration using a set of 175 known or predicted pathogenic and benign variants included within the variant library, 486 variants were characterized as functionally abnormal (n = 277 abnormal and n = 209 likely abnormal), of which 439 (90.3%) were missense. We identified divergent membrane expression occurring at specific residues, indicating that loss of the wild-type residue itself does not drive pathogenicity, a finding supported by structural modeling data and with broad implications for clinical variant classification both for Alagille syndrome and globally across other disease genes. Of 144 uncertain variants reported in patients undergoing clinical or research testing, 27 had functionally abnormal membrane expression, and inclusion of our data resulted in the reclassification of 26 to likely pathogenic. Functional evidence augments the classification of genomic variants, reducing uncertainty and improving diagnostics. Inclusion of this repository of functional evidence during JAG1 variant reclassification will significantly affect resolution of variant pathogenicity, making a critical impact on the molecular diagnosis of Alagille syndrome.


Subject(s)
Alagille Syndrome , Jagged-1 Protein , Mutation, Missense , Alagille Syndrome/genetics , Jagged-1 Protein/genetics , Humans , Exons/genetics
2.
Proc Natl Acad Sci U S A ; 119(50): e2201097119, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36469766

ABSTRACT

Despite the robust healing capacity of the liver, regenerative failure underlies numerous hepatic diseases, including the JAG1 haploinsufficient disorder, Alagille syndrome (ALGS). Cholestasis due to intrahepatic duct (IHD) paucity resolves in certain ALGS cases but fails in most with no clear mechanisms or therapeutic interventions. We find that modulating jag1b and jag2b allele dosage is sufficient to stratify these distinct outcomes, which can be either exacerbated or rescued with genetic manipulation of Notch signaling, demonstrating that perturbations of Jag/Notch signaling may be causal for the spectrum of ALGS liver severities. Although regenerating IHD cells proliferate, they remain clustered in mutants that fail to recover due to a blunted elevation of Notch signaling in the distal-most IHD cells. Increased Notch signaling is required for regenerating IHD cells to branch and segregate into the peripheral region of the growing liver, where biliary paucity is commonly observed in ALGS. Mosaic loss- and-gain-of-function analysis reveals Sox9b to be a key Notch transcriptional effector required cell autonomously to regulate these cellular dynamics during IHD regeneration. Treatment with a small-molecule putative Notch agonist stimulates Sox9 expression in ALGS patient fibroblasts and enhances hepatic sox9b expression, rescues IHD paucity and cholestasis, and increases survival in zebrafish mutants, thereby providing a proof-of-concept therapeutic avenue for this disorder.


Subject(s)
Alagille Syndrome , Bile Ducts, Intrahepatic , Signal Transduction , Animals , Humans , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Mosaicism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Regeneration , Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/pathology , Fibroblasts
3.
Nat Immunol ; 13(12): 1213-21, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23086448

ABSTRACT

CD46 is a complement regulator with important roles related to the immune response. CD46 functions as a pathogen receptor and is a potent costimulator for the induction of interferon-γ (IFN-γ)-secreting effector T helper type 1 (T(H)1) cells and their subsequent switch into interleukin 10 (IL-10)-producing regulatory T cells. Here we identified the Notch family member Jagged1 as a physiological ligand for CD46. Furthermore, we found that CD46 regulated the expression of Notch receptors and ligands during T cell activation and that disturbance of the CD46-Notch crosstalk impeded induction of IFN-γ and switching to IL-10. Notably, CD4(+) T cells from CD46-deficient patients and patients with hypomorphic mutations in the gene encoding Jagged1 (Alagille syndrome) failed to mount appropriate T(H)1 responses in vitro and in vivo, which suggested that CD46-Jagged1 crosstalk is responsible for the recurrent infections in subpopulations of these patients.


Subject(s)
Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymphocyte Activation , Membrane Cofactor Protein/metabolism , Membrane Proteins/metabolism , Th1 Cells/immunology , Adult , Alagille Syndrome/genetics , Alagille Syndrome/immunology , Animals , Cells, Cultured , Child , Child, Preschool , Humans , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Jagged-1 Protein , Mice , Mice, SCID , Mice, Transgenic , RNA Interference , RNA, Small Interfering , Serrate-Jagged Proteins , Th1 Cells/metabolism , alpha Catenin/genetics
4.
Hepatology ; 78(5): 1337-1351, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37021797

