Next Article in Journal
Deciphering miRNAs’ Action through miRNA Editing
Next Article in Special Issue
Genetic and Physiological Dissection of Photosynthesis in Barley Exposed to Drought Stress
Previous Article in Journal
A Computer Model of Oxygen Dynamics in the Cortex of the Rat Kidney at the Cell-Tissue Level
Previous Article in Special Issue
The 1,10-Phenanthroline Ligand Enhances the Antiproliferative Activity of DNA-Intercalating Thiourea-Pd(II) and -Pt(II) Complexes Against Cisplatin-Sensitive and -Resistant Human Ovarian Cancer Cell Lines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

Novel JAG1 Deletion Variant in Patient with Atypical Alagille Syndrome

1
Arrhythmology and Clinical Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Milan, Italy
2
Department of Congenital Heart Surgery, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Milan, Italy
3
Division of Genetics and Cell Biology, Unit of Genomics for diagnosis of human diseases IRCCS San Raffaele Sci. Inst., 20132 Milan, Italy
4
Laboratory of Clinical Molecular Biology and Cytogenetics, IRCCS San Raffaele Sci. Inst., 20132 Milan, Italy
5
San Raffaele Vita-Salute University, 20132 Milan, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(24), 6247; https://doi.org/10.3390/ijms20246247
Submission received: 21 October 2019 / Revised: 25 November 2019 / Accepted: 6 December 2019 / Published: 11 December 2019
(This article belongs to the Collection Feature Papers in Molecular Genetics and Genomics)

Abstract

:
Alagille syndrome (AGS) is an autosomal-dominant disorder characterized by various degrees of abnormalities in the liver, heart, eyes, vertebrae, kidneys, face, vasculature, skeleton, and pancreas. This case report describes a newborn child exhibiting a congenital neural tube defect and peculiar craniofacial appearance characterized by a prominent forehead, deep-set eyes, bulbous nasal tip, and subtle upper lip. Just a few hours after birth, congenital heart disease was suspected for cyanosis and confirmed by heart evaluation. In particular, echocardiography indicated pulmonary atresia with ventricular septal defect with severe hypoplasia of the pulmonary branches (1.5 mm), large patent ductus arteriosus and several major aortopulmonary collateral arteries. Due to the association of peculiar craniofacial appearance and congenital heart disease, a form of Alagille syndrome was suspected. In addition, on the fifth day after birth, the patient developed jaundice, had acholic stools, and high levels of conjugated bilirubin and gamma-glutamyltransferase (GGT) were detected in the blood. Genetic testing revealed the novel variant c.802del in a single copy of the JAG1 gene. No variants in the NOTCH2 gene were detected. To the best of our knowledge, this is the first clinical description of a congenital neural tube defect in a molecularly confirmed Alagille patient. This work demonstrates a novel pathogenic heterozygous JAG1 mutation is associated with an atypical form of Alagille syndrome, suggesting an increased risk for neural tube defects compared to other Alagille patients.

1. Introduction

Alagille syndrome (AGS) is an autosomal-dominant disorder characterized by various degrees of abnormalities in the liver, heart, eyes, vertebrae, kidneys, face, vasculature, skeleton, and pancreas [1,2]. The estimated frequency is one in 30,000 live births [3]. Alagille syndrome has been defined as the paucity of the interlobular bile ducts together with three to five of the following: chronic cholestasis, cardiac disease, skeletal abnormalities, ocular abnormalities, and a characteristic facial phenotype [4].
The syndrome received its name from the French pediatrician Daniel Alagille (1925–2005), who first described it in 1969, focusing on hepatic pathologies [5]. Since this discovery, many other authors characterized AGS cases by hepatic involvement and various other congenital abnormalities, all with the same genetic causes [6].
The diagnosis of AGS is difficult because of the variability in the clinical presentation, even between family members, despite almost complete penetrance [2,4]. However, genetic testing can aid in the diagnosis, as the majority of cases are due to haploinsufficiency of the JAG1 gene, mapping on the short arm of chromosome 20 (20p11.2–20p12), either because of point mutations, exon and whole gene deletions or the microdeletion of 20p12 [7]. JAG1 mutations account for around 94% of cases, while NOTCH2 mutations are less common [1]. Both of these genes are involved in the Notch signaling pathway and play an important role in cell fate determination [8]. New mutations commonly occur (about 60%), and the rate of germline mosaicism may also be relatively high [7]. Prenatal testing is not able to predict the severity of the phenotype because of a reduced penetrance (47% [9]). Due to the variability in clinical presentation, early genetic testing is needed to confirm the syndrome so that preventative measures can be taken to prevent complications in multiple organs.

