187 results on '"Bert B.A. de Vries"'
Search Results
2. GenIDA, a participatory patient registry for genetic forms of intellectual disability provides detailed caregiver-reported information on 237 individuals with Koolen-de Vries syndrome
- Author
-
Florent Colin, Pauline Burger, Timothée Mazzucotelli, Axelle Strehle, Joost Kummeling, Nicole Collot, Elyette Broly, Angela T. Morgan, Kenneth A. Myers, Agnès Bloch-Zupan, Charlotte W. Ockeloen, Bert B.A. de Vries, Tjitske Kleefstra, Pierre Parrend, David A. Koolen, and Jean-Louis Mandel
- Subjects
GenIDA ,Intellectual disability ,Koolen-de Vries syndrome ,Neurodevelopmental disorders ,Patient registry ,Genetics ,QH426-470 ,Medicine - Abstract
Purpose: GenIDA is an international patient registry for individuals diagnosed with intellectual disability, autism spectrum disorder, and/or epilepsy, which is based on an online questionnaire that is completed by parent caregivers. In this study, the GenIDA data on Koolen-de Vries syndrome (KdVS) was analyzed illustrating the value of GenIDA and patient/caregiver participation in rare genetic neurodevelopmental disorders (NDDs). Methods: Recruitment was done on the GenIDA website from November 2016 to February 2022. Clinical information on individuals with KdVS was extracted for in-depth analysis and for comparison with the GenIDA data of individuals diagnosed with other NDDs. Results: A total of 1417 patients/caregivers across 35 genetic conditions answered to the GenIDA questionnaire, including caregivers of 237 individuals with KdVS. GenIDA findings on KdVS were consistent with the existing literature, and there were no significant differences between individuals with a 17q21.31 microdeletion and those with a pathogenic variant in the KANSL1 gene. GenIDA provided detailed clinical information including features that are over-represented in KdVS compared with other NDDs (eg, laryngomalacia). Modeling of the natural history showed a positive development of speech and language over time and relatively good reading ability in KdVS. Valproate and oxcarbazepine were reported as effective antiepileptic drugs, and responses to open-ended questions indicated that childhood recurrent pneumonia and asthma are clinically relevant comorbidities that were not described in KdVS before. Conclusion: GenIDA is a powerful registry to collect and harness valuable data on rare NDDs. The study shows that caregiver-driven data collection is effective in terms of global recruitment and centralization of clinical data.
- Published
- 2023
- Full Text
- View/download PDF
3. TMEM218 dysfunction causes ciliopathies, including Joubert and Meckel syndromes
- Author
-
Julie C. Van De Weghe, Jessica L. Giordano, Inge B. Mathijssen, Majid Mojarrad, Dorien Lugtenberg, Caitlin V. Miller, Jennifer C. Dempsey, Mahsa Sadat Asl Mohajeri, Elizabeth van Leeuwen, Eva Pajkrt, Caroline C.W. Klaver, Henry Houlden, Atieh Eslahi, Aoife M. Waters, Michael J. Bamshad, Deborah A. Nickerson, Vimla S. Aggarwal, Bert B.A. de Vries, Reza Maroofian, and Dan Doherty
- Subjects
TMEM218 ,Joubert syndrome ,Meckel syndrome ,ciliopathy ,cilia ,primary cilia ,Genetics ,QH426-470 - Abstract
Summary: The Joubert-Meckel syndrome spectrum is a continuum of recessive ciliopathy conditions caused by primary cilium dysfunction. The primary cilium is a microtubule-based, antenna-like organelle that projects from the surface of most human cell types, allowing them to respond to extracellular signals. The cilium is partitioned from the cell body by the transition zone, a known hotspot for ciliopathy-related proteins. Despite years of Joubert syndrome (JBTS) gene discovery, the genetic cause cannot be identified in up to 30% of individuals with JBTS, depending on the cohort, sequencing method, and criteria for pathogenic variants. Using exome and targeted sequencing of 655 families with JBTS, we identified three individuals from two families harboring biallelic, rare, predicted-deleterious missense TMEM218 variants. Via MatchMaker Exchange, we identified biallelic TMEM218 variants in four additional families with ciliopathy phenotypes. Of note, four of the six families carry missense variants affecting the same highly conserved amino acid position 115. Clinical features included the molar tooth sign (N = 2), occipital encephalocele (N = 5, all fetuses), retinal dystrophy (N = 4, all living individuals), polycystic kidneys (N = 2), and polydactyly (N = 2), without liver involvement. Combined with existing functional data linking TMEM218 to ciliary transition zone function, our human genetic data make a strong case for TMEM218 dysfunction as a cause of ciliopathy phenotypes including JBTS with retinal dystrophy and Meckel syndrome. Identifying all genetic causes of the Joubert-Meckel spectrum enables diagnostic testing, prognostic and recurrence risk counseling, and medical monitoring, as well as work to delineate the underlying biological mechanisms and identify targets for future therapies.
- Published
- 2021
- Full Text
- View/download PDF
4. Loss-of-function variants in SRRM2 cause a neurodevelopmental disorder
- Author
-
Silvestre Cuinat, Mathilde Nizon, Bertrand Isidor, Alexander Stegmann, Richard H. van Jaarsveld, Koen L. van Gassen, Jasper J. van der Smagt, Catharina M.L. Volker-Touw, Sjoerd J.B. Holwerda, Paulien A. Terhal, Sarah Schuhmann, Georgia Vasileiou, Mohamed Khalifa, Alaa A. Nugud, Hemad Yasaei, Lilian Bomme Ousager, Charlotte Brasch-Andersen, Wallid Deb, Thomas Besnard, Marleen E.H. Simon, Karin Huijsdens-van Amsterdam, Nienke E. Verbeek, Dena Matalon, Natalie Dykzeul, Shana White, Elizabeth Spiteri, Koen Devriendt, Anneleen Boogaerts, Marjolein Willemsen, Han G. Brunner, Margje Sinnema, Bert B.A. De Vries, Erica H. Gerkes, Rolph Pfundt, Kosuke Izumi, Ian D. Krantz, Zhou L. Xu, Jill R. Murrell, Irene Valenzuela, Ivon Cusco, Eulàlia Rovira-Moreno, Yaping Yang, Varoona Bizaoui, Olivier Patat, Laurence Faivre, Frederic Tran-Mau-Them, Antonio Vitobello, Anne-Sophie Denommé-Pichon, Christophe Philippe, Stéphane Bezieau, Benjamin Cogné, MUMC+: DA KG Lab Specialisten (9), MUMC+: DA KG Lab Centraal Lab (9), MUMC+: DA KG Polikliniek (9), MUMC+: DA Klinische Genetica (5), Klinische Genetica, and RS: GROW - R4 - Reproductive and Perinatal Medicine
- Subjects
Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Developmental Disabilities ,Neurodevelopment ,Intellectual disability ,RNA-Binding Proteins ,SRRM2 ,Phenotype ,Spliceosome ,Neurodevelopmental Disorders ,Humans ,Muscle Hypotonia ,Molecular genetics ,Child ,Genetics (clinical) - Abstract
Contains fulltext : 282702.pdf (Publisher’s version ) (Open Access) PURPOSE: SRRM2 encodes the SRm300 protein, a splicing factor of the SR-related protein family characterized by its serine- and arginine-enriched domains. It promotes interactions between messenger RNA and the spliceosome catalytic machinery. This gene, predicted to be highly intolerant to loss of function (LoF) and very conserved through evolution, has not been previously reported in constitutive human disease. METHODS: Among the 1000 probands studied with developmental delay and intellectual disability in our database, we found 2 patients with de novo LoF variants in SRRM2. Additional families were identified through GeneMatcher. RESULTS: Here, we report on 22 patients with LoF variants in SRRM2 and provide a description of the phenotype. Molecular analysis identified 12 frameshift variants, 8 nonsense variants, and 2 microdeletions of 66 kb and 270 kb. The patients presented with a mild developmental delay, predominant speech delay, autistic or attention-deficit/hyperactivity disorder features, overfriendliness, generalized hypotonia, overweight, and dysmorphic facial features. Intellectual disability was variable and mild when present. CONCLUSION: We established SRRM2 as a gene responsible for a rare neurodevelopmental disease.
- Published
- 2022
5. Inherited variants in CHD3 show variable expressivity in Snijders Blok-Campeau syndrome
- Author
-
Jet van der Spek, Joery den Hoed, Lot Snijders Blok, Alexander J.M. Dingemans, Dick Schijven, Christoffer Nellaker, Hanka Venselaar, Galuh D.N. Astuti, Tahsin Stefan Barakat, E. Martina Bebin, Stefanie Beck-Wödl, Gea Beunders, Natasha J. Brown, Theresa Brunet, Han G. Brunner, Philippe M. Campeau, Goran Čuturilo, Christian Gilissen, Tobias B. Haack, Irina Hüning, Ralf A. Husain, Benjamin Kamien, Sze Chern Lim, Luca Lovrecic, Janine Magg, Ales Maver, Valancy Miranda, Danielle C. Monteil, Charlotte W. Ockeloen, Lynn S. Pais, Vasilica Plaiasu, Laura Raiti, Christopher Richmond, Angelika Rieß, Eva M.C. Schwaibold, Marleen E.H. Simon, Stephanie Spranger, Tiong Yang Tan, Michelle L. Thompson, Bert B.A. de Vries, Ella J. Wilkins, Marjolein H. Willemsen, Clyde Francks, Lisenka E.L.M. Vissers, Simon E. Fisher, Tjitske Kleefstra, Clinical Genetics, MUMC+: DA Klinische Genetica (5), Klinische Genetica, and RS: GROW - R4 - Reproductive and Perinatal Medicine
- Subjects
Neuroinformatics ,Heterozygote ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,DNA Helicases ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Syndrome ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,CHD3 ,Sensory disorders Donders Center for Medical Neuroscience [Radboudumc 12] ,Reduced penetrance ,Phenotype ,All institutes and research themes of the Radboud University Medical Center ,RARE ,SDG 3 - Good Health and Well-being ,Neurodevelopmental disorder ,Neurodevelopmental Disorders ,Variable expressivity ,Humans ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,Genetics (clinical) ,Mi-2 Nucleosome Remodeling and Deacetylase Complex ,Inherited variants - Abstract
PURPOSE: Common diagnostic next-generation sequencing strategies are not optimized to identify inherited variants in genes associated with dominant neurodevelopmental disorders as causal when the transmitting parent is clinically unaffected, leaving a significant number of cases with neurodevelopmental disorders undiagnosed.METHODS: We characterized 21 families with inherited heterozygous missense or protein-truncating variants in CHD3, a gene in which de novo variants cause Snijders Blok-Campeau syndrome.RESULTS: Computational facial and Human Phenotype Ontology-based comparisons showed that the phenotype of probands with inherited CHD3 variants overlaps with the phenotype previously associated with de novo CHD3 variants, whereas heterozygote parents are mildly or not affected, suggesting variable expressivity. In addition, similarly reduced expression levels of CHD3 protein in cells of an affected proband and of healthy family members with a CHD3 protein-truncating variant suggested that compensation of expression from the wild-type allele is unlikely to be an underlying mechanism. Notably, most inherited CHD3 variants were maternally transmitted.CONCLUSION: Our results point to a significant role of inherited variation in Snijders Blok-Campeau syndrome, a finding that is critical for correct variant interpretation and genetic counseling and warrants further investigation toward understanding the broader contributions of such variation to the landscape of human disease.
- Published
- 2022
6. TAB2 deletions and variants cause a highly recognisable syndrome with mitral valve disease, cardiomyopathy, short stature and hypermobility
- Author
-
Aafke Engwerda, Maarten P. van den Berg, Wilhelmina S. Kerstjens-Frederikse, Yvonne J. Vos, Bert B.A. de Vries, Tuula Rinne, Marc T R Roofthooft, Paulien A Terhal, Patrick Deelen, Conny M. A. van Ravenswaaij-Arts, Barbara Frentz, Katharina Löhner, Trijnie Dijkhuizen, Erika Leenders, Cardiovascular Centre (CVC), and Clinical Cognitive Neuropsychiatry Research Program (CCNP)
- Subjects
Joint Instability ,CONGENITAL HEART-DEFECTS ,Pathology ,medicine.medical_specialty ,TAK1 ,Heart Valve Diseases ,Cardiomyopathy ,GROWTH FAILURE ,Dwarfism ,Disease ,Short stature ,Article ,Mitral valve ,Genetics ,medicine ,Humans ,MICRODELETION ,DYSPLASIA ,Genetics (clinical) ,Exome sequencing ,Adaptor Proteins, Signal Transducing ,business.industry ,Syndrome ,medicine.disease ,Phenotype ,DELINEATION ,medicine.anatomical_structure ,Mitral Valve ,Noonan syndrome ,Chromosomes, Human, Pair 6 ,medicine.symptom ,Cardiomyopathies ,Haploinsufficiency ,business ,Gene Deletion - Abstract
Deletions that include the gene TAB2 and TAB2 loss-of-function variants have previously been associated with congenital heart defects and cardiomyopathy. However, other features, including short stature, facial dysmorphisms, connective tissue abnormalities and a variable degree of developmental delay, have only been mentioned occasionally in literature and thus far not linked to TAB2. In a large-scale, social media-based chromosome 6 study, we observed a shared phenotype in patients with a 6q25.1 deletion that includes TAB2. To confirm if this phenotype is caused by haploinsufficiency of TAB2 and to delineate a TAB2-related phenotype, we subsequently sequenced TAB2 in patients with matching phenotypes and recruited patients with pathogenic TAB2 variants detected by exome sequencing. This identified 11 patients with a deletion containing TAB2 (size 1.68-14.31 Mb) and 14 patients from six families with novel truncating TAB2 variants. Twenty (80%) patients had cardiac disease, often mitral valve defects and/or cardiomyopathy, 18 (72%) had short stature and 18 (72%) had hypermobility. Twenty patients (80%) had facial features suggestive for Noonan syndrome. No substantial phenotypic differences were noted between patients with deletions and those with intragenic variants. We then compared our patients to 45 patients from the literature. All literature patients had cardiac diseases, but syndromic features were reported infrequently. Our study shows that the phenotype in 6q25.1 deletions is caused by haploinsufficiency of TAB2 and that TAB2 is associated not just with cardiac disease, but also with a distinct phenotype, with features overlapping with Noonan syndrome. We propose the name "TAB2-related syndrome".
- Published
- 2021
7. Delineating the molecular and phenotypic spectrum of the SETD1B-related syndrome
- Author
-
Zou Pan, Marielle E. van Gijn, Marjolein H. Willemsen, Mariet W. Elting, Susanne Koning, Daniel C. Koboldt, Rebecca Baud, Renzo Guerrini, Ghayda M. Mirzaa, Laurence E. Walsh, Kim L. McBride, Jenny Thies, Andrew E. Timms, Shaoping Huang, Gretchen E. Rosso, Joshua Scheck, Haley McConkey, Matthew A. Deardorff, Peter D. Turnpenny, Suzanne M. Leal, Sanjay M. Sisodiya, Lin Yang, Melissa Lees, Cacha M.P.C.D. Peeters-Scholte, Henry Houlden, Marielle Alders, J. Austin Hamm, Karla A. Peña-Guerra, Richard E. Person, Leena Lauronen, Hannah K. Robinson, Theresa Mihalic Mosher, Alexandra Garza-Flores, Victoria Harrison, Tuomo Määttä, Daniela Q.C.M. Barge-Schaapveld, James R. Lupski, Houda Zghal Elloumi, Francisco J. Guzmán-Vega, Tamison Jewett, Siddharth Banka, Barbara W. van Paassen, J. Lawrence Merritt, Angela Sun, Yana Lara-Taranchenko, Irma Järvelä, Ivan K. Chinn, Claudia A. L. Ruivenkamp, Nicholas M. Allen, Xiaodong Wang, Amy Crunk, Selina H. Banu, Maura R.Z. Ruzhnikov, Jeffery McGlothlin, Mashaya Zaman, Adam Jackson, Stefan T. Arold, Bert B.A. de Vries, Jing Peng, Lauren Schenck, Isabelle Schrauwen, Marjon van Slegtenhorst, Luis Alberto Pedroza, Bekim Sadikovic, Annalisa Vetro, Reshmi Ramakrishnan, Kristin G. Monaghan, Kelly J. Cardona-Londoño, Catherine Quindipan, Kristina Lanko, Rolph Pfundt, Caroline M. Kehoe, Martino Montomoli, Christian Gilissen, Hamid Galehdari, Yolande van Bever, Jennifer Keller-Ramey, Sadegheh Haghshenas, Neda Mazaheri, Stephanie Efthymiou, Reza Maroofian, Lewis Pang, Fleur Vansenne, Abeltje M. Polstra, Kara C. Klemp, Marjolein J.A. Weerts, Xi Lin, Julia Baptista, Tahsin Stefan Barakat, Anneke Kievit, Adi Reich, Stephen R. Braddock, Shehla Mohammed, Abbey M. Putnam, Jennifer Kerkhof, Matthew Pastore, Sally Ann Lynch, Graduate School, ANS - Neuroinfection & -inflammation, Human Genetics, ACS - Pulmonary hypertension & thrombosis, ARD - Amsterdam Reproduction and Development, MUMC+: DA KG Polikliniek (9), RS: FHML non-thematic output, Human genetics, VU University medical center, Amsterdam Reproduction & Development (AR&D), Clinical Genetics, Irma Järvelä / Principal Investigator, Medicum, Department of Medical and Clinical Genetics, HUS Medical Imaging Center, Clinicum, BioMag Laboratory, HUS Children and Adolescents, and Kliinisen neurofysiologian yksikkö
- Subjects
Male ,INTELLECTUAL DISABILITY ,GENES ,Language delay ,VARIANTS ,Biology ,Bioinformatics ,3124 Neurology and psychiatry ,Article ,12Q24.31 ,SETD1B ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Neurodevelopmental disorder ,Seizures ,Intellectual disability ,medicine ,Humans ,MICRODELETION ,Global developmental delay ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,3112 Neurosciences ,RECOGNITION ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Penetrance ,Human genetics ,Phenotype ,Neurodevelopmental Disorders ,MOTIF ,Autism ,METHYLTRANSFERASE ,3111 Biomedicine ,030217 neurology & neurosurgery - Abstract
Contains fulltext : 243955.pdf (Publisher’s version ) (Open Access) PURPOSE: Pathogenic variants in SETD1B have been associated with a syndromic neurodevelopmental disorder including intellectual disability, language delay, and seizures. To date, clinical features have been described for 11 patients with (likely) pathogenic SETD1B sequence variants. This study aims to further delineate the spectrum of the SETD1B-related syndrome based on characterizing an expanded patient cohort. METHODS: We perform an in-depth clinical characterization of a cohort of 36 unpublished individuals with SETD1B sequence variants, describing their molecular and phenotypic spectrum. Selected variants were functionally tested using in vitro and genome-wide methylation assays. RESULTS: Our data present evidence for a loss-of-function mechanism of SETD1B variants, resulting in a core clinical phenotype of global developmental delay, language delay including regression, intellectual disability, autism and other behavioral issues, and variable epilepsy phenotypes. Developmental delay appeared to precede seizure onset, suggesting SETD1B dysfunction impacts physiological neurodevelopment even in the absence of epileptic activity. Males are significantly overrepresented and more severely affected, and we speculate that sex-linked traits could affect susceptibility to penetrance and the clinical spectrum of SETD1B variants. CONCLUSION: Insights from this extensive cohort will facilitate the counseling regarding the molecular and phenotypic landscape of newly diagnosed patients with the SETD1B-related syndrome.
- Published
- 2021
8. Loss-of-function and missense variants in NSD2 cause decreased methylation activity and are associated with a distinct developmental phenotype
- Author
-
Newell Belnap, Bert B.A. de Vries, Austin Larson, Rolph Pfundt, Marijke R. Wevers, Valérie Benoit, Markus Zweier, Pascal Joset, Anita Rauch, Angela Bahr, Jeroen Mourmans, Patricia G Wheeler, Or Gozani, Marisa V. Andrews, Monica H. Wojcik, Didier Lacombe, Sarah Grotto, Marwan Shinawi, Lot Snijders Blok, Conny M. A. van Ravenswaaij-Arts, Keri Ramsey, Deepanwita Sengupta, Mariarosaria Lang-Muritano, Isabelle Maystadt, Katharina Steindl, Paolo Zanoni, Antonio Vitobello, Geoffroy Delplancq, Katrin Õunap, Tania Attié-Bitach, Heinrich Sticht, Giulia Petrilli, Laurence Faivre, Vassilis Tsatsaris, and Clinical Cognitive Neuropsychiatry Research Program (CCNP)
- Subjects
0301 basic medicine ,In silico ,Biology ,Article ,REGION ,03 medical and health sciences ,ROGERS-DANKS-SYNDROME ,0302 clinical medicine ,Missense mutation ,HISTONE H3 ,Gene ,Genetics (clinical) ,Loss function ,Genetics ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,DELETION ,DEFECTS ,Methylation ,Phenotype ,LYSINE 36 ,030104 developmental biology ,Molecular mechanism ,WOLF-HIRSCHHORN-SYNDROME ,030217 neurology & neurosurgery ,Function (biology) ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] - Abstract
Purpose Despite a few recent reports of patients harboring truncating variants in NSD2, a gene considered critical for the Wolf–Hirschhorn syndrome (WHS) phenotype, the clinical spectrum associated with NSD2 pathogenic variants remains poorly understood. Methods We collected a comprehensive series of 18 unpublished patients carrying heterozygous missense, elongating, or truncating NSD2 variants; compared their clinical data to the typical WHS phenotype after pooling them with ten previously described patients; and assessed the underlying molecular mechanism by structural modeling and measuring methylation activity in vitro. Results The core NSD2-associated phenotype includes mostly mild developmental delay, prenatal-onset growth retardation, low body mass index, and characteristic facial features distinct from WHS. Patients carrying missense variants were significantly taller and had more frequent behavioral/psychological issues compared with those harboring truncating variants. Structural in silico modeling suggested interference with NSD2’s folding and function for all missense variants in known structures. In vitro testing showed reduced methylation activity and failure to reconstitute H3K36me2 in NSD2 knockout cells for most missense variants. Conclusion NSD2 loss-of-function variants lead to a distinct, rather mild phenotype partially overlapping with WHS. To avoid confusion for patients, NSD2 deficiency may be named Rauch–Steindl syndrome after the delineators of this phenotype., Genetics in Medicine, 23 (8)
- Published
- 2021
9. Human disease genes website series: An international, open and dynamic library for up‐to‐date clinical information
- Author
-
Jeroen Ewals, Kim M G Truijen, Sandra Jansen, Jozef Gecz, Diante E Stremmelaar, David A. Koolen, Bert B.A. de Vries, Alexander J. M. Dingemans, Michel Verbruggen, Lisenka E.L.M. Vissers, Noraly Jonis, Han G. Brunner, Evan E. Eichler, Maria J. Nabais Sá, Angela van Remortele, Sam van de Ven, and Human genetics
- Subjects
phenotype ,Computer science ,HDG ,World Wide Web ,Human disease ,Resource (project management) ,HDG website series ,Clinical information ,Genetics ,medicine ,Humans ,Gene ,Genetics (clinical) ,Gene Library ,Internet ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Genome, Human ,Genetic Diseases, Inborn ,online resource ,Original Articles ,medicine.disease ,Digital library ,Identification (information) ,Autism ,Original Article ,clinical data ,Human genome ,HPO - Abstract
Since the introduction of next‐generation sequencing, an increasing number of disorders have been discovered to have genetic etiology. To address diverse clinical questions and coordinate research activities that arise with the identification of these rare disorders, we developed the Human Disease Genes website series (HDG website series): an international digital library that records detailed information on the clinical phenotype of novel genetic variants in the human genome (https://humandiseasegenes.info/). Each gene website is moderated by a dedicated team of clinicians and researchers, focused on specific genes, and provides up‐to‐date—including unpublished—clinical information. The HDG website series is expanding rapidly with 424 genes currently adopted by 325 moderators from across the globe. On average, a gene website has detailed phenotypic information of 14.4 patients. There are multiple examples of added value, one being the ARID1B gene website, which was recently utilized in research to collect clinical information of 81 new patients. Additionally, several gene websites have more data available than currently published in the literature. In conclusion, the HDG website series provides an easily accessible, open and up‐to‐date clinical data resource for patients with pathogenic variants of individual genes. This is a valuable resource not only for clinicians dealing with rare genetic disorders such as developmental delay and autism, but other professionals working in diagnostics and basic research. Since the HDG website series is a dynamic platform, its data also include the phenotype of yet unpublished patients curated by professionals providing higher quality clinical detail to improve management of these rare disorders.