ABSTRACT

BACKGROUND AND AIMS: Paucity of intrahepatic bile ducts (BDs) is caused by various etiologies and often leads to cholestatic liver disease. For example, in patients with Alagille syndrome (ALGS), which is a genetic disease primarily caused by mutations in jagged 1 ( JAG1) , BD paucity often results in severe cholestasis and liver damage. However, no mechanism-based therapy exists to restore the biliary system in ALGS or other diseases associated with BD paucity. Based on previous genetic observations, we investigated whether postnatal knockdown of the glycosyltransferase gene protein O -glucosyltransferase 1 ( Poglut1) can improve the ALGS liver phenotypes in several mouse models generated by removing one copy of Jag1 in the germline with or without reducing the gene dosage of sex-determining region Y-box 9 in the liver. APPROACH AND RESULTS: Using an ASO established in this study, we show that reducing Poglut1 levels in postnatal livers of ALGS mouse models with moderate to profound biliary abnormalities can significantly improve BD development and biliary tree formation. Importantly, ASO injections prevent liver damage in these models without adverse effects. Furthermore, ASO-mediated Poglut1 knockdown improves biliary tree formation in a different mouse model with no Jag1 mutations. Cell-based signaling assays indicate that reducing POGLUT1 levels or mutating POGLUT1 modification sites on JAG1 increases JAG1 protein level and JAG1-mediated signaling, suggesting a likely mechanism for the observed in vivo rescue. CONCLUSIONS: Our preclinical studies establish ASO-mediated POGLUT1 knockdown as a potential therapeutic strategy for ALGS liver disease and possibly other diseases associated with BD paucity.


Subject(s)
Alagille Syndrome , Glycosyltransferases , Liver , Oligonucleotides, Antisense , Animals , Mice , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Alagille Syndrome/pathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Calcium-Binding Proteins/genetics , Cholestasis/genetics , Cholestasis/metabolism , Gene Silencing , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Phenotype , Serrate-Jagged Proteins/genetics , Serrate-Jagged Proteins/metabolism
5.
Liver Int ; 44(2): 541-558, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38014627

ABSTRACT

BACKGROUND & AIMS: Alagille syndrome (ALGS) manifests with peripheral intrahepatic bile duct (IHBD) paucity, which can spontaneously resolve. In a model for ALGS, Jag1Ndr/Ndr mice, this occurs with distinct architectural mechanisms in hilar and peripheral IHBDs. Here, we investigated region-specific IHBD characteristics and addressed whether IGF1, a cholangiocyte mitogen that is downregulated in ALGS and in Jag1Ndr/Ndr mice, can improve biliary outcomes. METHODS: Intrahepatic cholangiocyte organoids (ICOs) were derived from hilar and peripheral adult Jag1+/+ and Jag1Ndr/Ndr livers (hICOs and pICOs, respectively). ICOs were grown in Matrigel or microwell arrays, and characterized using bulk RNA sequencing, immunofluorescence, and high throughput analyses of nuclear sizes. ICOs were treated with IGF1, followed by analyses of growth, proliferation, and death. CellProfiler and Python scripts were custom written for image analyses. Key results were validated in vivo by immunostaining. RESULTS: Cell growth assays and transcriptomics demonstrated that Jag1Ndr/Ndr ICOs were less proliferative than Jag1+/+ ICOs. IGF1 specifically rescued survival and growth of Jag1Ndr/Ndr pICOs. Jag1Ndr/Ndr hICOs were the least proliferative, with lower Notch signalling and an enrichment of hepatocyte signatures and IGF uptake/transport pathways. In vitro (Jag1Ndr/Ndr hICOs) and in vivo (Jag1Ndr/Ndr hilar portal tracts) analyses revealed ectopic HNF4a+ hepatocytes. CONCLUSIONS: Hilar and peripheral Jag1Ndr/Ndr ICOs exhibit differences in Notch signalling status, proliferation, and cholangiocyte commitment which may result in cholangiocyte-to-hepatocyte transdifferentiation. While Jag1Ndr/Ndr pICOs can be rescued by IGF1, hICOs are unresponsive, perhaps due to their hepatocyte-like state and/or expression of IGF transport components. IGF1 represents a potential therapeutic for peripheral bile ducts.