2. Case Presentation

Written informed consent of human subjects (or parents, in the case of minor children) included in this case series report was obtained for their participation in the study and for publication of this case report. The procedures employed were reviewed and approved by the local Ethics Committee.
The proband is a female child who was born in Romania from healthy, non-consanguineous parents with no remarkable medical history at 37 weeks of gestational age with a birth weight of 2500 g (around 50th centile corrected for gestational age).
During pregnancy, her mother had regular check-ups and she did not take any medication, except for prenatal vitamins (vitamin D, vitamin B6, folic acid) and supplements (iron and calcium). In particular, the folic acid dose taken was 0.4 mg per day for one month before conception and for the first trimester of gestation.
At birth, the baby was cyanotic. Echocardiography showed complex cyanotic heart disease and she was put under inotropic support and prostaglandin infusion. On the fifth day, the baby developed jaundice and acholic stools, with a high level of conjugated bilirubin and gamma-glutamyltransferase (GGT) on blood investigations (Figure 1). The abdominal ultrasound showed mild dilation of the cholecystic duct without any changes in the intra- or extrahepatic bile ducts. The viral screening was negative for Cytomegalovirus (CMV), Rubella, Epstein–Barr virus (EBV) and Herpes simplex virus 1 (HSV1).
At 12 days of age, the proband was transferred to the Department of Pediatric Congenital Heart Disease of IRCCS Policlinico San Donato, Milan, Italy. On admission, the baby exhibited a compromised general condition with both cyanosis and jaundice. She had an oxygen saturation of 81% (environmental air), a weight of 2400 g, and a height of 49 cm. Remarkably, no hepatomegaly was described during this first clinical evaluation.
Laboratory workup revealed a high level of conjugated bilirubin (9.65 mg/dL) with a moderate increase of the transaminase level (ALT 45 U/L, AST 75 U/L), and mild increases in the C reactive protein levels (0.8 mg/L) and white cells count (23,000/mmc). The cultural exams for common pathogens were negative. The thyroid function showed TSH suppression with normal levels of fT3 and fT4. Thereafter, a genetic evaluation was recommended, noting a peculiar craniofacial appearance, characterized by a prominent forehead, deep-set eyes, bulbous nasal tip, and subtle upper lip. Taking into consideration all these clues, the geneticist raised the suspicion of Alagille syndrome.
The case was discussed with our surgical team: a congenital neural tube defect was clinically found, due to the presence of sacral dimple with an indented patch of hyperpigmented skin. Then a neuro-surgical evaluation was performed, consistent with spina bifida occulta but without indication for spinal MRI, considering both the patient’s age and her precarious clinical condition. It was felt that, in view of the marked cholestasis and severely hypoplastic pulmonary branches in the setting of Alagille Syndrome, the risk to perform palliative surgery was too high. To verify the suspicion of Alagille Syndrome, molecular analysis was performed on the JAG1 and NOTCH2 genes. Next-Generation Sequencing, with an Illumina-TruSight enrichment on an Illumina-NextSeq500 platform, was performed on genomic DNA extracted from a peripheral blood sample of the proband and revealed the presence of the novel JAG1 heterozygous single-base deletion at position 802 of the cDNA (NM_000214.2:c.802delC, LOVD: https://databases.lovd.nl/shared/variants/0000579828#00000084). The presence of the variant was confirmed by Sanger Sequencing (Figure 2). No variants in the NOTCH2 gene were detected.
To exclude the possible presence of genomic rearrangement in the patient, an Array CGH on genomic DNA extracted from peripheral blood was also performed, resulting in the discovery of a 229 kb microduplication in chromosome 17 with the following genomic position: arr [GRCh37] 17q22(55644562_55873916). This microduplication encompasses the MSI2 gene (MIM *607897) expressing an oncogenic RNA-binding protein, required for blast crisis chronic myeloid leukemia [10]. Both parents, as well as the proband’s older brother, tested negative for both the JAG1 variant and the microduplication within chromosome 17, suggesting that both this mutation and the genetic rearrangement are de novo (Figure 3).
Since the #182940 entry for neural tube defects in OMIM [11] describes five genes (FUZ, MTHFR, VANGL1, VANGL2, and CCL2) associated with neural tube defects, including spina bifida occulta, we sequenced these genes with the NGS technique. No mutation nor low-frequency variants were found, with both a medium coverage of 100X for each gene and no zone with coverage <20X Thus, the approach used for genetic testing in this proband was NGS with a panel including the genes JAG1, NOTCH2, FUZ, MTHFR, VANGL1, VANGL2, and CCL2. The medium coverage was 100X for each tested gene without zone with coverage less than 20X. No homozygous rare polymorphism was identified, making us confident about the absence of a deletion in all the studied genes.
In the meanwhile, the proband developed severe jaundice, including yellowing of both sclerae and lips. This prompted a further liver evaluation, revealing an increase in alanine transaminase, aspartate transaminase, glutamate-oxaloacetate transaminase, and an even larger increase in r-glutamyl transpeptidase. Conjugated “direct” bilirubin was discovered in the blood. Due to liver failure, the proband received three concentrated red blood cells transfusions and she continued on lactulose pharmaceutical therapy. The discovery of the JAG1 variant allowed the diagnosis of Alagille syndrome, which in turn prompted further testing of thyroid, kidney, and eyes, which were all normal.
Echocardiography indicated pulmonary atresia with ventricular septal defect and severe hypoplasia of the pulmonary branches (1.5 mm), large patent ductus arteriosus and several major aortopulmonary collateral arteries (Figure 4A). In view of the hypoplasia of the pulmonary branches in a low-weight newborn, the decision was taken to perform cardiac catheterization and to implant a stent in the arterial duct. The procedure was carried out without any complications. Hypoplastic pulmonary artery branches were visualized during the catheterization that was performed before implanting the stent (Figure 4B). Eleven days after the procedure, the child was transferred back to the Romanian Hospital to continue the therapeutic management. Over the course of the next few months, the proband showed good weight gain, but with persistent cholestasis and worsening cyanosis. At three months of age, the child weighed 5 kg, with oxygen saturation between 70 and 75% and conjugated bilirubin of 10–12 mg/dL.

3. Discussion

It has been known since 2000 that the JAG1 gene is expressed in normal human neural tube [12]. In spite of this, clinical descriptions of neural tube defects in molecularly confirmed Alagille patients are limited. No functional data about the role of the JAG1 heterozygous mutation in neural tube defects are available to date. However, it has been demonstrated that apical/basal cell polarity is essential for neural tube closure [13]. In particular, it is very well known that JAG1 is part of the NOTCH cell pathway [14] and that NOTCH signaling has an essential impact on apical/basal cell polarity [13]. The most updated data indicate that Notch signaling plays a role in both differentiation and the organization of development in the central nervous system.
To the best of our knowledge, this is the first description of a neural tube defect as an onset sign of Alagille in a pediatric patient. The vast majority of children affected by this condition display an association between cholestasis (96%) and congenital heart malformation, while the most common kind of heart involvement is pulmonic stenosis (67% of patients [15]). Spina bifida occulta has already been described in patients clinically affected by Alagille syndrome [16]. In these cases, already published, no mention about mutation(s) nor genes involved has never been provided. Instead, here we describe for the first time ever a possible cause for this phenotype, including a case of spina bifida occulta in the apparent absence of other major environmental and genetic causes. It is remarkable that the same dose of folic acid had been taken by the mother for the first trimester of gestation in the previous pregnancy, which resulted in a healthy male newborn without any clinical signs of neural tube defect.
In the present case, the clinical picture was characterized at its onset by congenital spina bifida occulta and cyanotic heart disease: this is not a common association to lead the pediatrician to the suspicion of Alagille syndrome. This condition is more commonly suspected due to the association between cholestasis and congenital heart malformations.
The face of a patient is an important element, as well, to raise the clinical suspicion: it is a triangular-shaped face with a prominent forehead, hypertelorism, deep-set eyes, a pointed chin, and a bulbous nasal tip. Most of these features are actually present in our patient as well, and they are one of the elements that justified genetic testing as a diagnostic approach.
Alagille syndrome is characterized by a marked variability, even in the same family: some patients could display a serious clinical picture from the first hours of life (often due to cardiac congenital malformation) or early infancy. On the other hand, some patients might suffer from apparently isolated cholestasis of unknown cause or even have only a particular ocular malformation [17].
JAG1 is the major gene causing Alagille syndrome, [18] with more than 600 pathogenic variants identified to date [7]. These variants are known to be truncating in the majority of cases (49% frameshift, 16% nonsense, 9% splicing [15], 11% large gene rearrangements), and non-truncating in about 15% (missense) [15]. In 1–2% of affected patients, Alagille syndrome is caused by heterozygous pathogenic variants in NOTCH2 on chromosome 1p13.2. No pathogenic variants are identified in about 4% of affected patients, suggesting that some other genes might play a causative role for this subset of cases [19].
To date, there is not a clear correlation between genotype and phenotype, and no clinical pictures seem exclusively associated with JAG1 or NOTCH2. Both Jagged1 and NOTCH proteins belong to the Notch pathway, a highly conserved signaling system expressed in many multicellular organisms and specifying cell fate decisions [20]. In humans, Notch signaling is based on four different proteins (NOTCH1-4) that function all as receptors interacting with a family of transmembrane ligands expressed on the surface of adjacent cells. Among these, Jagged1 is the ligand of NOTCH1. Recent studies resolved the structure of the Notch-Jagged extracellular interacting complex, revealing a binding interface. Luca and coauthors demonstrated how this interaction is specific for binding, ligand discrimination, and Notch signaling [21]. This relationship regulates many development processes (in particular pulmonary veins, bile ducts, pancreas, and kidneys).
Skeletal development requires normal Notch signaling. In fact, skeletal defects are features shared between Alagille and Hajdu-Cheney syndromes [22]. According to current knowledge, a whole gene heterozygous deletion (regardless of whether in JAG1 or NOTCH2) does not provoke a somite-related phenotype, suggesting a role for Notch signaling in the early stages of osteogenesis [23]. This might be the reason why some Alagille patients are affected by the butterfly vertebrae, a persistent notochordal tissue between the anterior vertebral arches [24].
Our patient is the first case of Alagille syndrome with the novel heterozygous single nucleotide deletion at position 802 (c.802del) of the JAG1 gene (reference sequence NM_000214.3), resulting in frameshift and insertion of a premature stop of translation after 144 codons, in the putative protein (p.His268Thrfs*144). This specific truncating variant has not been reported to date in patients clinically affected by Alagille syndrome and involves a highly conserved region of the gene (GERP score 5.92). It is possible that the heterozygous state for this mutation explains the entire phenotype of our proband, because it is not present in the general population nor in the healthy parents (de novo hypothesis), the resulting putative JAG1 protein is truncated, and the patient does not carry any mutation in the currently known neural tube defect genes, according to OMIM [11].
The c.802delC variant causes the substitution of His268, a residue anchoring the interface between NOTCH1-EGF10 and Jag1-EGF1 domains, as recently described [21]. We do not know if the truncated protein is present or degraded, but evidence exists that the mutated region might be critical for the Notch-Jagged1 complex. Functional studies are required to confirm this hypothesis.
In addition, it is very well known that heterozygous frameshift mutations in either JAG1 or NOTCH2 genes cause Alagille phenotypes similar to the one displayed by our patient [25,26]. Variants in the NOTCH2 gene have been associated with neural tube defects [27], but the proband in this report was not found to have a NOTCH2 variant, because the massively parallel sequencing of this gene demonstrated a wild type sequence. This is an important clue suggesting that, in addition to heterozygous deletions in JAG1 and NOTCH2 described in other studies, the heterozygous state for the JAG1 mutation described in this case alone might be independently responsible for neural tube defects. We cannot exclude, however, that other genetic variants might have caused the neural tube defect, even in the presence of evidence that the Jagged 1 pathway is actually involved in the pathogenesis of such a condition.
It is remarkable that in the Online Mendelian Inheritance in Men website [11], there are some rare cases described of neural tube defects (even more serious than in our patient) caused by a heterozygous mutation in a gene that belongs to the Notch pathway. In particular, one study described a few patients [28] with neural tube defects harboring heterozygous mutations in the NOTCH3 gene. The protein codified by this gene shows similarity with NOTCH2 [29], so it is possible that neural tube defects might be caused also by mutations in other genes coding for proteins that belong to the Notch signaling pathway. This mechanism had been already described in many other genetic conditions such as Rasopathies [30], coesinopathy [31], and mTOR related conditions [32]. Consequently, other genes associated with neural tube defects [11], including spina bifida, were sequenced, with negative results.
In conclusion, we have to mention that to reach a better diagnostic classification, a CGH Array analysis from peripheral blood extracted genomic DNA was performed. The main reason is that, in some patients affected by neural tube defects, a submicroscopic genomic rearrangement has been demonstrated [33]. Our patient harbors a microduplication in the long arm of chromosome 17 with unknown inheritance, identified with the CGH Array technique. This microduplication encompasses the MSI2 gene (MIM *607897) known to be involved in TGF beta signaling, but not to date in the Notch pathway [34]. To date, no MSI2 mutation, deletion, nor duplication has been described in patients with neural tube defects. Taken together, these two statements strengthen our hypothesis that the JAG1 frameshift variant demonstrated in our patient might be considered at least the major cause of her phenotype. We recommend that Alagille syndrome should be considered as a possible diagnosis in the presence of congenital heart malformations and neural tube defects, even in the absence of liver involvement. Future studies are needed to understand this relationship further. In particular, it needs to be assessed whether neural tube defects might be present also in other conditions (for example Hajdu Cheney syndrome) caused by heterozygous mutations in genes coding for Notch pathway proteins.