- Published
- 2021
10. De novo variants in ATP2B1 lead to neurodevelopmental delay
- Author
-
Meer Jacob Rahimi, Nicole Urban, Meret Wegler, Heinrich Sticht, Michael Schaefer, Bernt Popp, Frank Gaunitz, Manuela Morleo, Vincenzo Nigro, Silvia Maitz, Grazia M.S. Mancini, Claudia Ruivenkamp, Eun-Kyung Suk, Tobias Bartolomaeus, Andreas Merkenschlager, Daniel Koboldt, Dennis Bartholomew, Alexander P.A. Stegmann, Margje Sinnema, Irma Duynisveld, Ramona Salvarinova, Simone Race, Bert B.A. de Vries, Aurélien Trimouille, Sophie Naudion, Daphna Marom, Uri Hamiel, Noa Henig, Florence Demurger, Nils Rahner, Enrika Bartels, J. Austin Hamm, Abbey M. Putnam, Richard Person, Rami Abou Jamra, Henry Oppermann, Rahimi, Meer Jacob, Urban, Nicole, Wegler, Meret, Sticht, Heinrich, Schaefer, Michael, Popp, Bernt, Gaunitz, Frank, Morleo, Manuela, Nigro, Vincenzo, Maitz, Silvia, Mancini, Grazia M S, Ruivenkamp, Claudia, Suk, Eun-Kyung, Bartolomaeus, Tobia, Merkenschlager, Andrea, Koboldt, Daniel, Bartholomew, Denni, Stegmann, Alexander P A, Sinnema, Margje, Duynisveld, Irma, Salvarinova, Ramona, Race, Simone, de Vries, Bert B A, Trimouille, Aurélien, Naudion, Sophie, Marom, Daphna, Hamiel, Uri, Henig, Noa, Demurger, Florence, Rahner, Nil, Bartels, Enrika, Hamm, J Austin, Putnam, Abbey M, Person, Richard, Abou Jamra, Rami, Oppermann, Henry, Clinical Genetics, MUMC+: DA KG Lab Specialisten (9), MUMC+: DA KG Lab Centraal Lab (9), MUMC+: DA KG Polikliniek (9), and RS: FHML non-thematic output
- Subjects
EXPRESSION ,HOMEOSTASIS ,de novo ,seizure ,HOUSEKEEPING FUNCTION ,calcium homeostasi ,Mutation, Missense ,PLASMA-MEMBRANE CA2+-ATPASE ,ISOFORMS ,ATP2B1 ,Nervous System Malformations ,development delay ,abnormal behavior ,Plasma Membrane Calcium-Transporting ATPases ,CA2+ ,Report ,BINDING ,Genetics ,Humans ,MUTATION ,Genetics (clinical) ,neurodevelopmental disorder ,HEK293 Cells ,Phenotype ,Neurodevelopmental Disorders ,intellectual disability - Abstract
Calcium (Ca2+) is a universal second messenger involved in synaptogenesis and cell survival; consequently, its regulation is important for neurons. ATPase plasma membrane Ca2+ transporting 1 (ATP2B1) belongs to the family of ATP-driven calmodulin-dependent Ca2+ pumps that participate in the regulation of intracellular free Ca2+. Here, we clinically describe a cohort of 12 unrelated individuals with variants in ATP2B1 and an overlapping phenotype of mild to moderate global development delay. Additional common symptoms include autism, seizures, and distal limb abnormalities. Nine probands harbor missense variants, seven of which were in specific functional domains, and three individuals have nonsense variants. 3D structural protein modeling suggested that the variants have a destabilizing effect on the protein. We performed Ca2+ imaging after introducing all nine missense variants in transfected HEK293 cells and showed that all variants lead to a significant decrease in Ca2+ export capacity compared with the wild-type construct, thus proving their pathogenicity. Furthermore, we observed for the same variant set an incorrect intracellular localization of ATP2B1. The genetic findings and the overlapping phenotype of the probands as well as the functional analyses imply that de novo variants in ATP2B1 lead to a monogenic form of neurodevelopmental disorder.
- Published
- 2022
11. Imbalanced autophagy causes synaptic deficits in a human model for neurodevelopmental disorders
- Author
-
Marina P Hommersom, Chantal Schoenmaker, Astrid R. Oudakker, Katrin Linda, Lynn Devilee, Elly Lewerissa, Nael Nadif Kasri, Anouk H.A. Verboven, Bert B.A. de Vries, Michele Gabriele, Giuseppe Testa, David A. Koolen, Hans van Bokhoven, Monica Frega, Teun M. Klein Gunnewiek, Edda Ulferts, Dirk Schubert, Clinical Neurophysiology, and TechMed Centre
- Subjects
Autophagosome ,iPSCs ,Chromatin remodeling ,Epigenesis, Genetic ,Superoxide Dismutase-1 ,Sequestosome 1 ,Koolen-de Vries syndrome ,Intellectual Disability ,Lysosome ,medicine ,Autophagy ,neuronal development ,Humans ,Abnormalities, Multiple ,education ,synaptic function ,Molecular Biology ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Sirolimus ,chemistry.chemical_classification ,reactive oxygen species ,Reactive oxygen species ,education.field_of_study ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,biology ,Lysine ,TOR Serine-Threonine Kinases ,MTOR ,Autophagosomes ,Cell Biology ,Cell biology ,medicine.anatomical_structure ,chemistry ,biology.protein ,Chromosome Deletion ,Lysosomes ,Chromosomes, Human, Pair 17 - Abstract
Contains fulltext : 248864.pdf (Publisher’s version ) (Open Access) Macroautophagy (hereafter referred to as autophagy) is a finely tuned process of programmed degradation and recycling of proteins and cellular components, which is crucial in neuronal function and synaptic integrity. Mounting evidence implicates chromatin remodeling in fine-tuning autophagy pathways. However, this epigenetic regulation is poorly understood in neurons. Here, we investigate the role in autophagy of KANSL1, a member of the nonspecific lethal complex, which acetylates histone H4 on lysine 16 (H4K16ac) to facilitate transcriptional activation. Loss-of-function of KANSL1 is strongly associated with the neurodevelopmental disorder Koolen-de Vries Syndrome (KdVS). Starting from KANSL1-deficient human induced-pluripotent stem cells, both from KdVS patients and genome-edited lines, we identified SOD1 (superoxide dismutase 1), an antioxidant enzyme, to be significantly decreased, leading to a subsequent increase in oxidative stress and autophagosome accumulation. In KANSL1-deficient neurons, autophagosome accumulation at excitatory synapses resulted in reduced synaptic density, reduced GRIA/AMPA receptor-mediated transmission and impaired neuronal network activity. Furthermore, we found that increased oxidative stress-mediated autophagosome accumulation leads to increased MTOR activation and decreased lysosome function, further preventing the clearing of autophagosomes. Finally, by pharmacologically reducing oxidative stress, we could rescue the aberrant autophagosome formation as well as synaptic and neuronal network activity in KANSL1-deficient neurons. Our findings thus point toward an important relation between oxidative stress-induced autophagy and synapse function, and demonstrate the importance of H4K16ac-mediated changes in chromatin structure to balance reactive oxygen species- and MTOR-dependent autophagy.Abbreviations: APO: apocynin; ATG: autophagy related; BAF: bafilomycin A(1); BSO: buthionine sulfoximine; CV: coefficient of variation; DIV: days in vitro; H4K16ac: histone 4 lysine 16 acetylation; iPSC: induced-pluripotent stem cell; KANSL1: KAT8 regulatory NSL complex subunit 1; KdVS: Koolen-de Vries Syndrome; LAMP1: lysosomal associated membrane protein 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEA: micro-electrode array; MTOR: mechanistic target of rapamycin kinase; NSL complex: nonspecific lethal complex; 8-oxo-dG: 8-hydroxydesoxyguanosine; RAP: rapamycin; ROS: reactive oxygen species; sEPSCs: spontaneous excitatory postsynaptic currents; SOD1: superoxide dismutase 1; SQSTM1/p62: sequestosome 1; SYN: synapsin; WRT: wortmannin.
- Published
- 2022
12. BCL11A intellectual developmental disorder: defining the clinical spectrum and genotype-phenotype correlations
- Author
-
Øyvind L. Busk, Kimberley Bradbury, Arjan Bouman, Philippe M. Campeau, Lynne M. Bird, Cornelia Kraus, Colleen Carlston, Rong Mao, Juliette Piard, Laurence Faivre, Amanda Openshaw, Catherine Ward Melver, Mohnish Suri, Christiane Zweier, François Guillemot, Rolph Pfundt, Janice C. Palumbos, Parthiv Haldipur, Jane A. Hurst, Kimberly McDonald, Margaux Serey-Gaut, Luitgard Graul-Neumann, Karen J. Low, Jenny Carmichael, Patrick Ferrerira, Birgit Elisabeth Kristiansen, Ange-Line Bruel, Constance Motter, Andrea Accogli, Darrah N. Haffner, Suhair Hanna, Ruta Marcinkute, Angela Peron, Marcella Zollino, Sofia Maia, James Lespinasse, Claire E. Turner, Sally Ann Lynch, Richard E. Person, Valeria Capra, Kimberly A. Aldinger, Constance Smith-Hicks, Gyri Aasland Gradek, Ingrid M. Wentzensen, Megha Desai, Manuela Morleo, Aditi Shah Parikh, Marcello Scala, Cristina Dias, Gunnar Houge, Telethon Undiagnosed Disease Program, Anne Slavotinek, Roberta Battini, Mary J. Green, Anna Chassevent, Tara Montgomery, David Viskochil, Tatiana Tvrdik, Dawn L. Earl, Karin Weiss, Felice D'Arco, William B. Dobyns, Ping Yee Billie Au, Daniah Beleford, Erica F. Andersen, Bert B.A. de Vries, Jill Clayton-Smith, Christophe Philippe, and Michael J. Bamshad
- Subjects
business.industry ,Postnatal microcephaly ,Microdeletion syndrome ,medicine.disease ,Bioinformatics ,Hypotonia ,Developmental disorder ,Autism spectrum disorder ,Intellectual disability ,Fetal hemoglobin ,medicine ,Missense mutation ,medicine.symptom ,business - Abstract
PurposeHeterozygous variants in BCL11A underlie an intellectual developmental disorder with persistence of fetal hemoglobin (BCL11A-IDD, a.k.a. Dias-Logan syndrome). We sought to delineate the genotypic and phenotypic spectrum of BCL11A-IDD.MethodsWe performed an in-depth analysis of 42 patients with BCL11A-IDD ascertained through a collaborative network of clinical and research colleagues. We also reviewed 33 additional affected individuals previously reported in the literature or available through public repositories with clinical information.ResultsMolecular and clinical data analysis of 75 patients with BCL11A-IDD identified 60 unique variants (30 frameshift, 7 missense, 6 splice-site, 17 stop-gain) and 8 unique CNVs (microdeletions involving BCL11A only). We redefined the most frequent manifestations of the condition: intellectual disability, hypotonia, behavioral abnormalities, postnatal microcephaly and autism spectrum disorder. Two thirds of patients have brain MRI abnormalities, and we identified a recurrent posterior fossa phenotype of vermian hypoplasia and/or small brainstem. Truncating BCL11A variants, particularly those affecting the long (BCL11A-L) and extra-long (-XL) isoforms, sparing the short (-S) isoform, were associated with increased severity.ConclusionsWe expand the clinical delineation of BCL11A-IDD and identify a potential isoform-specific genotype-phenotype correlation. We show that BCL11A-IDD is associated with posterior fossa anomalies and highlight the differences between BCL11A-IDD and 2p16.1p15 microdeletion syndrome.
- Published
- 2021
13. Genome-wide variant calling in reanalysis of exome sequencing data uncovered a pathogenic TUBB3 variant
- Author
-
Elke de Boer, Burcu Yaldiz, Anne-Sophie Denommé-Pichon, Leslie Matalonga, Steve Laurie, Wouter Steyaert, Rick de Reuver, Christian Gilissen, Michael Kwint, Rolph Pfundt, Alain Verloes, Michèl A.A.P. Willemsen, Bert B.A. de Vries, A. Vitobello, Tjitske Kleefstra, Lisenka E.L.M. Vissers, Enzo Cohen, Isabel Cuesta, Daniel Danis, Fei Gao, Rita Horvath, Mridul Johari, Lennart Johanson, Shuang Li, Heba Morsy, Isabelle Nelson, Ida Paramonov, Iris B.A.W. te Paske, Peter Robinson, Marco Savarese, Ana Töpf, Aurélien Trimouille, Joeri K. van der Velde, Jana Vandrovcova, Antonio Vitobello, Birte Zurek, Kristin M. Abbot, Siddharth Banka, Elisa Benetti, Giorgio Casari, Andrea Ciolfi, Jill Clayton-Smith, Bruno Dallapiccola, Kornelia Ellwanger, Laurence Faivre, Holm Graessner, Tobias B. Haack, Anna Hammarsjö, Marketa Havlovicova, Alexander Hoischen, Anne Hugon, Adam Jackson, Mieke Kerstjens, Anna Lindstrand, Estrella López Martín, Milan Macek, Isabelle Maystadt, Manuela Morleo, Vicenzo Nigro, Ann Nordgren, Maria Pettersson, Michele Pinelli, Simone Pizzi, Manuel Posada, Francesca C. Radio, Alessandra Renieri, Caroline Rooryck, Lukas Ryba, Gijs W.E. Santen, Martin Schwarz, Marco Tartaglia, Christel Thauvin, Annalaura Torella, Lisenka Vissers, Pavel Votypka, Klea Vyshka, Kristina Zguro, Dutch Research Council (Holanda), Unión Europea. Comisión Europea. H2020, Netherlands Organisation for Health Research and Development, de Boer, E., Yaldiz, B., Denomme-Pichon, A. -S., Matalonga, L., Laurie, S., Steyaert, W., de Reuver, R., Gilissen, C., Kwint, M., Pfundt, R., Verloes, A., Willemsen, M. A. A. P., de Vries, B. B. A., Vitobello, A., Kleefstra, T., Vissers, L. E. L. M., Nigro, V., Torella, A., and Banfi, S.
- Subjects
Proband ,Exome sequencing ,Adolescent ,Developmental Disabilities ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Mutation, Missense ,Computational biology ,Biology ,Genome ,Exon ,All institutes and research themes of the Radboud University Medical Center ,Tubulin ,Intellectual Disability ,Solve-RD ,Exome Sequencing ,Genetics ,Coding region ,Missense mutation ,Humans ,TUBB3 ,Gene ,Genetics (clinical) ,Sequence (medicine) ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,ERN ITHACA ,Brain ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,General Medicine ,Genome-wide variant calling ,Strabismus ,Face ,Microcephaly ,Female - Abstract
Almost half of all individuals affected by intellectual disability (ID) remain undiagnosed. In the Solve-RD project, exome sequencing (ES) datasets from unresolved individuals with (syndromic) ID (n = 1,472 probands) are systematically reanalyzed, starting from raw sequencing files, followed by genome-wide variant calling and new data interpretation. This strategy led to the identification of a disease-causing de novo missense variant in TUBB3 in a girl with severe developmental delay, secondary microcephaly, brain imaging abnormalities, high hypermetropia, strabismus and short stature. Interestingly, the TUBB3 variant could only be identified through reanalysis of ES data using a genome-wide variant calling approach, despite being located in protein coding sequence. More detailed analysis revealed that the position of the variant within exon 5 of TUBB3 was not targeted by the enrichment kit, although consistent high-quality coverage was obtained at this position, resulting from nearby targets that provide off-target coverage. In the initial analysis, variant calling was restricted to the exon targets ± 200 bases, allowing the variant to escape detection by the variant calling algorithm. This phenomenon may potentially occur more often, as we determined that 36 established ID genes have robust off-target coverage in coding sequence. Moreover, within these regions, for 17 genes (likely) pathogenic variants have been identified before. Therefore, this clinical report highlights that, although compute-intensive, performing genome-wide variant calling instead of target-based calling may lead to the detection of diagnostically relevant variants that would otherwise remain unnoticed. This work was financially supported by Aspasia grants of the Dutch Research Council (015.014.036 to TK and 015.014.066 to LELMV) and Netherlands Organization for Health Research and Development (917.183.10 to TK). The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation programme under grant agreement No 779257. Sí
- Published
- 2021
14. A Case Series of Familial ARID1B Variants Illustrating Variable Expression and Suggestions to Update the ACMG Criteria
- Author
-
Claudia A. L. Ruivenkamp, Isabelle Maystadt, Scott E. Hickey, Bert B.A. de Vries, Marielle Alders, Stéphanie Moortgat, Bregje W.M. van Bon, Jill A. Rosenfeld, Kareesma Parbhoo, Catherine Vincent-Delorme, Johan T. den Dunnen, Thomas Smol, Debra S. Regier, Pleuntje J. van der Sluijs, Gijs W. E. Santen, Alexander J. M. Dingemans, Betsy Schmalz, Erica H. Gerkes, Bekim Sadikovic, Kyra E. Stuurman, Dan Doherty, Jennifer C. Dempsey, Ilana M. Milller, Human Genetics, ACS - Pulmonary hypertension & thrombosis, ARD - Amsterdam Reproduction and Development, and Clinical Genetics
- Subjects
Male ,Inherited ,Coffin–Siris syndrome ,Intellectual disability ,QH426-470 ,PHENOTYPE ,Variable Expression ,Familial ,non-pathogenic ,ARID1B ,Loss of Function Mutation ,Child ,Genetics (clinical) ,Genetics ,RISK ,familial ,Middle Aged ,ACMG guidelines ,DNA-Binding Proteins ,intellectual disability ,DNA methylation ,Medical genetics ,Female ,Haploinsufficiency ,Hand Deformities, Congenital ,Adult ,medicine.medical_specialty ,GENES ,Adolescent ,Genomics ,Biology ,Non-pathogenic ,Article ,Young Adult ,All institutes and research themes of the Radboud University Medical Center ,medicine ,Humans ,Abnormalities, Multiple ,Genetic Predisposition to Disease ,AUTISM ,Gene ,Loss function ,Sequence (medicine) ,COMPLEX ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,DISABILITY ,COFFIN-SIRIS SYNDROME ,Variable expression ,DNA Methylation ,inherited ,variable expression ,Gene Expression Regulation ,Face ,Transcription Factors - Abstract
ARID1B is one of the most frequently mutated genes in intellectual disability (~1%). Most variants are readily classified, since they are de novo and are predicted to lead to loss of function, and therefore classified as pathogenic according to the American College of Medical Genetics and Genomics (ACMG) guidelines for the interpretation of sequence variants. However, familial loss-of-function variants can also occur and can be challenging to interpret. Such variants may be pathogenic with variable expression, causing only a mild phenotype in a parent. Alternatively, since some regions of the ARID1B gene seem to be lacking pathogenic variants, loss-of-function variants in those regions may not lead to ARID1B haploinsufficiency and may therefore be benign. We describe 12 families with potential loss-of-function variants, which were either familial or with unknown inheritance and were in regions where pathogenic variants have not been described or are otherwise challenging to interpret. We performed detailed clinical and DNA methylation studies, which allowed us to confidently classify most variants. In five families we observed transmission of pathogenic variants, confirming their highly variable expression. Our findings provide further evidence for an alternative translational start site and we suggest updates for the ACMG guidelines for the interpretation of sequence variants to incorporate DNA methylation studies and facial analyses.