Subject(s)
Alagille Syndrome , Biliary Tract , Mice , Animals , Alagille Syndrome/genetics , Bile Ducts , Bile Ducts, Intrahepatic , Organoids/metabolism
6.
Pharmacol Res ; 199: 107006, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38000562

ABSTRACT

Gallbladder and biliary diseases (GBDs) are one of the most common digestive diseases. The connections between GBDs and several organs other than the liver have gradually surfaced accompanied by the changes in people's diet structure and the continuous improvement of medical diagnosis technology. Among them, cholecardia syndrome that takes the heart as the important target of GBDs complications has been paid close attention. However, there are still no systematic report about its corresponding clinical manifestations and pathogenesis. This review summarized recent reported types of cholecardia syndrome and found that arrhythmia, myocardial injury, acute coronary syndrome and heart failure are common in the general population. Besides, the clinical diagnosis rate of intrahepatic cholestasis of pregnancy (ICP) and Alagille syndrome associated with gene mutation is also increasing. Accordingly, the underlying pathogenesis including abnormal secretion of bile acid, gene mutation, translocation and deletion (JAG1, NOTCH2, ABCG5/8 and CYP7A1), nerve reflex and autonomic neuropathy were further revealed. Finally, the potential treatment measures and clinical medication represented by ursodeoxycholic acid were summarized to provide assistance for clinical diagnosis and treatment.


Subject(s)
Alagille Syndrome , Cholestasis, Intrahepatic , Pregnancy Complications , Female , Pregnancy , Humans , Alagille Syndrome/complications , Alagille Syndrome/diagnosis , Alagille Syndrome/genetics , Cholestasis, Intrahepatic/complications , Cholestasis, Intrahepatic/drug therapy , Ursodeoxycholic Acid/therapeutic use
7.
Curr Opin Pediatr ; 36(5): 524-536, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38957097

ABSTRACT

PURPOSE OF REVIEW: To highlight recent advances in pediatric cholestatic liver disease, including promising novel prognostic markers and new therapies. FINDINGS: Additional genetic variants associated with the progressive familial intrahepatic cholestasis (PFIC) phenotype and new genetic cholangiopathies, with an emerging role of ciliopathy genes, are increasingly being identified. Genotype severity predicts outcomes in bile salt export pump (BSEP) deficiency, and post-biliary diversion serum bile acid levels significantly affect native liver survival in BSEP and progressive familial intrahepatic cholestasis type 1 (FIC1 deficiency) patients. Heterozygous variants in the MDR3 gene have been associated with various cholestatic liver disease phenotypes in adults. Ileal bile acid transporter (IBAT) inhibitors, approved for pruritus in PFIC and Alagille Syndrome (ALGS), have been associated with improved long-term quality of life and event-free survival. SUMMARY: Next-generation sequencing (NGS) technologies have revolutionized diagnostic approaches, while discovery of new intracellular signaling pathways show promise in identifying therapeutic targets and personalized strategies. Bile acids may play a significant role in hepatic damage progression, suggesting their monitoring could guide cholestatic liver disease management. IBAT inhibitors should be incorporated early into routine management algorithms for pruritus. Data are emerging as to whether IBAT inhibitors are impacting disease biology and modifying the natural history of the cholestasis.


Subject(s)
Cholestasis, Intrahepatic , Humans , Child , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/diagnosis , Cholestasis, Intrahepatic/therapy , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , Bile Acids and Salts/metabolism , Bile Acids and Salts/blood , Alagille Syndrome/diagnosis , Alagille Syndrome/genetics , Alagille Syndrome/therapy , Phenotype , Prognosis , High-Throughput Nucleotide Sequencing
8.
J Gastroenterol Hepatol ; 39(5): 964-974, 2024 May.
Article in English | MEDLINE | ID: mdl-38323732

ABSTRACT

BACKGROUND AND AIM: Advances in molecular genetics have uncovered causative genes responsible for neonatal cholestasis. Panel-based next-generation sequencing has been used clinically in infants with neonatal cholestasis. We aimed to evaluate the clinical application of single-gene testing and next-generation sequencing and to develop a diagnostic algorithm for neonatal intrahepatic cholestasis. METHODS: From January 2010 to July 2021, patients suspected of having neonatal intrahepatic cholestasis were tested at the Seoul National University Hospital. If there was a clinically suspected disease, single-gene testing was performed. Alternatively, if it was clinically difficult to differentiate, a neonatal cholestasis gene panel test containing 34 genes was performed. RESULTS: Of the total 148 patients examined, 49 (33.1%) were received a confirmed genetic diagnosis, including 14 with Alagille syndrome, 14 with neonatal intrahepatic cholestasis caused by citrin deficiency, 7 with Dubin-Johnson syndrome, 5 with arthrogryposis-renal dysfunction-cholestasis syndrome, 5 with progressive familial intrahepatic cholestasis type II, 1 with Rotor syndrome, 1 with Niemann-Pick disease type C, 1 with Kabuki syndrome, and 1 with Phenylalanyl-tRNA synthetase subunit alpha mutation. Sixteen novel pathogenic or likely pathogenic variants of neonatal cholestasis were observed in this study. Based on the clinical characteristics and laboratory findings, we developed a diagnostic algorithm for neonatal intrahepatic cholestasis by integrating single-gene testing and next-generation sequencing. CONCLUSIONS: Alagille syndrome and neonatal intrahepatic cholestasis caused by citrin deficiency were the most common diseases associated with genetic neonatal cholestasis. Single-gene testing and next-generation sequencing are important and complementary tools for the diagnosis of genetic neonatal cholestasis.