4. Concluding Remarks

This work demonstrates a novel pathogenic heterozygous JAG1 mutation is associated with an atypical Alagille phenotype. The clinical picture of this female patient is characterized by some common Alagille signs associated with a congenital spina bifida occulta. This association has never been described before to the best of our knowledge: the keywords “Alagille” and “spina bifida occulta” provide no result in Pubmed (https://www.ncbi.nlm.nih.gov/pubmed/?term=Alagille+AND+spina+bifida+occulta, as of October 21, 2019). Considering the major role of Notch signaling in neural tube defect pathogenesis, our case suggests that Alagille patients might have an increased risk for neural tube defects compared to the general population. However, further studies are required to confirm this finding.

Author Contributions

Conceptualization: E.M., A.A.A., M.M.M.; data curation: All authors; formal analysis: All authors; funding acquisition: A.G. and C.P.; investigation: All authors; resources: A.G. and C.P.; supervision: E.M., E.T.L. and M.C., M.F., A.G., and C.P.; visualization: A.A.A., P.C., M.M.M., B.P., and S.P.; writing—original draft preparation: E.M., A.A.A., P.C., E.T.L. and M.M.M.; All authors have reviewed and provided corrections, and have agreed to be co-authors on the submitted version of the paper.

Funding

This study was partially supported by Ricerca Corrente funding from Italian Ministry of Health to IRCCS Policlinico San Donato.

Acknowledgments

The authors thank the members of the family for their help and participation in the study.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Kim, J.; Yang, B.; Paik, N.; Choe, Y.H.; Paik, Y.H. A case of Alagille syndrome presenting with chronic cholestasis in an adult. Clin. Mol. Hepatol. 2017, 23, 260–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Jurkiewicz, D.; Gliwicz, D.; Ciara, E.; Gerfen, J.; Pelc, M.; Piekutowska-Abramczuk, D.; Kugaudo, M.; Chrzanowska, K.; Spinner, N.B.; Krajewska-Walasek, M. Spectrum of JAG1 gene mutations in Polish patients with Alagille syndrome. J. Appl. Genet. 2014, 55, 329–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Saleh, M.; Kamath, B.M.; Chitayat, D. Alagille syndrome: Clinical perspectives. Appl. Clin. Genet. 2016, 9, 75–82. [Google Scholar] [PubMed] [Green Version]
  4. Emerick, K.M.; Rand, E.B.; Goldmuntz, E.; Krantz, I.D.; Spinner, N.B.; Piccoli, D.A. Features of Alagille syndrome in 92 patients: Frequency and relation to prognosis. Hepatology 1999, 29, 822–829. [Google Scholar] [CrossRef] [PubMed]
  5. Alagille, D.; Borde, J.; Habib, E.C.; Thomassin, N. Surgical attempts in atresia of the intrahepatic bile ducts with permeable extrahepatic bile duct. Study of 14 cases in children. Arch. Fr. Pediatr. 1969, 26, 51–71. [Google Scholar] [PubMed]
  6. Di Pinto, D.; Adragna, M. Renal manifestations in children with Alagille syndrome. Arch. Argent. Pediatr. 2018, 116, 149–153. [Google Scholar]
  7. Turnpenny, P.D.; Ellard, S. Alagille syndrome: Pathogenesis, diagnosis and management. Eur. J. Hum. Genet. 2012, 20, 251–257. [Google Scholar] [CrossRef] [Green Version]
  8. Lin, H.C.; Le Hoang, P.; Hutchinson, A.; Chao, G.; Gerfen, J.; Loomes, K.M.; Krantz, I.; Kamath, B.M.; Spinner, N.B. Alagille syndrome in a Vietnamese cohort: Mutation analysis and assessment of facial features. Am. J. Med. Genet. A 2012, 158, 1005–1013. [Google Scholar] [CrossRef] [Green Version]
  9. Kamath, B.M.; Bason, L.; Piccoli, D.A.; Krantz, I.D.; Spinner, N.B. Consequences of JAG1 mutations. J. Med. Genet. 2003, 40, 891–895. [Google Scholar] [CrossRef] [Green Version]
  10. Hattori, A.; Tsunoda, M.; Konuma, T.; Kobayashi, M.; Nagy, T.; Glushka, J.; Tayyari, F.; McSkimming, D.; Kannan, N.; Tojo, A.; et al. Cancer progression by reprogrammed BCAA metabolism in myeloid leukaemia. Nature 2017, 545, 500–504. [Google Scholar] [CrossRef] [Green Version]
  11. Online Mendelian Inheritance in Man. Available online: https://www.omim.org/ (accessed on 21 November 2019).
  12. Jones, E.A.; Clement-Jones, M.; Wilson, D.I. JAGGED1 expression in human embryos: Correlation with the Alagille syndrome phenotype. J. Med. Genet. 2000, 37, 658–662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Main, H.; Radenkovic, J.; Jin, S.B.; Lendahl, U.; Andersson, E.R. Notch signaling maintains neural rosette polarity. PLoS ONE 2013, 8, e62959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Grochowski, C.M.; Loomes, K.M.; Spinner, N.B. Jagged1 (JAG1): Structure, expression, and disease associations. Gene 2016, 576, 381–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Wu, K.Y.; Treece, A.L.; Russo, P.A.; Wen, J.W. An Atypical Presentation of Alagille Syndrome. Pediatr. Dev. Pathol. 2018, 21, 79–83. [Google Scholar] [CrossRef] [PubMed]
  16. Penton, A.L.; Leonard, L.D.; Spinner, N.B. Notch signaling in human development and disease. Semin. Cell Dev. Biol. 2012, 23, 450–457. [Google Scholar] [CrossRef] [Green Version]
  17. Orssaud, C.; Robert, M.P.; Roche, O. Relevance of Identifying JAG1 Mutations in Patients with Isolated Posterior Embryotoxon. J. Glaucoma 2016, 25, 923–925. [Google Scholar] [CrossRef]
  18. Krantz, I.D.; Colliton, R.P.; Genin, A.; Rand, E.B.; Li, L.; Piccoli, D.A.; Spinner, N.B. Spectrum and frequency of jagged1 (JAG1) mutations in Alagille syndrome patients and their families. Am. J. Hum. Genet. 1998, 62, 1361–1369. [Google Scholar] [CrossRef] [Green Version]
  19. Grochowski, C.M.; Rajagopalan, R.; Falsey, A.M.; Loomes, K.M.; Piccoli, D.A.; Krantz, I.D.; Devoto, M.; Spinner, N.B. Exome sequencing reveals compound heterozygous mutations in ATP8B1 in a JAG1/NOTCH2 mutation-negative patient with clinically diagnosed Alagille syndrome. Am. J. Med. Genet. A 2015, 167, 891–893. [Google Scholar] [CrossRef]
  20. Masek, J.; Andersson, E.R. The developmental biology of genetic Notch disorders. Development 2017, 144, 1743–1763. [Google Scholar] [CrossRef] [Green Version]
  21. Luca, V.C.; Kim, B.C.; Ge, C.; Kakuda, S.; Wu, D.; Roein-Peikar, M.; Haltiwanger, R.S.; Zhu, C.; Ha, T.; Garcia, K.C. Notch-Jagged complex structure implicates a catch bond in tuning ligand sensitivity. Science 2017, 355, 1320–1324. [Google Scholar] [CrossRef] [Green Version]
  22. Canalis, E.; Zanotti, S. Hajdu-Cheney Syndrome, a Disease Associated with NOTCH2 Mutations. Curr. Osteoporos. Rep. 2016, 14, 126–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hamada, Y.; Kadokawa, Y.; Okabe, M.; Ikawa, M.; Coleman, J.R.; Tsujimoto, Y. Mutation in ankyrin repeats of the mouse Notch2 gene induces early embryonic lethality. Development 1999, 126, 3415–3424. [Google Scholar] [PubMed]
  24. Youngstrom, D.W.; Dishowitz, M.I.; Bales, C.B.; Carr, E.; Mutyaba, P.L.; Kozloff, K.M.; Shitaye, H.; Hankenson, K.D.; Loomes, K.M. Jagged1 expression by osteoblast-lineage cells regulates trabecular bone mass and periosteal expansion in mice. Bone 2016, 91, 64–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kola, S.; Koneti, N.R.; Golla, J.P.; Akka, J.; Gundimeda, S.D.; Mundluru, H.P. Mutational analysis of JAG1 gene in non-syndromic tetralogy of Fallot children. Clin. Chim. Acta 2011, 412, 2232–2236. [Google Scholar] [CrossRef] [PubMed]
  26. Kamath, B.M.; Bauer, R.C.; Loomes, K.M.; Chao, G.; Gerfen, J.; Hutchinson, A.; Hardikar, W.; Hirschfield, G.; Jara, P.; Krantz, I.D.; et al. NOTCH2 mutations in Alagille syndrome. J. Med. Genet. 2012, 49, 138–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Mead, T.J.; Yutzey, K.E. Notch pathway regulation of neural crest cell development in vivo. Dev. Dyn. 2012, 241, 376–389. [Google Scholar] [CrossRef] [Green Version]
  28. Chen, K.M.; Bird, L.; Barnes, P.; Barth, R.; Hudgins, L. Lateral meningocele syndrome: Vertical transmission and expansion of the phenotype. Am. J. Med. Genet. A 2005, 133, 115–121. [Google Scholar] [CrossRef]
  29. Gordon, W.R.; Arnett, K.L.; Blacklow, S.C. The molecular logic of Notch signaling—A structural and biochemical perspective. J. Cell Sci. 2008, 121, 3109–3119. [Google Scholar] [CrossRef] [Green Version]
  30. Gimenez, N.; Martinez-Trillos, A.; Montraveta, A.; Lopez-Guerra, M.; Rosich, L.; Nadeu, F.; Valero, J.G.; Aymerich, M.; Magnano, L.; Rozman, M.; et al. Mutations in RAS-BRAF-MAPK-ERK pathway define a specific subgroup of patients with adverse clinical features and provide new therapeutic options in chronic lymphocytic leukemia. Haematologica 2019, 104, 576–586. [Google Scholar] [CrossRef]
  31. Yuan, B.; Neira, J.; Pehlivan, D.; Santiago-Sim, T.; Song, X.; Rosenfeld, J.; Posey, J.E.; Patel, V.; Jin, W.; Adam, M.P.; et al. Clinical exome sequencing reveals locus heterogeneity and phenotypic variability of cohesinopathies. Genet. Med. 2019, 21, 663. [Google Scholar] [CrossRef]
  32. Pachow, D.; Wick, W.; Gutmann, D.H.; Mawrin, C. The mTOR signaling pathway as a treatment target for intracranial neoplasms. Neuro Oncol. 2015, 17, 189–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Puvabanditsin, S.; Gueye-Ndiaye, S.; Puthenpura, V.; Gengel, N.; Tam, V.; Mehta, R. Microduplication of 17p[Dup(17)(12p11.2)]: Report of a Neonate With a Spina Bifida and Cardiac Anomalies and a Literature Review. Genet. Couns 2016, 27, 503–507. [Google Scholar] [PubMed]
  34. Kudinov, A.E.; Deneka, A.; Nikonova, A.S.; Beck, T.N.; Ahn, Y.H.; Liu, X.; Martinez, C.F.; Schultz, F.A.; Reynolds, S.; Yang, D.H.; et al. Musashi-2 (MSI2) supports TGF-beta signaling and inhibits claudins to promote non-small cell lung cancer (NSCLC) metastasis. Proc. Natl. Acad. Sci. USA 2016, 113, 6955–6960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. (A) Serum direct bilirubin concentration. (B) Serum total bilirubin concentration. (C) Serum gamma-glutamyltransferase concentration.
Figure 1. (A) Serum direct bilirubin concentration. (B) Serum total bilirubin concentration. (C) Serum gamma-glutamyltransferase concentration.
Ijms 20 06247 g001
Figure 2. Sanger direct sequencing of c.802delC. The variation creates a frameshift at codon His268. The new reading frame ends in a stop codon at position 144; wt: wild type; mut: mutant. The letters with red box means the called basis when there is a frameshift in the called position. So the letters with red box must be read as follows: M = C or A, S= C or G, R= G or A, W= T or A, K = G or T.
Figure 2. Sanger direct sequencing of c.802delC. The variation creates a frameshift at codon His268. The new reading frame ends in a stop codon at position 144; wt: wild type; mut: mutant. The letters with red box means the called basis when there is a frameshift in the called position. So the letters with red box must be read as follows: M = C or A, S= C or G, R= G or A, W= T or A, K = G or T.
Ijms 20 06247 g002
Figure 3. Family pedigree. Proband identified with an arrow. y = years old at diagnosis.
Figure 3. Family pedigree. Proband identified with an arrow. y = years old at diagnosis.
Ijms 20 06247 g003
Figure 4. (A) Echocardiography indicated severe hypoplasia of the pulmonary branches. (B) Hypoplastic pulmonary artery branches were visualized during the catheterization that was performed before implanting the stent.
Figure 4. (A) Echocardiography indicated severe hypoplasia of the pulmonary branches. (B) Hypoplastic pulmonary artery branches were visualized during the catheterization that was performed before implanting the stent.
Ijms 20 06247 g004

Share and Cite

MDPI and ACS Style

Micaglio, E.; Andronache, A.A.; Carrera, P.; Monasky, M.M.; Locati, E.T.; Pirola, B.; Presi, S.; Carminati, M.; Ferrari, M.; Giamberti, A.; et al. Novel JAG1 Deletion Variant in Patient with Atypical Alagille Syndrome. Int. J. Mol. Sci. 2019, 20, 6247. https://doi.org/10.3390/ijms20246247

AMA Style

Micaglio E, Andronache AA, Carrera P, Monasky MM, Locati ET, Pirola B, Presi S, Carminati M, Ferrari M, Giamberti A, et al. Novel JAG1 Deletion Variant in Patient with Atypical Alagille Syndrome. International Journal of Molecular Sciences. 2019; 20(24):6247. https://doi.org/10.3390/ijms20246247

Chicago/Turabian Style

Micaglio, Emanuele, Andreea Alina Andronache, Paola Carrera, Michelle M. Monasky, Emanuela T. Locati, Barbara Pirola, Silvia Presi, Mario Carminati, Maurizio Ferrari, Alessandro Giamberti, and et al. 2019. "Novel JAG1 Deletion Variant in Patient with Atypical Alagille Syndrome" International Journal of Molecular Sciences 20, no. 24: 6247. https://doi.org/10.3390/ijms20246247

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop