126 results on '"John Pappas"'
Search Results
2. P454: Deletion of exons 10 and 11 in FGFR2: Mother and daughter with sagittal craniosynostosis and micrognathia
- Author
-
John Pappas, Seth Shanefield, Michelle Goodman, and Rachel Rabin
- Subjects
Genetics ,QH426-470 ,Medicine - Published
- 2024
- Full Text
- View/download PDF
3. Impaired protein hydroxylase activity causes replication stress and developmental abnormalities in humans
- Author
-
Sally C. Fletcher, Charlotte Hall, Tristan J. Kennedy, Sander Pajusalu, Monica H. Wojcik, Uncaar Boora, Chan Li, Kaisa Teele Oja, Eline Hendrix, Christian A.E. Westrip, Regina Andrijes, Sonia K. Piasecka, Mansi Singh, Mohammed E. El-Asrag, Anetta Ptasinska, Vallo Tillmann, Martin R. Higgs, Deanna A. Carere, Andrew D. Beggs, John Pappas, Rachel Rabin, Stephen J. Smerdon, Grant S. Stewart, Katrin Õunap, and Mathew L. Coleman
- Subjects
Cell biology ,Genetics ,Medicine - Abstract
Although protein hydroxylation is a relatively poorly characterized posttranslational modification, it has received significant recent attention following seminal work uncovering its role in oxygen sensing and hypoxia biology. Although the fundamental importance of protein hydroxylases in biology is becoming clear, the biochemical targets and cellular functions often remain enigmatic. JMJD5 is a “JmjC-only” protein hydroxylase that is essential for murine embryonic development and viability. However, no germline variants in JmjC-only hydroxylases, including JMJD5, have yet been described that are associated with any human pathology. Here we demonstrate that biallelic germline JMJD5 pathogenic variants are deleterious to JMJD5 mRNA splicing, protein stability, and hydroxylase activity, resulting in a human developmental disorder characterized by severe failure to thrive, intellectual disability, and facial dysmorphism. We show that the underlying cellular phenotype is associated with increased DNA replication stress and that this is critically dependent on the protein hydroxylase activity of JMJD5. This work contributes to our growing understanding of the role and importance of protein hydroxylases in human development and disease.
- Published
- 2023
- Full Text
- View/download PDF
4. Rare deleterious mutations of HNRNP genes result in shared neurodevelopmental disorders
- Author
-
Madelyn A. Gillentine, Tianyun Wang, Kendra Hoekzema, Jill Rosenfeld, Pengfei Liu, Hui Guo, Chang N. Kim, Bert B. A. De Vries, Lisenka E. L. M. Vissers, Magnus Nordenskjold, Malin Kvarnung, Anna Lindstrand, Ann Nordgren, Jozef Gecz, Maria Iascone, Anna Cereda, Agnese Scatigno, Silvia Maitz, Ginevra Zanni, Enrico Bertini, Christiane Zweier, Sarah Schuhmann, Antje Wiesener, Micah Pepper, Heena Panjwani, Erin Torti, Farida Abid, Irina Anselm, Siddharth Srivastava, Paldeep Atwal, Carlos A. Bacino, Gifty Bhat, Katherine Cobian, Lynne M. Bird, Jennifer Friedman, Meredith S. Wright, Bert Callewaert, Florence Petit, Sophie Mathieu, Alexandra Afenjar, Celenie K. Christensen, Kerry M. White, Orly Elpeleg, Itai Berger, Edward J. Espineli, Christina Fagerberg, Charlotte Brasch-Andersen, Lars Kjærsgaard Hansen, Timothy Feyma, Susan Hughes, Isabelle Thiffault, Bonnie Sullivan, Shuang Yan, Kory Keller, Boris Keren, Cyril Mignot, Frank Kooy, Marije Meuwissen, Alice Basinger, Mary Kukolich, Meredith Philips, Lucia Ortega, Margaret Drummond-Borg, Mathilde Lauridsen, Kristina Sorensen, Anna Lehman, CAUSES Study, Elena Lopez-Rangel, Paul Levy, Davor Lessel, Timothy Lotze, Suneeta Madan-Khetarpal, Jessica Sebastian, Jodie Vento, Divya Vats, L. Manace Benman, Shane Mckee, Ghayda M. Mirzaa, Candace Muss, John Pappas, Hilde Peeters, Corrado Romano, Maurizio Elia, Ornella Galesi, Marleen E. H. Simon, Koen L. I. van Gassen, Kara Simpson, Robert Stratton, Sabeen Syed, Julien Thevenon, Irene Valenzuela Palafoll, Antonio Vitobello, Marie Bournez, Laurence Faivre, Kun Xia, SPARK Consortium, Rachel K. Earl, Tomasz Nowakowski, Raphael A. Bernier, and Evan E. Eichler
- Subjects
Neurodevelopmental disorders ,hnRNPs ,Cortex development ,Gene families ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background With the increasing number of genomic sequencing studies, hundreds of genes have been implicated in neurodevelopmental disorders (NDDs). The rate of gene discovery far outpaces our understanding of genotype–phenotype correlations, with clinical characterization remaining a bottleneck for understanding NDDs. Most disease-associated Mendelian genes are members of gene families, and we hypothesize that those with related molecular function share clinical presentations. Methods We tested our hypothesis by considering gene families that have multiple members with an enrichment of de novo variants among NDDs, as determined by previous meta-analyses. One of these gene families is the heterogeneous nuclear ribonucleoproteins (hnRNPs), which has 33 members, five of which have been recently identified as NDD genes (HNRNPK, HNRNPU, HNRNPH1, HNRNPH2, and HNRNPR) and two of which have significant enrichment in our previous meta-analysis of probands with NDDs (HNRNPU and SYNCRIP). Utilizing protein homology, mutation analyses, gene expression analyses, and phenotypic characterization, we provide evidence for variation in 12 HNRNP genes as candidates for NDDs. Seven are potentially novel while the remaining genes in the family likely do not significantly contribute to NDD risk. Results We report 119 new NDD cases (64 de novo variants) through sequencing and international collaborations and combined with published clinical case reports. We consider 235 cases with gene-disruptive single-nucleotide variants or indels and 15 cases with small copy number variants. Three hnRNP-encoding genes reach nominal or exome-wide significance for de novo variant enrichment, while nine are candidates for pathogenic mutations. Comparison of HNRNP gene expression shows a pattern consistent with a role in cerebral cortical development with enriched expression among radial glial progenitors. Clinical assessment of probands (n = 188–221) expands the phenotypes associated with HNRNP rare variants, and phenotypes associated with variation in the HNRNP genes distinguishes them as a subgroup of NDDs. Conclusions Overall, our novel approach of exploiting gene families in NDDs identifies new HNRNP-related disorders, expands the phenotypes of known HNRNP-related disorders, strongly implicates disruption of the hnRNPs as a whole in NDDs, and supports that NDD subtypes likely have shared molecular pathogenesis. To date, this is the first study to identify novel genetic disorders based on the presence of disorders in related genes. We also perform the first phenotypic analyses focusing on related genes. Finally, we show that radial glial expression of these genes is likely critical during neurodevelopment. This is important for diagnostics, as well as developing strategies to best study these genes for the development of therapeutics.
- Published
- 2021
- Full Text
- View/download PDF
5. EIF3F-related neurodevelopmental disorder: refining the phenotypic and expanding the molecular spectrum
- Author
-
Ulrike Hüffmeier, Cornelia Kraus, Miriam S. Reuter, Steffen Uebe, Mary-Alice Abbott, Syed A. Ahmed, Kristyn L. Rawson, Eileen Barr, Hong Li, Ange-Line Bruel, Laurence Faivre, Frédéric Tran Mau-Them, Christina Botti, Susan Brooks, Kaitlyn Burns, D. Isum Ward, Marina Dutra-Clarke, Julian A. Martinez-Agosto, Hane Lee, Stanley F. Nelson, UCLA California Center for Rare Disease, Pia Zacher, Rami Abou Jamra, Chiara Klöckner, Julie McGaughran, Jürgen Kohlhase, Sarah Schuhmann, Ellen Moran, John Pappas, Annick Raas-Rothschild, Maria J. Guillen Sacoto, Lindsay B. Henderson, Timothy Blake Palculict, Sureni V. Mullegama, Houda Zghal Elloumi, Adi Reich, Samantha A. Schrier Vergano, Erica Wahl, André Reis, and Christiane Zweier
- Subjects
EIF3F gene ,Neurodevelopmental disorder ,Short stature ,Deafness ,Behavioral difficulties ,Altered muscular tone ,Medicine - Abstract
Abstract Background An identical homozygous missense variant in EIF3F, identified through a large-scale genome-wide sequencing approach, was reported as causative in nine individuals with a neurodevelopmental disorder, characterized by variable intellectual disability, epilepsy, behavioral problems and sensorineural hearing-loss. To refine the phenotypic and molecular spectrum of EIF3F-related neurodevelopmental disorder, we examined independent patients. Results 21 patients were homozygous and one compound heterozygous for c.694T>G/p.(Phe232Val) in EIF3F. Haplotype analyses in 15 families suggested that c.694T>G/p.(Phe232Val) was a founder variant. All affected individuals had developmental delays including delayed speech development. About half of the affected individuals had behavioral problems, altered muscular tone, hearing loss, and short stature. Moreover, this study suggests that microcephaly, reduced sensitivity to pain, cleft lip/palate, gastrointestinal symptoms and ophthalmological symptoms are part of the phenotypic spectrum. Minor dysmorphic features were observed, although neither the individuals’ facial nor general appearance were obviously distinctive. Symptoms in the compound heterozygous individual with an additional truncating variant were at the severe end of the spectrum in regard to motor milestones, speech delay, organic problems and pre- and postnatal growth of body and head, suggesting some genotype–phenotype correlation. Conclusions Our study refines the phenotypic and expands the molecular spectrum of EIF3F-related syndromic neurodevelopmental disorder.
- Published
- 2021
- Full Text
- View/download PDF
6. Germline AGO2 mutations impair RNA interference and human neurological development
- Author
-
Davor Lessel, Daniela M. Zeitler, Margot R. F. Reijnders, Andriy Kazantsev, Fatemeh Hassani Nia, Alexander Bartholomäus, Victoria Martens, Astrid Bruckmann, Veronika Graus, Allyn McConkie-Rosell, Marie McDonald, Bernarda Lozic, Ee-Shien Tan, Erica Gerkes, Jessika Johannsen, Jonas Denecke, Aida Telegrafi, Evelien Zonneveld-Huijssoon, Henny H. Lemmink, Breana W. M. Cham, Tanja Kovacevic, Linda Ramsdell, Kimberly Foss, Diana Le Duc, Diana Mitter, Steffen Syrbe, Andreas Merkenschlager, Margje Sinnema, Bianca Panis, Joanna Lazier, Matthew Osmond, Taila Hartley, Jeremie Mortreux, Tiffany Busa, Chantal Missirian, Pankaj Prasun, Sabine Lüttgen, Ilaria Mannucci, Ivana Lessel, Claudia Schob, Stefan Kindler, John Pappas, Rachel Rabin, Marjolein Willemsen, Thatjana Gardeitchik, Katharina Löhner, Patrick Rump, Kerith-Rae Dias, Carey-Anne Evans, Peter Ian Andrews, Tony Roscioli, Han G. Brunner, Chieko Chijiwa, M. E. Suzanne Lewis, Rami Abou Jamra, David A. Dyment, Kym M. Boycott, Alexander P. A. Stegmann, Christian Kubisch, Ene-Choo Tan, Ghayda M. Mirzaa, Kirsty McWalter, Tjitske Kleefstra, Rolph Pfundt, Zoya Ignatova, Gunter Meister, and Hans-Jürgen Kreienkamp
- Subjects
Science - Abstract
AGO2 binds to miRNAs to repress expression of cognate target mRNAs. Here the authors report that heterozygous AGO2 mutations result in defects in neurological development and impair RNA interference.