Subject(s)
Algorithms , Cholestasis, Intrahepatic , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/diagnosis , High-Throughput Nucleotide Sequencing/methods , Infant, Newborn , Genetic Testing/methods , Male , Female , Alagille Syndrome/genetics , Alagille Syndrome/diagnosis , Infant
9.
BMC Pediatr ; 24(1): 532, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164659

ABSTRACT

BACKGROUND: Alagille syndrome (ALGS) is a multisystem genetic disorder frequently characterized by hepatic manifestations. This study analyzed the clinical, pathological, and molecular genetic features of ALGS to improve the efficiency of clinical diagnosis. METHODS: We retrospectively analyzed the clinical manifestations, pathological examination findings, and genetic testing results of 17 children diagnosed with ALGS based on the revised criteria and hospitalized at our center from January 2012 to January 2022. RESULTS: The clinical manifestations are as follows: Cholestasis (16/17, 94%), characteristic facies (15/17, 88%), heart disease (12/16, 75%), butterfly vertebrae (12/17, 71%) and posterior embryotoxon (7/12, 58%). Among the 15 patients who underwent liver pathology examination, 13 (87%) were found to have varying degrees of bile duct paucity. Genetic testing was performed on 15 children, and pathogenic variants of the jagged canonical Notch ligand 1 (JAG1) gene were identified in 13 individuals, including 4 novel variants. No pathogenic variant in the notch homolog 2 (NOTCH2) gene were identified, and 2 children exhibited none of the aforementioned gene pathogenic variants. The median follow-up duration was 7 years. Of the remaining 15 patients (excluding 2 lost to follow-up), 11 remained stable, 4 deteriorated, and no patient died during the follow-up period. CONCLUSIONS: Among children diagnosed with ALGS, cholestasis stands as the most common feature. To minimize the risk of misdiagnosis, genetic testing should be performed on children exhibiting cholestasis, followed by the application of the revised diagnostic criteria for ALGS. While pharmacological therapy has shown effectiveness for ALGS patients, liver transplantation may be considered in instances of severe pruritus.


Subject(s)
Alagille Syndrome , Genetic Testing , Jagged-1 Protein , Humans , Alagille Syndrome/genetics , Alagille Syndrome/diagnosis , Male , Female , Retrospective Studies , Child, Preschool , Infant , Jagged-1 Protein/genetics , Child , Cholestasis/genetics
10.
Hepatology ; 75(3): 567-583, 2022 03.
Article in English | MEDLINE | ID: mdl-34569629

ABSTRACT

BACKGROUND AND AIMS: Alagille Syndrome (ALGS) is a congenital disorder caused by mutations in the Notch ligand gene JAGGED1, leading to neonatal loss of intrahepatic duct (IHD) cells and cholestasis. Cholestasis can resolve in certain patients with ALGS, suggesting regeneration of IHD cells. However, the mechanisms driving IHD cell regeneration following Jagged loss remains unclear. Here, we show that cholestasis due to developmental loss of IHD cells can be consistently phenocopied in zebrafish with compound jagged1b and jagged2b mutations or knockdown. APPROACH AND RESULTS: Leveraging the transience of jagged knockdown in juvenile zebrafish, we find that resumption of Jagged expression leads to robust regeneration of IHD cells through a Notch-dependent mechanism. Combining multiple lineage tracing strategies with whole-liver three-dimensional imaging, we demonstrate that the extrahepatic duct (EHD) is the primary source of multipotent progenitors that contribute to the regeneration, but not to the development, of IHD cells. Hepatocyte-to-IHD cell transdifferentiation is possible but rarely detected. Progenitors in the EHD proliferate and migrate into the liver with Notch signaling loss and differentiate into IHD cells if Notch signaling increases. Tissue-specific mosaic analysis with an inducible dominant-negative Fgf receptor suggests that Fgf signaling from the surrounding mesenchymal cells maintains this extrahepatic niche by directly preventing premature differentiation and allocation of EHD progenitors to the liver. Indeed, transcriptional profiling and functional analysis of adult mouse EHD organoids uncover their distinct differentiation and proliferative potential relative to IHD organoids. CONCLUSIONS: Our data show that IHD cells regenerate upon resumption of Jagged/Notch signaling, from multipotent progenitors originating from an Fgf-dependent extrahepatic stem cell niche. We posit that if Jagged/Notch signaling is augmented, through normal stochastic variation, gene therapy, or a Notch agonist, regeneration of IHD cells in patients with ALGS may be enhanced.