- Published
- 2021
- Full Text
- View/download PDF
15. Truncating SRCAP variants outside the Floating-Harbor syndrome locus cause a distinct neurodevelopmental disorder with a specific DNA methylation signature
- Author
-
Ian R. Berry, Martin R. Larsen, Ann M. Neumeyer, Lilian Bomme Ousager, Leah J. Rowe, Richard E. Person, Chanika Phornphutkul, David A. Koolen, Constance T. R. M. Stumpel, Konrad Platzer, Elizabeth J. Bhoj, Eric Chater-Diehl, Jason Bunn, Erika Leenders, Koen L.I. van Gassen, Joshua Charkow, Rosanna Weksberg, Ny Hoang, Roos Cuperus, Davor Lessel, Rolph Pfundt, Oana Caluseriu, Sarah J. Goodman, Leandra Folk, Fanggeng Zou, Michelle T. Siu, David Chitayat, Dmitrijs Rots, Jeroen R. Vermeulen, Shuxi Liu, Cheryl Cytrynbaum, Elin Tønne, Hein Brackel, Mareike Mertens, Jennifer Campbell, Jonathan B. Strober, Maja Hempel, Tjitske Kleefstra, Małgorzata J.M. Nowaczyk, Amy Crunk, Marta Pacio-Míguez, Fernando Santos-Simarro, Nicola Brunetti-Pierri, Christa de Geus, María Palomares-Bralo, Lisenka E.L.M. Vissers, Sander Pajusalu, Peter Kannu, Sanaa Choufani, Kristin Lindstrom, Margarita Saenz, Berkley Schmidt, Daniëlle G.M. Bosch, Han G. Brunner, Arie van Haeringen, Ellen van Binsbergen, Brianna Pruniski, Claudia A. L. Ruivenkamp, William G. Wilson, Servi J. C. Stevens, Susan Walker, Kristian Tveten, Zain Awamleh, Gerarda Cappuccio, Alexander J. M. Dingemans, Michael Kwint, Ebba Alkhunaizi, Jonas Denecke, Alyssa Ritter, Eric W. Klee, Bert B.A. de Vries, Jeske V.T. van Harssel, Stephen Meyn, A. Chantal Deden, Francisca Millan, Eva Morava, Ingrid M. Wentzensen, Anne Slavotinek, Stephen W. Scherer, Katrin Õunap, Tuula Rinne, Jessica A. Radley, Yili Xie, Thatjana Gardeitchik, Laura Schultz-Rogers, Karit Reinson, Ronald D. Cohn, Hui Yang, RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Polikliniek (9), Klinische Genetica, MUMC+: DA KG Lab Centraal Lab (9), Klinische Neurowetenschappen, MUMC+: MA Med Staf Spec Neurologie (9), RS: MHeNs - R1 - Cognitive Neuropsychiatry and Clinical Neuroscience, MUMC+: DA Klinische Genetica (5), Rots, Dmitrij, Chater-Diehl, Eric, Dingemans, Alexander J M, Goodman, Sarah J, Siu, Michelle T, Cytrynbaum, Cheryl, Choufani, Sanaa, Hoang, Ny, Walker, Susan, Awamleh, Zain, Charkow, Joshua, Meyn, Stephen, Pfundt, Rolph, Rinne, Tuula, Gardeitchik, Thatjana, de Vries, Bert B A, Deden, A Chantal, Leenders, Erika, Kwint, Michael, Stumpel, Constance T R M, Stevens, Servi J C, Vermeulen, Jeroen R, van Harssel, Jeske V T, Bosch, Danielle G M, van Gassen, Koen L I, van Binsbergen, Ellen, de Geus, Christa M, Brackel, Hein, Hempel, Maja, Lessel, Davor, Denecke, Jona, Slavotinek, Anne, Strober, Jonathan, Crunk, Amy, Folk, Leandra, Wentzensen, Ingrid M, Yang, Hui, Zou, Fanggeng, Millan, Francisca, Person, Richard, Xie, Yili, Liu, Shuxi, Ousager, Lilian B, Larsen, Martin, Schultz-Rogers, Laura, Morava, Eva, Klee, Eric W, Berry, Ian R, Campbell, Jennifer, Lindstrom, Kristin, Pruniski, Brianna, Neumeyer, Ann M, Radley, Jessica A, Phornphutkul, Chanika, Schmidt, Berkley, Wilson, William G, Õunap, Katrin, Reinson, Karit, Pajusalu, Sander, van Haeringen, Arie, Ruivenkamp, Claudia, Cuperus, Roo, Santos-Simarro, Fernando, Palomares-Bralo, María, Pacio-Míguez, Marta, Ritter, Alyssa, Bhoj, Elizabeth, Tønne, Elin, Tveten, Kristian, Cappuccio, Gerarda, Brunetti-Pierri, Nicola, Rowe, Leah, Bunn, Jason, Saenz, Margarita, Platzer, Konrad, Mertens, Mareike, Caluseriu, Oana, Nowaczyk, Małgorzata J M, Cohn, Ronald D, Kannu, Peter, Alkhunaizi, Ebba, Chitayat, David, Scherer, Stephen W, Brunner, Han G, Vissers, Lisenka E L M, Kleefstra, Tjitske, Koolen, David A, and Weksberg, Rosanna
- Subjects
0301 basic medicine ,Heart Septal Defects, Ventricular ,Male ,DNA methylation signature ,nonsense-mediated decay ,speech delay ,PROTEIN ,030105 genetics & heredity ,PHENOTYPE ,epigenomic ,Medical and Health Sciences ,Epigenesis, Genetic ,Craniofacial Abnormalities ,Cohort Studies ,Neurodevelopmental disorder ,2.1 Biological and endogenous factors ,Aetiology ,Genetics (clinical) ,Growth Disorders ,Epigenomics ,non-FLHS SRCAP-related NDD ,Genetics ,Adenosine Triphosphatases ,Genetics & Heredity ,neurodevelopmental disorders ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,SOTOS-LIKE ,Biological Sciences ,SRCAP ,Hypotonia ,AT-HOOK ,3. Good health ,Phenotype ,Mental Health ,intellectual disability ,Speech delay ,DNA methylation ,Female ,medicine.symptom ,Abnormalities ,Multiple ,EXON 34 ,Intellectual and Developmental Disabilities (IDD) ,Locus (genetics) ,Biology ,genotype-phenotype correlation ,DIAGNOSIS ,Article ,03 medical and health sciences ,Genetic ,Clinical Research ,medicine ,Humans ,Abnormalities, Multiple ,Genetic Predisposition to Disease ,Floating-Harbor syndrome ,SPECTRUM ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,MUTATIONS ,Heart Septal Defects ,Infant, Newborn ,Ventricular ,dNaM ,Infant ,DNA Methylation ,medicine.disease ,Newborn ,neurodevelopmental disorder ,GENE ,Brain Disorders ,030104 developmental biology ,Floating–Harbor syndrome ,Case-Control Studies ,Mutation ,epigenomics ,EPISIGNATURES ,Epigenesis - Abstract
Contains fulltext : 234078.pdf (Publisher’s version ) (Open Access) Truncating variants in exons 33 and 34 of the SNF2-related CREBBP activator protein (SRCAP) gene cause the neurodevelopmental disorder (NDD) Floating-Harbor syndrome (FLHS), characterized by short stature, speech delay, and facial dysmorphism. Here, we present a cohort of 33 individuals with clinical features distinct from FLHS and truncating (mostly de novo) SRCAP variants either proximal (n = 28) or distal (n = 5) to the FLHS locus. Detailed clinical characterization of the proximal SRCAP individuals identified shared characteristics: developmental delay with or without intellectual disability, behavioral and psychiatric problems, non-specific facial features, musculoskeletal issues, and hypotonia. Because FLHS is known to be associated with a unique set of DNA methylation (DNAm) changes in blood, a DNAm signature, we investigated whether there was a distinct signature associated with our affected individuals. A machine-learning model, based on the FLHS DNAm signature, negatively classified all our tested subjects. Comparing proximal variants with typically developing controls, we identified a DNAm signature distinct from the FLHS signature. Based on the DNAm and clinical data, we refer to the condition as "non-FLHS SRCAP-related NDD." All five distal variants classified negatively using the FLHS DNAm model while two classified positively using the proximal model. This suggests divergent pathogenicity of these variants, though clinically the distal group presented with NDD, similar to the proximal SRCAP group. In summary, for SRCAP, there is a clear relationship between variant location, DNAm profile, and clinical phenotype. These results highlight the power of combined epigenetic, molecular, and clinical studies to identify and characterize genotype-epigenotype-phenotype correlations.
- Published
- 2021
16. The ARID1B spectrum in 143 patients
- Author
-
Catherine Vincent-Delorme, Claudia A. L. Ruivenkamp, Marjan De Rademaeker, Francisco Martínez, Tracy Dudding-Byth, Marianne McGuire, Bert B.A. de Vries, Mitsuhiro Kato, Levinus A. Bok, Hülya Kayserili, Jeff M. Milunsky, Suzanne C E H Sallevelt, Alwin F. J. Brouwer, Jill Clayton-Smith, Emilia K. Bijlsma, Miranda Splitt, Patricia G. Wheeler, Philippe M. Campeau, Fatma Mujgan Sonmez, Kylin Lammers, Stefanie Beck-Wödl, Caroline Rooryck, Louise C. Wilson, Evan E. Eichler, Sarina G. Kant, Johanna C. Herkert, Karin R. Heitink, Eyyup Uctepe, Pleuntje J. van der Sluijs, Miho Adachi-Fukuda, Lone W. Laulund, Sandra Jansen, Nicolette S. den Hollander, Damien Lederer, Tomoki Kosho, Constance T. R. M. Stumpel, Saskia M. Maas, Esra Kılıç, Erica H. Gerkes, Duco Steenbeek, Melissa Lees, Kay Metcalfe, Karin Dahan, Ineke van der Burgt, Isabelle Maystadt, Christian Netzer, Ute Grasshoff, Carmen Orellana, Mahmut Şamil Sağıroğlu, Gijs W. E. Santen, Pelin Ozlem Simsek-Kiper, Mónica Roselló, Gabriela Soares, Alexander P.A. Stegmann, Stephen P. Robertson, Adila Al-Kindy, Maian Roifman, Saori Tanabe, Vera Riehmer, Brain H Y Chung, Arie van Haeringen, G. Eda Utine, Yasemin Alanay, Rogier Kersseboom, John B. Moeschler, Barbara Oehl-Jaschkowitz, Katherine Berry, Denise Horn, Alice Gardham, Shane McKee, Anwar Baban, Amparo Sanchis Calvo, Golder N. Wilson, Krystyna H. Chrzanowska, G. M. S. Mancini, Ellen R. Elias, Małgorzata Krajewska-Walasek, Rolph Pfundt, Sarju G. Mehta, Fabienne G. Ropers, Seiji Mizuno, David Hunt, Caroline Pottinger, Dagmar Wieczorek, Yoyo W. Y. Chu, Laurent Pasquier, Bernd Wollnik, Nobuhiko Okamoto, Sunita Venkateswaran, Vanesa López-González, Natalie Canham, Blanca Gener, Anne Destree, Christina Fagerberg, Rachel K. Earl, Sharon N M Olminkhof, Nursel Elcioglu, Charlotte W. Ockeloen, Carlo Marcelis, Samantha A. Vergano, Hermine E. Veenstra-Knol, Anneke T. Vulto-van Silfhout, Allan Bayat, Catheline Vilain, Lucia Solaeche, MUMC+: DA KG Polikliniek (9), RS: GROW - R4 - Reproductive and Perinatal Medicine, Genetica & Celbiologie, MUMC+: DA KG Lab Centraal Lab (9), MUMC+: DA Pat Cytologie (9), Klinische Genetica, van der Sluijs, Pleuntje J., Jansen, Sandra, Vergano, Samantha A., Adachi-Fukuda, Miho, Alanay, Yasemin, AlKindy, Adila, Baban, Anwar, Bayat, Allan, Beck-Woedl, Stefanie, Berry, Katherine, Bijlsma, Emilia K., Bok, Levinus A., Brouwer, Alwin F. J., van der Burgt, Ineke, Campeau, Philippe M., Canham, Natalie, Chrzanowska, Krystyna, Chu, Yoyo W. Y., Chung, Brain H. Y., Dahan, Karin, De Rademaeker, Marjan, Destree, Anne, Dudding-Byth, Tracy, Earl, Rachel, Elcioglu, Nursel, Elias, Ellen R., Fagerberg, Christina, Gardham, Alice, Gener, Blanca, Gerkes, Erica H., Grasshoff, Ute, van Haeringen, Arie, Heitink, Karin R., Herkert, Johanna C., den Hollander, Nicolette S., Horn, Denise, Hunt, David, Kant, Sarina G., Kato, Mitsuhiro, Kayserili, Hulya, Kersseboom, Rogier, Kilic, Esra, Krajewska-Walasek, Malgorzata, Lammers, Kylin, Laulund, Lone W., Lederer, Damien, Lees, Melissa, Lopez-Gonzalez, Vanesa, Maas, Saskia, Mancini, Grazia M. S., Marcelis, Carlo, Martinez, Francisco, Maystadt, Isabelle, McGuire, Marianne, McKee, Shane, Mehta, Sarju, Metcalfe, Kay, Milunsky, Jeff, Mizuno, Seiji, Moeschler, John B., Netzer, Christian, Ockeloen, Charlotte W., Oehl-Jaschkowitz, Barbara, Okamoto, Nobuhiko, Olminkhof, Sharon N. M., Orellana, Carmen, Pasquier, Laurent, Pottinger, Caroline, Riehmer, Vera, Robertson, Stephen P., Roifman, Maian, Rooryck, Caroline, Ropers, Fabienne G., Rosello, Monica, Ruivenkamp, Claudia A. L., Sagiroglu, Mahmut S., Sallevelt, Suzanne C. E. H., Sanchis Calvo, Amparo, Simsek-Kiper, Pelin O., Soares, Gabriela, Solaeche, Lucia, Sonmez, Fatma Mujgan, Splitt, Miranda, Steenbeek, Duco, Stegmann, Alexander P. A., Stumpel, Constance T. R. M., Tanabe, Saori, Uctepe, Eyyup, Utine, G. Eda, Veenstra-Knol, Hermine E., Venkateswaran, Sunita, Vilain, Catheline, Vincent-Delorme, Catherine, Vulto-van Silfhout, Anneke T., Wheeler, Patricia, Wilson, Golder N., Wilson, Louise C., Wollnik, Bernd, Kosho, Tomoki, Wieczorek, Dagmar, Eichler, Evan, Pfundt, Rolph, de Vries, Bert B. A., Clayton-Smith, Jill, Santen, Gijs W. E., Erasmus MC other, Clinical Genetics, Human Genetics, and Acibadem University Dspace
- Subjects
Male ,0301 basic medicine ,Hypertrichosis ,Pediatrics ,cuello ,bias ,Coffin–Siris syndrome ,Chromosomal Proteins, Non-Histone ,humanos ,adolescente ,Penetrance ,PHENOTYPE ,0302 clinical medicine ,Genotype ,Intellectual disability ,Exome ,Coffin-Siris syndrome ,Child ,mediana edad ,Genetics (clinical) ,Exome sequencing ,factores de transcripción ,adulto ,Middle Aged ,estudios de asociación genética ,3. Good health ,DNA-Binding Proteins ,intellectual disability ,Child, Preschool ,discapacidad intelectual ,penetrancia ,Female ,Hand Deformities, Congenital ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Adult ,medicine.medical_specialty ,Adolescent ,Micrognathism ,Article ,CHROMATIN-REMODELING COMPLEX ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,cara ,micrognatismo ,Human Phenotype Ontology ,medicine ,Humans ,Abnormalities, Multiple ,mutación ,Long eyelashes ,Genetic Association Studies ,lactante ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,business.industry ,MUTATIONS ,proteínas de unión al ADN ,Infant, Newborn ,Genetic Variation ,Infant ,ARID1B ,Hand Deformities ,Phalanx ,medicine.disease ,variación genética ,deformidades de la mano ,exoma ,030104 developmental biology ,Face ,Mutation ,business ,Neck ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Purpose: Pathogenic variants in ARID1B are one of the most frequent causes of intellectual disability (ID) as determined by large-scale exome sequencing studies. Most studies published thus far describe clinically diagnosed Coffin-Siris patients (ARID1BCSS) and it is unclear whether these data are representative for patients identified through sequencing of unbiased ID cohorts (ARID1B-ID). We therefore sought to determine genotypic and phenotypic differences between ARID1B-ID and ARID1B-CSS. In parallel, we investigated the effect of different methods of phenotype reporting. Methods: Clinicians entered clinical data in an extensive webbased survey. Results: 79 ARID1B-CSS and 64 ARID1B-ID patients were included. CSS-associated dysmorphic features, such as thick eyebrows, long eyelashes, thick alae nasi, long and/or broad philtrum, small nails and small or absent fifth distal phalanx and hypertrichosis, were observed significantly more often (p < 0.001) in ARID1B-CSS patients. No other significant differences were identified. Conclusion: There are only minor differences between ARID1BID and ARID1B-CSS patients. ARID1B-related disorders seem to consist of a spectrum, and patients should be managed similarly. We demonstrated that data collection methods without an explicit option to report the absence of a feature (such as most Human Phenotype Ontology-based methods) tended to underestimate gene-related features., We are grateful for the assistance of Pepijn Cox in setting up the website www.arid1bgene.com. This study has made use of data generated by the Human Disease Genes website series, www.humandiseasegenes.com. This work was financially supported by grants from the Netherlands Organisation for Health Research and Development (917-86-319 to B.B.A.d.V., 912-12-109 to B.B.A.d.V.)
- Published
- 2019
17. De novo variants in FBXO11 cause a syndromic form of intellectual disability with behavioral problems and dysmorphisms
- Author
-
Paulien A Terhal, Rosa Pettinato, Jessica Jackson, Maria J. Guillen Sacoto, R. Frank Kooy, Rolph Pfundt, Grace E. VanNoy, Asbjørg Stray-Pedersen, Elizabeth Judd, Tuula Rinne, Rhonda E. Schnur, Marie José H. Van Den Boogaard, Jolien S. Klein Wassink-Ruiter, Paldeep S. Atwal, David A. Sweetser, Ilse J. Anderson, Jessica L. Waxler, Ilse M. van der Werf, Kristian Tveten, Alexander P.A. Stegmann, Petra de Vries, Alexandra Afenjar, Lisenka E.L.M. Vissers, Anke Van Dijck, Sonja A. de Munnik, Anthonie J. van Essen, Ivan Iossifov, Marcia C. Willing, Charu Kaiwar, Charlotte W. Ockeloen, Joris A. Veltman, Mieke M. van Haelst, Diane Doummar, Sandra Jansen, Marije Meuwissen, Eric W. Klee, Pankaj B. Agrawal, Ellen van Binsbergen, Bert B.A. de Vries, Victoria R. Sanders, A. Micheil Innes, Kristin G. Monaghan, Hilary Racher, Corrado Romano, Zeynep Coban-Akdemir, Albertien M. van Eerde, Eric J. Smeets, Caroline Nava, Lucia Castiglia, Boris Keren, Koen L.I. van Gassen, Human genetics, Amsterdam Neuroscience - Complex Trait Genetics, Amsterdam Reproduction & Development (AR&D), MUMC+: DA KG Polikliniek (9), MUMC+: DA KG Lab Centraal Lab (9), and RS: FHML non-thematic output
- Subjects
Protein-Arginine N-Methyltransferases ,media_common.quotation_subject ,Nonsense ,Biology ,PHENOTYPE ,Article ,DNA sequencing ,Frameshift mutation ,GENEMATCHER ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,E3 ,Intellectual Disability ,Intellectual disability ,Genetics ,medicine ,Journal Article ,UBIQUITIN LIGASES ,Missense mutation ,Humans ,Abnormalities, Multiple ,Gene ,Genetics (clinical) ,media_common ,0303 health sciences ,Behavior ,SPECTRUM ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,IDENTIFICATION ,MUTATIONS ,F-Box Proteins ,030305 genetics & heredity ,Genetic Variation ,Syndrome ,medicine.disease ,GENE ,CANCER ,Hypotonia ,FAMILY ,Chemistry ,Autism spectrum disorder ,Human medicine ,medicine.symptom ,Abnormalities ,Multiple ,Gene Deletion ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] - Abstract
Determining pathogenicity of genomic variation identified by next-generation sequencing techniques can be supported by recurrent disruptive variants in the same gene in phenotypically similar individuals. However, interpretation of novel variants in a specific gene in individuals with mild-moderate intellectual disability (ID) without recognizable syndromic features can be challenging and reverse phenotyping is often required. We describe 24 individuals with a de novo disease-causing variant in, or partial deletion of, the F-box only protein 11 gene (FBXO11, also known as VIT1 and PRMT9). FBXO11 is part of the SCF (SKP1-cullin-F-box) complex, a multi-protein E3 ubiquitin-ligase complex catalyzing the ubiquitination of proteins destined for proteasomal degradation. Twenty-two variants were identified by next-generation sequencing, comprising 2 in-frame deletions, 11 missense variants, 1 canonical splice site variant, and 8 nonsense or frameshift variants leading to a truncated protein or degraded transcript. The remaining two variants were identified by array-comparative genomic hybridization and consisted of a partial deletion of FBXO11. All individuals had borderline to severe ID and behavioral problems (autism spectrum disorder, attention-deficit/hyperactivity disorder, anxiety, aggression) were observed in most of them. The most relevant common facial features included a thin upper lip and a broad prominent space between the paramedian peaks of the upper lip. Other features were hypotonia and hyperlaxity of the joints. We show that de novo variants in FBXO11 cause a syndromic form of ID. The current series show the power of reverse phenotyping in the interpretation of novel genetic variances in individuals who initially did not appear to have a clear recognizable phenotype.
- Published
- 2019
18. De Novo and Inherited Pathogenic Variants in KDM3B Cause Intellectual Disability, Short Stature, and Facial Dysmorphism
- Author
-
Laurence Perrin, Marjolijn C.J. Jongmans, Isabelle Thiffault, Han G. Brunner, Bethany Peri, Sarah K. Bartz, Alexandra Afenjar, Kristina Baltrunaite, Esmé Waanders, Jayne Y. Hehir-Kwa, Illja J. Diets, Boris Keren, Alexander J. M. Dingemans, Gea Beunders, Roland P. Kuiper, Anneke T. Vulto-van Silfhout, Laurens Wiel, Bert Callewaert, Andrew Dauber, Margot R.F. Reijnders, Tjitske Kleefstra, Vivian Hwa, Caroline Nava, Matias Wagner, Matthias Griese, Lina Huerta-Saenz, Roos van der Donk, Rolph Pfundt, Nicoline Hoogerbrugge, Christian Gilissen, Maxime Cadieux-Dion, Julia Vodopiutz, Koen L.I. van Gassen, Annekatrien Boel, Julien Thevenon, Nienke E. Verbeek, Bert B.A. de Vries, Vassiliki Konstantopoulou, Human genetics, MUMC+: DA KG Polikliniek (9), MUMC+: DA Klinische Genetica (5), Klinische Genetica, and RS: GROW - R4 - Reproductive and Perinatal Medicine
- Subjects
0301 basic medicine ,Male ,Jumonji Domain-Containing Histone Demethylases ,Developmental Disabilities ,WEAVER SYNDROME ,PROTEIN ,Haploinsufficiency ,Craniofacial Abnormalities ,Histones ,0302 clinical medicine ,Intellectual disability ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Missense mutation ,DEMETHYLASE KDM3B ,Exome ,Child ,Genetics (clinical) ,Exome sequencing ,Genetics ,RUBINSTEIN-TAYBI SYNDROME ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Phenotype ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Joint hypermobility ,GENETICS ,JMJD1C ,Mutation, Missense ,Dwarfism ,Biology ,Short stature ,Kdm3b ,Cancer Predisposition ,Developmental Delay ,Facial Recognition ,Intellectual Disability ,Leukemia ,Lymphoma ,Short Stature ,03 medical and health sciences ,Report ,medicine ,Humans ,MYELOID-LEUKEMIA ,Genetic Association Studies ,Germ-Line Mutation ,Weaver syndrome ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Rubinstein–Taybi syndrome ,MUTATIONS ,DELETION ,Genetic Variation ,medicine.disease ,Body Height ,Musculoskeletal Abnormalities ,INDIVIDUALS ,030104 developmental biology ,Face ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] - Abstract
Contains fulltext : 202646.pdf (Publisher’s version ) (Open Access) By using exome sequencing and a gene matching approach, we identified de novo and inherited pathogenic variants in KDM3B in 14 unrelated individuals and three affected parents with varying degrees of intellectual disability (ID) or developmental delay (DD) and short stature. The individuals share additional phenotypic features that include feeding difficulties in infancy, joint hypermobility, and characteristic facial features such as a wide mouth, a pointed chin, long ears, and a low columella. Notably, two individuals developed cancer, acute myeloid leukemia and Hodgkin lymphoma, in childhood. KDM3B encodes for a histone demethylase and is involved in H3K9 demethylation, a crucial part of chromatin modification required for transcriptional regulation. We identified missense and truncating variants, suggesting that KDM3B haploinsufficiency is the underlying mechanism for this syndrome. By using a hybrid facial-recognition model, we show that individuals with a pathogenic variant in KDM3B have a facial gestalt, and that they show significant facial similarity compared to control individuals with ID. In conclusion, pathogenic variants in KDM3B cause a syndrome characterized by ID, short stature, and facial dysmorphism.
- Published
- 2019
19. De novo and biallelic DEAF1 variants cause a phenotypic spectrum
- Author
-
Elysa J. Marco, Heather C Mefford, Stacey McGee, Christèle Dubourg, Edmund Cauley, Randi J Hagerman, Maria J. Nabais Sá, Bert B.A. de Vries, Rüdiger Lorenz, Elizabeth E. Palmer, Michael J. Parker, Arjan P.M. de Brouwer, Hester Y. Kroes, M. Chiara Manzini, Abbey A. Scott, Tara Montgomery, Naama Orenstein, Jeanne Amiel, Delphine Héron, Leonie A. Menke, Jonathan Berg, Sylvie Odent, Rachel Harrison, Philip J. Jensik, Rani Sachdev, Miranda Splitt, Tyler Mark Pierson, Jan Maarten Cobben, Ehsan Ghayoor Karimiani, Anneke T. Vulto-vanSilfhout, Roberto Colombo, Nayana Lahiri, Julian A. Martinez-Agosto, Evan P. McNeil, Boris Keren, John M. Graham, Chanika Phornphutkul, Reza Maroofian, Radboud University Medical Center [Nijmegen], Southern Illinois University [Carbondale] (SIU), Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, St George's, University of London, Geisel School of Medicine at Dartmouth, University Medical Center [Utrecht], University of California [Davis] (UC Davis), University of California, Nottingham University Hospitals NHS Trust, Northern Genetics Service, Newcastle University [Newcastle], University of New South Wales [Sydney] (UNSW), Department of Pediatrics [Seattle, WA, USA] (Division of Genetic Medicine), University of Washington [Seattle]-Seattle Children’s Hospital, University of California [Los Angeles] (UCLA), Tel Aviv University [Tel Aviv], University of Dundee, Imagine - Institut des maladies génétiques (IMAGINE - U1163), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de Génétique Cytogénétique et Embryologie [CHU Pitié-Salpêtrière], CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), University of Amsterdam [Amsterdam] (UvA), Cedars-Sinai Medical Center, The George Washington University (GW), Fondazione 'Policlinico Universitario A. Gemelli' [Rome], CHU Pontchaillou [Rennes], Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Centre National de la Recherche Scientifique (CNRS)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Warren Alpert Medical School of Brown University, University of California (UC), Nottingham University Hospitals NHS Trust (NUH), Department of Pediatrics [Seattle, WA, USA], Tel Aviv University (TAU), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Université de Rennes (UR)-Centre National de la Recherche Scientifique (CNRS)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), General Paediatrics, Paediatric Genetics, Amsterdam Reproduction & Development (AR&D), Sheffield Children's Hospital, St George‘s, University of London, University of Washington [Seattle]-Seattle Children’s Hospital [Seattle, WA, USA], Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-CHU Pitié-Salpêtrière [APHP], George Washington University (GW), and AMS - Sports
- Subjects
Adult ,Male ,Microcephaly ,Adolescent ,phenotype ,Developmental Disabilities ,genotype ,[SDV]Life Sciences [q-bio] ,Mutation, Missense ,Biology ,Young Adult ,03 medical and health sciences ,Neurodevelopmental disorder ,All institutes and research themes of the Radboud University Medical Center ,Genotype ,medicine ,Humans ,Exome ,Language Development Disorders ,Autistic Disorder ,Allele ,Child ,Alleles ,Genetic Association Studies ,Genetics (clinical) ,030304 developmental biology ,Genetics ,0303 health sciences ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,030305 genetics & heredity ,medicine.disease ,Phenotype ,neurodevelopmental disorder ,Human genetics ,DEAF1 ,DNA-Binding Proteins ,intellectual disability ,Child, Preschool ,Speech delay ,Autism ,Female ,medicine.symptom ,Transcription Factors - Abstract
International audience; Purpose To investigate the effect of different DEAF1 variants on the phenotype of patients with autosomal dominant and recessive inheritance patterns and on DEAF1 activity in vitro. Methods We assembled a cohort of 23 patients with de novo and biallelic DEAF1 variants, described the genotype–phenotype correlation, and investigated the differential effect of de novo and recessive variants on transcription assays using DEAF1 and Eif4g3 promoter luciferase constructs. Results The proportion of the most prevalent phenotypic features, including intellectual disability, speech delay, motor delay, autism, sleep disturbances, and a high pain threshold, were not significantly different in patients with biallelic and pathogenic de novo DEAF1 variants. However, microcephaly was exclusively observed in patients with recessive variants (p < 0.0001). Conclusion We propose that different variants in the DEAF1 gene result in a phenotypic spectrum centered around neurodevelopmental delay. While a pathogenic de novo dominant variant would also incapacitate the product of the wild-type allele and result in a dominant-negative effect, a combination of two recessive variants would result in a partial loss of function. Because the clinical picture can be nonspecific, detailed phenotype information, segregation, and functional analysis are fundamental to determine the pathogenicity of novel variants and to improve the care of these patients. © 2019, American College of Medical Genetics and Genomics.
- Published
- 2019
20. Establishing the phenotypic spectrum of ZTTK syndrome by analysis of 52 individuals with variants in SON
- Author
-
Ingrid M.B.H. van de Laar, Eline A. Verberne, Kim M G Truijen, Marlon E F Wilsterman, Mieke M. van Haelst, Jung-Hyun Kim, Margje Sinnema, Erica H. Gerkes, Kathleen M Collins, Zahide Alaçam, Kristin Lindstrom, Alexander J. M. Dingemans, Hermine E. Veenstra-Knol, Mathilde Nizon, Lisenka E.L.M. Vissers, Anneke T. Vulto-van Silfhout, Astrid S Plomp, Laurence Faivre, Bert B.A. de Vries, Edyta Heropolitańska-Pliszka, Rani Sachdev, James Pauling, Eun-Young Erin Ahn, Jon Skranes, Caroline Racine, Human Genetics, Graduate School, MUMC+: DA KG Polikliniek (9), RS: FHML non-thematic output, Clinical Genetics, Human genetics, Amsterdam Neuroscience - Complex Trait Genetics, and Amsterdam Reproduction & Development (AR&D)
- Subjects
Developmental Disabilities ,Mutation, Missense ,Biology ,Article ,INTELLECTUAL-DISABILITY ,Minor Histocompatibility Antigens ,SDG 3 - Good Health and Well-being ,Intellectual Disability ,Human Phenotype Ontology ,Intellectual disability ,Genetics ,medicine ,Missense mutation ,Humans ,Child ,Gene ,Genetics (clinical) ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Comment ,Phenotypic trait ,Cognitive artificial intelligence ,Syndrome ,medicine.disease ,Phenotype ,DNA-Binding Proteins ,RNA splicing ,Haploinsufficiency - Abstract
Contains fulltext : 248367.pdf (Publisher’s version ) (Closed access) Zhu-Tokita-Takenouchi-Kim (ZTTK) syndrome, an intellectual disability syndrome first described in 2016, is caused by heterozygous loss-of-function variants in SON. Its encoded protein promotes pre-mRNA splicing of many genes essential for development. Whereas individual phenotypic traits have previously been linked to erroneous splicing of SON target genes, the phenotypic spectrum and the pathogenicity of missense variants have not been further evaluated. We present the phenotypic abnormalities in 52 individuals, including 17 individuals who have not been reported before. In total, loss-of-function variants were detected in 49 individuals (de novo in 47, inheritance unknown in 2), and in 3, a missense variant was observed (2 de novo, 1 inheritance unknown). Phenotypic abnormalities, systematically collected and analyzed in Human Phenotype Ontology, were found in all organ systems. Significant inter-individual phenotypic variability was observed, even in individuals with the same recurrent variant (n = 13). SON haploinsufficiency was previously shown to lead to downregulation of downstream genes, contributing to specific phenotypic features. Similar functional analysis for one missense variant, however, suggests a different mechanism than for heterozygous loss-of-function. Although small in numbers and while pathogenicity of these variants is not certain, these data allow for speculation whether de novo missense variants cause ZTTK syndrome via another mechanism, or a separate overlapping syndrome. In conclusion, heterozygous loss-of-function variants in SON define a recognizable syndrome, ZTTK, associated with a broad, severe phenotypic spectrum, characterized by a large inter-individual variability. These observations provide essential information for affected individuals, parents, and healthcare professionals to ensure appropriate clinical management. 11 p.
- Published
- 2021
21. SPEN haploinsufficiency causes a neurodevelopmental disorder overlapping proximal 1p36 deletion syndrome with an episignature of X chromosomes in females
- Author
-
Gilles Morin, Krista Bluske, Nathaniel H. Robin, Laurence Faivre, Manuela Priolo, Dihong Zhou, Evangeline Kurtz-Nelson, Tianyun Wang, Omar Sherbini, Daryl A. Scott, Karen Stals, Fabíola Paoli Monteiro, Kaifang Pang, Sara Cabet, Francesca Clementina Radio, Bruno Dallapiccola, Marjon van Slegtenhorst, Rachel K. Earl, Katheryn Grand, Maria Iascone, Alice S. Brooks, Angelo Selicorni, July K. Jean Cuevas, Paolo Gasparini, Maria Lisa Dentici, Marialetizia Motta, Britt-Marie Anderlid, Kristin Lindstrom, Berrin Monteleone, Andrea Ciolfi, Karin Weiss, Katharina Steindl, Kirsty McWalter, Rosalba Carrozzo, Ruben Boers, Helen Kingston, Kym M. Boycott, Bekim Sadikovic, Laura Schultz-Rogers, Evan E. Eichler, Laura A Cross, Alison M R Castle, Louisa Kalsner, Lucia Pedace, Marijke R. Wevers, John M. Graham, Jessica Sebastian, Antonio Vitobello, Gaetan Lesca, Alexander P.A. Stegmann, Suneeta Madan-Khetarpal, Tahsin Stefan Barakat, Abdallah F. Elias, Teresa Robert Finestra, Adeline Vanderver, Peter D. Turnpenny, Bregje W.M. van Bon, Aida Telegrafi, David J. Amor, Deepali N. Shinde, Pedro A. Sanchez-Lara, Lisenka E.L.M. Vissers, Adam Jackson, Rolph Pfundt, Alessandro Bruselles, Andres Hernandez-Garcia, Karin E. M. Diderich, Flavio Faletra, Dana H. Goodloe, Joanne Baez, Sarit Ravid, Romano Tenconi, Sarah L. Sawyer, Lynn Pais, Bronwyn Kerr, Joost Gribnau, Lauren Carter, Melissa T. Carter, Zhandong Liu, Jennifer L. Kemppainen, Jennifer MacKenzie, Jimmy Holder, Elke de Boer, Margaret Au, Taila Hartley, Carol J Saunders, Luciana Musante, Bert B.A. de Vries, Tania Vertemati Secches, Haley McConkey, Willow Sheehan, Francesca Pantaleoni, Caterina Zanus, Christophe Philippe, Chelsea Roadhouse, Stefania Lo Cicero, Sian Ellard, R. Tanner Hagelstrom, Megha Desai, Fernando Kok, Joset Pascal, Marco Tartaglia, Eric W. Klee, Eva Morava, Michael A. Levy, Peggy Kulch, Lyndon Gallacher, Erica L. Macke, Emilia Stellacci, Siddharth Banka, Kristin G. Monaghan, Anita Rauch, Meghan C. Towne, Kate Chandler, Clinical Genetics, Developmental Biology, Radio, F. C., Pang, K., Ciolfi, A., Levy, M. A., Hernandez-Garcia, A., Pedace, L., Pantaleoni, F., Liu, Z., de Boer, E., Jackson, A., Bruselles, A., Mcconkey, H., Stellacci, E., Lo Cicero, S., Motta, M., Carrozzo, R., Dentici, M. L., Mcwalter, K., Desai, M., Monaghan, K. G., Telegrafi, A., Philippe, C., Vitobello, A., Au, M., Grand, K., Sanchez-Lara, P. A., Baez, J., Lindstrom, K., Kulch, P., Sebastian, J., Madan-Khetarpal, S., Roadhouse, C., Mackenzie, J. J., Monteleone, B., Saunders, C. J., Jean Cuevas, J. K., Cross, L., Zhou, D., Hartley, T., Sawyer, S. L., Monteiro, F. P., Secches, T. V., Kok, F., Schultz-Rogers, L. E., Macke, E. L., Morava, E., Klee, E. W., Kemppainen, J., Iascone, M., Selicorni, A., Tenconi, R., Amor, D. J., Pais, L., Gallacher, L., Turnpenny, P. D., Stals, K., Ellard, S., Cabet, S., Lesca, G., Pascal, J., Steindl, K., Ravid, S., Weiss, K., Castle, A. M. R., Carter, M. T., Kalsner, L., de Vries, B. B. A., van Bon, B. W., Wevers, M. R., Pfundt, R., Stegmann, A. P. A., Kerr, B., Kingston, H. M., Chandler, K. E., Sheehan, W., Elias, A. F., Shinde, D. N., Towne, M. C., Robin, N. H., Goodloe, D., Vanderver, A., Sherbini, O., Bluske, K., Hagelstrom, R. T., Zanus, C., Faletra, F., Musante, L., Kurtz-Nelson, E. C., Earl, R. K., Anderlid, B. -M., Morin, G., van Slegtenhorst, M., Diderich, K. E. M., Brooks, A. S., Gribnau, J., Boers, R. G., Finestra, T. R., Carter, L. B., Rauch, A., Gasparini, P., Boycott, K. M., Barakat, T. S., Graham, J. M., Faivre, L., Banka, S., Wang, T., Eichler, E. E., Priolo, M., Dallapiccola, B., Vissers, L. E. L. M., Sadikovic, B., Scott, D. A., Holder, J. L., Tartaglia, M., MUMC+: DA KG Lab Centraal Lab (9), and RS: FHML non-thematic output
- Subjects
0301 basic medicine ,SHARP ,Male ,obesity ,genotype-phenotype correlations ,Autism Spectrum Disorder ,PROTEIN ,Chromosome Disorders ,Haploinsufficiency ,RNA-Binding Protein ,PHENOTYPE CORRELATIONS ,1p36 ,distal 1p36 deletion syndrome ,DNA methylome analysis ,episignature ,neurodevelopmental disorder ,proximal 1p36 deletion syndrome ,SPEN ,X chromosome ,Adolescent ,Child ,Child, Preschool ,Chromosome Deletion ,Chromosomes, Human, Pair 1 ,Chromosomes, Human, X ,DNA Methylation ,DNA-Binding Proteins ,Epigenesis, Genetic ,Female ,Humans ,Intellectual Disability ,Neurodevelopmental Disorders ,Phenotype ,RNA-Binding Proteins ,Young Adult ,0302 clinical medicine ,Neurodevelopmental disorder ,Neurodevelopmental Disorder ,Intellectual disability ,MOLECULAR CHARACTERIZATION ,Genetics (clinical) ,Genetics ,DNA methylome analysi ,SPLIT-ENDS ,Hypotonia ,Autism spectrum disorder ,MONOSOMY 1P36 ,Pair 1 ,medicine.symptom ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Human ,DNA-Binding Protein ,Biology ,genotype-phenotype correlation ,Chromosomes ,03 medical and health sciences ,Genetic ,SDG 3 - Good Health and Well-being ,Report ,REVEALS ,medicine ,Epigenetics ,Preschool ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,1p36 deletion syndrome ,IDENTIFICATION ,MUTATIONS ,medicine.disease ,GENE ,030104 developmental biology ,Chromosome Disorder ,030217 neurology & neurosurgery ,Epigenesis - Abstract
Contains fulltext : 231702.pdf (Publisher’s version ) (Closed access) Deletion 1p36 (del1p36) syndrome is the most common human disorder resulting from a terminal autosomal deletion. This condition is molecularly and clinically heterogeneous. Deletions involving two non-overlapping regions, known as the distal (telomeric) and proximal (centromeric) critical regions, are sufficient to cause the majority of the recurrent clinical features, although with different facial features and dysmorphisms. SPEN encodes a transcriptional repressor commonly deleted in proximal del1p36 syndrome and is located centromeric to the proximal 1p36 critical region. Here, we used clinical data from 34 individuals with truncating variants in SPEN to define a neurodevelopmental disorder presenting with features that overlap considerably with those of proximal del1p36 syndrome. The clinical profile of this disease includes developmental delay/intellectual disability, autism spectrum disorder, anxiety, aggressive behavior, attention deficit disorder, hypotonia, brain and spine anomalies, congenital heart defects, high/narrow palate, facial dysmorphisms, and obesity/increased BMI, especially in females. SPEN also emerges as a relevant gene for del1p36 syndrome by co-expression analyses. Finally, we show that haploinsufficiency of SPEN is associated with a distinctive DNA methylation episignature of the X chromosome in affected females, providing further evidence of a specific contribution of the protein to the epigenetic control of this chromosome, and a paradigm of an X chromosome-specific episignature that classifies syndromic traits. We conclude that SPEN is required for multiple developmental processes and SPEN haploinsufficiency is a major contributor to a disorder associated with deletions centromeric to the previously established 1p36 critical regions.
- Published
- 2021
22. Quantitative facial phenotyping for Koolen-de Vries and 22q11.2 deletion syndrome
- Author
-
Diante E Stremmelaar, Bert B.A. de Vries, Patrick Rump, Jayne Y. Hehir-Kwa, Lisenka E.L.M. Vissers, Alexander J. M. Dingemans, Roos van der Donk, and David A. Koolen
- Subjects
Adult ,Male ,22q11 Deletion Syndrome ,Adolescent ,Bioinformatics ,Article ,Diagnosis, Differential ,03 medical and health sciences ,Unknown Significance ,Sex Factors ,Intellectual Disability ,Intellectual disability ,Genetics ,medicine ,Humans ,Deletion syndrome ,Clinical significance ,Abnormalities, Multiple ,Child ,Genetics (clinical) ,0303 health sciences ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,business.industry ,030305 genetics & heredity ,Age Factors ,Infant ,Nuclear Proteins ,medicine.disease ,Pathogenicity ,Phenotype ,Child, Preschool ,Face ,Gestalt psychology ,Female ,Chromosome Deletion ,business ,Hybrid model ,Chromosomes, Human, Pair 17 - Abstract
Contains fulltext : 237899.pdf (Publisher’s version ) (Closed access) The Koolen-de Vries syndrome (KdVS) is a multisystem syndrome with variable facial features caused by a 17q21.31 microdeletion or KANSL1 truncating variant. As the facial gestalt of KdVS has resemblance with the gestalt of the 22q11.2 deletion syndrome (22q11.2DS), we assessed whether our previously described hybrid quantitative facial phenotyping algorithm could distinguish between these two syndromes, and whether there is a facial difference between the molecular KdVS subtypes. We applied our algorithm to 2D photographs of 97 patients with KdVS (78 microdeletions, 19 truncating variants (likely) causing KdVS) and 48 patients with 22q11.2DS as well as age, gender and ethnicity matched controls with intellectual disability (n = 145). The facial gestalts of KdVS and 22q11.2DS were both recognisable through significant clustering by the hybrid model, yet different from one another (p = 7.5 × 10(-10) and p = 0.0052, respectively). Furthermore, the facial gestalts of KdVS caused by a 17q21.31 microdeletion and KANSL1 truncating variant (likely) causing KdVS were indistinguishable (p = 0.981 and p = 0.130). Further application to three patients with a variant of unknown significance in KANSL1 showed that these faces do not match KdVS. Our data highlight quantitative facial phenotyping not only as a powerful tool to distinguish syndromes with overlapping facial dysmorphisms but also to establish pathogenicity of variants of unknown clinical significance.
- Published
- 2021
23. DLG4-related synaptopathy: a new rare brain disorder
- Author
-
Edgard Verdura, Alex MacKenzie, Rolph Pfundt, Tobias B. Haack, Ange Line Bruel, Paulino Gómez-Puertas, Anna C.E. Hurst, Bert B.A. de Vries, Stella A. de Man, Maria Johansson Soller, Bregje W.M. van Bon, Elisabeth Sarrazin, Agustí Rodríguez-Palmero, Stephan Waldmüller, Melanie O’Leary, Anne Sophie Denommé-Pichon, Bitten Schönewolf-Greulich, Joseph T. Shieh, V. A. Bjerregaard, Vahid Bahrambeigi, Malin Kvarnung, Agatha Schlüter, Anne Marie Bisgaard, Ingrid M.B.H. van de Laar, Elisa Giorgio, Lars Feuk, Mieke M. van Haelst, Thomas D. Challman, Ineke van de Burgt, Sulagna Kushary, Simone F. Reiter, David B. Everman, Zeynep Tümer, Giorgia Mandrile, Conny M. A. van Ravenswaaij-Arts, Charles Shaw-Smith, Juliane Hoyer, Chad R. Haldeman-Englert, Lotte Kleinendorst, Bryce A. Mendelsohn, Anna Lindstrand, Christine Coubes, Gea Beunders, Sixto García-Miñaur, Antonio Vitobello, Melissa Maria Boerrigter, Alysia Kern Lovgren, Anya Revah-Politi, Carlos E. Prada, Bertrand Isidor, Elena Repnikova, Stephanie Spranger, Esmée van Drie, Frédéric Tran Mau-Them, Zohra Shad, Ben Pode-Shakked, Aurora Pujol, Christiane Zweier, Bjørn Ivar Haukanes, David Gómez-Andrés, Kathleen A. Leppig, Marta Pacio-Míguez, Motti Shohat, Yuval Landau, Benjamin Cogné, Frances Elmslie, Kimberly A. Aldinger, Anita Rauch, Juliann M. Savatt, Nicolas Gruchy, Sharon Whiting, William B. Dobyns, Thomas J. Dye, Sebastien Moutton, Heidi Thiese, Setareh Moghadasi, Iñigo Marcos-Alcalde, Jenny Morton, Sumit Parikh, María Palomares-Bralo, Stéphanie Arpin, Tracy S. Gertler, Meredith J. Ross, Bernt Popp, Amelie J. Müller, Claudia A. L. Ruivenkamp, Institut d'Investigació Biomèdica de Bellvitge [Barcelone] (IDIBELL), CIBER de Enfermedades Raras (CIBERER), Hospital Universitario Germans Trias I Pujol, Vall d'Hebron University Hospital [Barcelona], Center for Integrative Brain Research [Seattle, WA, USA], University of Washington [Seattle]-Seattle Children's Research Institute, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Service de génétique [Tours], Centre Hospitalier Régional Universitaire de Tours (CHRU Tours)-Hôpital Bretonneau, Department of Molecular and Human Genetics (Baylor College of Medicine), Baylor College of Medecine, Biologie, génétique et thérapies ostéoarticulaires et respiratoires (BIOTARGEN), Université de Caen Normandie (UNICAEN), Normandie Université (NU)-Normandie Université (NU), Hôpital Bretonneau-Centre Hospitalier Régional Universitaire de Tours (CHRU Tours), Clinical Genetics, Human Genetics, Graduate School, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, ARD - Amsterdam Reproduction and Development, Clinical Cognitive Neuropsychiatry Research Program (CCNP), Human genetics, Amsterdam Neuroscience - Complex Trait Genetics, and Amsterdam Reproduction & Development (AR&D)
- Subjects
0301 basic medicine ,Autism Spectrum Disorder ,[SDV]Life Sciences [q-bio] ,030105 genetics & heredity ,Biology ,03 medical and health sciences ,Intellectual Disability ,Intellectual disability ,medicine ,Missense mutation ,Humans ,Global developmental delay ,Exome ,Genetics (clinical) ,Genetics ,Brain Diseases ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Brain ,medicine.disease ,030104 developmental biology ,Phenotype ,Renal disorders Radboud Institute for Molecular Life Sciences [Radboudumc 11] ,Autism spectrum disorder ,Neurodevelopmental Disorders ,Synaptopathy ,DLG4 ,Postsynaptic density ,Disks Large Homolog 4 Protein - Abstract
Contains fulltext : 245031.pdf (Publisher’s version ) (Closed access) PURPOSE: Postsynaptic density protein-95 (PSD-95), encoded by DLG4, regulates excitatory synaptic function in the brain. Here we present the clinical and genetic features of 53 patients (42 previously unpublished) with DLG4 variants. METHODS: The clinical and genetic information were collected through GeneMatcher collaboration. All the individuals were investigated by local clinicians and the gene variants were identified by clinical exome/genome sequencing. RESULTS: The clinical picture was predominated by early onset global developmental delay, intellectual disability, autism spectrum disorder, and attention deficit-hyperactivity disorder, all of which point to a brain disorder. Marfanoid habitus, which was previously suggested to be a characteristic feature of DLG4-related phenotypes, was found in only nine individuals and despite some overlapping features, a distinct facial dysmorphism could not be established. Of the 45 different DLG4 variants, 39 were predicted to lead to loss of protein function and the majority occurred de novo (four with unknown origin). The six missense variants identified were suggested to lead to structural or functional changes by protein modeling studies. CONCLUSION: The present study shows that clinical manifestations associated with DLG4 overlap with those found in other neurodevelopmental disorders of synaptic dysfunction; thus, we designate this group of disorders as DLG4-related synaptopathy.
- Published
- 2021
24. Delineating the molecular and phenotypic spectrum of the SETD1B-related syndrome
- Author
-
Reshmi Ramakrishnan, Catherine Quindipan, Claudia A. L. Ruivenkamp, Nicholas M. Allen, Mashaya Zaman, Daniela Q.C.M. Barge-Schaapveld, Annalisa Vetro, Stephanie Efthymiou, James R. Lupski, Kara C. Klemp, Zou Pan, Adam Jackson, Marielle E. van Gijn, Joshua Scheck, Marielle Alders, Mariet W. Elting, Karla A. Peña-Guerra, Stephen R. Braddock, Rolph Pfundt, Ivan K. Chinn, Lin Yang, Lauren Schenck, Xiaodong Wang, Melissa Lees, Houda Zghal Elloumi, Shehla Mohammed, Sally Ann Lynch, Henry Houlden, Jennifer Keller-Ramey, Stefan T. Arold, Anneke Kievit, Jefferey McGlothlin, Marjon van Slegtenhorst, Marjolein H. Willemsen, Hannah K. Robinson, Bert B.A. de Vries, Irma Järvelä, Kelly J. Cardona-Londoño, Yolande van Bever, Abeltje M. Polstra, Neda Mazaheri, Barbara W. van Paassen, Maura R.Z. Ruzhnikov, Lewis Pang, Theresa Mihalic Mosher, J. Lawrence Merritt, Jing Peng, Sadegheh Haghshenas, Amy Crunk, Christian Gilissen, Fleur Vansenne, Cacha M.P.C.D. Peeters-Scholte, Richard E. Person, Hamid Galehdari, Leena Lauronen, Abbey M. Putnam, Jennifer Kerkhof, Matthew Pastore, Angela Sun, Caroline M. Kehoe, Alexandra Garza-Flores, Julia Baptista, Martino Montomoli, Selina H. Banu, Tahsin Stefan Barakat, Adi Reich, Luis Alberto Pedroza, Laurence E. Walsh, Renzo Guerrini, Ghayda M. Mirzaa, Peter D. Turnpenny, J. Austin Hamm, Xi Lin, Kristina Lanko, Reza Maroofian, Tuomo Määttä, Yana Lara-Taranchenko, Kim L. McBride, Jenny Thies, Andrew E. Timms, Shaoping Huang, Suzanne M. Leal, Daniel C. Koboldt, Rebecca Baud, Gretchen E. Rosso, Haley McConkey, Matthew A. Deardorff, Marjolein J.A. Weerts, Francisco J. Guzmán-Vega, Tamison Jewett, Siddharth Banka, Kristin G. Monaghan, Isabelle Schrauwen, Bekim Sadikovic, Sanjay M. Sisodiya, Victoria Harrison, and Susanne Koning
- Subjects
Genetics ,0303 health sciences ,Language delay ,Mechanism (biology) ,Biology ,medicine.disease ,Penetrance ,03 medical and health sciences ,0302 clinical medicine ,Neurodevelopmental disorder ,Intellectual disability ,medicine ,Autism ,Global developmental delay ,Epigenetics ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Pathogenic variants in SETD1B have been associated with a syndromic neurodevelopmental disorder including intellectual disability, language delay and seizures. To date, clinical features have been described for eleven patients with (likely) pathogenic SETD1B sequence variants. We perform an in-depth clinical characterization of a cohort of 36 unpublished individuals with SETD1B sequence variants, describing their molecular and phenotypic spectrum. Selected variants were functionally tested using in vitro and genome-wide methylation assays. Our data present evidence for a loss-of-function mechanism of SETD1B variants, resulting in a core clinical phenotype of global developmental delay, language delay including regression, intellectual disability, autism and other behavioral issues, and variable epilepsy phenotypes. Developmental delay appeared to precede seizure onset, suggesting SETD1B dysfunction impacts physiological neurodevelopment even in the absence of epileptic activity. Interestingly, males are significantly overrepresented and more severely affected, and we speculate that sex-linked traits could affect susceptibility to penetrance and the clinical spectrum of SETD1B variants. Finally, despite the possibility of non-redundant contributions of SETD1B and its paralogue SETD1A to epigenetic control, the clinical phenotypes of the related disorders share many similarities, indicating that elucidating shared and divergent downstream targets of both genes will help to understand the mechanism leading to the neurobehavioral phenotypes. Insights from this extensive cohort will facilitate the counseling regarding the molecular and phenotypic landscape of newly diagnosed patients with the SETD1B-related syndrome.
- Published
- 2021
25. TMEM218 dysfunction causes ciliopathies, including Joubert and Meckel syndromes
- Author
-
Henry Houlden, Bert B.A. de Vries, Eva Pajkrt, Julie C. Van De Weghe, Caroline C W Klaver, Michael J. Bamshad, Inge B. Mathijssen, Majid Mojarrad, Deborah A. Nickerson, Jennifer C. Dempsey, Elizabeth van Leeuwen, Atieh Eslahi, Dan Doherty, Reza Maroofian, Mahsa Mohajeri, Dorien Lugtenberg, Caitlin V. Miller, Vimla Aggarwal, Aoife M. Waters, Jessica Giordano, Human genetics, Obstetrics and gynaecology, Human Genetics, Obstetrics and Gynaecology, Amsterdam Reproduction & Development (AR&D), APH - Personalized Medicine, APH - Quality of Care, and Ophthalmology
- Subjects
Meckel syndrome ,Biology ,QH426-470 ,Ciliopathies ,Joubert syndrome ,Sensory disorders Donders Center for Medical Neuroscience [Radboudumc 12] ,Article ,primary cilia ,medicine ,Genetics ,retinal dystrophy ,Exome ,TMEM218 ,Genetics (clinical) ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Polydactyly ,Cilium ,cilia ,Ciliary transition zone ,medicine.disease ,Ciliopathy ,ciliopathy ,Molecular Medicine - Abstract
Contains fulltext : 235063.pdf (Publisher’s version ) (Open Access) The Joubert-Meckel syndrome spectrum is a continuum of recessive ciliopathy conditions caused by primary cilium dysfunction. The primary cilium is a microtubule-based, antenna-like organelle that projects from the surface of most human cell types, allowing them to respond to extracellular signals. The cilium is partitioned from the cell body by the transition zone, a known hotspot for ciliopathy-related proteins. Despite years of Joubert syndrome (JBTS) gene discovery, the genetic cause cannot be identified in up to 30% of individuals with JBTS, depending on the cohort, sequencing method, and criteria for pathogenic variants. Using exome and targeted sequencing of 655 families with JBTS, we identified three individuals from two families harboring biallelic, rare, predicted-deleterious missense TMEM218 variants. Via MatchMaker Exchange, we identified biallelic TMEM218 variants in four additional families with ciliopathy phenotypes. Of note, four of the six families carry missense variants affecting the same highly conserved amino acid position 115. Clinical features included the molar tooth sign (N = 2), occipital encephalocele (N = 5, all fetuses), retinal dystrophy (N = 4, all living individuals), polycystic kidneys (N = 2), and polydactyly (N = 2), without liver involvement. Combined with existing functional data linking TMEM218 to ciliary transition zone function, our human genetic data make a strong case for TMEM218 dysfunction as a cause of ciliopathy phenotypes including JBTS with retinal dystrophy and Meckel syndrome. Identifying all genetic causes of the Joubert-Meckel spectrum enables diagnostic testing, prognostic and recurrence risk counseling, and medical monitoring, as well as work to delineate the underlying biological mechanisms and identify targets for future therapies.
- Published
- 2021
26. Alternative genomic diagnoses for individuals with a clinical diagnosis of Dubowitz syndrome
- Author
-
Sixto Garcia Minaur, Pankaj B. Agrawal, A. Micheil Innes, Catherine A. Brownstein, David S. Wargowski, Brenda McInnes, Isaac Wong, Albert E. Chudley, Jennifer E. Posey, Francesc López-Giráldez, Ping-Yee B Au, Alper Gezdirici, Kyrieckos A. Aleck, Eric Boerwinkle, Paolo Prontera, Bilgen Bilge Geçkinli, Yeting Zhang, An Nguyen, David A. Dyment, Jukka S. Moilanen, Alan H. Beggs, Nara Sobreira, Hatip Aydin, Elizabeth E. Blue, Kathryn Dunn, Gerald F. Cox, Bernard N. Chodirker, Harrison Brand, Jinchuan Xing, Hind Al Sharhan, Bert B.A. de Vries, Maria Juliana Rodovalho Doriqui, Davut Pehlivan, Shalini N. Jhangiani, Centers for Mendelian Genomics, Katrin Õunap, Cheryl R. Greenberg, Kaya Bilguvar, Carol L. Clericuzio, Cynthia J. Curry, Taila Hartley, Julie Lauzon, Michael J. Bamshad, Timothy Poterba, R. Brian Lowry, Jill A. Fahrner, Cullen M. Dutmer, M. E. Suzanne Lewis, Steve Buyske, Ender Karaca, Aziz Mhanni, William T. Gibson, Valentina Stanley, April Hall, Elke de Boer, Kristin D. Kernohan, Joseph G. Gleeson, P. Dane Witmer, Jungmin Choi, Danny Antaki, Małgorzata J.M. Nowaczyk, Sander Pajusalu, Anne H. O’Donnell-Luria, Sarah L. Sawyer, Zeynep Coban Akdemir, Tara C. Matise, Jennifer McEvoy-Venneri, Casie A. Genetti, Kym M. Boycott, Lynette S. Penney, Ada Hamosh, Eleina M. England, Deniz Torun, Maha S. Zaki, Deborah A. Nickerson, Dyment, David A., O'Donnell-Luria, Anne, Agrawal, Pankaj B., Coban Akdemir, Zeynep, Aleck, Kyrieckos A., Antaki, Danny, Al Sharhan, Hind, Au, Ping-Yee B., Aydin, Hatip, Beggs, Alan H., Bilguvar, Kaya, Boerwinkle, Eric, Brand, Harrison, Brownstein, Catherine A., Buyske, Steve, Chodirker, Bernard, Choi, Jungmin, Chudley, Albert E., Clericuzio, Carol L., Cox, Gerald F., Curry, Cynthia, de Boer, Elke, de Vries, Bert B. A., Dunn, Kathryn, Dutmer, Cullen M., England, Eleina M., Fahrner, Jill A., Geckinli, Bilgen B., Genetti, Casie A., Gezdirici, Alper, Gibson, William T., Gleeson, Joseph G., Greenberg, Cheryl R., Hall, April, Hamosh, Ada, Hartley, Taila, Jhangiani, Shalini N., Karaca, Ender, Kernohan, Kristin, Lauzon, Julie L., Lewis, M. E. Suzanne, Lowry, R. Brian, Lopez-Giraldez, Francesc, Matise, Tara C., McEvoy-Venneri, Jennifer, McInnes, Brenda, Mhanni, Aziz, Garcia Minaur, Sixto, Moilanen, Jukka, Nguyen, An, Nowaczyk, Malgorzata J. M., Posey, Jennifer E., Ounap, Katrin, Pehlivan, Davut, Pajusalu, Sander, Penney, Lynette S., Poterba, Timothy, Prontera, Paolo, Doriqui, Maria Juliana Rodovalho, Sawyer, Sarah L., Sobreira, Nara, Stanley, Valentina, Torun, Deniz, Wargowski, David, Witmer, P. Dane, Wong, Isaac, Xing, Jinchuan, Zaki, Maha S., Zhang, Yeting, Boycott, Kym M., Bamshad, Michael J., Nickerson, Deborah A., Blue, Elizabeth E., and Innes, A. Micheil
- Subjects
0301 basic medicine ,Male ,ANOMALIES ,INTELLECTUAL DISABILITY ,Eczema ,030105 genetics & heredity ,PHENOTYPE ,genetic heterogeneity ,Locus heterogeneity ,Dubowitz syndrome ,Exome ,Child ,Genetics (clinical) ,Exome sequencing ,Growth Disorders ,Genetics ,FRAMESHIFT ,Genomics ,3. Good health ,VPS13B ,genome sequencing ,LOSS-OF-FUNCTION ,Child, Preschool ,symbols ,Microcephaly ,Female ,microarray ,Adolescent ,DNA Copy Number Variations ,Biology ,NSUN2 ,PATIENT ,DNA sequencing ,Histone Deacetylases ,Article ,03 medical and health sciences ,symbols.namesake ,medicine ,Humans ,Genetic Predisposition to Disease ,ANEMIA ,Genetic heterogeneity ,Genome, Human ,MUTATIONS ,Facies ,Infant ,PLATFORM ,medicine.disease ,Repressor Proteins ,030104 developmental biology ,Mendelian inheritance ,exome sequencing - Abstract
Dubowitz syndrome (DubS) is considered a recognizable syndrome characterized by a distinctive facial appearance and deficits in growth and development. There have been over 200 individuals reported with Dubowitz or a "Dubowitz-like" condition, although no single gene has been implicated as responsible for its cause. We have performed exome (ES) or genome sequencing (GS) for 31 individuals clinically diagnosed with DubS. After genome-wide sequencing, rare variant filtering and computational and Mendelian genomic analyses, a presumptive molecular diagnosis was made in 13/27 (48%) families. The molecular diagnoses included biallelic variants in SKIV2L, SLC35C1, BRCA1, NSUN2; de novo variants in ARID1B, ARID1A, CREBBP, POGZ, TAF1, HDAC8, and copy-number variation at1p36.11(ARID1A), 8q22.2(VPS13B), Xp22, and Xq13(HDAC8). Variants of unknown significance in known disease genes, and also in genes of uncertain significance, were observed in 7/27 (26%) additional families. Only one gene, HDAC8, could explain the phenotype in more than one family (N = 2). All but two of the genomic diagnoses were for genes discovered, or for conditions recognized, since the introduction of next-generation sequencing. Overall, the DubS-like clinical phenotype is associated with extensive locus heterogeneity and the molecular diagnoses made are for emerging clinical conditions sharing characteristic features that overlap the DubS phenotype.
- Published
- 2021
27. Genetic convergence of developmental and epileptic encephalopathies and intellectual disability
- Author
-
Bert B.A. de Vries, Gemma L. Carvill, Sandra Jansen, Ingrid E. Scheffer, Matthew Zemel, Heather C Mefford, Han G. Brunner, Michele G. Mehaffey, Amy Lacroix, Lisenka E.L.M. Vissers, and Petra de Vries
- Subjects
Male ,medicine.medical_specialty ,Adolescent ,Receptors, N-Methyl-D-Aspartate ,Epilepsy ,Developmental Neuroscience ,Intellectual Disability ,Intellectual disability ,medicine ,Humans ,Exome ,Child ,Psychiatry ,NAV1.2 Voltage-Gated Sodium Channel ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Genetic heterogeneity ,business.industry ,Infant ,Odds ratio ,medicine.disease ,Phenotype ,Clinical research ,Mutation ,Pediatrics, Perinatology and Child Health ,Cohort ,Autism ,Female ,Neurology (clinical) ,Convergence (relationship) ,business ,Spasms, Infantile - Abstract
Contains fulltext : 243998.pdf (Publisher’s version ) (Open Access) AIM: To determine whether genes that cause developmental and epileptic encephalopathies (DEEs) are more commonly implicated in intellectual disability with epilepsy as a comorbid feature than in intellectual disability only. METHOD: We performed targeted resequencing of 18 genes commonly implicated in DEEs in a cohort of 830 patients with intellectual disability (59% male) and 393 patients with DEEs (52% male). RESULTS: We observed a significant enrichment of pathogenic/likely pathogenic variants in patients with epilepsy and intellectual disability (16 out of 159 in seven genes) compared with intellectual disability only (2 out of 671) (p
- Published
- 2021
28. Disruption of RFX family transcription factors causes autism, attention-deficit/hyperactivity disorder, intellectual disability, and dysregulated behavior
- Author
-
Tomi L. Toler, Timothy W. Yu, William B. Dobyns, Marcia C. Willing, Karen W. Gripp, Rolph Pfundt, Muhammad Iqbal, Xiadong Wang, Lance H. Rodan, Ada Hamosh, Cynthia S. Gubbels, Janice Baker, Thatjana Gardeitchik, Jenny Lai, André Reis, Fleur Vansenne, Jennifer E. Posey, Paranchai Boonsawat, Mathilde Nizon, Sébastien Küry, Jill R. Murrell, Julian L. Ambrus, Yunhong Wu, Laura A. Baker, Aubrie Soucy, Severine Audebert-Bellanger, Ellen van Binsbergen, Thomas Courtin, Guiseppe Zampino, Caleb P. Bupp, Holly K. Harris, Alan H. Beggs, Giulia Pascolini, Catharina (Nienke) M.L. Volker-Touw, Bert B.A. de Vries, Casie A. Genetti, La Donna L. Immken, Paola Grammatico, Martin Jakob Larsen, Sylvia Redon, Kévin Uguen, Reza Asadollahi, Madeleine Fannemel, Catherine Buchanan, Boris Keren, George E. Tiller, Lilian L. Cohen, Tojo Nakayama, Laurence E. Walsh, Iqra Ghulam Rasool, Audrey Labalme, Koen L.I. van Gassen, Pankaj B. Agrawal, Boxun Zhao, Gaetan Lesca, Steffan Syrbe, Kimberly A. Aldinger, Emanuele Agolini, Maria Kibaek, Muhammad Yasir Zahoor, Peter D. Turnpenny, Antonio Novelli, Ines Brösse, Claude Férec, Jorune Balciuniene, Nikoleta Argyrou, Victoria Suslovitch, Alice Poisson, Anita Rauch, Katelyn Payne, Christina Fagerberg, Cyril Mignot, Christopher Gray, Anne Blomhoff, Carolyn D. Applegate, Cornelia Kraus, Rami Abou Jamra, Marleen Simon, Martin Broly, Cara M. Skraban, and Emily Fassi
- Subjects
Adult ,0301 basic medicine ,Autism Spectrum Disorder ,Regulatory Factor X Transcription Factors ,030105 genetics & heredity ,Biology ,Article ,FRX ,autism ,intellectual disability ,03 medical and health sciences ,Intellectual Disability ,Ciliogenesis ,Intellectual disability ,mental disorders ,medicine ,Humans ,Attention deficit hyperactivity disorder ,Autistic Disorder ,Gene ,Genetics (clinical) ,Genetics ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,medicine.disease ,Phenotype ,Renal disorders Radboud Institute for Molecular Life Sciences [Radboudumc 11] ,030104 developmental biology ,Attention Deficit Disorder with Hyperactivity ,Autism spectrum disorder ,Autism ,RFX3 ,Transcription Factors - Abstract
Contains fulltext : 234024.pdf (Publisher’s version ) (Closed access) PURPOSE: We describe a novel neurobehavioral phenotype of autism spectrum disorder (ASD), intellectual disability, and/or attention-deficit/hyperactivity disorder (ADHD) associated with de novo or inherited deleterious variants in members of the RFX family of genes. RFX genes are evolutionarily conserved transcription factors that act as master regulators of central nervous system development and ciliogenesis. METHODS: We assembled a cohort of 38 individuals (from 33 unrelated families) with de novo variants in RFX3, RFX4, and RFX7. We describe their common clinical phenotypes and present bioinformatic analyses of expression patterns and downstream targets of these genes as they relate to other neurodevelopmental risk genes. RESULTS: These individuals share neurobehavioral features including ASD, intellectual disability, and/or ADHD; other frequent features include hypersensitivity to sensory stimuli and sleep problems. RFX3, RFX4, and RFX7 are strongly expressed in developing and adult human brain, and X-box binding motifs as well as RFX ChIP-seq peaks are enriched in the cis-regulatory regions of known ASD risk genes. CONCLUSION: These results establish a likely role of deleterious variation in RFX3, RFX4, and RFX7 in cases of monogenic intellectual disability, ADHD and ASD, and position these genes as potentially critical transcriptional regulators of neurobiological pathways associated with neurodevelopmental disease pathogenesis.
- Published
- 2021
29. Sonlicromanol improves neuronal network dysfunction and transcriptome changes linked to m.3243A>G heteroplasmy in iPSC-derived neurons
- Author
-
Teun M. Klein Gunnewiek, Jan A.M. Smeitink, Chantal Schoenmaker, Herma Renkema, Bert B.A. de Vries, Nael Nadif Kasri, Julien Beyrath, Iris Pelgrim, Peter-Bram A.C. ’t Hoen, Anouk H.A. Verboven, Tamas Kozicz, and Mark Hogeweg
- Subjects
Nervous system ,Mitochondrial encephalomyopathy ,RNA, Transfer, Leu ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Stress-related disorders Donders Center for Medical Neuroscience [Radboudumc 13] ,neurons ,Mitochondrion ,Biology ,Heteroplasmy ,DNA, Mitochondrial ,Biochemistry ,Article ,iPSC-derived neurons ,Transcriptome ,Mitochondrial Encephalomyopathies ,Genetics ,medicine ,Animals ,Humans ,sonlicromanol ,Genetic Predisposition to Disease ,Chromans ,Induced pluripotent stem cell ,Gene ,Cells, Cultured ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Gene Expression Profiling ,Cell Differentiation ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Cell Biology ,medicine.disease ,Rats ,Cell biology ,mitochondria ,Disease Models, Animal ,Phenotype ,medicine.anatomical_structure ,Gene Expression Regulation ,Astrocytes ,Lactic acidosis ,MELAS ,micro-electrode arrays ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,Developmental Biology - Abstract
Summary Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) is often caused by an adenine to guanine variant at m.3243 (m.3243A>G) of the MT-TL1 gene. To understand how this pathogenic variant affects the nervous system, we differentiated human induced pluripotent stem cells (iPSCs) into excitatory neurons with normal (low heteroplasmy) and impaired (high heteroplasmy) mitochondrial function from MELAS patients with the m.3243A>G pathogenic variant. We combined micro-electrode array (MEA) measurements with RNA sequencing (MEA-seq) and found reduced expression of genes involved in mitochondrial respiration and presynaptic function, as well as non-cell autonomous processes in co-cultured astrocytes. Finally, we show that the clinical phase II drug sonlicromanol can improve neuronal network activity when treatment is initiated early in development. This was intricately linked with changes in the neuronal transcriptome. Overall, we provide insight in transcriptomic changes in iPSC-derived neurons with high m.3243A>G heteroplasmy, and show the pathology is partially reversible by sonlicromanol., Highlights • High m.3243A>G heteroplasmy leads to neuronal network dysfunction • Expression of genes involved in mitochondrial and synaptic function are affected • Gene expression in co-cultured rat astrocytes is non-cell autonomously affected • Sonlicromanol partially rescues the network activity and transcriptome changes, Using human induced pluripotent stem cell-derived neurons with high levels of m.3243A>G heteroplasmy, Klein Gunnewiek et al. report on transcriptome changes underlying the functional neuronal network phenotype and show that sonlicromanol can partially improve both the neuronal network phenotype and the transcriptome changes.
- Published
- 2021
30. De Novo variants in EEF2 cause a neurodevelopmental disorder with benign external hydrocephalus
- Author
-
Rolph Pfundt, Jonathan D. Dinman, Arjan P.M. de Brouwer, Grace Yoon, Maria J. Nabais Sá, Alexandra Schneider, Michèl A.A.P. Willemsen, Graeme A.M. Nimmo, Alexandra N. Olson, Christopher M. Gomez, Bert B.A. de Vries, and Francisca Millan
- Subjects
Elongation Factor 2 Kinase ,Male ,Heterozygote ,Biology ,EEF2 ,03 medical and health sciences ,Neurodevelopmental disorder ,Exome Sequencing ,Genetics ,medicine ,Humans ,Missense mutation ,Exome ,Child ,Molecular Biology ,Gene ,Genetics (clinical) ,Exome sequencing ,030304 developmental biology ,0303 health sciences ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Cerebellar ataxia ,030305 genetics & heredity ,Translation (biology) ,General Medicine ,medicine.disease ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Phenotype ,Neurodevelopmental Disorders ,Child, Preschool ,Mutation ,General Article ,medicine.symptom ,Hydrocephalus ,Ventriculomegaly - Abstract
Contains fulltext : 231523.pdf (Publisher’s version ) (Closed access) Eukaryotic translation elongation factor 2 (eEF2) is a key regulatory factor in gene expression that catalyzes the elongation stage of translation. A functionally impaired eEF2, due to a heterozygous missense variant in the EEF2 gene, was previously reported in one family with spinocerebellar ataxia-26 (SCA26), an autosomal dominant adult-onset pure cerebellar ataxia. Clinical exome sequencing identified de novo EEF2 variants in three unrelated children presenting with a neurodevelopmental disorder (NDD). Individuals shared a mild phenotype comprising motor delay and relative macrocephaly associated with ventriculomegaly. Populational data and bioinformatic analysis underscored the pathogenicity of all de novo missense variants. The eEF2 yeast model strains demonstrated that patient-derived variants affect cellular growth, sensitivity to translation inhibitors and translational fidelity. Consequently, we propose that pathogenic variants in the EEF2 gene, so far exclusively associated with late-onset SCA26, can cause a broader spectrum of neurologic disorders, including childhood-onset NDDs and benign external hydrocephalus.
- Published
- 2021
31. A case series of familial ARID1B variants illustrating variable expression and suggestions to update the ACMG criteria
- Author
-
Pleuntje J. van der Sluijs, Mariëlle Alders, Alexander J.M. Dingemans, Kareesma Parbhoo, Bregje W. van Bon, Jennifer C. Dempsey, Dan Doherty, Johan T. Den Dunnen, Erica H. Gerkes, Ilana M. Milller, Stephanie Moortgat, Debra S. Regier, Claudia Ruivenkamp, Betsy Schmalz, Thomas Smol, K.E. (Kyra) Stuurman, Catherine Vincent-Delorme, Bert B.A. de Vries, Bekim Sadikovic, Scott E. Hickey, Jill A. Rosenfeld, Isabelle Maystadt, Gijs W.E. Santen, Pleuntje J. van der Sluijs, Mariëlle Alders, Alexander J.M. Dingemans, Kareesma Parbhoo, Bregje W. van Bon, Jennifer C. Dempsey, Dan Doherty, Johan T. Den Dunnen, Erica H. Gerkes, Ilana M. Milller, Stephanie Moortgat, Debra S. Regier, Claudia Ruivenkamp, Betsy Schmalz, Thomas Smol, K.E. (Kyra) Stuurman, Catherine Vincent-Delorme, Bert B.A. de Vries, Bekim Sadikovic, Scott E. Hickey, Jill A. Rosenfeld, Isabelle Maystadt, and Gijs W.E. Santen
- Abstract
ARID1B is one of the most frequently mutated genes in intellectual disability (~1%). Most variants are readily classified, since they are de novo and are predicted to lead to loss of function, and therefore classified as pathogenic according to the American College of Medical Genetics and Genomics (ACMG) guidelines for the interpretation of sequence variants. However, familial loss-of-function variants can also occur and can be challenging to interpret. Such variants may be pathogenic with variable expression, causing only a mild phenotype in a parent. Alternatively, since some regions of the ARID1B gene seem to be lacking pathogenic variants, loss-of-function variants in those regions may not lead to ARID1B haploinsufficiency and may therefore be benign. We describe 12 families with potential loss-of-function variants, which were either familial or with unknown inheritance and were in regions where pathogenic variants have not been described or are otherwise challenging to interpret. We performed detailed clinical and DNA methylation studies, which allowed us to confidently classify most variants. In five families we observed transmission of pathogenic variants, confirming their highly variable expression. Our findings provide further evidence for an alternative translational start site and we suggest updates for the ACMG guidelines for the interpretation of seque
- Published
- 2021
- Full Text
- View/download PDF
32. Sonlicromanol improves neuronal network dysfunction and transcriptome changes linked to m.3243A>G heteroplasmy in iPSC-derived neurons
- Author
-
Peter A C 't Hoen, Tamas Kozicz, Teun M. Klein Gunnewiek, Chantal Schoenmaker, Nael Nadif Kasri, Jan A.M. Smeitink, Herma Renkema, Anouk H.A. Verboven, Bert B.A. de Vries, Julien Beyrath, and Mark Hogeweg
- Subjects
Mitochondrial encephalomyopathy ,Nervous system ,Transcriptome ,medicine.anatomical_structure ,Lactic acidosis ,Mutation (genetic algorithm) ,medicine ,Biological neural network ,Biology ,medicine.disease ,Gene ,Heteroplasmy ,Cell biology - Abstract
SummaryMitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes (MELAS) is often caused by an adenine to guanine mutation at m.3243 (m.3243A>G) of the MT-TL1 gene (tRNAleu(UUR)). To understand how this mutation affects the nervous system, we differentiated human induced-pluripotent stem cells (iPSCs) into excitatory neurons with normal (low heteroplasmy) and impaired (high heteroplasmy) mitochondrial function from MELAS patients with the m.3243A>G mutation. We combined micro-electrode array (MEA) measurements with RNA sequencing (MEA-seq) and found that the m.3243A>G mutation affects expression of genes involved in mitochondrial respiration- and presynaptic function, as well as non-cell autonomous processes in co-cultured astrocytes. Finally, we show that the clinical II stage drug sonlicromanol (KH176) improved neuronal network activity in a patient-specific manner when treatment is initiated early in development. This was intricately linked with changes in the neural transcriptome. Overall, MEA-seq is a powerful approach to identify mechanisms underlying the m.3243A>G mutation and to study the effect of pharmacological interventions in iPSC-derived neurons.Highlights- High m.3243A>G heteroplasmy leads to lower neuronal network activity and synchronicity- High heteroplasmy affects expression of genes involved in mitochondrial ATP production and the synaptic function / the presynaptic vesicle cycle- High neuronal heteroplasmy non cell autonomously affects gene expression in healthy co-cultured astrocytes- Sonlicromanol partially rescues neuronal network activity and transcriptome changes induced by high heteroplasmyeTOC BlurbUsing human inducible pluripotent stem cell-derived neurons with high levels of m.3243A>G heteroplasmy, Klein Gunnewiek et al. show transcriptome changes underlying the functional neuronal network phenotype, and how sonlicromanol can partially improve both this neuronal network phenotype, and the transcriptome changes, in a patient-specific manner.
- Published
- 2020
33. KANSL1 Deficiency Causes Neuronal Dysfunction by Oxidative Stress-Induced Autophagy
- Author
-
Lynn Devilee, Giuseppe Testa, Dirk Schubert, EIly I. Lewerissa, Nael Nadif Kasri, Monica Frega, Edda Ulferts, Michele Gabriele, Hans van Bokhoven, Katrin Linda, Chantal Schoenmaker, Teun M. Klein Gunnewiek, Anouk H.A. Verboven, Bert B.A. de Vries, David A. Koolen, and Astrid R. Oudakker
- Subjects
Autophagosome ,chemistry.chemical_classification ,Reactive oxygen species ,biology ,Chemistry ,Autophagy ,medicine.disease_cause ,Cell biology ,Synapse ,Superoxide dismutase ,medicine.anatomical_structure ,Lysosome ,medicine ,biology.protein ,Oxidative stress ,PI3K/AKT/mTOR pathway - Abstract
Autophagy is a finely tuned process of programmed degradation and recycling of proteins and cellular components, which is crucial in neuronal function and synaptic integrity. Mounting evidence implicates chromatin remodelling in fine-tuning autophagy pathways. However, this epigenetic regulation is poorly understood in neurons. Here, we investigate the role in autophagy of KANSL1, a member of the nonspecific lethal complex, which acetylates histone H4 on lysine 16 (H4K16ac) to facilitate transcriptional activation. Loss-of-function of KANSL1 is strongly associated with the neurodevelopmental disorder Koolen-de Vries Syndrome (KdVS).Starting from KANSL1-deficient human induced-pluripotent stem cells, both from KdVS patients and genome-edited lines, we identified superoxide dismutase 1, an antioxidant enzyme, to be significantly decreased, leading to a subsequent increase in oxidative stress and autophagosome accumulation. In KANSL1-deficient neurons, autophagosome accumulation at excitatory synapses resulted in reduced synaptic density, reduced AMPA receptor-mediated transmission and impaired neuronal network activity. Furthermore, we found that increased oxidative stress-mediated autophagosome accumulation leads to increased mTOR activation and decreased lysosome function, further preventing the clearing of autophagosomes. Finally, by pharmacologically reducing oxidative stress, we could rescue the aberrant autophagosome formation as well as synaptic and neuronal network activity in KANSL1-deficient neurons. Our findings thus point towards an important relation between oxidative stress-induced autophagy and synapse function, and demonstrate the importance of H4K16ac-mediated changes in chromatin structure to balance reactive oxygen species- and mTOR-dependent autophagy.
- Published
- 2020
34. De novo CLTC variants are associated with a variable phenotype from mild to severe intellectual disability, microcephaly, hypoplasia of the corpus callosum, and epilepsy
- Author
-
Michael C. Kruer, Hanka Venselaar, Arjan P.M. de Brouwer, Eulalie Lasseaux, Rolph Pfundt, Emilia K. Bijlsma, Marieke F. van Dooren, Amélie Piton, Christian Gilissen, Maria J. Nabais Sá, Sophie Naudion, Renzo Guerrini, Bert B.A. de Vries, Didier Lacombe, André Reis, Michèl A.A.P. Willemsen, Laurens Wiel, Elisabeth Gabau, Annalisa Vetro, Regina Trollmann, Anna Ruiz, Catherine Vincent-Delorme, David A. Koolen, Aurélien Trimouille, Christiane Zweier, Monica S Cooper, David J. Amor, Tahsin Stefan Barakat, Somayeh Bakhtiari, Marjon van Slegtenhorst, Clinical Genetics, Radboud University Medical Center [Nijmegen], Radboud Institute for Molecular Life Sciences [Nijmegen, the Netherlands], Radboud Institute for Molecular Life Sciences (RIMLS), CHU Bordeaux [Bordeaux], Laboratoire Maladies Rares: Génétique et Métabolisme (Bordeaux) (U1211 INSERM/MRGM), Université de Bordeaux (UB)-Groupe hospitalier Pellegrin-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Hôpital Jeanne de Flandre [Lille], Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Universitat Autònoma de Barcelona (UAB), Università degli Studi di Firenze = University of Florence [Firenze] (UNIFI), University of Arizona, Murdoch Children's Research Institute (MCRI), University of Melbourne, Royal Children's Hospital Melbourne, Leiden University Medical Center (LUMC), and Erasmus University Medical Center [Rotterdam] (Erasmus MC)
- Subjects
Microcephaly ,media_common.quotation_subject ,Population ,Nonsense ,Biology ,Corpus Callosum ,Frameshift mutation ,CLTC ,medicine ,Humans ,Missense mutation ,education ,Genetics (clinical) ,media_common ,Genetics ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,education.field_of_study ,Epilepsy ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,medicine.disease ,neurodevelopmental disorder ,Phenotype ,Biological Variation, Population ,intellectual disability ,Allelic heterogeneity ,Haploinsufficiency ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] - Abstract
Contains fulltext : 218155.pdf (Publisher’s version ) (Closed access) PURPOSE: To delineate the genotype-phenotype correlation in individuals with likely pathogenic variants in the CLTC gene. METHODS: We describe 13 individuals with de novo CLTC variants. Causality of variants was determined by using the tolerance landscape of CLTC and computer-assisted molecular modeling where applicable. Phenotypic abnormalities observed in the individuals identified with missense and in-frame variants were compared with those with nonsense or frameshift variants in CLTC. RESULTS: All de novo variants were judged to be causal. Combining our data with that of 14 previously reported affected individuals (n = 27), all had intellectual disability (ID), ranging from mild to moderate/severe, with or without additional neurologic, behavioral, craniofacial, ophthalmologic, and gastrointestinal features. Microcephaly, hypoplasia of the corpus callosum, and epilepsy were more frequently observed in individuals with missense and in-frame variants than in those with nonsense and frameshift variants. However, this difference was not significant. CONCLUSIONS: The wide phenotypic variability associated with likely pathogenic CLTC variants seems to be associated with allelic heterogeneity. The detailed clinical characterization of a larger cohort of individuals with pathogenic CLTC variants is warranted to support the hypothesis that missense and in-frame variants exert a dominant-negative effect, whereas the nonsense and frameshift variants would result in haploinsufficiency.
- Published
- 2020
35. Opposite modulation of RAC1 by mutations in TRIO is associated with distinct, domain specific neurodevelopmental disorders
- Author
-
Marcie A. Steeves, Marjorlaine Willems, Siddharth Banka, Yline Capri, Michael J. Parker, Stephanie Greville-Heygate, Emma Clement, David Goudie, Vincent Cantagrel, Diana Rodriguez, Marlène Rio, Matthew Guille, Htoo A Wai, Anne Debant, Ajoy Sarkar, Fleur Vansenne, Frédéric Tran Mau-Them, Peter D Turnpenny, Audrey Putoux, Christine Fagotto-Kaufmann, Karine Siquier-Pernet, Bert B.A. de Vries, Boris Keren, Maxime Bonnet, Lydie Burglen, Sébastien Moutton, Marion Gérard, Susanne Schmidt, Diana Baralle, Sónia Barbosa, Benjamin Cogné, Damien Laouteouet, Amélie Piton, Helen Cox, Rebecca Mawby, Marie Vincent, Annie Godwin, Andrey V. Kajava, Sarju G. Mehta, Alexander J. M. Dingemans, Jozef Hertecant, Jayne Y. Hehir-Kwa, Gaetan Lesca, Centre de recherche en Biologie Cellulaire (CRBM), and Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Université Montpellier 1 (UM1)
- Subjects
Male ,rac1 GTP-Binding Protein ,0301 basic medicine ,Microcephaly ,INTELLECTUAL DISABILITY ,[SDV]Life Sciences [q-bio] ,APC-PAID ,medicine.disease_cause ,Cohort Studies ,PATHWAY ,0302 clinical medicine ,Guanine Nucleotide Exchange Factors ,Missense mutation ,Spectrin ,microcephaly ,GEF ,ComputingMilieux_MISCELLANEOUS ,Genetics (clinical) ,Genetics ,Mutation ,Spectrin repeat ,Phenotype ,intellectual disability ,BBSRC ,Female ,Nonsense mutation ,autism ,Protein Serine-Threonine Kinases ,Biology ,macrocephaly ,RHO-GTPASES ,Article ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,medicine ,Humans ,Amino Acid Sequence ,BB/R014841/1 ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Sequence Homology, Amino Acid ,RCUK ,medicine.disease ,HEK293 Cells ,030104 developmental biology ,XENOPUS ,Neurodevelopmental Disorders ,Axon guidance ,030217 neurology & neurosurgery ,SPERM - Abstract
The Rho-guanine nucleotide exchange factor (RhoGEF) TRIO acts as a key regulator of neuronal migration, axonal outgrowth, axon guidance, and synaptogenesis by activating the GTPase RAC1 and modulating actin cytoskeleton remodeling. Pathogenic variants in TRIO are associated with neurodevelopmental diseases, including intellectual disability (ID) and autism spectrum disorders (ASD). Here, we report the largest international cohort of 24 individuals with confirmed pathogenic missense or nonsense variants in TRIO. The nonsense mutations are spread along the TRIO sequence, and affected individuals show variable neurodevelopmental phenotypes. In contrast, missense variants cluster into two mutational hotspots in the TRIO sequence, one in the seventh spectrin repeat and one in the RAC1-activating GEFD1. Although all individuals in this cohort present with developmental delay and a neuro-behavioral phenotype, individuals with a pathogenic variant in the seventh spectrin repeat have a more severe ID associated with macrocephaly than do most individuals with GEFD1 variants, who display milder ID and microcephaly. Functional studies show that the spectrin and GEFD1 variants cause a TRIO-mediated hyper- or hypo-activation of RAC1, respectively, and we observe a striking correlation between RAC1 activation levels and the head size of the affected individuals. In addition, truncations in TRIO GEFD1 in the vertebrate model X. tropicalis induce defects that are concordant with the human phenotype. This work demonstrates distinct clinical and molecular disorders clustering in the GEFD1 and seventh spectrin repeat domains and highlights the importance of tight control of TRIO-RAC1 signaling in neuronal development.
- Published
- 2020
36. KMT2B-related disorders: expansion of the phenotypic spectrum and long-term efficacy of deep brain stimulation
- Author
-
Nihr BioResource, F. Lucy Raymond, Shekeeb S. Mohammad, TD Graves, Susan J. Hayflick, Bert B.A. de Vries, Katy Barwick, Conor Fearon, Dora Steel, Mark Hallett, Asif Doja, Emilie Chan Seng, Camilo Toro, Fiona Stewart, Diane Demailly, Suh Young Jeong, Claudio M. de Gusmao, Frédérique Pavillard, Nutan Sharma, Fabienne Cyprien, Juan C Pallais, Brent L. Fogel, David R. FitzPatrick, Lucia Abela, Russell C. Dale, Bettina Balint, Natalie Trump, Michel Tchan, Sony Malhotra, Swasti Pal, Maya Topf, Manju A. Kurian, Michelle Sahagian, Julia Rankin, Laila Selim, Jeff L. Waugh, Sidney Krystal, Gustavo Polo, Caleb Rogers, Michel Mondain, Kailash P. Bhatia, Ishwar C. Verma, Marisela Dy-Hollins, Kelly A. Mills, Derek Wong, Laura Cif, William A. Gahl, Meredith W Allain, Sanaz Attaripour Isfahani, Agathe Roubertie, Jenny L. Wilson, Allison Gregory, Victoria Gonzalez, Carolyn D. Applegate, Nathalie Dorison, Jennifer A. Bassetti, Catherine Blanchet, Ada Hamosh, Deciphering Developmental Disorders Study, Hane Lee, Julien Baleine, Emma L. Baple, Gaetan Lesca, Anna Znaczko, Thomas Roujeau, Mario Sa, Laurence Lion François, Neil Mahant, Diane Doummar, Sandra Jansen, Marie Hully, Christine Coubes, Eva B. Forman, Victor S.C. Fung, Gaëtan Poulen, Raghda Zaitoun, Serena Galosi, Timothy Lynch, Xavier Vasques, Elise Schaefer, Richard Selway, Adeline Ngoh, Tuula Rinne, Philippe Coubes, Elizabeth L. Fieg, Rachel Fox, Jennifer Friedman, Andrea K. Petersen, Hugo Morales-Briceño, Rebecca Signer, Luis Rohena, Sandra Chantot Bastaraud, Chloé Laurencin, Kishore R. Kumar, Julian A. Martinez-Agosto, Ellyn Farrelly, Kathleen M. Gorman, Esther Meyer, Joel B. Krier, Ariane Soldatos, Lydie Burglen, Jean-Pierre Lin, Pierre-François Perrigault, Dolly Zhen, Harutomo Hasegawa, Mary D. King, Alba Sanchis-Juan, David A. Stevenson, Gilles Cambonie, Wui K. Chong, Christophe Milési, Vincent d'Hardemare, John R. Østergaard, Laboratoire de Recherche en Neurosciences Cliniques, IBM Systems Group, Service de Neurochirurgie [Montpellier], Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier)-CHU Gui de Chauliac [Montpellier], Evelina London Children's Hospital, Institute of Child Health [London], University College of London [London] (UCL), Birkbeck College [University of London], Great Ormond Street Hospital for Children [London] (GOSH), Cambridge University Hospitals - NHS (CUH), University of Cambridge [UK] (CAM), Royal Devon and Exeter NHS Foundation Trust [UK], and Stanford University
- Subjects
0301 basic medicine ,Male ,Pediatrics ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,Deep Brain Stimulation ,DYT1 DYSTONIA ,GENERALIZED DYSTONIA ,VARIANTS ,Cohort Studies ,0302 clinical medicine ,genetics ,[MATH]Mathematics [math] ,Deep brain stimulation (DBS) ,Child ,Laryngeal dystonia ,ComputingMilieux_MISCELLANEOUS ,Dystonia ,Fetal Growth Retardation ,neurodevelopment ,Parkinsonism ,KMT2B ,3. Good health ,INSIGHTS ,Phenotype ,Treatment Outcome ,Dystonic Disorders ,Child, Preschool ,Cohort ,Disease Progression ,deep brain stimulation (DBS) ,dystonia ,Female ,Chromosome Deletion ,Adult ,medicine.medical_specialty ,Deep brain stimulation ,Adolescent ,DATABASE ,Mutation, Missense ,Endocrine System Diseases ,Laryngeal Diseases ,03 medical and health sciences ,Young Adult ,All institutes and research themes of the Radboud University Medical Center ,medicine ,Genetics ,Humans ,Computer Simulation ,Gait Disorders, Neurologic ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,business.industry ,MUTATIONS ,Histone-Lysine N-Methyltransferase ,Original Articles ,medicine.disease ,Status dystonicus ,030104 developmental biology ,Disease Presentation ,Brain stimulation ,Mutation ,Quality of Life ,Neurology (clinical) ,business ,FOLLOW-UP ,GAIT ,030217 neurology & neurosurgery ,SYSTEM - Abstract
Heterozygous mutations in KMT2B are associated with an early-onset, progressive and often complex dystonia (DYT28). Key characteristics of typical disease include focal motor features at disease presentation, evolving through a caudocranial pattern into generalized dystonia, with prominent oromandibular, laryngeal and cervical involvement. Although KMT2B-related disease is emerging as one of the most common causes of early-onset genetic dystonia, much remains to be understood about the full spectrum of the disease. We describe a cohort of 53 patients with KMT2B mutations, with detailed delineation of their clinical phenotype and molecular genetic features. We report new disease presentations, including atypical patterns of dystonia evolution and a subgroup of patients with a non-dystonic neurodevelopmental phenotype. In addition to the previously reported systemic features, our study has identified co-morbidities, including the risk of status dystonicus, intrauterine growth retardation, and endocrinopathies. Analysis of this study cohort (n = 53) in tandem with published cases (n = 80) revealed that patients with chromosomal deletions and protein truncating variants had a significantly higher burden of systemic disease (with earlier onset of dystonia) than those with missense variants. Eighteen individuals had detailed longitudinal data available after insertion of deep brain stimulation for medically refractory dystonia. Median age at deep brain stimulation was 11.5 years (range: 4.5–37.0 years). Follow-up after deep brain stimulation ranged from 0.25 to 22 years. Significant improvement of motor function and disability (as assessed by the Burke Fahn Marsden’s Dystonia Rating Scales, BFMDRS-M and BFMDRS-D) was evident at 6 months, 1 year and last follow-up (motor, P = 0.001, P = 0.004, and P = 0.012; disability, P = 0.009, P = 0.002 and P = 0.012). At 1 year post-deep brain stimulation, >50% of subjects showed BFMDRS-M and BFMDRS-D improvements of >30%. In the long-term deep brain stimulation cohort (deep brain stimulation inserted for >5 years, n = 8), improvement of >30% was maintained in 5/8 and 3/8 subjects for the BFMDRS-M and BFMDRS-D, respectively. The greatest BFMDRS-M improvements were observed for trunk (53.2%) and cervical (50.5%) dystonia, with less clinical impact on laryngeal dystonia. Improvements in gait dystonia decreased from 20.9% at 1 year to 16.2% at last assessment; no patient maintained a fully independent gait. Reduction of BFMDRS-D was maintained for swallowing (52.9%). Five patients developed mild parkinsonism following deep brain stimulation. KMT2B-related disease comprises an expanding continuum from infancy to adulthood, with early evidence of genotype-phenotype correlations. Except for laryngeal dysphonia, deep brain stimulation provides a significant improvement in quality of life and function with sustained clinical benefit depending on symptoms distribution.
- Published
- 2020
37. De Novo Variants in SPOP Cause Two Clinically Distinct Neurodevelopmental Disorders
- Author
-
Hanka Venselaar, Jennifer Keller-Ramey, Arjan P.M. de Brouwer, Tiziano Bernasocchi, Amber Begtrup, Michael Parker, Margot I. Van Allen, Koen L.I. van Gassen, Christian Gilissen, Maria J. Nabais Sá, Lisenka E.L.M. Vissers, Ellen R. Elias, Daniela del Gaudio, Sarah L. Sawyer, Bert B.A. de Vries, Farah Kanani, Jean-Philippe Theurillat, Klaas J. Wierenga, Marie-José H. van den Boogaard, Gabriela Purcarin, Rolph Pfundt, Laurens Wiel, and Geniver El Tekle
- Subjects
Male ,Microcephaly ,Adolescent ,Mutation, Missense ,SPOP ,Biology ,03 medical and health sciences ,Young Adult ,All institutes and research themes of the Radboud University Medical Center ,0302 clinical medicine ,Germline mutation ,Neurodevelopmental disorder ,Report ,Intellectual Disability ,Genetics ,medicine ,Missense mutation ,Humans ,Hypertelorism ,Child ,Genetics (clinical) ,Exome sequencing ,030304 developmental biology ,0303 health sciences ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Skull ,Macrocephaly ,Facies ,Infant ,Nuclear Proteins ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,medicine.disease ,Repressor Proteins ,Neurodevelopmental Disorders ,Child, Preschool ,Female ,medicine.symptom ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,030217 neurology & neurosurgery - Abstract
Recurrent somatic variants in SPOP are cancer specific; endometrial and prostate cancers result from gain-of-function and dominant-negative effects toward BET proteins, respectively. By using clinical exome sequencing, we identified six de novo pathogenic missense variants in SPOP in seven individuals with developmental delay and/or intellectual disability, facial dysmorphisms, and congenital anomalies. Two individuals shared craniofacial dysmorphisms, including congenital microcephaly, that were strikingly different from those of the other five individuals, who had (relative) macrocephaly and hypertelorism. We measured the effect of SPOP variants on BET protein amounts in human Ishikawa endometrial cancer cells and patient-derived cell lines because we hypothesized that variants would lead to functional divergent effects on BET proteins. The de novo variants c.362G>A (p.Arg121Gln) and c. 430G>A (p.Asp144Asn), identified in the first two individuals, resulted in a gain of function, and conversely, the c.73A>G (p.Thr25Ala), c.248A>G (p.Tyr83Cys), c.395G>T (p.Gly132Val), and c.412C>T (p.Arg138Cys) variants resulted in a dominant-negative effect. Our findings suggest that these opposite functional effects caused by the variants in SPOP result in two distinct and clinically recognizable syndromic forms of intellectual disability with contrasting craniofacial dysmorphisms.
- Published
- 2020
38. A genotype-first approach identifies an intellectual disability-overweight syndrome caused by PHIP haploinsufficiency
- Author
-
Rolph Pfundt, Bregje W.M. van Bon, Petra de Vries, Carl Baker, Marloes Steehouwer, Bradley P. Coe, Tjitske Kleefstra, Lisenka E.L.M. Vissers, Han G. Brunner, David A. Koolen, Caroline Nava, Daniëlle G. M. Bosch, Anna Hackett, Hilde Van Esch, Fleur Vansenne, Alexander Hoischen, Christian Gilissen, Kali Witherspoon, Heather C Mefford, Martin Jakob Larsen, Janneke H M Schuurs-Hoeijmakers, Malin Kvarnung, Gemma L. Carvill, Bert B.A. de Vries, Fiona Haslam McKenzie, Maartje van de Vorst, Mirella Vinci, Jozef Gecz, Carlo Marcelis, Sandra Jansen, Marijke Bauters, Raphael Bernier, Joris A. Veltman, Corrado Romano, Evan E. Eichler, Ulla A Andersen, Hedi L Claahsen-van der Grinten, Connie T.R.M. Stumpel, Lucia Grillo, Marie Lorraine Monin, Servi J. C. Stevens, MUMC+: DA KG Lab Centraal Lab (9), RS: GROW - R4 - Reproductive and Perinatal Medicine, Klinische Genetica, MUMC+: DA KG Polikliniek (9), and MUMC+: DA Klinische Genetica (5)
- Subjects
Male ,0301 basic medicine ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,AUTISM SPECTRUM DISORDERS ,Haploinsufficiency ,Disease ,DOMAIN-INTERACTING PROTEIN ,0302 clinical medicine ,Intellectual disability ,Genotype ,OF-FUNCTION MUTATIONS ,SCHIZOPHRENIA ,Child ,Genetics (clinical) ,Genetics ,DEVELOPMENTAL DELAY ,Genotype-first approach ,Intracellular Signaling Peptides and Proteins ,Vascular damage Radboud Institute for Molecular Life Sciences [Radboudumc 16] ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Syndrome ,Phenotype ,Sequence Analysis, DNA/methods ,Female ,Sequence Analysis ,Genetic Testing/methods ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Adult ,GENES ,Adolescent ,Biology ,Article ,SIGNALING PATHWAYS ,UBIQUITIN LIGASE ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,Intellectual Disability ,medicine ,Humans ,Genetic Testing ,Overweight/genetics ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Reproducibility of Results ,LINKED MENTAL-RETARDATION ,DNA ,Sequence Analysis, DNA ,Overweight ,medicine.disease ,Intellectual Disability/genetics ,Human genetics ,030104 developmental biology ,DE-NOVO MUTATIONS ,Autism ,Intracellular Signaling Peptides and Proteins/genetics ,030217 neurology & neurosurgery - Abstract
Genotype-first combined with reverse phenotyping has shown to be a powerful tool in human genetics, especially in the era of next generation sequencing. This combines the identification of individuals with mutations in the same gene and linking these to consistent (endo)phenotypes to establish disease causality. We have performed a MIP (molecular inversion probe)-based targeted re-sequencing study in 3,275 individuals with intellectual disability (ID) to facilitate a genotype-first approach for 24 genes previously implicated in ID.Combining our data with data from a publicly available database, we confirmed 11 of these 24 genes to be relevant for ID. Amongst these, PHIP was shown to have an enrichment of disruptive mutations in the individuals with ID (5 out of 3,275). Through international collaboration, we identified a total of 23 individuals with PHIP mutations and elucidated the associated phenotype. Remarkably, all 23 individuals had developmental delay/ID and the majority were overweight or obese. Other features comprised behavioral problems (hyperactivity, aggression, features of autism and/or mood disorder) and dysmorphisms (full eyebrows and/or synophrys, upturned nose, large ears and tapering fingers). Interestingly, PHIP encodes two protein-isoforms, PHIP/DCAF14 and NDRP, each involved in neurodevelopmental processes, including E3 ubiquitination and neuronal differentiation. Detailed genotype-phenotype analysis points towards haploinsufficiency of PHIP/DCAF14, and not NDRP, as the underlying cause of the phenotype.Thus, we demonstrated the use of large scale re-sequencing by MIPs, followed by reverse phenotyping, as a constructive approach to verify candidate disease genes and identify novel syndromes, highlighted by PHIP haploinsufficiency causing an ID-overweight syndrome. ispartof: European Journal of Human Genetics vol:26 issue:1 pages:54-63 ispartof: location:England status: published
- Published
- 2018
39. Heterozygous variants inACTL6A, encoding a component of the BAF complex, are associated with intellectual disability
- Author
-
Alyssa A. Tran, Bert B.A. de Vries, Brendan Lee, Chester W. Brown, Philippe M. Campeau, Ronit Marom, Brett H. Graham, Ralitza H. Gavrilova, Mahim Jain, Marwan Shinawi, Justine Rousseau, Richard A. Gibbs, Andrew T. Gunter, I-Wen Song, Christine M. Eng, James T. Lu, Yangjin Bae, Jill A. Rosenfeld, Yuqing Chen, Yaping Yang, Servi J. C. Stevens, Julie D. Kaplan, Alexander P.A. Stegmann, and Lindsay C. Burrage
- Subjects
0301 basic medicine ,Genetics ,Biology ,medicine.disease ,Chromatin remodeling ,03 medical and health sciences ,030104 developmental biology ,RNA splicing ,Intellectual disability ,Homologous chromosome ,medicine ,Mutation testing ,Missense mutation ,Gene ,Coffin–Siris syndrome ,Genetics (clinical) - Abstract
Pathogenic variants in genes encoding components of the BRG1-associated factor (BAF) chromatin remodeling complex have been associated with intellectual disability syndromes. We identified heterozygous, novel variants in ACTL6A, a gene encoding a component of the BAF complex, in three subjects with varying degrees of intellectual disability. Two subjects have missense variants affecting highly conserved amino acid residues within the actin-like domain. Missense mutations in the homologous region in yeast actin were previously reported to be dominant lethal and were associated with impaired binding of the human ACTL6A to β-actin and BRG1. A third subject has a splicing variant that creates an in-frame deletion. Our findings suggest that the variants identified in our subjects may have a deleterious effect on the function of the protein by disturbing the integrity of the BAF complex. Thus, ACTL6A gene mutation analysis should be considered in patients with intellectual disability, learning disabilities, or developmental language disorder.
- Published
- 2017
40. Quantification of Phenotype Information Aids the Identification of Novel Disease Genes
- Author
-
Bregje W.M. van Bon, Christian Gilissen, Han G. Brunner, David A. Koolen, Bert B.A. de Vries, Arjan P.M. de Brouwer, Jelle J. Goeman, Claudia J.M. van Amen-Hellebrekers, Sandra Jansen, Anneke T. Vulto-van Silfhout, Erik A. Sistermans, MUMC+: DA Klinische Genetica (5), Klinische Genetica, RS: FHML non-thematic output, Amsterdam Reproduction & Development (AR&D), Human genetics, and Amsterdam Neuroscience - Complex Trait Genetics
- Subjects
0301 basic medicine ,Mutation rate ,INTELLECTUAL DISABILITY ,Genotype ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,systematic phenotyping ,AUTISM SPECTRUM DISORDERS ,de novo mutations ,Disease ,030105 genetics & heredity ,Biology ,03 medical and health sciences ,Intellectual disability ,Genetics ,medicine ,Humans ,statistical approach ,Genetic Predisposition to Disease ,phenotype features ,Gene ,Genetics (clinical) ,Genetic Association Studies ,patient cohorts ,Disease gene ,Models, Statistical ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Models, Genetic ,DEVELOPMENTAL DELAY ,Reproducibility of Results ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,medicine.disease ,Phenotype ,3. Good health ,ONTOLOGY ,030104 developmental biology ,DE-NOVO MUTATIONS ,DISCOVERY ,Cohort ,IMPLICATE ,Identification (biology) ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] - Abstract
Contains fulltext : 174800.pdf (Publisher’s version ) (Open Access) Next-generation sequencing led to the identification of many potential novel disease genes. The presence of mutations in the same gene in multiple unrelated patients is, however, a priori insufficient to establish that these genes are truly involved in the respective disease. Here, we show how phenotype information can be incorporated within statistical approaches to provide additional evidence for the causality of mutations. We developed a broadly applicable statistical model that integrates gene-specific mutation rates, cohort size, mutation type, and phenotype frequency information to assess the chance of identifying de novo mutations affecting the same gene in multiple patients with shared phenotype features. We demonstrate our approach based on the frequency of phenotype features present in a unique cohort of 6,149 patients with intellectual disability. We show that our combined approach can decrease the number of patients required to identify novel disease genes, especially for patients with combinations of rare phenotypes. In conclusion, we show how integrating genotype-phenotype information can aid significantly in the interpretation of de novo mutations in potential novel disease genes.
- Published
- 2017
41. YY1 Haploinsufficiency Causes an Intellectual Disability Syndrome Featuring Transcriptional and Chromatin Dysfunction
- Author
-
Asbjørg Stray-Pedersen, Anne Ronan, Yunru Shao, Eric Haan, Katharina Steindl, Zöe Powis, Perla Thulin, Giuseppe Testa, Janneke H M Schuurs-Hoeijmakers, William J. Craigen, Raman Kumar, David Rodriguez-Buritica, Michele Gabriele, Laura S. Farach, Susanne Kjaergaard, Rolph Pfundt, Jillian Nicholl, Jozef Gecz, Petter Strømme, Stefan H. Lelieveld, Kenjiro Kosaki, Sally Ann Lynch, Kimberly M. Nugent, Willy M. Nillesen, Bregje W.M. van Bon, Jill A. Rosenfeld, Charlotte Brasch-Andersen, Eirik Frengen, Lisenka E.L.M. Vissers, Scott D. McLean, Evelyn Douglas, Joris Andrieux, David A. Koolen, Anneke T. Vulto-van Silfhout, Han G. Brunner, Arie van Haeringen, Jenny Morton, Sophie Patrier, Anita Rauch, Christeen Ramane J. Pedurupillay, Pierre-Luc Germain, Peter J. Anderson, Christian Gilissen, Christian P. Schaaf, Alessandro Vitriolo, Jennifer Friedman, Toshiki Takenouchi, Pascal Chambon, Bert B.A. de Vries, Doriana Misceo, Pernille Mathiesen Tørring, Family Medicine, MUMC+: DA Klinische Genetica (5), Klinische Genetica, and RS: FHML non-thematic output
- Subjects
Male ,Models, Molecular ,0301 basic medicine ,Transcription, Genetic ,Haploinsufficiency ,Cohort Studies ,Histones ,0302 clinical medicine ,Missense mutation ,Lymphocytes ,YY1 Transcription Factor ,Genetics (clinical) ,GENE-EXPRESSION ,Genetics ,KANSL1 CAUSE ,Acetylation ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,DEMETHYLASE JMJD3 ,Chromatin ,Enhancer Elements, Genetic ,Child, Preschool ,embryonic structures ,Female ,Neurodevelopmental disorders Radboud Institute for Molecular Life Sciences [Radboudumc 7] ,STEM-CELLS ,Protein Binding ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Chromatin Immunoprecipitation ,LIQUID WATER ,Adolescent ,Mutation, Missense ,Repressor ,Biology ,Methylation ,Article ,17Q21.31 MICRODELETION SYNDROME ,03 medical and health sciences ,ADENOVIRUS E1A ,Protein Domains ,Intellectual Disability ,Journal Article ,Humans ,Enhancer ,Transcription factor ,Hemizygote ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Base Sequence ,POTENTIAL FUNCTIONS ,Activator (genetics) ,YY1 ,MUTATIONS ,Gene Ontology ,030104 developmental biology ,Haplotypes ,030217 neurology & neurosurgery ,MENTAL-RETARDATION - Abstract
Contains fulltext : 174704.pdf (Publisher’s version ) (Open Access) Yin and yang 1 (YY1) is a well-known zinc-finger transcription factor with crucial roles in normal development and malignancy. YY1 acts both as a repressor and as an activator of gene expression. We have identified 23 individuals with de novo mutations or deletions of YY1 and phenotypic features that define a syndrome of cognitive impairment, behavioral alterations, intrauterine growth restriction, feeding problems, and various congenital malformations. Our combined clinical and molecular data define "YY1 syndrome" as a haploinsufficiency syndrome. Through immunoprecipitation of YY1-bound chromatin from affected individuals' cells with antibodies recognizing both ends of the protein, we show that YY1 deletions and missense mutations lead to a global loss of YY1 binding with a preferential retention at high-occupancy sites. Finally, we uncover a widespread loss of H3K27 acetylation in particular on the YY1-bound enhancers, underscoring a crucial role for YY1 in enhancer regulation. Collectively, these results define a clinical syndrome caused by haploinsufficiency of YY1 through dysregulation of key transcriptional regulators.
- Published
- 2017
42. Loss-of-Function Mutations in YY1AP1 Lead to Grange Syndrome and a Fibromuscular Dysplasia-Like Vascular Disease
- Author
-
Xue Yan Duan, Bert B.A. de Vries, Michael J. Bamshad, Dorothy K. Grange, Callie S. Kwartler, Deborah A. Nickerson, Dongchuan Guo, Dieter Kotzot, Xuetong Shen, Ellen S. Regalado, Rolph Pfundt, Dianna M. Milewicz, Kenneth Lieberman, Alan C. Braverman, Heather L. Gornik, Lauren Mellor-Crummey, Albert Schinzel, Min-Lee Yang, Santhi K. Ganesh, Dong H. Kim, University of Zurich, and Milewicz, Dianna M
- Subjects
Adult ,Male ,0301 basic medicine ,Heterozygote ,2716 Genetics (clinical) ,medicine.medical_specialty ,Vascular smooth muscle ,Adolescent ,10039 Institute of Medical Genetics ,DNA repair ,Cell Cycle Proteins ,Genes, Recessive ,610 Medicine & health ,Fibromuscular dysplasia ,Biology ,Compound heterozygosity ,Article ,Bone and Bones ,Muscle, Smooth, Vascular ,Frameshift mutation ,03 medical and health sciences ,1311 Genetics ,Internal medicine ,Genetics ,medicine ,Fibromuscular Dysplasia ,Humans ,Exome ,Genetics (clinical) ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Learning Disabilities ,Vascular disease ,Brachydactyly ,Homozygote ,Nuclear Proteins ,Grange syndrome ,Cell Cycle Checkpoints ,Syndrome ,Middle Aged ,Cell cycle ,medicine.disease ,Pedigree ,030104 developmental biology ,Endocrinology ,Mutation ,570 Life sciences ,biology ,Female ,Syndactyly ,Transcription Factors - Abstract
Contains fulltext : 169736.pdf (Publisher’s version ) (Closed access) Fibromuscular dysplasia (FMD) is a heterogeneous group of non-atherosclerotic and non-inflammatory arterial diseases that primarily involves the renal and cerebrovascular arteries. Grange syndrome is an autosomal-recessive condition characterized by severe and early-onset vascular disease similar to FMD and variable penetrance of brachydactyly, syndactyly, bone fragility, and learning disabilities. Exome-sequencing analysis of DNA from three affected siblings with Grange syndrome identified compound heterozygous nonsense variants in YY1AP1, and homozygous nonsense or frameshift YY1AP1 variants were subsequently identified in additional unrelated probands with Grange syndrome. YY1AP1 encodes yin yang 1 (YY1)-associated protein 1 and is an activator of the YY1 transcription factor. We determined that YY1AP1 localizes to the nucleus and is a component of the INO80 chromatin remodeling complex, which is responsible for transcriptional regulation, DNA repair, and replication. Molecular studies revealed that loss of YY1AP1 in vascular smooth muscle cells leads to cell cycle arrest with decreased proliferation and increased levels of the cell cycle regulator p21/WAF/CDKN1A and disrupts TGF-beta-driven differentiation of smooth muscle cells. Identification of YY1AP1 mutations as a cause of FMD indicates that this condition can result from underlying genetic variants that significantly alter the phenotype of vascular smooth muscle cells.
- Published
- 2017
43. Overrepresentation of genetic variation in the AnkyrinG interactome is related to a range of neurodevelopmental disorders
- Author
-
Ilse M. van der Werf, Christian Gilissen, Alexander Hoischen, Lisenka E.L.M. Vissers, Geert Vandeweyer, Anke Van Dijck, Bert B.A. de Vries, Amber Gerstmans, Sandra Jansen, Maartje van de Vorst, Petra de Vries, and R. Frank Kooy
- Subjects
Ankyrins ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Gene regulatory network ,Biology ,Interactome ,Article ,03 medical and health sciences ,Interaction network ,Genetic variation ,Databases, Genetic ,Genetics ,Humans ,Gene Regulatory Networks ,Protein Interaction Maps ,Gene ,Genetics (clinical) ,0303 health sciences ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Polymorphism, Genetic ,030305 genetics & heredity ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Large cohort ,Chemistry ,Neurodevelopmental Disorders ,Human medicine ,Protein network ,Protein Interaction Map - Abstract
Contains fulltext : 229283.pdf (Publisher’s version ) (Closed access) Upon the discovery of numerous genes involved in the pathogenesis of neurodevelopmental disorders, several studies showed that a significant proportion of these genes converge on common pathways and protein networks. Here, we used a reversed approach, by screening the AnkyrinG protein-protein interaction network for genetic variation in a large cohort of 1009 cases with neurodevelopmental disorders. We identified a significant enrichment of de novo potentially disease-causing variants in this network, confirming that this protein network plays an important role in the emergence of several neurodevelopmental disorders.
- Published
- 2019
44. KIF1A variants are a frequent cause of autosomal dominant hereditary spastic paraplegia
- Author
-
Leonard H. van den Berg, Mariet W. Elting, Marjolein Kriek, Jiddeke M. van de Kamp, Maartje Pennings, Judith van Gaalen, Jacques Bruijn, Eva H. Brilstra, Michael A. van Es, Bart P.C. van de Warrenburg, Dick M.H. Zuidgeest, Floor A. M. Duijkers, Kalliopi Sofou, Susanne T. de Bot, Jolanda H. Schieving, Rowdy Meijer, Christine E. M. de Die-Smulders, Bert B.A. de Vries, Bregje Jaeger, Karin Y. van Spaendonck-Zwarts, Meyke Schouten, Corien Verschuuren, Christiaan G J Saris, Erik-Jan Kamsteeg, Faculteit Medische Wetenschappen/UMCG, Human Genetics, Neurology, Paediatric Neurology, Human genetics, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Neuroscience - Complex Trait Genetics, Amsterdam Reproduction & Development (AR&D), RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Polikliniek (9), and Klinische Genetica
- Subjects
Male ,DIMERIZATION ,Molecular biology ,Kinesins ,Sensory disorders Donders Center for Medical Neuroscience [Radboudumc 12] ,Motor domain ,Spastic ,Missense mutation ,Child ,Genetics (clinical) ,Exome sequencing ,KIF1A ,Genes, Dominant ,Genetics ,0303 health sciences ,030305 genetics & heredity ,Middle Aged ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,musculoskeletal system ,Pedigree ,Child, Preschool ,Female ,Haploinsufficiency ,Paraplegia ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,musculoskeletal diseases ,Adult ,congenital, hereditary, and neonatal diseases and abnormalities ,MOTOR DOMAIN ,Adolescent ,Mutation, Missense ,Biology ,MONOMERIC MOTOR ,Article ,MECHANISMS ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,Protein Domains ,medicine ,Humans ,Allele ,Movement disorders ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,MUTATIONS ,Spastic Paraplegia, Hereditary ,NEUROPATHY ,Infant ,medicine.disease ,TRANSPORT ,nervous system diseases - Abstract
Variants in the KIF1A gene can cause autosomal recessive spastic paraplegia 30, autosomal recessive hereditary sensory neuropathy, or autosomal (de novo) dominant mental retardation type 9. More recently, variants in KIF1A have also been described in a few cases with autosomal dominant spastic paraplegia. Here, we describe 20 KIF1A variants in 24 patients from a clinical exome sequencing cohort of 347 individuals with a mostly 'pure' spastic paraplegia. In these patients, spastic paraplegia was slowly progressive and mostly pure, but with a highly variable disease onset (0-57 years). Segregation analyses showed a de novo occurrence in seven cases, and a dominant inheritance pattern in 11 families. The motor domain of KIF1A is a hotspot for disease causing variants in autosomal dominant spastic paraplegia, similar to mental retardation type 9 and recessive spastic paraplegia type 30. However, unlike these allelic disorders, dominant spastic paraplegia was also caused by loss-of-function variants outside this domain in six families. Finally, three missense variants were outside the motor domain and need further characterization. In conclusion, KIF1A variants are a frequent cause of autosomal dominant spastic paraplegia in our cohort (6-7%). The identification of KIF1A loss-of-function variants suggests haploinsufficiency as a possible mechanism in autosomal dominant spastic paraplegia.
- Published
- 2019
45. Expanding the phenotype of intellectual disability caused by HIVEP2 variants
- Author
-
Maria J. N. Sá, Rolph Pfundt, Mark Williams, Helen Heussler, Bert B.A. de Vries, Anna Wells, Heidi Goldsmith, Melissa Buckman, and Himanshu Goel
- Subjects
Male ,Microcephaly ,Genotype ,All institutes and research themes of the Radboud University Medical Center ,Ocular Motility Disorders ,Intellectual Disability ,Intellectual disability ,Exome Sequencing ,Genetics ,medicine ,Humans ,Child ,Gene ,Genetics (clinical) ,Exome sequencing ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Epilepsy ,business.industry ,Genetic Diseases, X-Linked ,medicine.disease ,Body Dysmorphic Disorders ,Phenotype ,Hypotonia ,DNA-Binding Proteins ,Neurodevelopmental Disorders ,Muscle Hypotonia ,Ataxia ,Female ,medicine.symptom ,Differential diagnosis ,business ,Transcription Factors - Abstract
De novo pathogenic variants in the human immunodeficiency virus enhancer type I binding protein 2 (HIVEP2) gene, a large transcription factor predominantly expressed in the brain have previously been associated with intellectual disability (ID) and dysmorphic features in nine patients. We describe the phenotype and genotype of two additional patients with novel de novo pathogenic HIVEP2 variants, who have previously unreported features, including hyperphagia and Angelman-like features. Exome sequencing was utilized in the investigation of the patients who had previously incurred a rigorous genetic workup for their neurodevelopmental delay, and in whom no genetic cause had been detected. Information pertaining to phenotype and genotype for new patients was collated along with data from previous reports, showing that the phenotypic spectrum of patients with HIVEP2 variants is broader than first noted. Additional characteristics are: an increased body mass index; and features of Angelman-like syndromes including: ID, limited speech, post-natal microcephaly, and hypotonia. Dysmorphic features vary between patients. As yet, no clear association between the type of gene aberration and phenotype can be concluded. HIVEP2-related ID needs to be considered in the differential diagnosis of patients with Angelman-like phenotypes and hyperphagia, and whole-exome sequencing should be considered in the genetic diagnostic armamentarium for patients with ID of inconclusive etiology.
- Published
- 2019
46. A de novo variant in the X-linked gene CNKSR2 is associated with seizures and mild intellectual disability in a female patient
- Author
-
Hans van Bokhoven, Daniel L. Polla, Bert B.A. de Vries, Arjan P.M. de Brouwer, and Harriet R Saunders
- Subjects
0301 basic medicine ,de novo ,lcsh:QH426-470 ,media_common.quotation_subject ,Nonsense ,030105 genetics & heredity ,Clinical Reports ,Frameshift mutation ,03 medical and health sciences ,Epilepsy ,symbols.namesake ,Seizures ,X Chromosome Inactivation ,Genetic linkage ,Obligate carrier ,Genetics ,medicine ,Humans ,Child ,Molecular Biology ,Genetics (clinical) ,Exome sequencing ,Adaptor Proteins, Signal Transducing ,media_common ,Sanger sequencing ,Clinical Report ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,business.industry ,CNKSR2 ,Brain ,medicine.disease ,Magnetic Resonance Imaging ,X‐linked ,lcsh:Genetics ,030104 developmental biology ,Codon, Nonsense ,intellectual disability ,symbols ,Female ,business ,exome sequencing - Abstract
Contains fulltext : 215374.pdf (Publisher’s version ) (Open Access) BACKGROUND: Eight different deletions and point variants of the X-chromosomal gene CNKSR2 have been reported in families with males presenting intellectual disability (ID) and epilepsy. Obligate carrier females with a frameshift variant in the N-terminal protein coding part of CNKSR2 or with a deletion of the complete gene are not affected. Only for one C-terminal nonsense variant, two carrier females were mildly affected by seizures without or with mild motor and language delay. METHODS: Exome sequencing was performed in one female child of a Dutch family, presenting seizures, mild ID, facial dysmorphisms, and abnormalities of the extremities. Potential causative variants were validated by Sanger sequencing. X-chromosome-inactivation (XCI) analysis was performed by methylation-sensitive PCR and fragment-length analysis of the androgen-receptor CAG repeat polymorphism. RESULTS: We identified a de novo variant, c.2304G>A (p.(Trp768*)), in the C-terminal protein coding part of the X-chromosomal gene CNKSR2 in a female patient with seizures and mild ID. Sanger sequencing confirmed the presence of this nonsense variant. XCI analysis showed a mild skewing of X inactivation (20:80) in the blood of our patient. Our variant is the second C-terminal-affecting CNKSR2 variant described in neurologically affected females. CONCLUSION: Our results indicate that CNKSR2 nonsense variants in the C-terminal coding part can result in ID with seizures in female variant carriers.
- Published
- 2019
47. De Novo Mutations Affecting the Catalytic Calpha Subunit of PP2A, PPP2CA, Cause Syndromic Intellectual Disability Resembling Other PP2A-Related Neurodevelopmental Disorders
- Author
-
Ellen Macnamara, Marlène Rio, Nicole Revencu, Saleem Malik, Siska Van Belle, Maura R.Z. Ruzhnikov, Jolanda H. Schieving, Lisenka E.L.M. Vissers, Barak Tziperman, Hilde M.H. Braakman, Alma Kuechler, Susan Sell, Philip Harrer, Ernie M.H.F. Bongers, Marjolein Kriek, Dagmar Wieczorek, Bert B.A. de Vries, Christopher T. Gordon, Jeanne Amiel, Matias Wagner, Dorien Haesen, Roger L. Ladda, Koen L.I. van Gassen, Elise Brimble, Sandra Jansen, Sonja Henry, Carlo Marcelis, Paulien A Terhal, Nienke E. Verbeek, Ortal Barel, Sara Reynhout, Carlos Ferreira, Jessica Scott Schwoerer, Veerle Janssens, Heather M. McLaughlin, Sonja A. de Munnik, Cacha M.P.C.D. Peeters-Scholte, UCL - (SLuc) Centre de génétique médicale UCL, UCL - (SLuc) Centre de malformations vasculaires congénitales, and UCL - SSS/IREC/SLUC - Pôle St.-Luc
- Subjects
Male ,0301 basic medicine ,DNA Mutational Analysis ,Medizin ,Haploinsufficiency ,0302 clinical medicine ,De Novo Mutation ,Epilepsy ,Intellectual Disability ,Pp2a ,Pp2a-related Neurodevelopmental Disorders ,Ppp2ca ,Syndrome ,Intellectual disability ,Gene duplication ,Missense mutation ,Genetics(clinical) ,Protein Phosphatase 2 ,Child ,Genetics (clinical) ,Genetics ,0303 health sciences ,030302 biochemistry & molecular biology ,syndrome ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Hypotonia ,PP2A ,intellectual disability ,Child, Preschool ,Female ,medicine.symptom ,Protein Binding ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Adolescent ,Protein subunit ,Nonsense mutation ,Biology ,Article ,PP2A-related neurodevelopmental disorders ,Frameshift mutation ,03 medical and health sciences ,medicine ,Humans ,Gene ,De novo mutations ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Correction ,medicine.disease ,Human genetics ,PPP2CA ,de novo mutation ,Protein Subunits ,HEK293 Cells ,030104 developmental biology ,Mutation ,epilepsy ,030217 neurology & neurosurgery - Abstract
Type 2A protein phosphatases (PP2As) are highly expressed in the brain and regulate neuronal signaling by catalyzing phospho-Ser/Thr dephosphorylations in diverse substrates. PP2A holoenzymes comprise catalytic C-, scaffolding A-, and regulatory B-type subunits, which determine substrate specificity and physiological function. Interestingly, de novo mutations in genes encoding A- and B-type subunits have recently been implicated in intellectual disability (ID) and developmental delay (DD). We now report 16 individuals with mild to profound ID and DD and a de novo mutation in PPP2CA, encoding the catalytic Cα subunit. Other frequently observed features were severe language delay (71%), hypotonia (69%), epilepsy (63%), and brain abnormalities such as ventriculomegaly and a small corpus callosum (67%). Behavioral problems, including autism spectrum disorders, were reported in 47% of individuals, and three individuals had a congenital heart defect. PPP2CA de novo mutations included a partial gene deletion, a frameshift, three nonsense mutations, a single amino acid duplication, a recurrent mutation, and eight non-recurrent missense mutations. Functional studies showed complete PP2A dysfunction in four individuals with seemingly milder ID, hinting at haploinsufficiency. Ten other individuals showed mutation-specific biochemical distortions, including poor expression, altered binding to the A subunit and specific B-type subunits, and impaired phosphatase activity and C-terminal methylation. Four were suspected to have a dominant-negative mechanism, which correlated with severe ID. Two missense variants affecting the same residue largely behaved as wild-type in our functional assays. Overall, we found that pathogenic PPP2CA variants impair PP2A-B56(δ) functionality, suggesting that PP2A-related neurodevelopmental disorders constitute functionally converging ID syndromes. ispartof: American Journal Of Human Genetics vol:104 issue:1 pages:139-156 ispartof: location:United States status: published
- Published
- 2019
48. Disruptive variants of CSDE1 associate with autism and interfere with neuronal development and synaptic transmission
- Author
-
Corrado Romano, Magnus Nordenskjöld, Tianyun Wang, Min Long, Suneeta Madan-Khetarpal, Evan E. Eichler, Jingping Zhao, Mengzhu Ou, Wei Xie, Yu Zhang, Kirsty McWalter, Chanika Phornphutkul, Kristin G. Monaghan, Koen L.I. van Gassen, Grazia M.S. Mancini, Zhengmao Hu, Madelyn A. Gillentine, Jessica Sebastian, Ying Li, Yaowen Zhang, Jieqiong Tan, Robert J. Hopkin, Kendra Hoekzema, Jozef Gecz, Lu Shen, Meilin Chen, Zhi-Qing David Xu, Carlos E. Prada, Alexander P.A. Stegmann, Judith D. Ranells, Hailun Ni, Ting Bai, Kuokuo Li, Tengfei Zhu, Joseph T. Shieh, Robert B. Hufnagel, Darius J. Adams, Lijuan Liu, Anna Lindstrand, Daryl A. Scott, Huidan Wu, Yingting Quan, Kun Xia, Melissa Racobaldo, J Peng, Mahshid Azamian, Raphael Bernier, Rongjuan Zhao, E. Haan, Fan Xia, Pengwei Peng, Nan Pang, Malin Kvarnung, Honghui Li, Xiangbin Jia, Seema R. Lalani, Jill A. Rosenfeld, Qiumeng Zhang, Susie Ball, Lin Han, Hui Guo, Ikeoluwa A. Osei-Owusu, Giuseppe Calabrese, Ornella Galesi, Tao Xu, Xiaobing Zou, Ann Nordgren, Yaning Liu, Pengfei Liu, Cenying Liu, Jonathan Pevsner, Bert B.A. de Vries, Peter M. van Hasselt, Clinical Genetics, MUMC+: DA KG Lab Centraal Lab (9), and RS: FHML non-thematic output
- Subjects
Male ,Dendritic spine ,Diseases and Disorders ,RNA-binding protein ,Synaptic Transmission ,Synapse ,Mice ,0302 clinical medicine ,Child ,Non-U.S. Gov't ,Research Articles ,Neurons ,RISK ,0303 health sciences ,Gene knockdown ,ARCHITECTURE ,Multidisciplinary ,Research Support, Non-U.S. Gov't ,SciAdv r-articles ,RNA-Binding Proteins ,MENTAL-RETARDATION PROTEIN ,3. Good health ,Pedigree ,Fragile X syndrome ,DNA-Binding Proteins ,DROSOPHILA ,Phenotype ,Child, Preschool ,Female ,MESSENGER-RNA ,Research Article ,GENES ,Neurite ,Adolescent ,Neurogenesis ,Neurotransmission ,Biology ,Research Support ,behavioral disciplines and activities ,N.I.H ,03 medical and health sciences ,Young Adult ,Research Support, N.I.H., Extramural ,mental disorders ,medicine ,Journal Article ,Animals ,Humans ,Genetic Predisposition to Disease ,FRAGILE-X-SYNDROME ,Autistic Disorder ,Preschool ,Genetic Association Studies ,030304 developmental biology ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,IDENTIFICATION ,Animal ,Genetic Variation ,Extramural ,Human Genetics ,medicine.disease ,Disease Models, Animal ,DE-NOVO MUTATION ,Genetic Loci ,Synapses ,Disease Models ,Autism ,TRANSLATION ,Neuroscience ,030217 neurology & neurosurgery - Abstract
CSDE1 disruptive mutations are associated with autism., RNA binding proteins are key players in posttranscriptional regulation and have been implicated in neurodevelopmental and neuropsychiatric disorders. Here, we report a significant burden of heterozygous, likely gene-disrupting variants in CSDE1 (encoding a highly constrained RNA binding protein) among patients with autism and related neurodevelopmental disabilities. Analysis of 17 patients identifies common phenotypes including autism, intellectual disability, language and motor delay, seizures, macrocephaly, and variable ocular abnormalities. HITS-CLIP revealed that Csde1-binding targets are enriched in autism-associated gene sets, especially FMRP targets, and in neuronal development and synaptic plasticity–related pathways. Csde1 knockdown in primary mouse cortical neurons leads to an overgrowth of the neurites and abnormal dendritic spine morphology/synapse formation and impaired synaptic transmission, whereas mutant and knockdown experiments in Drosophila result in defects in synapse growth and synaptic transmission. Our study defines a new autism-related syndrome and highlights the functional role of CSDE1 in synapse development and synaptic transmission.
- Published
- 2019
49. Biallelicframeshift mutation in RIN2 in a patient with intellectual disability and cataract, without RIN2 syndrome
- Author
-
Bert B.A. de Vries, Claudia J.M. van Amen-Hellebrekers, Sandra Jansen, Alexander P.A. Stegmann, Rolph Pfundt, Servi J. C. Stevens, MUMC+: DA KG Lab Centraal Lab (9), and RS: FHML non-thematic output
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,MACS SYNDROME ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,business.industry ,medicine.disease ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,030104 developmental biology ,Intellectual disability ,Mutation (genetic algorithm) ,Genetics ,Medicine ,business ,Genetics (clinical) - Abstract
Contains fulltext : 179551.pdf (Publisher’s version ) (Closed access)
- Published
- 2017
50. Next-generation phenotyping using computer vision algorithms in rare genomic neurodevelopmental disorders
- Author
-
Lia C. M. J. Goltstein, Janneke H M Schuurs-Hoeijmakers, Patrick Kemmeren, Christoffer Nellåker, Bert B.A. de Vries, Jayne Y. Hehir-Kwa, Lisenka E.L.M. Vissers, David A. Koolen, Roos van der Donk, Han G. Brunner, Alexander Hoischen, Sandra Jansen, and Human genetics
- Subjects
Male ,0301 basic medicine ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Vesicular Transport Proteins ,Facial recognition system ,Craniofacial Abnormalities ,0302 clinical medicine ,Intellectual disability ,Image Processing, Computer-Assisted ,Missense mutation ,Child ,Genetics (clinical) ,education.field_of_study ,Intracellular Signaling Peptides and Proteins ,Genomics ,Middle Aged ,Phenotype ,Protein Phosphatase 2C ,Muscular Atrophy ,Child, Preschool ,facial phenotyping ,Female ,Chromosome Deletion ,Facial Recognition ,Hybrid model ,Algorithms ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Adult ,phenotyping ,Adolescent ,Population ,Mutation, Missense ,Other Research Radboud Institute for Molecular Life Sciences [Radboudumc 0] ,Computational biology ,Article ,Young Adult ,03 medical and health sciences ,Facial dysmorphism ,All institutes and research themes of the Radboud University Medical Center ,Intellectual Disability ,medicine ,Humans ,Computer vision algorithms ,Abnormalities, Multiple ,education ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,business.industry ,Infant ,facial image processing ,medicine.disease ,Musculoskeletal Abnormalities ,030104 developmental biology ,Neurodevelopmental Disorders ,business ,030217 neurology & neurosurgery ,Chromosomes, Human, Pair 17 - Abstract
Purpose The interpretation of genetic variants after genome-wide analysis is complex in heterogeneous disorders such as intellectual disability (ID). We investigate whether algorithms can be used to detect if a facial gestalt is present for three novel ID syndromes and if these techniques can help interpret variants of uncertain significance. Methods Facial features were extracted from photos of ID patients harboring a pathogenic variant in three novel ID genes (PACS1, PPM1D, and PHIP) using algorithms that model human facial dysmorphism, and facial recognition. The resulting features were combined into a hybrid model to compare the three cohorts against a background ID population. Results We validated our model using images from 71 individuals with Koolen–de Vries syndrome, and then show that facial gestalts are present for individuals with a pathogenic variant in PACS1 (p = 8 × 10−4), PPM1D (p = 4.65 × 10−2), and PHIP (p = 6.3 × 10−3). Moreover, two individuals with a de novo missense variant of uncertain significance in PHIP have significant similarity to the expected facial phenotype of PHIP patients (p Conclusion Our results show that analysis of facial photos can be used to detect previously unknown facial gestalts for novel ID syndromes, which will facilitate both clinical and molecular diagnosis of rare and novel syndromes.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.