- Published
- 2020
- Full Text
- View/download PDF
7. Transcriptome sequencing identifies a noncoding, deep intronic variant in CLCN7 causing autosomal recessive osteopetrosis
- Author
-
Odelia Chorin, Naomi Yachelevich, Khaled Mohamed, Ilana Moscatelli, John Pappas, Kim Henriksen, and Gilad D. Evrony
- Subjects
CLCN7 ,osteopetrosis ,RNA‐sequencing ,transcriptomics ,undiagnosed diseases ,Genetics ,QH426-470 - Abstract
Abstract Background Over half of children with rare genetic diseases remain undiagnosed despite maximal clinical evaluation and DNA‐based genetic testing. As part of an Undiagnosed Diseases Program applying transcriptome (RNA) sequencing to identify the causes of these unsolved cases, we studied a child with severe infantile osteopetrosis leading to cranial nerve palsies, bone deformities, and bone marrow failure, for whom whole‐genome sequencing was nondiagnostic. Methods We performed transcriptome (RNA) sequencing of whole blood followed by analysis of aberrant transcript isoforms and osteoclast functional studies. Results We identified a pathogenic deep intronic variant in CLCN7 creating an unexpected, frameshifting pseudoexon causing complete loss of function. Functional studies, including osteoclastogenesis and bone resorption assays, confirmed normal osteoclast differentiation but loss of osteoclast function. Conclusion This is the first report of a pathogenic deep intronic variant in CLCN7, and our approach provides a model for systematic identification of noncoding variants causing osteopetrosis—a disease for which molecular‐genetic diagnosis can be pivotal for potentially curative hematopoietic stem cell transplantation. Our work illustrates that cryptic splice variants may elude DNA‐only sequencing and supports broad first‐line use of transcriptome sequencing for children with undiagnosed diseases.
- Published
- 2020
- Full Text
- View/download PDF
8. Quantitative Systems Pharmacology Modeling of Acid Sphingomyelinase Deficiency and the Enzyme Replacement Therapy Olipudase Alfa Is an Innovative Tool for Linking Pathophysiology and Pharmacology
- Author
-
Chanchala D. Kaddi, Bradley Niesner, Rena Baek, Paul Jasper, John Pappas, John Tolsma, Jing Li, Zachary van Rijn, Mengdi Tao, Catherine Ortemann‐Renon, Rachael Easton, Sharon Tan, Ana Cristina Puga, Edward H. Schuchman, Jeffrey S. Barrett, and Karim Azer
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
Acid sphingomyelinase deficiency (ASMD) is a rare lysosomal storage disorder with heterogeneous clinical manifestations, including hepatosplenomegaly and infiltrative pulmonary disease, and is associated with significant morbidity and mortality. Olipudase alfa (recombinant human acid sphingomyelinase) is an enzyme replacement therapy under development for the non‐neurological manifestations of ASMD. We present a quantitative systems pharmacology (QSP) model supporting the clinical development of olipudase alfa. The model is multiscale and mechanistic, linking the enzymatic deficiency driving the disease to molecular‐level, cellular‐level, and organ‐level effects. Model development was informed by natural history, and preclinical and clinical studies. By considering patient‐specific pharmacokinetic (PK) profiles and indicators of disease severity, the model describes pharmacodynamic (PD) and clinical end points for individual patients. The ASMD QSP model provides a platform for quantitatively assessing systemic pharmacological effects in adult and pediatric patients, and explaining variability within and across these patient populations, thereby supporting the extrapolation of treatment response from adults to pediatrics.
- Published
- 2018
- Full Text
- View/download PDF
9. Clinical Phenotype in a Toddler with a Novel Heterozygous Mutation of the Vitamin D Receptor
- Author
-
Preneet Cheema Brar, Elena Dingle, John Pappas, and Manish Raisingani
- Subjects
Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
We present the clinical phenotype of a toddler who presented with vitamin D-resistant rickets, with one of the highest initial levels of alkaline phosphatase and parathyroid hormone (PTH) levels reported in the literature. The toddler had novel compound heterozygous mutations in the ligand-binding site of the vitamin D receptor and had an excellent response to calcitriol (1,25(OH)2D).
- Published
- 2017
- Full Text
- View/download PDF
10. Expanding the phenotypic spectrum of <scp> COLEC10 ‐Related 3MC </scp> syndrome: A glimpse into <scp> COLEC10 ‐Related 3MC </scp> syndrome in the Ashkenazi Jewish population
- Author
-
Rachel Rabin, Yoel Hirsch, Wendy K. Chung, Josef Ekstein, Ephrat Levy‐Lahad, Shachar Zuckerman, Hagar Mor‐Shaked, Vardiella Meiner, Kevin T. Booth, and John Pappas
- Subjects
Genetics ,Genetics (clinical) - Published
- 2022
11. Clinical Heterogeneity and Different Phenotypes in Patients with SETD2 Variants: 18 New Patients and Review of the Literature
- Author
-
Lapunzina, Alejandro Parra, Rachel Rabin, John Pappas, Patricia Pascual, Mario Cazalla, Pedro Arias, Natalia Gallego-Zazo, Alfredo Santana, Ignacio Arroyo, Mercè Artigas, Harry Pachajoa, Yasemin Alanay, Ozlem Akgun-Dogan, Lyse Ruaud, Nathalie Couque, Jonathan Levy, Gloria Liliana Porras-Hurtado, Fernando Santos-Simarro, Maria Juliana Ballesta-Martinez, Encarna Guillén-Navarro, Hugo Muñoz-Hernández, Julián Nevado, Spanish OverGrowth Registry Initiative Spanish OverGrowth Registry Initiative, Jair Tenorio-Castano, and Pablo
- Subjects
SETD2 ,Luscan–Lumish syndrome ,Rabin–Pappas syndrome ,intellectual developmental disorder ,autosomal dominant 70 ,overgrowth ,intellectual disability ,autism spectrum disorder ,MRD70 ,LLS ,RAPAS - Abstract
SETD2 belongs to the family of histone methyltransferase proteins and has been associated with three nosologically distinct entities with different clinical and molecular features: Luscan–Lumish syndrome (LLS), intellectual developmental disorder, autosomal dominant 70 (MRD70), and Rabin–Pappas syndrome (RAPAS). LLS [MIM #616831] is an overgrowth disorder with multisystem involvement including intellectual disability, speech delay, autism spectrum disorder (ASD), macrocephaly, tall stature, and motor delay. RAPAS [MIM #6201551] is a recently reported multisystemic disorder characterized by severely impaired global and intellectual development, hypotonia, feeding difficulties with failure to thrive, microcephaly, and dysmorphic facial features. Other neurologic findings may include seizures, hearing loss, ophthalmologic defects, and brain imaging abnormalities. There is variable involvement of other organ systems, including skeletal, genitourinary, cardiac, and potentially endocrine. Three patients who carried the missense variant p.Arg1740Gln in SETD2 were reported with a moderately impaired intellectual disability, speech difficulties, and behavioral abnormalities. More variable findings included hypotonia and dysmorphic features. Due to the differences with the two previous phenotypes, this association was then named intellectual developmental disorder, autosomal dominant 70 [MIM 620157]. These three disorders seem to be allelic and are caused either by loss-of-function, gain-of-function, or missense variants in the SETD2 gene. Here we describe 18 new patients with variants in SETD2, most of them with the LLS phenotype, and reviewed 33 additional patients with variants in SETD2 that have been previously reported in the scientific literature. This article offers an expansion of the number of reported individuals with LLS and highlights the clinical features and the similarities and differences among the three phenotypes associated with SETD2.
- Published
- 2023
- Full Text
- View/download PDF
12. HERACLITIAN DYNAMICS IN THE ANTIGONE AND THE FALLACY OF THE RIGHT OF THE STRONGEST
- Author
-
Demetra Asimakopoulou and John Pappas
- Abstract
The endless debates on Sophocles' Antigone reflect different analytical perspectives as to the multiple and concurrent dualities intertwined in the drama, like legality and legitimacy, lawfulness and morality, expediency and tradition, humans and the divine. Still, subjective perspectives notwithstanding, a conceptually and aesthetically prevalent duality in the Antigone pertains to the head-on conflict between (king Creon's) material power and (Antigone's) moral strength: Adult Creon's reasoning for enforcing his deadly kingly edict on adolescent Antigone is well founded with respect to the imperative need to maintain law and order in his dominion, as a necessary condition for reinstituting socio-political stability and ensuring the security and independence of his polis, especially in extreme conditions of civil warfare; nevertheless his aesthetically hubristic exertion of power leads the monarch's own family to self-destruction. In this mythological twist of fate, the Antigone brings to the fore artistically (and barely disputably) the limitations of the so-called right of the strongest, because the strongest is never strong enough to be always the master, unless he transforms strength into right, and obedience into duty.
- Published
- 2022
13. Heterozygous variants in CTR9, which encodes a major component of the PAF1 complex, are associated with a neurodevelopmental disorder
- Author
-
Marije Meuwissen, Aline Verstraeten, Emmanuelle Ranza, Justyna Iwaszkiewicz, Maaike Bastiaansen, Ligia Mateiu, Merlijn Nemegeer, Josephina A.N. Meester, Alexandra Afenjar, Michelle Amaral, Diana Ballhausen, Sarah Barnett, Magalie Barth, Bob Asselbergh, Katrien Spaas, Bavo Heeman, Jennifer Bassetti, Patrick Blackburn, Marie Schaer, Xavier Blanc, Vincent Zoete, Kari Casas, Thomas Courtin, Diane Doummar, Frédéric Guerry, Boris Keren, John Pappas, Rachel Rabin, Amber Begtrup, Marwan Shinawi, Anneke T. Vulto-van Silfhout, Tjitske Kleefstra, Matias Wagner, Alban Ziegler, Elise Schaefer, Benedicte Gerard, Charlotte I. De Bie, Sjoerd J.B. Holwerda, Mary Alice Abbot, Stylianos E. Antonarakis, and Bart Loeys
- Subjects
Heterozygote ,All institutes and research themes of the Radboud University Medical Center ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Gene Expression Regulation ,Autism Spectrum Disorder ,Neurodevelopmental Disorders ,Intellectual Disability ,Humans ,Human medicine ,Phosphoproteins ,Genetics (clinical) ,Transcription Factors - Abstract
PURPOSE CTR9 is a subunit of the PAF1 complex (PAF1C) that plays a crucial role in transcription regulation by binding CTR9 to RNA polymerase II. It is involved in transcription-coupled histone modification through promoting H3K4 and H3K36 methylation. We describe the clinical and molecular studies in 13 probands, harboring likely pathogenic CTR9 missense variants, collected through GeneMatcher. METHODS Exome sequencing was performed in all individuals. CTR9 variants were assessed through 3-dimensional modeling of the activated human transcription complex Pol II-DSIF-PAF-SPT6 and the PAF1/CTR9 complex. H3K4/H3K36 methylation analysis, mitophagy assessment based on tetramethylrhodamine ethyl ester perchlorate immunofluorescence, and RNA-sequencing in skin fibroblasts from 4 patients was performed. RESULTS Common clinical findings were variable degrees of intellectual disability, hypotonia, joint hyperlaxity, speech delay, coordination problems, tremor, and autism spectrum disorder. Mild dysmorphism and cardiac anomalies were less frequent. For 11 CTR9 variants, de novo occurrence was shown. Three-dimensional modeling predicted a likely disruptive effect of the variants on local CTR9 structure and protein interaction. Additional studies in fibroblasts did not unveil the downstream functional consequences of the identified variants. CONCLUSION We describe a neurodevelopmental disorder caused by (mainly) de novo variants in CTR9, likely affecting PAF1C function.
- Published
- 2022
14. Bi-allelic variants in NAE1 cause intellectual disability, ischiopubic hypoplasia, stress-mediated lymphopenia and neurodegeneration
- Author
-
Irena J.J. Muffels, Imre F. Schene, Holger Rehmann, Maarten P.G. Massink, Maria M. van der Wal, Corinna Bauder, Martha Labeur, Natalia G. Armando, Maarten H. Lequin, Michiel L. Houben, Jaques C. Giltay, Saskia Haitjema, Albert Huisman, Fleur Vansenne, Judith Bluvstein, John Pappas, Lala V. Shailee, Yuri A. Zarate, Michal Mokry, Gijs W. van Haaften, Edward E.S. Nieuwenhuis, Damian Refojo, Femke van Wijk, Sabine A. Fuchs, and Peter M. van Hasselt
- Subjects
Genetics ,Lymphopenia ,Nae1 ,Neddylation ,Neurodegeneration ,Ocurrence Ratio ,Phenotypic Specificity ,Post-translational Protein Modification ,Proteasome ,Ubiquitination ,Article ,Genetics (clinical) - Abstract
Neddylation has been implicated in various cellular pathways and in the pathophysiology of numerous diseases. We identified four individuals with bi-allelic variants in NAE1, which encodes the neddylation E1 enzyme. Pathogenicity was supported by decreased NAE1 abundance and overlapping clinical and cellular phenotypes. To delineate how cellular consequences of NAE1 deficiency would lead to the clinical phenotype, we focused primarily on the rarest phenotypic features, based on the assumption that these would best reflect the pathophysiology at stake. Two of the rarest features, neuronal loss and lymphopenia worsening during infections, suggest that NAE1 is required during cellular stress caused by infections to protect against cell death. In support, we found that stressing the proteasome system with MG132-requiring upregulation of neddylation to restore proteasomal function and proteasomal stress-led to increased cell death in fibroblasts of individuals with NAE1 genetic variants. Additionally, we found decreased lymphocyte counts after CD3/CD28 stimulation and decreased NF-κB translocation in individuals with NAE1 variants. The rarest phenotypic feature-delayed closure of the ischiopubic rami-correlated with significant downregulation of RUN2X and SOX9 expression in transcriptomic data of fibroblasts. Both genes are involved in the pathophysiology of ischiopubic hypoplasia. Thus, we show that NAE1 plays a major role in (skeletal) development and cellular homeostasis during stress. Our approach suggests that a focus on rare phenotypic features is able to provide significant pathophysiological insights in diseases caused by mutations in genes with pleiotropic effects.
- Published
- 2023
15. Biochemical characterization of two novel mutations in the human high-affinity choline transporter 1 identified in a patient with congenital myasthenic syndrome
- Author
-
Midhat Rizvi, Tina K Truong, Janet Zhou, Manav Batta, Ellen S Moran, John Pappas, Mary Lynn Chu, Oana Caluseriu, Gilad D Evrony, Elaine M Leslie, and Emmanuelle Cordat
- Subjects
Genetics ,General Medicine ,Molecular Biology ,Genetics (clinical) - Abstract
Congenital myasthenic syndrome (CMS) is a heterogeneous condition associated with 34 different genes, including SLC5A7, which encodes the high-affinity choline transporter 1 (CHT1). CHT1 is expressed in presynaptic neurons of the neuromuscular junction where it uses the inward sodium gradient to reuptake choline. Biallelic CHT1 mutations often lead to neonatal lethality, and less commonly to non-lethal motor weakness and developmental delays. Here, we report detailed biochemical characterization of two novel mutations in CHT1, p.I294T and p.D349N, which we identified in an 11-year-old patient with a history of neonatal respiratory distress, and subsequent hypotonia and global developmental delay. Heterologous expression of each CHT1 mutant in human embryonic kidney cells showed two different mechanisms of reduced protein function. The p.I294T CHT1 mutant transporter function was detectable, but its abundance and half-life were significantly reduced. In contrast, the p.D349N CHT1 mutant was abundantly expressed at the cell membrane, but transporter function was absent. The residual function of the p.I294T CHT1 mutant may explain the non-lethal form of CMS in this patient, and the divergent mechanisms of reduced CHT1 function that we identified may guide future functional studies of the CHT1 myasthenic syndrome. Based on these in vitro studies that provided a diagnosis, treatment with cholinesterase inhibitor together with physical and occupational therapy significantly improved the patient’s strength and quality of life.
- Published
- 2022
16. Expanding the phenotype of <scp> ASXL3 </scp> ‐related syndrome: A comprehensive description of 45 unpublished individuals with inherited and de novo pathogenic variants in <scp> ASXL3 </scp>
- Author
-
Katherine Bergstrom, Nichola Foulds, Yue Si, Anne Slavotinek, John Dean, Evan Reid, Ruth Armstrong, Charlotte W. Ockeloen, Richard Fisher, Maria J. Guillen Sacoto, Dayna Morel, Fowzan S. Alkuraya, Costa Cinzia, Thomas D. Challman, Samantha A. Schrier Vergano, Francisca Milan Zamora, Naomi Meeks, John Pappas, Katheryn Grand, Abhijit Dixit, Julie S. Cohen, Ddd Study, Marjolein H. Willemsen, Serwet Demirdas, Rachel Harrison, Usha Kini, Bertrand Isidor, Patricia Blanchet, Emily Palen, Arjan Bouman, Jagdeep S. Walia, Ruth Newbury-Ecob, Rachel Rabin, Shadi Albaba, Diana Johnson, Paolo Prontera, Paula Girotto, Ange-Line Bruel, Meena Balasubramanian, Nicola K. Ragge, Schaida Schirwani, Deborah L. Renaud, Christopher Cunniff, John M. Graham, Natalie Dykzeul, Swati Naik, Valerie Slegesky, Hessa F Albassam, Maria Giovanna Tedesco, Sally Ann Lynch, Julie Vogt, Natalie Hauser, Dong Li, Deanna Alexis Carere, and Benjamin Cogné
- Subjects
Genetics ,Biology ,medicine.disease ,Phenotype ,Hypotonia ,Natural history ,Neurodevelopmental disorder ,Intellectual disability ,medicine ,Missense mutation ,Hypertelorism ,medicine.symptom ,Genetics (clinical) ,Sequence (medicine) - Abstract
The study aimed at widening the clinical and genetic spectrum of ASXL3-related syndrome, a neurodevelopmental disorder, caused by truncating variants in the ASXL3 gene. In this international collaborative study, we have undertaken a detailed clinical and molecular analysis of 45 previously unpublished individuals with ASXL3-related syndrome, as well as a review of all previously published individuals. We have reviewed the rather limited functional characterization of pathogenic variants in ASXL3 and discuss current understanding of the consequences of the different ASXL3 variants. In this comprehensive analysis of ASXL3-related syndrome, we define its natural history and clinical evolution occurring with age. We report familial ASXL3 pathogenic variants, characterize the phenotype in mildly affected individuals and discuss nonpenetrance. We also discuss the role of missense variants in ASXL3. We delineate a variable but consistent phenotype. The most characteristic features are neurodevelopmental delay with consistently limited speech, significant neuro-behavioral issues, hypotonia, and feeding difficulties. Distinctive features include downslanting palpebral fissures, hypertelorism, tubular nose with a prominent nasal bridge, and low-hanging columella. The presented data will inform clinical management of individuals with ASXL3-related syndrome and improve interpretation of new ASXL3 sequence variants.
- Published
- 2021
17. CDK19-related disorder results from both loss-of-function and gain-of-function de novo missense variants
- Author
-
Denise Horn, Anna-Elina Lehesjoki, Janina Gburek-Augustat, Amarilis Sanchez-Valle, Kenjiro Kosaki, Katherine Anderson, Anna-Kaisa Anttonen, Tohru Ishitani, Katja Kloth, Manuel Holtgrewe, Sora Harako, Rhonda E. Schnur, Maria J. Guillen Sacoto, Yutaka Hirose, Seiji Mizuno, Shizuka Ishitani, Kota Abe, Tadashi Kaname, Yusaku Kaido, Johannes Luppe, Nadja Ehmke, Michelle M. Morrow, John Pappas, Tatjana Bierhals, Masayuki Oginuma, David Viskochil, Yoshiaki Ohkuma, Tomoko Uehara, Konrad Platzer, Courtney L. Edgar-Zarate, Rachel Rabin, Yuri A. Zarate, Mikko Muona, Nobuhiko Okamoto, Department of Medical and Clinical Genetics, University of Helsinki, Medicum, HUSLAB, Anna-Elina Lehesjoki / Principal Investigator, Neuroscience Center, and Helsinki University Hospital Area
- Subjects
0301 basic medicine ,GENES ,CDK8 ,Mutation, Missense ,PROTEIN ,030105 genetics & heredity ,SEQUENCE ,03 medical and health sciences ,Neurodevelopmental disorder ,Intellectual Disability ,medicine ,Animals ,Humans ,Missense mutation ,Kinase activity ,Zebrafish ,Genetics (clinical) ,Loss function ,MEDIATOR COMPLEX ,Genetics ,biology ,Autophosphorylation ,Infant ,biology.organism_classification ,medicine.disease ,Cyclin-Dependent Kinases ,Hypotonia ,MED12 ,030104 developmental biology ,Protein kinase domain ,Neurodevelopmental Disorders ,Gain of Function Mutation ,3111 Biomedicine ,medicine.symptom - Abstract
Purpose To expand the recent description of a new neurodevelopmental syndrome related to alterations in CDK19. Methods Individuals were identified through international collaboration. Functional studies included autophosphorylation assays for CDK19 Gly28Arg and Tyr32His variants and in vivo zebrafish assays of the CDK19(G28R) and CDK19(Y32H). Results We describe 11 unrelated individuals (age range: 9 months to 14 years) with de novo missense variants mapped to the kinase domain of CDK19, including two recurrent changes at residues Tyr32 and Gly28. In vitro autophosphorylation and substrate phosphorylation assays revealed that kinase activity of protein was lower for p.Gly28Arg and higher for p.Tyr32His substitutions compared with that of the wild-type protein. Injection of CDK19 messenger RNA (mRNA) with either the Tyr32His or the Gly28Arg variants using in vivo zebrafish model significantly increased fraction of embryos with morphological abnormalities. Overall, the phenotype of the now 14 individuals with CDK19-related disorder includes universal developmental delay and facial dysmorphism, hypotonia (79%), seizures (64%), ophthalmologic anomalies (64%), and autism/autistic traits (56%). Conclusion CDK19 de novo missense variants are responsible for a novel neurodevelopmental disorder. Both kinase assay and zebrafish experiments showed that the pathogenetic mechanism may be more diverse than previously thought.
- Published
- 2021
18. Severe epileptic encephalopathy associated with compound heterozygosity of <scp>THG1L</scp> variants in the Ashkenazi Jewish population
- Author
-
Rachel Rabin, Yoel Hirsch, Ahron Ekstein, Martin Johansson, Joseph Ekstein, and John Pappas
- Subjects
Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,education.field_of_study ,TRNA modification ,Microcephaly ,Ataxia ,Cerebellar ataxia ,business.industry ,Population ,Mitochondrial tRNA modification ,Compound heterozygosity ,medicine.disease ,medicine ,medicine.symptom ,education ,business ,Cerebellar hypoplasia ,Genetics (clinical) - Abstract
THG1L-associated autosomal recessive ataxia belongs to a group of disorders that occur due to abnormal mitochondrial tRNA modification. The product of THG1L is the tRNA-histidine guanylyltransferase 1-like enzyme that catalyzes the 3'-5"addition of guanine to the 5"-end of tRNA-histidine in the mitochondrion. To date, five individuals with homozygosity for p.(Val55Ala) in THG1L have been reported and presented with mild delays or normal development and cerebellar dysfunction. We present seven individuals with biallelic variants in THG1L. Three individuals were compound heterozygous for the p.(Cys51Trp) and p.(Val55Ala) variants and presented with profound developmental delays, microcephaly, intractable epilepsy, and cerebellar hypoplasia. Four siblings were homozygous for the p.(Val55Ala) variant and presented with cerebellar ataxia with cerebellar vermis hypoplasia, dysarthria, mild developmental delays, and normal/near-normal cognition. All seven patients were of Ashkenazi Jewish descent. Carrier rates for the two variants were calculated in a cohort of 26,731 Ashkenazi Jewish individuals tested by the Dor Yeshorim screening program. The p.(Cys51Trp) variant is novel and was found in 40 of the Ashkenazi Jewish individuals tested, with a carrier rate of 1 in 668 (0.15%). The p.(Val55Ala) variant was found in 229 of the Ashkenazi Jewish individuals tested, with a carrier rate of 1 in 117 (0.85%). The individuals with compound heterozygosity of the p.(Val55Ala) and p.(Cys51Trp) variants expand the phenotypic spectrum of THG1L-related disorders to include severe epileptic encephalopathy. The individuals with homozygosity of the p.(V55A) variant further establish the associated mild and slowly progressive or nonprogressive neurodevelopmental phenotype.
- Published
- 2021
19. A synonymous variant in MYO15A enriched in the Ashkenazi Jewish population causes autosomal recessive hearing loss due to abnormal splicing
- Author
-
Michal Macarov, Yoel Hirsch, Karen B. Avraham, Josef Ekstein, Cynthia C. Morton, Richard J.H. Smith, Devorah Yefet, Jun Shen, Tzvi Weiden, Chantal Farra, Diana L. Kolbe, Kevin T. Booth, John Pappas, Rachel Rabin, Carla Nishimura, Minjie Luo, Chayada Tangshewinsirikul, Andrea M. Oza, Zippora Brownstein, Adina Quint, Katherine A Lafferty, Kathy L. Frees, Sami S. Amr, Sholem Y. Scher, Margaret A. Kenna, Bella Davidov, Hela Azaiez, and Heidi L. Rehm
- Subjects
Adult ,Male ,MYO15A ,Adolescent ,Hearing loss ,RNA Splicing ,Population ,Genes, Recessive ,Myosins ,Biology ,Compound heterozygosity ,Article ,03 medical and health sciences ,Gene Frequency ,Genetics ,medicine ,Humans ,Child ,Hearing Loss ,education ,Genetics (clinical) ,Genetic testing ,0303 health sciences ,education.field_of_study ,medicine.diagnostic_test ,Genetic heterogeneity ,030305 genetics & heredity ,Infant ,medicine.disease ,Phenotype ,Pedigree ,Child, Preschool ,Jews ,Mutation ,Female ,Sensorineural hearing loss ,medicine.symptom - Abstract
Nonsyndromic hearing loss is genetically heterogeneous. Despite comprehensive genetic testing, many cases remain unsolved because the clinical significance of identified variants is uncertain or because biallelic pathogenic variants are not identified for presumed autosomal recessive cases. Common synonymous variants are often disregarded. Determining the pathogenicity of synonymous variants may improve genetic diagnosis. We report a synonymous variant c.9861 C > T/p.(Gly3287=) in MYO15A in homozygosity or compound heterozygosity with another pathogenic or likely pathogenic MYO15A variant in 10 unrelated families with nonsyndromic sensorineural hearing loss. Biallelic variants in MYO15A were identified in 21 affected and were absent in 22 unaffected siblings. A mini-gene assay confirms that the synonymous variant leads to abnormal splicing. The variant is enriched in the Ashkenazi Jewish population. Individuals carrying biallelic variants involving c.9861 C > T often exhibit progressive post-lingual hearing loss distinct from the congenital profound deafness typically associated with biallelic loss-of-function MYO15A variants. This study establishes the pathogenicity of the c.9861 C > T variant in MYO15A and expands the phenotypic spectrum of MYO15A-related hearing loss. Our work also highlights the importance of multicenter collaboration and data sharing to establish the pathogenicity of a relatively common synonymous variant for improved diagnosis and management of hearing loss.
- Published
- 2021
20. Genotype-phenotype correlation at codon 1740 ofSETD2
- Author
-
Bernt Popp, Shelby Romoser, Lara Menzies, Stacey A. Bélanger, Alireza Radmanesh, Kimberly A. Aldinger, Jennifer Keller-Ramey, Janice Baker, Jane A. Hurst, William B. Dobyns, Schahram Akbarian, Sébastien Jacquemont, Jan Maarten Cobben, Larissa Kerecuk, Kelly Radtke, Joseph T. Shieh, Khadije Jizi, Ian A. Glass, Patrick Watts, Nicola Foulds, Jerica Lenberg, Sumit Punj, George E. Hoganson, Nancy J. Mendelsohn, Rachel Rabin, Ina Sorge, Katarzyna A. Ellsworth, Katharina Löhner, Manuela Siekmeyer, Jennifer Burton, Leah Dowsett, John A. Bernat, Hannah Bombei, John Pappas, Henny H. Lemmink, Francis H. Sansbury, Ingrid M. Wentzensen, Kirsty McWalter, Deborah Osio, Pamela Trapane, Hermine E. Veenstra-Knol, General Paediatrics, Paediatric Genetics, and ANS - Complex Trait Genetics
- Subjects
Male ,Microcephaly ,Mutation, Missense ,Biology ,Nervous System Malformations ,Epigenesis, Genetic ,Histone H3 ,Loss of Function Mutation ,Tubulin ,SETD2 ,Intellectual Disability ,Genetics ,medicine ,Humans ,Missense mutation ,Genetic Predisposition to Disease ,histone modification ,Epigenetics ,AUTISM ,Child ,Codon ,Genetic Association Studies ,Genetics (clinical) ,Loss function ,HYPB/SETD2 ,MARK ,IDENTIFICATION ,MUTATIONS ,METHYLATION ,Infant ,Histone-Lysine N-Methyltransferase ,Methylation ,neurodevelopmental ,medicine.disease ,Histone ,genotype phenotype ,Neurodevelopmental Disorders ,Child, Preschool ,biology.protein ,Female ,clinical genetics - Abstract
The SET domain containing 2, histone lysine methyltransferase encoded by SETD2 is a dual-function methyltransferase for histones and microtubules and plays an important role for transcriptional regulation, genomic stability, and cytoskeletal functions. Specifically, SETD2 is associated with trimethylation of histone H3 at lysine 36 (H3K36me3) and methylation of α-tubulin at lysine 40. Heterozygous loss of function and missense variants have previously been described with Luscan-Lumish syndrome (LLS), which is characterized by overgrowth, neurodevelopmental features, and absence of overt congenital anomalies. We have identified 15 individuals with de novo variants in codon 1740 of SETD2 whose features differ from those with LLS. Group 1 consists of 12 individuals with heterozygous variant c.5218C>T p.(Arg1740Trp) and Group 2 consists of 3 individuals with heterozygous variant c.5219G>A p.(Arg1740Gln). The phenotype of Group 1 includes microcephaly, profound intellectual disability, congenital anomalies affecting several organ systems, and similar facial features. Individuals in Group 2 had moderate to severe intellectual disability, low normal head circumference, and absence of additional major congenital anomalies. While LLS is likely due to loss of function of SETD2, the clinical features seen in individuals with variants affecting codon 1740 are more severe suggesting an alternative mechanism, such as gain of function, effects on epigenetic regulation, or posttranslational modification of the cytoskeleton. Our report is a prime example of different mutations in the same gene causing diverging phenotypes and the features observed in Group 1 suggest a new clinically recognizable syndrome uniquely associated with the heterozygous variant c.5218C>T p.(Arg1740Trp) in SETD2.
- Published
- 2020
21. De Novo Variants in the ATPase Module of MORC2 Cause a Neurodevelopmental Disorder with Growth Retardation and Variable Craniofacial Dysmorphism
- Author
-
Rosanna Weksberg, Courtney Kiss, Marta Szybowska, Nina Ekhilevitch, Irina Anselm, David A. Sweetser, Michael Marble, Kristin Lindstrom, Cara Forster, Haley Streff, Renata C. Gallagher, John Pappas, Jessica Nance, Patricia G Wheeler, Melissa A. Walker, Grace Yoon, Carl E. Stafstrom, Weiyi Mu, Mary Kay Koenig, Wei Wang, Jane Juusola, Lauren C. Briere, Eric Muller, Julie S. Cohen, E. Hallie Andrew, Frances A. High, Cheryl Cytrynbaum, Jamie L. Fraser, Joel B. Krier, Hannah Meddaugh, Ali Fatemi, Robert E. Kingston, Kristin W. Barañano, Bridget Ostrem, Maria J. Guillen Sacoto, Ellen Moran, Marvin R. Natowicz, Karin Weiss, Erin Torti, Iva A. Tchasovnikarova, William J. Craigen, Cara Inglese, Andrea Guerin, Matthew J. Elrick, Devon Haynes, Chantal F. Morel, and Adam L. Numis
- Subjects
Adult ,Male ,0301 basic medicine ,Heterozygote ,Microcephaly ,Adolescent ,Biology ,Chromatin remodeling ,Craniofacial Abnormalities ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Neurodevelopmental disorder ,Intellectual Disability ,Report ,Intellectual disability ,Genetics ,medicine ,Humans ,Child ,Growth Disorders ,Genetics (clinical) ,Exome sequencing ,Adenosine Triphosphatases ,Genetic Diseases, Inborn ,Infant ,Spinal muscular atrophy ,Hyporeflexia ,Middle Aged ,medicine.disease ,Phenotype ,030104 developmental biology ,Neurodevelopmental Disorders ,Child, Preschool ,Mutation ,Female ,medicine.symptom ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
MORC2 encodes an ATPase that plays a role in chromatin remodeling, DNA repair, and transcriptional regulation. Heterozygous variants in MORC2 have been reported in individuals with autosomal-dominant Charcot-Marie-Tooth disease type 2Z and spinal muscular atrophy, and the onset of symptoms ranges from infancy to the second decade of life. Here, we present a cohort of 20 individuals referred for exome sequencing who harbor pathogenic variants in the ATPase module of MORC2. Individuals presented with a similar phenotype consisting of developmental delay, intellectual disability, growth retardation, microcephaly, and variable craniofacial dysmorphism. Weakness, hyporeflexia, and electrophysiologic abnormalities suggestive of neuropathy were frequently observed but were not the predominant feature. Five of 18 individuals for whom brain imaging was available had lesions reminiscent of those observed in Leigh syndrome, and five of six individuals who had dilated eye exams had retinal pigmentary abnormalities. Functional assays revealed that these MORC2 variants result in hyperactivation of epigenetic silencing by the HUSH complex, supporting their pathogenicity. The described set of morphological, growth, developmental, and neurological findings and medical concerns expands the spectrum of genetic disorders resulting from pathogenic variants in MORC2.
- Published
- 2020
22. Severe Acute Systemic Reaction After the First Injections of Ixekizumab
- Author
-
John, Pappas, Maryam, Liaqat, and Analisa V, Halpern
- Subjects
Adult ,Treatment Outcome ,Arthritis, Psoriatic ,Graft vs Host Disease ,Humans ,Psoriasis ,Female ,Antibodies, Monoclonal, Humanized ,Severity of Illness Index - Abstract
We report the case of a 39-year-old woman who presented with generalized malaise; lymphadenopathy; arthritis; dactylitis; ecchymosis; acute onycholysis; and a red, nonpruritic, nonscaly, mottled rash on the right breast 24 hours after the first injections of ixekizumab for psoriasis and psoriatic arthritis. Ixekizumab is a humanized IgG4 monoclonal antibody that binds to IL-17A. Adverse events of ixeki-zumab include infection, inflammatory bowel disease, candidiasis and tinea infections, severe injection-site reactions, arthralgia, headache, infections, neutropenia, and thrombocytopenia. Other biologics, specifically tumor necrosis factor (TNF) inhibitors, have been reported to cause onycholysis attributed to immune dysregulation. We propose that ixekizumab alters the inflammatory cascade that underlies the induction of acute onycholysis and arthritis.
- Published
- 2022
23. ODP380 Case of Skeletal Dysplasia in Post-Menarchal Pediatric Patient due to Novel Mutation of CSGALNACT1
- Author
-
Merilyn Baby, Emily Breidbart, Brenda Kohn, and John Pappas
- Subjects
Endocrinology, Diabetes and Metabolism - Abstract
We report the case of a 14 year old Yemenite female diagnosed with autosomal recessive skeletal dysplasia secondary to a novel mutation of CSGALNACT1. She was initially referred to pediatric endocrine for poor linear growth at the age of 11 year 7 months. At that time, her height was well below the growth curve with z-score of -3.51. She had no reported history of intellectual disability. She had a normal MRI of the brain as well as normal growth factors, growth hormone stimulation test, thyroid function, IgA celiac antibodies, and a normal karyotype (46,XX). SHOX and microarray were negative. Her bone age was slightly delayed at 10-11 years with a chronological age of 11y8m. On initial evaluation by genetics, she was normocephalic with no dysmorphic facial features. She had proportionate limbs but was noted to have mild lumbar lordosis, increased joint laxity, and flattened feet with fifth toe clinodactyly. Notably there was a family history of consanguinity (parents are first cousins once removed). A skeletal dysplasia gene panel was not thought to be indicated, and she was referred back to endocrine. She was lost to follow up with endocrine but returned to clinic after 1 year when she was post-menarchal. She continued to have poor linear growth. Upon reevaluation, she was noted to additionally have a high-arched palate and shortened hallux. A panel for skeletal dysplasia was sent which detected a novel homozygous mutation of CSGALNACT1 with autosomal recessive inheritance. CSGALNACT1 encodes chondroitin sulfate N-acetylgalactosaminyl transferase which is crucial for chondroitin sulfate chain biosynthesis and glycosaminoglycan synthesis. Congenital disorders of glycosylation are genetically inherited conditions due to abnormal glycan biosynthesis. Glycosaminoglycans (GAGs) are vital in normal development of cartilage and the brain. We performed a PubMed search and saw four reported cases of individuals with mutations of CSGALNACT1. Clinically these individuals had relative macrocephaly, rhizomelia, hyperlordosis, joint laxity, and mild neurodevelopmental delay. Reported radiographic findings of affected individuals include advanced bone ages, flattened acetabular roofs, and vertebral anomalies. Our patient in contrast did not have overt dysmorphic features on exam nor reported intellectual disability to prompt a more immediate workup for skeletal dysplasia. There have been only a handful of cases with CSGALNACT1 mutations and skeletal dysplasia. These individuals more typically have findings of dysmorphia on exam including macrocephaly, lordosis, and joint laxity. Our patient lacked these typical features that are more normally associated with skeletal dysplasia yet was revealed to have a novel mutation of CSGALNACT1. Based on her initial genetic evaluation, her clinical findings were subtle and did not indicate a workup for skeletal dysplasia. Her case however shows that even in the absence of dysmorphic features, skeletal dysplasia should be considered in individuals with severe short stature. Presentation: No date and time listed
- Published
- 2022
24. eP206: Novel variant in ARSA associated with late infantile metachromatic leukodystrophy and heterozygote rate in individuals of Ashkenazi Jewish ancestry
- Author
-
John Pappas, Rachel Rabin, Pramod Mistry, Yoel Hirsch, and Naomi Yachelevich
- Subjects
Genetics (clinical) - Published
- 2022
25. Effects of Processing Conditions on Laser Direct Deposited Alumina Ceramics
- Author
-
Xiangyang Dong and John Pappas
- Published
- 2021
26. Clinical spectrum of individuals with pathogenic N F1 missense variants affecting p.Met1149, p.Arg1276, and p.Lys1423: genotype–phenotype study in neurofibromatosis type 1
- Author
-
Giulio Piluso, Katharina Wimmer, Veronica Saletti, Eniko K. Pivnick, Geraldine Kelly-Mancuso, Karen W. Gripp, Cristin Griffis, Louanne Hudgins, Alessandro De Luca, Michael F. Wangler, M. Daniela D'Agostino, Marica Eoli, Cynthia M. Powell, Laura A. Baker, Mayra Martinez Ojeda, Silvia Esposito, Elizabeth A. Sellars, Kory Keller, David D. Weaver, James T. Bennett, Nicole J. Ullrich, Allison L. Goetsch, Donald Basel, Bruce R. Korf, Stephanie Fox, Katelyn Hodge, Laura Dosa, Robert S. Greenwood, Mario Bengala, Andrea M. Lewis, Ruth Sheffer, Valentina Pinna, Fanny Cortés, Dusica Babovic-Vuksanovic, Aaina Kochhar, Rosemarie Smith, Concepción Hernández-Chico, Elizabeth Siqveland, Robert Listernick, Lola K. Clarkson, Punita Gupta, E. Haan, Martin B. Delatycki, Amy Theos, Noa Ruhrman Shahar, Teresa Giugliano, Carey McDougall, Mitch Cunningham, David W. Stockton, Tom Callens, Maria Cristina Digilio, Yunjia Chen, Ludwine Messiaen, Eva Trevisson, Samantha A. Schrier Vergano, Caleb Rogers, Magdalena Koczkowska, Kathleen Claes, Christine Fauth, Jan Liebelt, Pamela Trapane, Eric Johns, John M. Slopis, Chelsea Chambers, Tamara L. Haygarth, Lesley K. McGregor, Alberto Spalice, Małgorzata J.M. Nowaczyk, Mary Ella M Pierpont, Kaleb Yohay, Alicia Gomes, Vickie Zurcher, Gail E. Tomlinson, Angie W. Lichty, Stephanie E Wallace, Rachel K. Hachen, Isabelle Maystadt, S. Lane Rutledge, Yael Goldberg, Grace Tran, Ulrich A. Schatz, Allison Schreiber, Jenneke van den Ende, Michael J. Lyons, Mary Louise Freckmann, Kim Armfield Uhas, Alesha D. Hicks, Maurizio Clementi, Haley Streff, June Ortenberg, John Pappas, Nancy J. Mendelsohn, Sandra Janssens, Karin Panzer, Yolanda Martin, Elaine H. Zackai, Sandra Giustini, Linlea Armstrong, Katherine A. Bosanko, Angela Sharp, Daryl A. Scott, Jonathan Zonana, Robert J. Hopkin, Eric Legius, Dinel A. Pond, Daniela Melis, Claudia Santoro, and Sarah A. Sandaradura
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,0303 health sciences ,medicine.medical_specialty ,education.field_of_study ,Pulmonic stenosis ,030305 genetics & heredity ,Population ,Spinal neurofibromas ,Biology ,medicine.disease ,Phenotype ,Gastroenterology ,nervous system diseases ,03 medical and health sciences ,Internal medicine ,Cohort ,Genetics ,medicine ,Missense mutation ,Noonan syndrome ,Neurofibromatosis ,education ,Genetics (clinical) ,030304 developmental biology - Abstract
We report 281 individuals carrying a pathogenic recurrent NF1 missense variant at p.Met1149, p.Arg1276, or p.Lys1423, representing three nontruncating NF1 hotspots in the University of Alabama at Birmingham (UAB) cohort, together identified in 1.8% of unrelated NF1 individuals. About 25% (95% confidence interval: 20.5-31.2%) of individuals heterozygous for a pathogenic NF1 p.Met1149, p.Arg1276, or p.Lys1423 missense variant had a Noonan-like phenotype, which is significantly more compared with the "classic" NF1-affected cohorts (all p < .0001). Furthermore, p.Arg1276 and p.Lys1423 pathogenic missense variants were associated with a high prevalence of cardiovascular abnormalities, including pulmonic stenosis (all p < .0001), while p.Arg1276 variants had a high prevalence of symptomatic spinal neurofibromas (p < .0001) compared with "classic" NF1-affected cohorts. However, p.Met1149-positive individuals had a mild phenotype, characterized mainly by pigmentary manifestations without externally visible plexiform neurofibromas, symptomatic spinal neurofibromas or symptomatic optic pathway gliomas. As up to 0.4% of unrelated individuals in the UAB cohort carries a p.Met1149 missense variant, this finding will contribute to more accurate stratification of a significant number of NF1 individuals. Although clinically relevant genotype-phenotype correlations are rare in NF1, each affecting only a small percentage of individuals, together they impact counseling and management of a significant number of the NF1 population.
- Published
- 2019
27. In vivo epigenetic editing of Sema6a promoter reverses transcallosal dysconnectivity caused by C11orf46/Arl14ep risk gene
- Author
-
Cyril J. Peter, Atsushi Saito, John Pappas, Tariq Fayyad, Sergio Espeso-Gil, Joan C. Han, Schahram Akbarian, Atsushi Kamiya, Achla Gupta, Mohika Nagpal, Gabriel Perez, Lakshmi A. Devi, John A. Butman, Aslihan Dincer, Francois Lalonde, Yuya Tanaka, Emily Alway, Chana Ratner, and Yuto Hasegawa
- Subjects
0301 basic medicine ,General Physics and Astronomy ,Semaphorins ,Biochemistry ,Corpus Callosum ,Epigenesis, Genetic ,Epigenome ,0302 clinical medicine ,lcsh:Science ,Promoter Regions, Genetic ,Gene Editing ,Regulation of gene expression ,Gene knockdown ,Multidisciplinary ,Histocytochemistry ,Nuclear Proteins ,Biogeochemistry ,Chromatin ,Phenotype ,Haploinsufficiency ,Protein Binding ,Science ,Repressor ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,03 medical and health sciences ,Neurites ,Animals ,Humans ,Genetic Predisposition to Disease ,Protein Methyltransferases ,Epigenetics ,Histone-Lysine N-Methyltransferase ,General Chemistry ,Axons ,Mice, Inbred C57BL ,Adaptor Proteins, Vesicular Transport ,HEK293 Cells ,030104 developmental biology ,Gene Expression Regulation ,Mutagenesis ,RNAi ,lcsh:Q ,Nerve Net ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Many neuropsychiatric risk genes contribute to epigenetic regulation but little is known about specific chromatin-associated mechanisms governing the formation of neuronal connectivity. Here we show that transcallosal connectivity is critically dependent on C11orf46, a nuclear protein encoded in the chromosome 11p13 WAGR risk locus. C11orf46 haploinsufficiency was associated with hypoplasia of the corpus callosum. C11orf46 knockdown disrupted transcallosal projections and was rescued by wild type C11orf46 but not the C11orf46R236H mutant associated with intellectual disability. Multiple genes encoding key regulators of axonal development, including Sema6a, were hyperexpressed in C11orf46-knockdown neurons. RNA-guided epigenetic editing of Sema6a gene promoters via a dCas9-SunTag system with C11orf46 binding normalized SEMA6A expression and rescued transcallosal dysconnectivity via repressive chromatin remodeling by the SETDB1 repressor complex. Our study demonstrates that interhemispheric communication is sensitive to locus-specific remodeling of neuronal chromatin, revealing the therapeutic potential for shaping the brain’s connectome via gene-targeted designer activators and repressor proteins., Although many neuropsychiatric risk genes are known to contribute to epigenetic regulation of gene expression, very little is known about specific chromatin-associated mechanisms that govern the formation and maintenance of neuronal connectivity. Here, the authors report that transcallosal connectivity is critically dependent on C11orf46/ARL14EP, a nuclear protein encoded in the chromosome 11p13 WAGR risk locus, and that RNA-guided epigenetic editing of hyperexpressed Sema6a gene promoters in C11orf46-knockdown neurons resulted in normalization of expression and rescue of transcallosal dysconnectivity via repressive chromatin remodeling.
- Published
- 2019
28. HIST1H1E heterozygous protein‐truncating variants cause a recognizable syndrome with intellectual disability and distinctive facial gestalt: A study to clarify the HIST1H1E syndrome phenotype in 30 individuals
- Author
-
John M. Graham, Anna Ardissone, Dieter Kotzot, Paul R. Mark, Anna Zachariou, Guillermo Lay-Son, Allyn McConkie-Rosell, John Pappas, Karen Low, Fiona Stewart, Chey Loveday, Brian G. Skotko, Melissa Lees, Helen Stewart, Ho Ming Luk, Cheryl Cytrynbaum, Rachel Horton, Siddharth Banka, Gerard Marion, Deborah J. Shears, Marie T. McDonald, Ricardo A. Verdugo, Christine Coubes, Yuri A. Zarate, Christophe Phillipe, Katrina Tatton-Brown, Clare Allen, Deepika D.Cunha Burkardt, Rosanna Weksberg, I. Karen Temple, Alexia Bourgois, David J. Amor, Frédéric Tran Mau-Them, Laurence Faivre, Case Western Reserve University [Cleveland], The institute of cancer research [London], University College London Hospitals (UCLH), Murdoch Children's Research Institute (MCRI), University of Melbourne, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', University of Manchester [Manchester], Manchester University NHS Foundation Trust (MFT), Service de Génétique [CHU Caen], CHU Caen, Normandie Université (NU)-Tumorothèque de Caen Basse-Normandie (TCBN)-Normandie Université (NU)-Tumorothèque de Caen Basse-Normandie (TCBN)-Université de Caen Normandie (UNICAEN), Normandie Université (NU), Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), The Hospital for sick children [Toronto] (SickKids), Hôpital d'Enfants [CHU Dijon], Hôpital du Bocage, Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand (CHU Dijon)-Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand (CHU Dijon), Biologie, génétique et thérapies ostéoarticulaires et respiratoires (BIOTARGEN), Université de Caen Normandie (UNICAEN), Normandie Université (NU)-Normandie Université (NU), University Hospital Southampton NHS Foundation Trust, Paracelsus Medizinische Privatuniversität = Paracelsus Medical University (PMU), Pontificia Universidad Católica de Chile (UC), Great Ormond Street Hospital for Children [London] (GOSH), University Hospitals Bristol, Department of Health Clinical Genetic Service Centre, Spectrum Health [Grand Rapids], Department of Molecular Genetics and Microbiology [Durham] (MGM), Duke University [Durham], New York University School of Medicine (NYU), New York University School of Medicine, NYU System (NYU)-NYU System (NYU), Lipides - Nutrition - Cancer [Dijon - U1231] (LNC), Université de Bourgogne (UB)-Institut National de la Santé et de la Recherche Médicale (INSERM)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement, Department of Clinical Genetics [Churchill Hospital], Churchill Hospital Oxford Centre for Haematology, Harvard Medical School [Boston] (HMS), Belfast City Hospital, Oxford University Hospitals NHS Trust, University of Oxford [Oxford], University of Southampton, Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand (CHU Dijon), Universitad de Chile, Arkansas Children's Hospital, Cedars-Sinai Medical Center, St George’s University Hospitals, and Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
Heterozygote ,Bioinformatics ,Corpus callosum ,Rahman syndrome ,Histones ,03 medical and health sciences ,Frontal Bossing ,0302 clinical medicine ,HIST1H1E ,Gene cluster ,Intellectual disability ,Genetics ,Humans ,Learning ,Medicine ,Epigenetics ,Genetics (clinical) ,030304 developmental biology ,Behavior ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,0303 health sciences ,epigenetic regulator gene ,biology ,business.industry ,Facies ,Heterozygote advantage ,Syndrome ,medicine.disease ,Phenotype ,Histone ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,intellectual disability ,Mutation ,biology.protein ,Growth and Development ,business ,030217 neurology & neurosurgery - Abstract
International audience; Histone Gene Cluster 1 Member E, HIST1H1E, encodes Histone H1.4, is one of a family of epigenetic regulator genes, acts as a linker histone protein, and is responsible for higher order chromatin structure. HIST1H1E syndrome (also known as Rahman syndrome, OMIM #617537) is a recently described intellectual disability (ID) syndrome. Since the initial description of five unrelated individuals with three different heterozygous protein-truncating variants (PTVs) in the HIST1H1E gene in 2017, we have recruited 30 patients, all with HIST1H1E PTVs that result in the same shift in frame and that cluster to a 94-base pair region in the HIST1H1E carboxy terminal domain. The identification of 30 patients with HIST1H1E variants has allowed the clarification of the HIST1H1E syndrome phenotype. Major findings include an ID and a recognizable facial appearance. ID was reported in all patients and is most frequently of moderate severity. The facial gestalt consists of a high frontal hairline and full lower cheeks in early childhood and, in later childhood and adulthood, affected individuals have a strikingly high frontal hairline, frontal bossing, and deep-set eyes. Other associated clinical features include hypothyroidism, abnormal dentition, behavioral issues, cryptorchidism, skeletal anomalies, and cardiac anomalies. Brain magnetic resonance imaging (MRI) is frequently abnormal with a slender corpus callosum a frequent finding.
- Published
- 2019
29. Study of carrier frequency of Warsaw breakage syndrome in the Ashkenazi Jewish population and presentation of two cases
- Author
-
Martin Johansson, David A. Zeevi, Yoel Hirsch, Joseph Ekstein, Elaine H. Zackai, John Pappas, Beth Keena, and Rachel Rabin
- Subjects
Male ,0301 basic medicine ,Heterozygote ,congenital, hereditary, and neonatal diseases and abnormalities ,Microcephaly ,Adolescent ,RNA Splicing ,Population ,030105 genetics & heredity ,Biology ,Frameshift mutation ,Young Adult ,03 medical and health sciences ,DDX11 ,Intellectual disability ,Genetics ,medicine ,Humans ,Abnormalities, Multiple ,Genetic Testing ,Child ,education ,Gene ,Genetics (clinical) ,education.field_of_study ,Base Sequence ,Infant, Newborn ,Infant ,Syndrome ,medicine.disease ,Hypoplasia ,Phenotype ,030104 developmental biology ,Child, Preschool ,Jews ,Female ,Sensorineural hearing loss - Abstract
Warsaw breakage syndrome (WABS), caused by bi-allelic variants in the DDX11 gene, is a rare cohesinopathy characterized by pre- and postnatal growth retardation, microcephaly, intellectual disability, facial dysmorphia, and sensorineural hearing loss due to cochlear hypoplasia. The DDX11 gene codes for an iron-sulfur DNA helicase in the Superfamily 2 helicases and plays an important role in genomic stability and maintenance. Fourteen individuals with WABS have been previously reported in the medical literature. Affected individuals have been of various ethnic backgrounds with different pathogenic variants. We report two unrelated individuals of Ashkenazi Jewish descent affected with WABS, who are homozygous for the c.1763-1G>C variant in the DDX11 gene. Their phenotype is consistent with previously reported individuals. RNA studies showed that this variant causes an alternative splice acceptor site leading to a frameshift in the open reading frame. Carrier screening of the c.1763-1G>C variant in the Jewish population revealed a high carrier frequency of 1 in 68 in the Ashkenazi Jewish population. Due to the high carrier frequency and the low number of affected individuals, we hypothesize a high rate of miscarriage of homozygous fetuses and/or subfertility for carrier couples. If the carrier frequency is reproducible in additional Ashkenazi Jewish populations, we suggest including DDX11 to Ashkenazi Jewish carrier screening panels.
- Published
- 2019
30. Hyponatremic Seizures and Adrenal Hypoplasia Congenita in a Neonate with Congenital Diaphragmatic Hernia
- Author
-
Sheryl Purrier, Sourabh Verma, John Pappas, Pradeep V. Mally, Tara M. Randis, and Emily Breidbart
- Subjects
Pediatrics ,medicine.medical_specialty ,Hyperkalemia ,business.industry ,Fludrocortisone ,lcsh:RJ1-570 ,Congenital diaphragmatic hernia ,lcsh:Pediatrics ,Case Report ,030209 endocrinology & metabolism ,General Medicine ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,X-linked adrenal hypoplasia congenita ,medicine ,Adrenal insufficiency ,030212 general & internal medicine ,medicine.symptom ,business ,Hyponatremia ,Neonatal seizure ,Hydrocortisone ,medicine.drug - Abstract
Congenital diaphragmatic hernia (CDH) in neonates may occur as an isolated finding, in association with other anomalies, or as part of a genetic syndrome. We report the first case of an infant with CDH who presented with hyponatremic seizures due to adrenal hypoplasia congenita (AHC). The patient underwent repair of CDH defect. After an uncomplicated postoperative course while on discharge planning, he developed a seizure episode associated with severe hyponatremia and hyperkalemia. Extensive diagnostic workup revealed an NR0B1 gene variant confirming the diagnosis of X-linked AHC. The patient was eventually discharged home on hydrocortisone, fludrocortisone, and salt supplements. There are a few case reports of adrenal insufficiency in neonates with CDH, manifesting with symptoms before and immediately after reparative surgery. Clinical presentation of our patient was unique in manifesting as neonatal seizure secondary to severe hyponatremia after a stable postoperative phase. The patient’s electrolytes and hemodynamic status remained stable before, during, and after surgery for CDH. This case underlines the importance of taking detailed family history and continued vigilance for signs and symptoms of adrenal insufficiency in infants with repaired CDH by pediatricians and intensivists.
- Published
- 2019
31. Abstract 5423: Translational PK/PD/efficacy modeling and efficacious human dose prediction for a first-in-class MUC1-EGFR (M1231) bispecific antibody drug conjugate
- Author
-
Anup Zutshi, Berend Neuteboom, Seema Kumar, Willem Sloot, Christine Knuehl, Julia Dotterweich, Jianguo Ma, Christiane Amendt, Karthik Venkatakrishnan, Taeshin Park, John Pappas, and Kyoung-Ae Kim
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: M1231 is a first-in-class bispecific antibody drug conjugate (ADC) targeting mucin-1 (MUC1) and epidermal growth factor receptor (EGFR), that is conjugated with a novel hemiasterlin-related microtubule inhibitor payload. Following dose, M1231 binds to EGFR and co-binds to tumor-associated hypoglycosylated MUC1, internalizes into the tumor cell and traffics to lysosomes where the payload is enzymatically released to affect cell viability. Methods: A Multi-scale Systems Pharmacology model was developed to account for the ADC’s disposition and interactions with the underlying physiologic system, resulting in intracellular payload release in tumor cells to drive tumor growth inhibition (TGI). An in vitro model quantified ADC internalization and lysosomal trafficking in the MUC1 and EGFR-expressing cancer cell lines MDA-MB-468 and OVCAR-3. TGI was assessed in mice bearing MUC1-tumors from the squamous NSCLC patient-derived xenograft model, LUX003. Pharmacokinetics in cynomolgus monkeys was modeled using a Target Mediated Drug Disposition model and was allometrically scaled to humans. Data Summary: All preclinical modeling results were integrated and scaled to simulate plasma M1231 concentrations to predict the human dose and corresponding exposures. Tumor stasis was estimated to begin at a dose of 2.4 mg/kg every 3 weeks (Q3W) with a maximum tumor regression achieved at a dose of 4.3 mg/kg Q3W. Conclusions: The quantitative systems pharmacology model-based efficacious dose prediction range of 2.4 mg/kg to 4.3 mg/kg dosed Q3W informed the design of the ongoing M1231 first-in-human trial (NCT04695847). Citation Format: Anup Zutshi, Berend Neuteboom, Seema Kumar, Willem Sloot, Christine Knuehl, Julia Dotterweich, Jianguo Ma, Christiane Amendt, Karthik Venkatakrishnan, Taeshin Park, John Pappas, Kyoung-Ae Kim. Translational PK/PD/efficacy modeling and efficacious human dose prediction for a first-in-class MUC1-EGFR (M1231) bispecific antibody drug conjugate [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5423.
- Published
- 2022
32. PPP3CA truncating variants clustered in the regulatory domain cause early-onset refractory epilepsy
- Author
-
Wenmiao Zhu, Haley Streff, Saadet Mercimek-Andrews, John Pappas, Vann Chau, Fan Xia, Seema R. Lalani, Rachel Rabin, Christine M. Eng, Yaping Yang, Weimin Bi, Sugi Panneerselvam, Ddd Study, Lisa Emrick, Karen J. Low, Rui Xiao, Claudia Soler-Alfonso, Jill A. Rosenfeld, Julia Wang, Anne E. Anderson, and Hongzheng Dai
- Subjects
0301 basic medicine ,Male ,Drug Resistant Epilepsy ,Adolescent ,Protein subunit ,Phosphatase ,Gene Expression ,030105 genetics & heredity ,Biology ,Frameshift mutation ,03 medical and health sciences ,Epilepsy ,Mutant protein ,Genetics ,medicine ,Missense mutation ,Humans ,Child ,Genetics (clinical) ,Loss function ,Sequence Analysis, RNA ,Calcineurin ,medicine.disease ,030104 developmental biology ,Child, Preschool ,Mutation ,Female - Abstract
PPP3CA encodes the catalytic subunit of calcineurin, a calcium-calmodulin-regulated serine-threonine phosphatase. Loss-of-function (LoF) variants in the catalytic domain have been associated with epilepsy, while gain-of-function (GoF) variants in the auto-inhibitory domain cause multiple congenital abnormalities. We herein report five new patients with de novo PPP3CA variants. Interestingly, the two frameshift variants in this study and the six truncating variants reported previously are all located within a 26-amino acid region in the regulatory domain (RD). Patients with a truncating variant had more severe earlier onset seizures compared to patients with a LoF missense variant, while autism spectrum disorder was a more frequent feature in the latter. Expression studies of a truncating variant showed apparent RNA expression from the mutant allele, but no detectable mutant protein. Our data suggest that PPP3CA truncating variants clustered in the RD, causing more severe early-onset refractory epilepsy and representing a type of variants distinct from LoF or GoF missense variants.
- Published
- 2021
33. Author response for 'PPP3CA truncating variants clustered in the regulatory domain cause early-onset refractory epilepsy'
- Author
-
Jill A. Rosenfeld, Wenmiao Zhu, Julia Wang, Anne E. Anderson, Fan Xia, Vann Chau, Yaping Yang, Lisa Emrick, Rachel Rabin, Saadet Mercimek-Andrews, Christine M. Eng, Weimin Bi, Hongzheng Dai, Sugi Panneerselvam, Karen J. Low, Seema R. Lalani, Rui Xiao, Claudia Soler-Alfonso, Haley Streff, and John Pappas
- Subjects
business.industry ,Refractory epilepsy ,Medicine ,business ,Neuroscience ,Early onset ,Domain (software engineering) - Published
- 2021
34. Mobile Technology in a Clinical Setting.
- Author
-
Kara Holleran, John Pappas, Hong Lou, Pat Rubalcaba, Rita Lee, Steve Clay, Janice Cutone, Steve Flammini, Gilad J. Kuperman, and Blackford Middleton
- Published
- 2003
35. Designing Enterprise-wide Tools to Improve User Accountability for Protected Health Information: a Pilot Study.
- Author
-
Jonathan S. Wald, Joan Gallagher, Pat Rubalcaba, John Pappas, Debra A. Mikels, Karen G. Grant, Sally Millar, and Cynthia Spurr
- Published
- 2002
36. eP220: Expanding the phenotypic spectrum of COLEC10-related 3MC syndrome: A glimpse into COLEC10-related 3MC syndrome in the Ashkenazi Jewish population
- Author
-
Rachel Rabin, Yoel Hirsch, Wendy Chung, Josef Ekstein, Kevin Booth, and John Pappas
- Subjects
Genetics (clinical) - Published
- 2022
37. Germline AGO2 mutations impair RNA interference and human neurological development
- Author
-
Ee-Shien Tan, John Pappas, Tiffany Busa, Marjolein H. Willemsen, Erica H. Gerkes, Astrid Bruckmann, Linda Ramsdell, Davor Lessel, Chieko Chijiwa, Diana Mitter, Ghayda M. Mirzaa, Peter Ian Andrews, Daniela M. Zeitler, Claudia Schob, Pankaj Prasun, M. E. Suzanne Lewis, Rami Abou Jamra, Andriy Kazantsev, Aida Telegrafi, Steffen Syrbe, Chantal Missirian, Victoria Martens, Kimberly Foss, Margot R.F. Reijnders, Bianca Panis, Ilaria Mannucci, Bernarda Lozić, Fatemeh Hassani Nia, Rolph Pfundt, Tanja Kovacevic, Han G. Brunner, Christian Kubisch, Allyn McConkie-Rosell, Breana Cham, Gunter Meister, Jessika Johannsen, Veronika Graus, Henny H. Lemmink, Stefan Kindler, Jonas Denecke, Kirsty McWalter, Ivana Lessel, Zoya Ignatova, Hans-Jürgen Kreienkamp, Matthew Osmond, Thatjana Gardeitchik, Alexander P.A. Stegmann, Rachel Rabin, Alexander Bartholomäus, Jérémie Mortreux, Katharina Löhner, Tony Roscioli, Tjitske Kleefstra, Taila Hartley, Andreas Merkenschlager, Patrick Rump, Marie T. McDonald, Kym M. Boycott, Evelien Zonneveld-Huijssoon, David A. Dyment, Carey-Anne Evans, Margje Sinnema, Sabine Lüttgen, Joanna Lazier, Diana Le Duc, Kerith-Rae Dias, Ene-Choo Tan, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Département de génétique médicale [Hôpital de la Timone - APHM], Aix Marseille Université (AMU)-Assistance Publique - Hôpitaux de Marseille (APHM)- Hôpital de la Timone [CHU - APHM] (TIMONE)-Institut National de la Santé et de la Recherche Médicale (INSERM), ANR-17-CE16-0025,MicroRNAct,Identification de complexes microARN/mARN fonctionnels dans le cerveau antérieur de souris: de la neurogenèse au comportement et à la pathologie(2017), MUMC+: DA KG Polikliniek (9), and RS: FHML non-thematic output
- Subjects
0301 basic medicine ,INTELLECTUAL DISABILITY ,MICRORNAS ,[SDV]Life Sciences [q-bio] ,molecular-dynamics ,General Physics and Astronomy ,ARGONAUTE-2, ARGONAUTE-2 ,Hippocampus ,Nervous System ,Transcriptome ,Mice ,0302 clinical medicine ,RNA interference ,Cluster Analysis ,Phosphorylation ,RNA, Small Interfering ,lcsh:Science ,Child ,Regulation of gene expression ,Neurons ,Multidisciplinary ,Molecular medicine ,Neurodevelopmental disorders ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,3. Good health ,Cell biology ,Child, Preschool ,Argonaute Proteins ,RNA Interference ,STRUCTURAL BASIS ,Adolescent ,Science ,Biology ,Molecular Dynamics Simulation ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Protein Domains ,microRNA ,Gene silencing ,Animals ,Humans ,RNA-Induced Silencing Complex ,Gene Silencing ,RNA, Messenger ,Messenger RNA ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,CLEAVAGE ,HEK 293 cells ,RECOGNITION ,crystal-structure ,RNA ,General Chemistry ,Dendrites ,Fibroblasts ,GENE ,Rats ,030104 developmental biology ,Germ Cells ,HEK293 Cells ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Mutation ,lcsh:Q ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,030217 neurology & neurosurgery - Abstract
ARGONAUTE-2 and associated miRNAs form the RNA-induced silencing complex (RISC), which targets mRNAs for translational silencing and degradation as part of the RNA interference pathway. Despite the essential nature of this process for cellular function, there is little information on the role of RISC components in human development and organ function. We identify 13 heterozygous mutations in AGO2 in 21 patients affected by disturbances in neurological development. Each of the identified single amino acid mutations result in impaired shRNA-mediated silencing. We observe either impaired RISC formation or increased binding of AGO2 to mRNA targets as mutation specific functional consequences. The latter is supported by decreased phosphorylation of a C-terminal serine cluster involved in mRNA target release, increased formation of dendritic P-bodies in neurons and global transcriptome alterations in patient-derived primary fibroblasts. Our data emphasize the importance of gene expression regulation through the dynamic AGO2-RNA association for human neuronal development., AGO2 binds to miRNAs to repress expression of cognate target mRNAs. Here the authors report that heterozygous AGO2 mutations result in defects in neurological development and impair RNA interference.
- Published
- 2020
38. EIF3F-related neurodevelopmental disorder: delineating and expanding the phenotypic and molecular spectrum
- Author
-
Ulrike Hueffmeier, Cornelia Kraus, Miriam Reuter, Steffen Uebe, Mary-Alice Abbott, Syed Ahmed, Kristyn Rawson, Eileen Barr, Hong Li, ange-line bruel, Laurence Faivre, Frederic Tran Mau Them, Christina Botti, Susan Brooks, Kaitlyn Burns, Isum Ward, Marina Dutra-Clarke, Julian Martinez-Agosto, Hane Lee, Stanley Nelson, UCLA California Center for Rare Disease, Pia Zacher, Rami Abou Jamra, Chiara Klöckner, Julie MacGaughran, Juergen Kohlhase, Sarah Schuhmann, Ellen Moran, John Pappas, Annick Rothschild, Maria Guillen Sacoto, Lindsay Henderson, Timothy Palculict, Sureni Mullegama, Houda Elloumi, Adi Reich, Samantha Schrier Vergano, Erica Wahl, André Reis, and Christiane Zweier
- Published
- 2020
39. Transcriptome sequencing identifies a noncoding, deep intronic variant in CLCN7 causing autosomal recessive osteopetrosis
- Author
-
Ilana Moscatelli, Kim Henriksen, Odelia Chorin, Naomi Yachelevich, Gilad D. Evrony, John Pappas, and Khaled Mohamed
- Subjects
0301 basic medicine ,medicine.medical_specialty ,lcsh:QH426-470 ,medicine.medical_treatment ,RNA Splicing ,Osteoclasts ,Genes, Recessive ,Hematopoietic stem cell transplantation ,RNA‐sequencing ,030105 genetics & heredity ,Biology ,Clinical Reports ,Transcriptome ,03 medical and health sciences ,transcriptomics ,Osteoclast ,Chloride Channels ,Genetics ,medicine ,Humans ,Genetic Testing ,RNA-Seq ,Molecular Biology ,Genetics (clinical) ,Loss function ,Genetic testing ,Clinical Report ,medicine.diagnostic_test ,Osteopetrosis ,medicine.disease ,undiagnosed diseases ,Introns ,3. Good health ,lcsh:Genetics ,030104 developmental biology ,medicine.anatomical_structure ,Child, Preschool ,biology.protein ,Medical genetics ,Female ,CLCN7 - Abstract
Background Over half of children with rare genetic diseases remain undiagnosed despite maximal clinical evaluation and DNA‐based genetic testing. As part of an Undiagnosed Diseases Program applying transcriptome (RNA) sequencing to identify the causes of these unsolved cases, we studied a child with severe infantile osteopetrosis leading to cranial nerve palsies, bone deformities, and bone marrow failure, for whom whole‐genome sequencing was nondiagnostic. Methods We performed transcriptome (RNA) sequencing of whole blood followed by analysis of aberrant transcript isoforms and osteoclast functional studies. Results We identified a pathogenic deep intronic variant in CLCN7 creating an unexpected, frameshifting pseudoexon causing complete loss of function. Functional studies, including osteoclastogenesis and bone resorption assays, confirmed normal osteoclast differentiation but loss of osteoclast function. Conclusion This is the first report of a pathogenic deep intronic variant in CLCN7, and our approach provides a model for systematic identification of noncoding variants causing osteopetrosis—a disease for which molecular‐genetic diagnosis can be pivotal for potentially curative hematopoietic stem cell transplantation. Our work illustrates that cryptic splice variants may elude DNA‐only sequencing and supports broad first‐line use of transcriptome sequencing for children with undiagnosed diseases., Over half of children with rare genetic diseases remain undiagnosed despite maximal DNA‐based genetic testing. Using transcriptome (RNA) sequencing, we identified a deep intronic variant in CLCN7 as the cause of a child's previously undiagnosed severe osteopetrosis syndrome. This work illustrates that cryptic splice variants may elude DNA‐only sequencing and supports first‐line use of transcriptome sequencing for undiagnosed diseases.
- Published
- 2020
40. A software architecture to support a large-scale, multi-tier clinical information system.
- Author
-
Joel A. Yungton, Dean F. Sittig, P. Reilly, John Pappas, Steve Flammini, Henry C. Chueh, and Jonathan M. Teich
- Published
- 1998
41. Intellectual disability due to monoallelic variant in GATAD2B and mosaicism in unaffected parent
- Author
-
Juan Cabrera-Luque, Francisca Millan, Rachel Rabin, and John Pappas
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Genetic counseling ,Biology ,GATA Transcription Factors ,03 medical and health sciences ,Neurodevelopmental disorder ,Pregnancy ,Intellectual Disability ,Intellectual disability ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Alleles ,Genetic Association Studies ,Genetics (clinical) ,Exome sequencing ,Genetic testing ,medicine.diagnostic_test ,Mosaicism ,Facies ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Infant ,Electroencephalography ,medicine.disease ,Magnetic Resonance Imaging ,Hypotonia ,Repressor Proteins ,030104 developmental biology ,GATAD2B ,Medical genetics ,Female ,medicine.symptom - Abstract
The GATA zing finger domain-containing protein 2B (GATAD2B) gene encodes the p66beta protein, a subunit of the MeCP1-Mi2/ nucleosome remodeling and deacetylase complex, which is involved in transcription regulation and chromatin remodeling. Pathogenic variants in the GATAD2B gene have recently been associated with a recognizable neurodevelopmental syndrome, characterized by intellectual disability, limited speech, childhood hypotonia, and dysmorphic features. The majority of reported patients resulted from de novo loss of function (LOF) variants. We report a patient identified through whole exome sequencing analysis where a healthy mother was found to be low level mosaic for the pathogenic LOF variant found in her child, who is affected with GATAD2B-associated neurodevelopmental disorder (GAND). This variant was only found with the use of next generation sequencing technology in the mother and confirmed by digital droplet PCR. We summarize additional patients with GATAD2B LOF variants from a literature review and with our patient we contribute to delineate the phenotypic spectrum of GAND. We highlight the importance of detailed genetic testing, testing method, and counseling for cases of somatic mosaicism in an unaffected parent of children with GAND. This inheritance may be underreported and has a direct impact on reproductive planning and prenatal diagnosis.
- Published
- 2018
42. Whose Turn Is It, Anyway? Online Board Gaming and Libraries
- Author
-
John Pappas; Bebo and John Pappas; Bebo
- Abstract
This webinar explores platforms for online board gaming, how to moderate meetups, and facilitate discussions among participants.
- Published
- 2020
43. Whose Turn Is It, Anyway? Online Board Gaming and Libraries
- Author
-
John Pappas, Bebo, John Pappas, and Bebo
- Abstract
This webinar explores platforms for online board gaming, how to moderate meetups, and facilitate discussions among participants.
- Published
- 2020
44. Quantitative Systems Pharmacology Modeling of Acid Sphingomyelinase Deficiency and the Enzyme Replacement Therapy Olipudase Alfa Is an Innovative Tool for Linking Pathophysiology and Pharmacology
- Author
-
Catherine Ortemann-Renon, Zachary van Rijn, Chanchala D. Kaddi, John E. Tolsma, Mengdi Tao, Edward H. Schuchman, Ana Cristina Puga, Rachael Easton, Bradley Niesner, John Pappas, Paul Jasper, Karim Azer, Jeffrey S. Barrett, Sharon Tan, Rena Baek, and Jing Li
- Subjects
0301 basic medicine ,Hepatosplenomegaly ,Disease ,Bioinformatics ,Models, Biological ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Animals ,Humans ,Medicine ,Enzyme Replacement Therapy ,Pharmacology (medical) ,Mice, Knockout ,Niemann-Pick Diseases ,business.industry ,Research ,lcsh:RM1-950 ,Enzyme replacement therapy ,Recombinant Proteins ,Pathophysiology ,Sphingomyelin Phosphodiesterase ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,Modeling and Simulation ,Pharmacodynamics ,Calibration ,medicine.symptom ,Acid sphingomyelinase ,business ,030217 neurology & neurosurgery ,medicine.drug ,Systems pharmacology - Abstract
Acid sphingomyelinase deficiency (ASMD) is a rare lysosomal storage disorder with heterogeneous clinical manifestations, including hepatosplenomegaly and infiltrative pulmonary disease, and is associated with significant morbidity and mortality. Olipudase alfa (recombinant human acid sphingomyelinase) is an enzyme replacement therapy under development for the non-neurological manifestations of ASMD. We present a quantitative systems pharmacology (QSP) model supporting the clinical development of olipudase alfa. The model is multiscale and mechanistic, linking the enzymatic deficiency driving the disease to molecular-level, cellular-level, and organ-level effects. Model development was informed by natural history, and preclinical and clinical studies. By considering patient-specific pharmacokinetic (PK) profiles and indicators of disease severity, the model describes pharmacodynamic (PD) and clinical end points for individual patients. The ASMD QSP model provides a platform for quantitatively assessing systemic pharmacological effects in adult and pediatric patients, and explaining variability within and across these patient populations, thereby supporting the extrapolation of treatment response from adults to pediatrics.
- Published
- 2018
45. Genotype-phenotype correlations in individuals with pathogenicREREvariants
- Author
-
John A. Bernat, Elliott H. Sherr, Weimin Bi, Tugce B. Balci, Ronald J. Wapner, Jessica L. Giordano, Jill A. Rosenfeld, Donna M. Martin, Brieana Fregeau, Valerie K. Jordan, Annika M. Dries, Amanda Moccia, Colette DeFilippo, Jennefer N. Kohler, Yaping Yang, Vincent Cantagrel, Andrea M. Lewis, Willa Thorson, Xiaoyan Ge, Anshika Srivastava, Stephanie L. Bielas, Jonathan A. Bernstein, Melissa T. Carter, Marlène Rio, Nathalie Boddaert, John Pappas, Melissa D. Svoboda, Fernando Scaglia, and Daryl A. Scott
- Subjects
Male ,0301 basic medicine ,Adolescent ,Biology ,1p36 deletion syndrome ,Article ,genotype–phenotype correlations ,CHD7 ,Young Adult ,03 medical and health sciences ,CHARGE syndrome ,Fatal Outcome ,Neurodevelopmental disorder ,Gene duplication ,RERE ,Genetics ,medicine ,Humans ,Gene ,Genetic Association Studies ,Genetics (clinical) ,Point mutation ,Infant ,medicine.disease ,Phenotype ,030104 developmental biology ,Child, Preschool ,Mutation ,Female ,Carrier Proteins ,NEDBEH ,Haploinsufficiency - Abstract
Heterozygous variants in the arginine-glutamic acid dipeptide repeats gene (RERE) have been shown to cause neurodevelopmental disorder with or without anomalies of the brain, eye, or heart (NEDBEH). Here, we report nine individuals with NEDBEH who carry partial deletions or deleterious sequence variants in RERE. These variants were found to be de novo in all cases in which parental samples were available. An analysis of data from individuals with NEDBEH suggests that point mutations affecting the Atrophin-1 domain of RERE are associated with an increased risk of structural eye defects, congenital heart defects, renal anomalies, and sensorineural hearing loss when compared with loss-of-function variants that are likely to lead to haploinsufficiency. A high percentage of RERE pathogenic variants affect a histidine-rich region in the Atrophin-1 domain. We have also identified a recurrent two-amino-acid duplication in this region that is associated with the development of a CHARGE syndrome-like phenotype. We conclude that mutations affecting RERE result in a spectrum of clinical phenotypes. Genotype–phenotype correlations exist and can be used to guide medical decision making. Consideration should also be given to screening for RERE variants in individuals who fulfill diagnostic criteria for CHARGE syndrome but do not carry pathogenic variants in CHD7.
- Published
- 2018
46. Kufor-Rakeb Syndrome Due to a Novel ATP13A2 Mutation in 2 Chinese-American Brothers
- Author
-
Wendy Alcaraz, Natalie Hellmers, Evan Noch, Mary Lynn Chu, John Pappas, Ellen Moran, Claire Henchcliffe, and Harini Sarva
- Subjects
0301 basic medicine ,Genetics ,Parkinson's disease ,Movement disorders ,business.industry ,Case Reports ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Neurology ,Kufor Rakeb syndrome ,Mutation (genetic algorithm) ,medicine ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Chinese americans - Published
- 2017
47. Deletion of TBL1XR1: neurodevelopmental disorder with characteristic facial features
- Author
-
John Pappas and Rachel Rabin
- Subjects
Endocrinology ,Neurodevelopmental disorder ,business.industry ,Endocrinology, Diabetes and Metabolism ,Genetics ,Medicine ,business ,medicine.disease ,Molecular Biology ,Biochemistry ,Neuroscience - Published
- 2021
48. Recessive Mutations in AP1B1 Cause Ichthyosis, Deafness, and Photophobia
- Author
-
Claire E. Hamilton, John Pappas, Lihi Atzmony, Rachel Rabin, Shawn M. Ferguson, Lynn M. Boyden, Keith A. Choate, Richard P. Lifton, Julie S. Prendiville, Young H. Lim, Jing Zhou, Ronghua Hu, Yoel Hirsch, and Joseph Ekstien
- Subjects
0301 basic medicine ,Male ,Photophobia ,Adaptor Protein Complex 1 ,Genes, Recessive ,Biology ,Deafness ,Cell junction ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Report ,Genetics ,medicine ,Humans ,Endomembrane system ,Adaptor Protein Complex beta Subunits ,Hearing Loss ,Gene ,Genetics (clinical) ,Cell Proliferation ,Ichthyosis ,Signal transducing adaptor protein ,Cell Differentiation ,medicine.disease ,Phenotype ,Thrombocytopenia ,Cell biology ,Protein Subunits ,Protein Transport ,030104 developmental biology ,Failure to thrive ,Mutation ,Female ,medicine.symptom - Abstract
We describe unrelated individuals with ichthyosis, failure to thrive, thrombocytopenia, photophobia, and progressive hearing loss. Each have bi-allelic mutations in AP1B1, the gene encoding the β subunit of heterotetrameric adaptor protein 1 (AP-1) complexes, which mediate endomembrane polarization, sorting, and transport. In affected keratinocytes the AP-1 β subunit is lost, and the γ subunit is greatly reduced, demonstrating destabilization of the AP-1 complex. Affected cells and tissue contain an abundance of abnormal vesicles and show hyperproliferation, abnormal epidermal differentiation, and derangement of intercellular junction proteins. Transduction of affected cells with wild-type AP1B1 rescues the vesicular phenotype, conclusively establishing that loss of AP1B1 function causes this disorder.
- Published
- 2019
49. Comparing Characteristics of Patients Who Connect Their iPhones to an Electronic Health Records System Versus Patients Who Connect Without Personal Devices: Cohort Study
- Author
-
Sara Silacci, William J. Gordon, John Pappas, David W. Bates, Daniel Fuchs, and Adam B. Landman
- Subjects
Adult ,Male ,medicine.medical_specialty ,020205 medical informatics ,Adolescent ,Download ,Health Informatics ,02 engineering and technology ,Health records ,health information interoperability ,Cohort Studies ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Patient Portals ,information technology ,Health care ,0202 electrical engineering, electronic engineering, information engineering ,Medicine ,Electronic Health Records ,Humans ,030212 general & internal medicine ,Patient participation ,mobile health ,Male gender ,Aged ,Aged, 80 and over ,Original Paper ,business.industry ,Patient portal ,Middle Aged ,Quartile ,Family medicine ,Female ,patient participation ,business ,Cell Phone ,Cohort study - Abstract
Background While individual access to health records has traditionally been through paper and other physical media, there has been a recent push toward digitizing this process. Direct patient access to health data through application programming interfaces (APIs) is an important part of current United States policy initiatives, and Apple has created the product “Health Records on iPhone” to leverage APIs for this purpose. Objective The objective of this study was to examine the characteristics of patients at our institution who connected their personal iPhone devices to our electronic health records (EHRs) system through “Health Records on iPhone”, as compared to patients at our institution who used our patient portal but did not connect a personal device to our system. Methods We examined adult patients at our institution who had authorized an iPhone device to download their health data from the Partners HealthCare EHR via APIs through “Health Records on iPhone” from February 18, 2018 (the date this feature was enabled at our health system) until February 17, 2019. We compared these patients to adult patients who used our portal at least once during this period but did not authorize an iPhone device to download their data via APIs. Results Variables associated with an increased likelihood of using “Health Records on iPhone” included male gender (adjusted OR 3.36; 95% CI 3.11-3.62; P Conclusions Early results from the implementation of patient-facing APIs at a single institution suggest that there are opportunities for expanding these technologies to ensure all patients are aware of, and have access to, their health data on their personal devices. More work is needed on expanding these technologies to different patient populations.
- Published
- 2019
50. De Novo Mutations in 124 Cases of Sudden Unexplained Deaths in Childhood
- Author
-
Matthew Halvorsen, John Pappas, Peter T. Lin, David Goldstein, Xiaohan Wang, Rachel Rabin, Michael J. Ackerman, Orrin Devinsky, David J. Tester, Laura Crandall, and Richard W. Tsien
- Subjects
medicine.medical_specialty ,business.industry ,Public health ,Odds ratio ,Institutional review board ,medicine.disease ,Sudden death ,Sudden cardiac death ,Epilepsy ,Family medicine ,Cohort ,medicine ,Parental consent ,business - Abstract
Background: Sudden unexplained death in childhood (SUDC) is an understudied public health issue with multiple potential risk factors, including undiagnosed cardiac and seizure disorders, for which genetic risk factors can contribute. Methods: We studied whole exome sequence data from 124 consecutively ascertained trios (decedent child and unaffected parents) to test for excessive de novo mutations in cardiac, epilepsy and other genes. Findings: Among SUDC decedents, nonsynonymous mutations were enriched in genes associated with cardiac and seizure disorders relative to non-sudden death controls (odds ratio=9.76, p=2.15x10-4). Beyond these genes there was a trend for excess loss-of-function mutations in genes that are loss-of-function intolerant (odds ratio=2.34, p=0.08). In our cohort, two genes had recurrent missense mutations, RYR2 and CACNA1C. Both RYR2 mutations are known as pathogenic (p=1.7x10-7). Both CACNA1C mutations are within a short (104 nucleotide) exon (p=1.0x10-7). We found evidence for over-transmission of loss-of-function or pathogenic variants within cardiac and seizure disorder genes (11/14 transmitted, p=0.03). Interpretation: Eleven of 124 (8.9%) decedents carried likely pathogenic or pathogenic variants associated with sudden death via a lethal cardiac or seizure disorder. Of these, 7 were de novo in origin, 1 was a transmitted parental mosaic mutation, and 3 were transmitted from heterozygous parents. Funding Statement: This study was supported by funding from the SUDC Foundation and Finding A Cure for Epilepsy and Seizures (NYU). RWT was supported by NIH grants R01 DA040484 and R01 MH71739. MJA and DJT were supported by the Mayo Clinic Windland Smith Rice Comprehensive Sudden Cardiac Death Program. Declaration of Interests: MJA is a consultant for Audentes Therapeutics, Boston Scientific, Gilead Sciences, Invitae, Medtronic, Myokardia, and St. Jude Medical. MJA and Mayo Clinic have an equity/royalty relationship with AliveCor, Blue Ox Health, and StemoniX. However, none of these entities are related to study. DG reports he is cofounder and holds equity in Q State – Pairnomix and Praxis Therapeutics, and equity in Apostle Inc. He receives personal fees from AstraZeneca, outside the submitted work. RT reports his participation on the Scientific Advisory boards of Howard Hughes Medical Institute, the Institute of Neuroscience (Shanghai), and the Institute of Science and Technology (Vienna). RT also reports his participation in the Actelion Scientific Advisory Board (Basel) with no compensation during 3 year span. OD reports equity interest in Empatica and receive funding from NINDS on SUDEP research. MJA, OD and PL are members of the SUDC Foundation’s scientific advisory board. LGC is President of the SUDC Foundation. MJA, OD, PL and LGC do not participate in grant funding decisions or negotiations by the SUDC Foundation to NYU Langone Health. PL, DMH, RR, XW, JP have no declarations of interests. Ethics Approval Statement: Parental consent was obtained in this NYU Institutional Review Board approved study (# S14-01061).
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.