Subject(s)
Alagille Syndrome , Bile Ducts, Extrahepatic , Bile Ducts, Intrahepatic , Calcium-Binding Proteins , Jagged-1 Protein , Liver Regeneration/physiology , Receptors, Notch/metabolism , Zebrafish Proteins , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Animals , Bile Ducts, Extrahepatic/growth & development , Bile Ducts, Extrahepatic/physiology , Bile Ducts, Intrahepatic/growth & development , Bile Ducts, Intrahepatic/physiology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Transdifferentiation , Disease Models, Animal , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Liver/growth & development , Liver/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
11.
J Clin Gastroenterol ; 57(7): 686-693, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37022007

ABSTRACT

Children with cholestatic liver diseases are increasingly living into adulthood, thanks to innovations in medical and surgical therapies. The excellent outcomes observed in pediatric liver transplantation for diseases, such as biliary atresia, have transformed the life trajectory of children born with once-fatal liver diseases. The evolution of molecular genetic testing, has helped expedite the diagnosis of other cholestatic disorders, improving the clinical management, disease prognosis, and family planning for inherited disorders, such as progressive familial intrahepatic cholestasis and bile acid synthesis disorders. The expanding list of therapeutics, including bile acids and the newer ileal bile acid transport inhibitors, has also helped slow the progression of disease and improve the quality of life for certain diseases, like Alagille syndrome. More and more children with cholestatic disorders are expected to require care from adult providers familiar with the natural history and potential complications of these childhood diseases. The aim of this review is to bridge the gap between pediatric and adult care in children with cholestatic disorders. The present review addresses the epidemiology, clinical features, diagnostic testing, treatment, prognosis, and transplant outcomes of 4 hallmark childhood cholestatic liver diseases: biliary atresia, Alagille syndrome, progressive familial intrahepatic cholestasis, and bile acid synthesis disorders.


Subject(s)
Alagille Syndrome , Biliary Atresia , Cholestasis, Intrahepatic , Cholestasis , Gastroenterologists , Child , Adult , Humans , Biliary Atresia/diagnosis , Biliary Atresia/therapy , Alagille Syndrome/diagnosis , Alagille Syndrome/genetics , Alagille Syndrome/therapy , Quality of Life , Cholestasis/diagnosis , Cholestasis, Intrahepatic/diagnosis , Cholestasis, Intrahepatic/epidemiology , Cholestasis, Intrahepatic/genetics , Bile Acids and Salts
12.
Curr Gastroenterol Rep ; 25(11): 344-354, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37651067

ABSTRACT

PURPOSE OF REVIEW: Cholestasis is characterized by a conjugated hyperbilirubinemia secondary to impaired bile synthesis, transport, or excretion from the liver. It is always pathologic and can be indicative of an underlying hepatobiliary, genetic, or metabolic disorder, several of which require timely diagnosis to ensure proper management and optimal outcomes. This review provides an overview of the evaluation of cholestasis with a focus on current and emerging treatment strategies. RECENT FINDINGS: Increased accessibility of next generation sequencing (NGS) allows for utilization of genetic testing early in the diagnostic process. This may alter the clinical algorithm for diagnosis of cholestatic disorders. An enhanced understanding of the underlying pathophysiology may help guide future development of targeted therapies, such as ileal bile acid transporter (IBAT) inhibitors. These were recently approved for treatment of cholestatic pruritus in patients with Alagille syndrome and Progressive Familial Intrahepatic Cholestasis. Current management of cholestasis is aimed at the biochemical consequences of impaired bile flow, including malnutrition, pruritus, and progressive fibrosis. NGS has led to an enhanced understanding of biliary pathology and may guide development of future treatment modalities based on specific gene mutations. Rapid discernment of the underlying etiology is essential as new treatment modalities emerge.


Subject(s)
Alagille Syndrome , Cholestasis, Intrahepatic , Cholestasis , Humans , Child , Infant , Child, Preschool , Cholestasis/complications , Cholestasis/diagnosis , Cholestasis, Intrahepatic/diagnosis , Alagille Syndrome/complications , Alagille Syndrome/diagnosis , Alagille Syndrome/genetics , Pruritus/diagnosis , Pruritus/etiology , Pruritus/therapy
13.
PLoS Genet ; 16(11): e1008802, 2020 11.
Article in English | MEDLINE | ID: mdl-33226994

ABSTRACT

The clinical evaluation of a genetic syndrome relies upon recognition of a characteristic pattern of signs or symptoms to guide targeted genetic testing for confirmation of the diagnosis. However, individuals displaying a single phenotype of a complex syndrome may not meet criteria for clinical diagnosis or genetic testing. Here, we present a phenome-wide association study (PheWAS) approach to systematically explore the phenotypic expressivity of common and rare alleles in genes associated with four well-described syndromic diseases (Alagille (AS), Marfan (MS), DiGeorge (DS), and Noonan (NS) syndromes) in the general population. Using human phenotype ontology (HPO) terms, we systematically mapped 60 phenotypes related to AS, MS, DS and NS in 337,198 unrelated white British from the UK Biobank (UKBB) based on their hospital admission records, self-administrated questionnaires, and physiological measurements. We performed logistic regression adjusting for age, sex, and the first 5 genetic principal components, for each phenotype and each variant in the target genes (JAG1, NOTCH2 FBN1, PTPN1 and RAS-opathy genes, and genes in the 22q11.2 locus) and performed a gene burden test. Overall, we observed multiple phenotype-genotype correlations, such as the association between variation in JAG1, FBN1, PTPN11 and SOS2 with diastolic and systolic blood pressure; and pleiotropy among multiple variants in syndromic genes. For example, rs11066309 in PTPN11 was significantly associated with a lower body mass index, an increased risk of hypothyroidism and a smaller size for gestational age, all in concordance with NS-related phenotypes. Similarly, rs589668 in FBN1 was associated with an increase in body height and blood pressure, and a reduced body fat percentage as observed in Marfan syndrome. Our findings suggest that the spectrum of associations of common and rare variants in genes involved in syndromic diseases can be extended to individual phenotypes within the general population.


Subject(s)
Biological Variation, Population/genetics , Genetic Association Studies/methods , Genome-Wide Association Study/methods , Alagille Syndrome/genetics , Alleles , DiGeorge Syndrome/genetics , Female , Gene Frequency/genetics , Genetic Predisposition to Disease/genetics , Genetic Testing/methods , Genetic Variation/genetics , Humans , Male , Marfan Syndrome/genetics , Noonan Syndrome/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , United Kingdom , White People/genetics
14.
Int J Mol Sci ; 24(14)2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37511516

ABSTRACT

Alagille syndrome (ALGS) is a multisystem condition characterized by cholestasis and bile duct paucity on liver biopsy and variable involvement of the heart, skeleton, eyes, kidneys, and face and caused by pathogenic variants in the JAG1 or NOTCH2 gene. The variable expressivity of the clinical phenotype and the lack of genotype-phenotype correlations lead to significant diagnostic difficulties. Here we present an analysis of 18 patients with cholestasis who were diagnosed with ALGS. We used an NGS panel targeting coding exons of 52 genes, including the JAG1 and NOTCH2 genes. Sanger sequencing was used to verify the mutation in the affected individuals and family members. The specific facial phenotype was seen in 16/18 (88.9%). Heart defects were seen in 8/18 (44.4%) patients (pulmonary stenosis in 7/8). Butterfly vertebrae were seen in 5/14 (35.7%) patients. Renal involvement was detected in 2/18 (11.1%) cases-one patient had renal cysts, and one had obstructive hydronephrosis. An ophthalmology examination was performed on 12 children, and only one had posterior embryotoxon (8.3%). A percutaneous liver biopsy was performed in nine cases. Bile duct paucity was detected in six/nine cases (66.7%). Two patients required liver transplantation because of cirrhosis. We identified nine novel variants in the JAG1 gene-eight frameshift variants (c.1619_1622dupGCTA (p.Tyr541X), c.1160delG (p.Gly387fs), c.964dupT (p.C322fs), c.120delG (p.L40fs), c.1984dupG (p.Ala662Glyfs), c.3168_3169delAG (p.R1056Sfs*51), c.2688delG (p.896CysfsTer49), c.164dupG (p.Cys55fs)) and one missense variant, c.2806T > G (p.Cys936Gly). None of the patients presented with NOTCH2 variants. In accordance with the classical criteria, only six patients could meet the diagnostic criteria in our cohort without genetic analysis. Genetic testing is important in the diagnosis of ALGS and can help differentiate it from other types of cholestasis.


Subject(s)
Alagille Syndrome , Cholestasis , Humans , Alagille Syndrome/complications , Alagille Syndrome/genetics , Cholestasis/genetics , Mutation , Mutation, Missense , Phenotype , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism
15.
Gastroenterol Nurs ; 46(6): 436-444, 2023.
Article in English | MEDLINE | ID: mdl-37581873

ABSTRACT

Alagille syndrome is a rare and complex pleiotropic multisystem disorder caused by an autosomal dominant genetic mutation of JAG1 (90%) and NOTCH2 (1%-2%) genes located on the short arm of chromosome 20. This case is reported as per the CA se RE ports (CARE) guidelines (2013). A 14-year-old boy who is a known case of chronic cholestatic liver disease of neonatal onset, was diagnosed with Alagille syndrome as evident from a NOTCH 2 mutation in genetic analysis and paucity of intrahepatic bile ducts on biopsy. He presented with portal hypertension, growth failure, and persistent hyperbilirubinemia. This case highlights the gamut of multisystem dysfunctions faced by this child. He is currently on conservative management and worked up for liver transplantation. The condition is often rare and challenging due to the multisystem pathogenesis. Thus, the nursing care is also multifaceted. This case study identified relevant North American Nursing Diagnosis Association (NANDA) Classification, Nursing Interventions Classification (NIC), and Nursing Outcomes Classification (NOC) concepts to describe care of children with Alagille syndrome based on actual patient data.


Subject(s)
Alagille Syndrome , Standardized Nursing Terminology , Male , Child , Infant, Newborn , Humans , Adolescent , Alagille Syndrome/diagnosis , Alagille Syndrome/therapy , Alagille Syndrome/genetics , Nursing Diagnosis , Patient Care
16.
Hum Mutat ; 43(12): 1824-1828, 2022 12.
Article in English | MEDLINE | ID: mdl-35819173

ABSTRACT

Pathogenic variants in JAG1 are known to cause Alagille syndrome (ALGS), a disorder that primarily affects the liver, lung, kidney, and skeleton. Whereas cardiac symptoms are also frequently observed in ALGS, thoracic aortic aneurysms have only been reported sporadically in postmortem autopsies. We here report two families with segregating JAG1 variants that present with isolated aneurysmal disease, as well as the first histological evaluation of aortic aneurysm tissue of a JAG1 variant carrier. Our observations shed more light on the pathomechanisms behind aneurysm formation in JAG1 variant harboring individuals and underline the importance of cardiovascular imaging in the clinical follow-up of such individuals.


Subject(s)
Alagille Syndrome , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Alagille Syndrome/genetics , Heart , Calcium-Binding Proteins
17.
Liver Int ; 42(8): 1836-1848, 2022 08.
Article in English | MEDLINE | ID: mdl-35567760

ABSTRACT

BACKGROUND AND AIMS: Alagille syndrome (ALGS) type 2 caused by mutations in NOTCH2 has genotypic and phenotypic heterogeneity. Diagnosis in some atypical patients with isolated hepatic presentation could be missed. METHODS: Using 2087 patients with paediatric liver manifestations, NOTCH2 allele frequencies, in-silico prediction, protein domains and clinical features were analysed to define the pathogenicity of NOTCH2 variants for diagnosis of ALGS type 2. RESULTS: Among 2087 patients with paediatric liver manifestations, significantly more NOTCH2 variants were absent in gnomAD in patients with elevated γ-glutamyltransferase (GGT) (p = .041). Significantly more NOTCH2 variants which were absent in gnomAD were located in protein functional domains (p = .038). When missense variants were absent in gnomAD and predicted to be pathogenic by at least three out of seven in-silico tools, they were found to be significantly associated with liver manifestations with elevated GGT (p = .003). Comparing this to patients with likely benign (LB) variants, the patients with likely-pathogenic (LP) variants have significantly more liver manifestations with elevated GGT (p = .0001). Significantly more patients with LP variants had extra-hepatic phenotypes of ALGS compared with those patients with LB variants (p = .0004). CONCLUSION: When NOTCH2 variants are absent in gnomAD, null variants and missense variants which were predicted to be pathogenic by at least three in-silico tools could be considered pathogenic in patients with high GGT chronic liver diseases.


Subject(s)
Alagille Syndrome , Receptor, Notch2 , Alagille Syndrome/diagnosis , Alagille Syndrome/genetics , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Mutation , Phenotype , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Virulence
18.
J Pediatr Gastroenterol Nutr ; 74(1): 96-103, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34694263

ABSTRACT

OBJECTIVE: To evaluate neurodevelopmental status among children with inherited cholestatic liver diseases with native liver and variables predictive of impairment. METHODS: Participants with Alagille syndrome (ALGS), progressive familial intrahepatic cholestasis (PFIC), and alpha 1 antitrypsin deficiency (A1AT) enrolled in a longitudinal, multicenter study and completed the Wechsler Preschool and Primary Scale of Intelligence-III or Intelligence Scale for Children-IV. Full Scale Intelligence Quotient (FSIQ) was analyzed continuously and categorically (>100, 85-99, 70-84, <70). Univariate linear regression was performed to study association between FSIQ and risk factors, stratified by disease. RESULTS: Two hundred and fifteen completed testing (ALGS n = 70, PFIC n = 43, A1AT n = 102); median age was 7.6 years (3.0-16.9). Mean FSIQ in ALGS was lower than A1AT (94 vs 101, P = 0.01). Frequency of FSIQ < 85 (>1 standard deviation [SD] below average) was highest in ALGS (29%) versus 18.6% in PFIC and 12.8% in A1AT, and was greater than expected in ALGS based on normal distribution (29% vs 15.9%, P = 0.003). ALGS scored significantly lower than test norms in almost all Wechsler composites; A1AT scored lower on Working Memory and Processing Speed; PFIC was not different from test norms. Total bilirubin, alkaline phosphatase, albumin, hemoglobin, and parental education were significantly associated with FSIQ. CONCLUSIONS: Patients with ALGS are at increased risk of lower FSIQ, whereas our data suggest A1AT and PFIC are not. A1AT and ALGS appear vulnerable to working memory and processing speed deficits suggestive of attention/executive function impairment. Malnutrition, liver disease severity, and sociodemographic factors appear related to FSIQ deficits, potentially identifying targets for early interventions.


Subject(s)
Alagille Syndrome , Cholestasis, Intrahepatic , Cholestasis , Alagille Syndrome/complications , Alagille Syndrome/genetics , Child , Child, Preschool , Humans , Wechsler Scales
19.
BMC Pediatr ; 22(1): 688, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36447191

ABSTRACT

BACKGROUND: Alagille syndrome (ALGS) is a multisystem disorder with variable clinical penetrance. The genes responsible for this disease are JAGGED1 (JAG1) and NOTCH2. Clinical data of this disease are limited in China. The purpose of this study was to enrich the present data of Chinese children with Alagille syndrome by summarizing the clinical characteristics and genetic variations of these cases. From January 2011 to February 2022, 10 children were diagnosed with ALGS. The organs involved in ALGS were as follows: liver (10, 100%); heart (7, 70%); characteristic facial features (7, 70%); skeleton (4, 40%); brain (1,10%) and kidney (3, 30%). Four patients (40%) were small for gestational age. The main clinical manifestations were cholestasis, heart disease, and facial features. The median total bilirubin, direct bilirubin, and total bile acid levels were 138.75 µmol/L (normal, 3.4-20.5 µmol/L), 107.25 µmol/L (normal, 0-8.6 µmol/L), and 110.65 µmol/L (normal, 0.5-10.0 µmol/L), respectively. The median value of gamma-glutamyltranspeptidase was 223 U/L (normal, 9-64 U/L). Six (60%) children had hypercholesteremia. Eight different JAG1 gene variations and one NOTCH2 gene pathogenic variant in the 10 Chinese ALGS patients were identified. CONCLUSION: Cholestasis was the most common initial presenting symptom in Chinese ALGS pediatric patients. Pathogenic variants in JAG1 and NOTCH2 are the primary mutations in Chinese children with ALGS, but we had our own unique variant spectrum. ALGS should be considered for cholestasis in infants and young children, especially those with multiorgan abnormalities.


Subject(s)
Alagille Syndrome , Cholestasis , Infant , Humans , Child , Child, Preschool , Alagille Syndrome/diagnosis , Alagille Syndrome/genetics , Genetic Testing , Asian People/genetics , Bilirubin
20.
J Extra Corpor Technol ; 54(4): 338-342, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36742021

ABSTRACT

Alagille syndrome is an autosomal dominant disorder that is caused by heterozygous mutation of JAG1 or NOTCH2 gene that impacts several multisystem organs including but may not be limited to the liver, heart, musculoskeletal, skin, and the eyes. The most common congenital heart defect associated with Alagille syndrome is multilevel right ventricular outflow tract obstruction with multiple central and peripheral branch pulmonary arterial stenoses occurring in up to two-thirds of these patients. We report two cases of Alagille syndrome who underwent extensive pulmonary arterial branch rehabilitation and experienced unusual oxygenator failure during cardiopulmonary bypass (CPB). We present lessons learned from these two cases and the changes that we implemented in our practice that facilitated smooth conduct of CPB in other cases that we performed subsequently.


Subject(s)
Alagille Syndrome , Heart Defects, Congenital , Hypertension, Pulmonary , Humans , Alagille Syndrome/complications , Alagille Syndrome/surgery , Alagille Syndrome/genetics , Cardiopulmonary Bypass/adverse effects , Hypertension, Pulmonary/complications , Oxygenators/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL