212 results on '"Vera M. Kalscheuer"'
Search Results
2. Integration of Hi-C with short and long-read genome sequencing reveals the structure of germline rearranged genomes
- Author
-
Robert Schöpflin, Uirá Souto Melo, Hossein Moeinzadeh, David Heller, Verena Laupert, Jakob Hertzberg, Manuel Holtgrewe, Nico Alavi, Marius-Konstantin Klever, Julius Jungnitsch, Emel Comak, Seval Türkmen, Denise Horn, Yannis Duffourd, Laurence Faivre, Patrick Callier, Damien Sanlaville, Orsetta Zuffardi, Romano Tenconi, Nehir Edibe Kurtas, Sabrina Giglio, Bettina Prager, Anna Latos-Bielenska, Ida Vogel, Merete Bugge, Niels Tommerup, Malte Spielmann, Antonio Vitobello, Vera M. Kalscheuer, Martin Vingron, and Stefan Mundlos
- Subjects
Science - Abstract
Here the authors characterize structural variations (SVs) in a cohort of individuals with complex genomic rearrangements, identifying breakpoints by employing short- and long-read genome sequencing and investigate their impact on gene expression and the three-dimensional chromatin architecture. They find breakpoints are enriched in inactive regions and can result in chromatin domain fusions.
- Published
- 2022
- Full Text
- View/download PDF
3. Large-Scale Functional Assessment of Genes Involved in Rare Diseases with Intellectual Disabilities Unravels Unique Developmental and Behaviour Profiles in Mouse Models
- Author
-
Hamid Meziane, Marie-Christine Birling, Olivia Wendling, Sophie Leblanc, Aline Dubos, Mohammed Selloum, Guillaume Pavlovic, Tania Sorg, Vera M. Kalscheuer, Pierre Billuart, Frédéric Laumonnier, Jamel Chelly, Hans van Bokhoven, and Yann Herault
- Subjects
mouse model ,genetic modification ,intellectual disability ,behavior phenotypes ,Biology (General) ,QH301-705.5 - Abstract
Major progress has been made over the last decade in identifying novel genes involved in neurodevelopmental disorders, although the task of elucidating their corresponding molecular and pathophysiological mechanisms, which are an essential prerequisite for developing therapies, has fallen far behind. We selected 45 genes for intellectual disabilities to generate and characterize mouse models. Thirty-nine of them were based on the frequency of pathogenic variants in patients and literature reports, with several corresponding to de novo variants, and six other candidate genes. We used an extensive screen covering the development and adult stages, focusing specifically on behaviour and cognition to assess a wide range of functions and their pathologies, ranging from basic neurological reflexes to cognitive abilities. A heatmap of behaviour phenotypes was established, together with the results of selected mutants. Overall, three main classes of mutant lines were identified based on activity phenotypes, with which other motor or cognitive deficits were associated. These data showed the heterogeneity of phenotypes between mutation types, recapitulating several human features, and emphasizing the importance of such systematic approaches for both deciphering genetic etiological causes of ID and autism spectrum disorders, and for building appropriate therapeutic strategies.
- Published
- 2022
- Full Text
- View/download PDF
4. Whole genome sequencing identifies a duplicated region encompassing Xq13.2q13.3 in a large Iranian family with intellectual disability
- Author
-
Sepideh Mehvari, Farzaneh Larti, Hao Hu, Zohreh Fattahi, Maryam Beheshtian, Seyedeh Sedigheh Abedini, Sanaz Arzhangi, Hans‐Hilger Ropers, Vera M. Kalscheuer, Daniel Auld, Kimia Kahrizi, Yasser Riazalhosseini, and Hossein Najmabadi
- Subjects
intellectual disability ,whole genome sequencing ,Xq duplication ,Xq13.2q13.3 ,Genetics ,QH426-470 - Abstract
Abstract Background The X chromosome has historically been one of the most thoroughly investigated chromosomes regarding intellectual disability (ID), whose etiology is attributed to many factors including copy number variations (CNVs). Duplications of the long arm of the X chromosome have been reported in patients with ID, short stature, facial anomalies, and in many cases hypoplastic genitalia and/or behavioral abnormalities. Methods Here, we report on a large Iranian family with X‐linked ID caused by a duplication on the X chromosome identified by whole genome sequencing in combination with linkage analysis. Results Seven affected males in different branches of the family presented with ID, short stature, seizures, facial anomalies, behavioral abnormalities (aggressiveness, self‐injury, anxiety, impaired social interactions, and shyness), speech impairment, and micropenis. The duplication of the region Xq13.2q13.3, which is ~1.8 Mb in size, includes seven protein‐coding OMIM genes. Three of these genes, namely SLC16A2, RLIM, and NEXMIF, if impaired, can lead to syndromes presenting with ID. Of note, this duplicated region was located within a linkage interval with a LOD score >3. Conclusion Our report indicates that CNVs should be considered in multi‐affected families where no candidate gene defect has been identified in sequencing data analysis.
- Published
- 2020
- Full Text
- View/download PDF
5. X chromosome dosage and presence of SRY shape sex-specific differences in DNA methylation at an autosomal region in human cells
- Author
-
Bianca Ho, Keelin Greenlaw, Abeer Al Tuwaijri, Sanny Moussette, Francisco Martínez, Elisa Giorgio, Alfredo Brusco, Giovanni Battista Ferrero, Natália D. Linhares, Eugênia R. Valadares, Marta Svartman, Vera M. Kalscheuer, Germán Rodríguez Criado, Catherine Laprise, Celia M. T. Greenwood, and Anna K. Naumova
- Subjects
DNA methylation ,Sex ,X chromosome ,Y chromosome ,Medicine ,Physiology ,QP1-981 - Abstract
Abstract Background Sexual dimorphism in DNA methylation levels is a recurrent epigenetic feature in different human cell types and has been implicated in predisposition to disease, such as psychiatric and autoimmune disorders. To elucidate the genetic origins of sex-specific DNA methylation, we examined DNA methylation levels in fibroblast cell lines and blood cells from individuals with different combinations of sex chromosome complements and sex phenotypes focusing on a single autosomal region––the differentially methylated region (DMR) in the promoter of the zona pellucida binding protein 2 (ZPBP2) as a reporter. Results Our data show that the presence of the sex determining region Y (SRY) was associated with lower methylation levels, whereas higher X chromosome dosage in the absence of SRY led to an increase in DNA methylation levels at the ZPBP2 DMR. We mapped the X-linked modifier of DNA methylation to the long arm of chromosome X (Xq13-q21) and tested the impact of mutations in the ATRX and RLIM genes, located in this region, on methylation levels. Neither ATRX nor RLIM mutations influenced ZPBP2 methylation in female carriers. Conclusions We conclude that sex-specific methylation differences at the autosomal locus result from interaction between a Y-linked factor SRY and at least one X-linked factor that acts in a dose-dependent manner.
- Published
- 2018
- Full Text
- View/download PDF
6. Mutation p.R356Q in the Collybistin Phosphoinositide Binding Site Is Associated With Mild Intellectual Disability
- Author
-
Tzu-Ting Chiou, Philip Long, Alexandra Schumann-Gillett, Venkateswarlu Kanamarlapudi, Stefan A. Haas, Kirsten Harvey, Megan L. O’Mara, Angel L. De Blas, Vera M. Kalscheuer, and Robert J. Harvey
- Subjects
ARHGEF9 ,Collybistin ,Gephyrin ,PH domain ,PI3P ,XLID ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The recruitment of inhibitory GABAA receptors to neuronal synapses requires a complex interplay between receptors, neuroligins, the scaffolding protein gephyrin and the GDP-GTP exchange factor collybistin (CB). Collybistin is regulated by protein-protein interactions at the N-terminal SH3 domain, which can bind neuroligins 2/4 and the GABAAR α2 subunit. Collybistin also harbors a RhoGEF domain which mediates interactions with gephyrin and catalyzes GDP-GTP exchange on Cdc42. Lastly, collybistin has a pleckstrin homology (PH) domain, which binds phosphoinositides, such as phosphatidylinositol 3-phosphate (PI3P/PtdIns3P) and phosphatidylinositol 4-monophosphate (PI4P/PtdIns4P). PI3P located in early/sorting endosomes has recently been shown to regulate the postsynaptic clustering of gephyrin and GABAA receptors and consequently the strength of inhibitory synapses in cultured hippocampal neurons. This process is disrupted by mutations in the collybistin gene (ARHGEF9), which cause X-linked intellectual disability (XLID) by a variety of mechanisms converging on disrupted gephyrin and GABAA receptor clustering at central synapses. Here we report a novel missense mutation (chrX:62875607C>T, p.R356Q) in ARHGEF9 that affects one of the two paired arginine residues in the PH domain that were predicted to be vital for binding phosphoinositides. Functional assays revealed that recombinant collybistin CB3SH3-R356Q was deficient in PI3P binding and was not able to translocate EGFP-gephyrin to submembrane microaggregates in an in vitro clustering assay. Expression of the PI3P-binding mutants CB3SH3-R356Q and CB3SH3-R356N/R357N in cultured hippocampal neurones revealed that the mutant proteins did not accumulate at inhibitory synapses, but instead resulted in a clear decrease in the overall number of synaptic gephyrin clusters compared to controls. Molecular dynamics simulations suggest that the p.R356Q substitution influences PI3P binding by altering the range of structural conformations adopted by collybistin. Taken together, these results suggest that the p.R356Q mutation in ARHGEF9 is the underlying cause of XLID in the probands, disrupting gephyrin clustering at inhibitory GABAergic synapses via loss of collybistin PH domain phosphoinositide binding.
- Published
- 2019
- Full Text
- View/download PDF
7. Novel Missense Mutation A789V in IQSEC2 Underlies X-Linked Intellectual Disability in the MRX78 Family
- Author
-
Vera M. Kalscheuer, Victoria M. James, Miranda L. Himelright, Philip Long, Renske Oegema, Corinna Jensen, Melanie Bienek, Hao Hu, Stefan A. Haas, Maya Topf, A. Jeannette M. Hoogeboom, Kirsten Harvey, Randall Walikonis, and Robert J. Harvey
- Subjects
ArfGEF ,BRAG1 ,IQ-ArfGEF ,IQSEC2 ,MRX78 ,XLID ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Disease gene discovery in neurodevelopmental disorders, including X-linked intellectual disability (XLID) has recently been accelerated by next-generation DNA sequencing approaches. To date, more than 100 human X chromosome genes involved in neuronal signaling pathways and networks implicated in cognitive function have been identified. Despite these advances, the mutations underlying disease in a large number of XLID families remained unresolved. We report the resolution of MRX78, a large family with six affected males and seven affected females, showing X-linked inheritance. Although a previous linkage study had mapped the locus to the short arm of chromosome X (Xp11.4-p11.23), this region contained too many candidate genes to be analyzed using conventional approaches. However, our X-chromosome exome resequencing, bioinformatics analysis and inheritance testing revealed a missense mutation (c.C2366T, p.A789V) in IQSEC2, encoding a neuronal GDP-GTP exchange factor for Arf family GTPases (ArfGEF) previously implicated in XLID. Molecular modeling of IQSEC2 revealed that the A789V substitution results in the insertion of a larger side-chain into a hydrophobic pocket in the catalytic Sec7 domain of IQSEC2. The A789V change is predicted to result in numerous clashes with adjacent amino acids and disruption of local folding of the Sec7 domain. Consistent with this finding, functional assays revealed that recombinant IQSEC2A789V was not able to catalyze GDP-GTP exchange on Arf6 as efficiently as wild-type IQSEC2. Taken together, these results strongly suggest that the A789V mutation in IQSEC2 is the underlying cause of XLID in the MRX78 family.
- Published
- 2016
- Full Text
- View/download PDF
8. Aberrant phase separation and nucleolar dysfunction in rare genetic diseases
- Author
-
Martin A. Mensah, Henri Niskanen, Alexandre P. Magalhaes, Shaon Basu, Martin Kircher, Henrike L. Sczakiel, Alisa M. V. Reiter, Jonas Elsner, Peter Meinecke, Saskia Biskup, Brian H. Y. Chung, Gregor Dombrowsky, Christel Eckmann-Scholz, Marc Phillip Hitz, Alexander Hoischen, Paul-Martin Holterhus, Wiebke Hülsemann, Kimia Kahrizi, Vera M. Kalscheuer, Anita Kan, Mandy Krumbiegel, Ingo Kurth, Jonas Leubner, Ann Carolin Longardt, Jörg D. Moritz, Hossein Najmabadi, Karolina Skipalova, Lot Snijders Blok, Andreas Tzschach, Eberhard Wiedersberg, Martin Zenker, Carla Garcia-Cabau, René Buschow, Xavier Salvatella, Matthew L. Kraushar, Stefan Mundlos, Almuth Caliebe, Malte Spielmann, Denise Horn, and Denes Hnisz
- Subjects
All institutes and research themes of the Radboud University Medical Center ,Multidisciplinary ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Genetics ,Proteins ,Proteïnes ,Genètica - Abstract
Thousands of genetic variants in protein-coding genes have been linked to disease. However, the functional impact of most variants is unknown as they occur within intrinsically disordered protein regions that have poorly defined functions1–3. Intrinsically disordered regions can mediate phase separation and the formation of biomolecular condensates, such as the nucleolus4,5. This suggests that mutations in disordered proteins may alter condensate properties and function6–8. Here we show that a subset of disease-associated variants in disordered regions alter phase separation, cause mispartitioning into the nucleolus and disrupt nucleolar function. We discover de novo frameshift variants in HMGB1 that cause brachyphalangy, polydactyly and tibial aplasia syndrome, a rare complex malformation syndrome. The frameshifts replace the intrinsically disordered acidic tail of HMGB1 with an arginine-rich basic tail. The mutant tail alters HMGB1 phase separation, enhances its partitioning into the nucleolus and causes nucleolar dysfunction. We built a catalogue of more than 200,000 variants in disordered carboxy-terminal tails and identified more than 600 frameshifts that create arginine-rich basic tails in transcription factors and other proteins. For 12 out of the 13 disease-associated variants tested, the mutation enhanced partitioning into the nucleolus, and several variants altered rRNA biogenesis. These data identify the cause of a rare complex syndrome and suggest that a large number of genetic variants may dysregulate nucleoli and other biomolecular condensates in humans.
- Published
- 2023
9. Functional and clinical studies reveal pathophysiological complexity of CLCN4-related neurodevelopmental condition
- Author
-
Elizabeth E. Palmer, Michael Pusch, Alessandra Picollo, Caitlin Forwood, Matthew H. Nguyen, Vanessa Suckow, Jessica Gibbons, Alva Hoff, Lisa Sigfrid, Andre Megarbane, Mathilde Nizon, Benjamin Cogné, Claire Beneteau, Fowzan S. Alkuraya, Aziza Chedrawi, Mais O. Hashem, Hannah Stamberger, Sarah Weckhuysen, Arnaud Vanlander, Berten Ceulemans, Sulekha Rajagopalan, Kenneth Nunn, Stéphanie Arpin, Martine Raynaud, Constance S. Motter, Catherine Ward-Melver, Katrien Janssens, Marije Meuwissen, Diane Beysen, Nicola Dikow, Mona Grimmel, Tobias B. Haack, Emma Clement, Amy McTague, David Hunt, Sharron Townshend, Michelle Ward, Linda J. Richards, Cas Simons, Gregory Costain, Lucie Dupuis, Roberto Mendoza-Londono, Tracy Dudding-Byth, Jackie Boyle, Carol Saunders, Emily Fleming, Salima El Chehadeh, Marie-Aude Spitz, Amelie Piton, Bénédicte Gerard, Marie-Thérèse Abi Warde, Gillian Rea, Caoimhe McKenna, Sofia Douzgou, Siddharth Banka, Cigdem Akman, Jennifer M. Bain, Tristan T. Sands, Golder N. Wilson, Erin J. Silvertooth, Lauren Miller, Damien Lederer, Rani Sachdev, Rebecca Macintosh, Olivier Monestier, Deniz Karadurmus, Felicity Collins, Melissa Carter, Luis Rohena, Marjolein H. Willemsen, Charlotte W. Ockeloen, Rolph Pfundt, Sanne D. Kroft, Michael Field, Francisco E. R. Laranjeira, Ana M. Fortuna, Ana R. Soares, Vincent Michaud, Sophie Naudion, Sailaja Golla, David D. Weaver, Lynne M. Bird, Jennifer Friedman, Virginia Clowes, Shelagh Joss, Laura Pölsler, Philippe M. Campeau, Maria Blazo, Emilia K. Bijlsma, Jill A. Rosenfeld, Christian Beetz, Zöe Powis, Kirsty McWalter, Tracy Brandt, Erin Torti, Mikaël Mathot, Shekeeb S. Mohammad, Ruth Armstrong, Vera M. Kalscheuer, UCL - SSS/IREC/MONT - Pôle Mont Godinne, UCL - (MGD) Service de pédiatrie, Growth and Development, Pediatrics, Centre for Medical Genetics, Brussels Heritage Lab, and Medical Genetics
- Subjects
Male ,DISRUPTION ,Chloride Channels/genetics ,EXCHANGER ,Mutation, Missense ,LYSOSOMAL STORAGE DISEASE ,VARIANTS ,Neurodevelopmental Disorders/genetics ,PHENOTYPE ,Cellular and Molecular Neuroscience ,All institutes and research themes of the Radboud University Medical Center ,Genes, X-Linked ,CLC CHLORIDE ,Medicine and Health Sciences ,Humans ,Molecular Biology ,MUTATION ,Biology ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,CHANNELS ,LINKED MENTAL-RETARDATION ,ASSOCIATION ,GENE ,Psychiatry and Mental health ,Chemistry ,Female ,Human medicine - Abstract
Missense and truncating variants in the X-chromosome-linked CLCN4 gene, resulting in reduced or complete loss-of-function (LOF) of the encoded chloride/proton exchanger ClC-4, were recently demonstrated to cause a neurocognitive phenotype in both males and females. Through international clinical matchmaking and interrogation of public variant databases we assembled a database of 90 rare CLCN4 missense variants in 90 families: 41 unique and 18 recurrent variants in 49 families. For 43 families, including 22 males and 33 females, we collated detailed clinical and segregation data. To confirm causality of variants and to obtain insight into disease mechanisms, we investigated the effect on electrophysiological properties of 59 of the variants in Xenopus oocytes using extended voltage and pH ranges. Detailed analyses revealed new pathophysiological mechanisms: 25% (15/59) of variants demonstrated LOF, characterized by a “shift” of the voltage-dependent activation to more positive voltages, and nine variants resulted in a toxic gain-of-function, associated with a disrupted gate allowing inward transport at negative voltages. Functional results were not always in line with in silico pathogenicity scores, highlighting the complexity of pathogenicity assessment for accurate genetic counselling. The complex neurocognitive and psychiatric manifestations of this condition, and hitherto under-recognized impacts on growth, gastrointestinal function, and motor control are discussed. Including published cases, we summarize features in 122 individuals from 67 families with CLCN4-related neurodevelopmental condition and suggest future research directions with the aim of improving the integrated care for individuals with this diagnosis.
- Published
- 2023
- Full Text
- View/download PDF
10. Single cell, whole embryo phenotyping of pleiotropic disorders of mammalian development
- Author
-
Xingfan Huang, Jana Henck, Chengxiang Qiu, Varun K. A. Sreenivasan, Saranya Balachandran, Rose Behncke, Wing-Lee Chan, Alexandra Despang, Diane E. Dickel, Natja Haag, Rene Hägerling, Nils Hansmeier, Friederike Hennig, Cooper Marshall, Sudha Rajderkar, Alessa Ringel, Michael Robson, Lauren Saunders, Sanjay R. Srivatsan, Sascha Ulferts, Lars Wittler, Yiwen Zhu, Vera M. Kalscheuer, Daniel Ibrahim, Ingo Kurth, Uwe Kornak, David R. Beier, Axel Visel, Len A. Pennacchio, Cole Trapnell, Junyue Cao, Jay Shendure, and Malte Spielmann
- Abstract
Mouse models are a critical tool for studying human diseases, particularly developmental disorders, as well as for advancing our general understanding of mammalian biology. However, it has long been suspected that conventional approaches for phenotyping are insufficiently sensitive to detect subtle defects throughout the developing mouse. Here we set out to establish single cell RNA sequencing (sc-RNA-seq) of the whole embryo as a scalable platform for the systematic molecular and cellular phenotyping of mouse genetic models. We applied combinatorial indexing-based sc-RNA-seq to profile 101 embryos of 26 genotypes at embryonic stage E13.5, altogether profiling gene expression in over 1.6M nuclei. The 26 genotypes include 22 mouse mutants representing a range of anticipated severities, from established multisystem disorders to deletions of individual enhancers, as well as the 4 wildtype backgrounds on which these mutants reside. We developed and applied several analytical frameworks for detecting differences in composition and/or gene expression across 52 cell types or trajectories. Some mutants exhibited changes in dozens of trajectories (e.g., the pleiotropic consequences of altering the Sox9 regulatory landscape) whereas others showed phenotypes affecting specific subsets of cells. We also identify differences between widely used wildtype strains, compare phenotyping of gain vs. loss of function mutants, and characterise deletions of topological associating domain (TAD) boundaries. Intriguingly, even among these 22 mutants, some changes are shared by heretofore unrelated models, suggesting that developmental pleiotropy might be “decomposable” through further scaling of this approach. Overall, our findings show how single cell profiling of whole embryos can enable the systematic molecular and cellular phenotypic characterization of mouse mutants with unprecedented breadth and resolution.
- Published
- 2022
- Full Text
- View/download PDF
11. Detecting genomic indel variants with exact breakpoints in single- and paired-end sequencing data using SplazerS.
- Author
-
Anne-Katrin Emde, Marcel H. Schulz, David Weese, Ruping Sun, Martin Vingron, Vera M. Kalscheuer, Stefan A. Haas, and Knut Reinert
- Published
- 2012
- Full Text
- View/download PDF
12. Novel pathogenic <scp> EIF2S3 </scp> missense variants causing clinically variable <scp>MEHMO</scp> syndrome with impaired <scp>eIF2γ</scp> translational function, and literature review
- Author
-
Matias Wagner, Sara K. Young-Baird, Urania Kotzaeridou, Thomas E. Dever, Tim M. Strom, Vera M. Kalscheuer, Vanessa Suckow, Alexis G. Thornburg, Inga Harting, and Stine Christ
- Subjects
0301 basic medicine ,Genetics ,eIF2 ,Microcephaly ,Translation (biology) ,Hypopituitarism ,030105 genetics & heredity ,Biology ,medicine.disease ,Phenotype ,03 medical and health sciences ,030104 developmental biology ,Diabetes mellitus ,medicine ,Missense mutation ,EIF2S3 ,Genetics (clinical) - Abstract
Rare pathogenic EIF2S3 missense and terminal deletion variants cause the X-linked intellectual disability (ID) syndrome MEHMO, or a milder phenotype including pancreatic dysfunction and hypopituitarism. We present two unrelated male patients who carry novel EIF2S3 pathogenic missense variants (p.(Thr144Ile) and p.(Ile159Leu)) thereby broadening the limited genetic spectrum and underscoring clinically variable expressivity of MEHMO. While the affected male with p.(Thr144Ile) presented with severe motor delay, severe microcephaly, moderate ID, epileptic seizures responsive to treatments, hypogenitalism, central obesity, facial features, and diabetes, the affected male with p.(Ile159Leu) presented with moderate ID, mild motor delay, microcephaly, epileptic seizures resistant to treatment, central obesity, and mild facial features. Both variants are located in the highly conserved guanine nucleotide binding domain of the EIF2S3 encoded eIF2γ subunit of the heterotrimeric translation initiation factor 2 (eIF2) complex. Further, we investigated both variants in a structural model and in yeast. The reduced growth rates and lowered fidelity of translation with increased initiation at non-AUG codons observed for both mutants in these studies strongly support pathogenicity of the variants.
- Published
- 2020
- Full Text
- View/download PDF
13. A missense mutation in the CSTF2 gene that impairs the function of the RNA recognition motif and causes defects in 3′ end processing is associated with intellectual disability in humans
- Author
-
Pierre Billuart, Clinton C. MacDonald, Vera M. Kalscheuer, Thierry Bienvenu, Michael P. Latham, Petar N. Grozdanov, Marie-Ange Delrue, Elahe Masoumzadeh, Texas Tech University Health Sciences Center, Texas Tech University [Lubbock] (TTU), Max Planck Institute for Molecular Genetics (MPIMG), Max-Planck-Gesellschaft, Institut de psychiatrie et neurosciences de Paris (IPNP - U1266 Inserm), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), CHU Sainte Justine [Montréal], Martinez Rico, Clara, and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)
- Subjects
Male ,Polyadenylation ,AcademicSubjects/SCI00010 ,[SDV]Life Sciences [q-bio] ,Mutation, Missense ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Intellectual Disability ,Genetics ,Missense mutation ,Animals ,Humans ,Child ,Gene ,Molecular Biology ,3' Untranslated Regions ,030304 developmental biology ,0303 health sciences ,Messenger RNA ,RNA recognition motif ,MRNA cleavage ,Three prime untranslated region ,RNA ,Brain ,Pedigree ,[SDV] Life Sciences [q-bio] ,Mice, Inbred C57BL ,Cleavage Stimulation Factor ,Female ,030217 neurology & neurosurgery ,RNA Recognition Motif ,HeLa Cells ,Protein Binding - Abstract
CSTF2 encodes an RNA-binding protein that is essential for mRNA cleavage and polyadenylation (C/P). No disease-associated mutations have been described for this gene. Here, we report a mutation in the RNA recognition motif (RRM) of CSTF2 that changes an aspartic acid at position 50 to alanine (p.D50A), resulting in intellectual disability in male patients. In mice, this mutation was sufficient to alter polyadenylation sites in over 1300 genes critical for brain development. Using a reporter gene assay, we demonstrated that C/P efficiency of CSTF2D50A was lower than wild type. To account for this, we determined that p.D50A changed locations of amino acid side chains altering RNA binding sites in the RRM. The changes modified the electrostatic potential of the RRM leading to a greater affinity for RNA. These results highlight the significance of 3′ end mRNA processing in expression of genes important for brain plasticity and neuronal development.
- Published
- 2020
- Full Text
- View/download PDF
14. Hi-C Identifies Complex Genomic Rearrangements and TAD-Shuffling in Developmental Diseases
- Author
-
Wiebke Hülsemann, Sérgio B. Sousa, Seval Türkmen, Pedro Louro, Vera M. Kalscheuer, Martin Vingron, Anna Latos-Bielenska, Marius-Konstantin Klever, Stefan Mundlos, Manuel Holtgrewe, Andreas Dufke, Björn Fischer-Zirnsak, Malte Spielmann, Fabiola Quintero-Rivera, Martin A. Mensah, Rocio Acuna-Hidalgo, Verena Heinrich, Eunice Matoso, Ilina D. Pluym, Uirá Souto Melo, Monika Cohen, and Robert Schöpflin
- Subjects
Developmental Disabilities ,Molecular Conformation ,Chromosomal translocation ,ectopic enhancer-promoter interactions ,Medical and Health Sciences ,cytogenetics ,Translocation, Genetic ,Cohort Studies ,Chromosome conformation capture ,Chromosome Breakpoints ,Segmental Duplications, Genomic ,0302 clinical medicine ,Hi-C ,Chromosomes, Human ,Genetics (clinical) ,Genetics & Heredity ,0303 health sciences ,Genome ,SOX9 Transcription Factor ,Biological Sciences ,Phenotype ,Segmental Duplications ,chromosome conformation capture ,Human ,Biotechnology ,medicine.medical_specialty ,Translocation ,Locus (genetics) ,Computational biology ,topologically associating domains ,Biology ,Chromosomes ,Article ,03 medical and health sciences ,Genetic ,neo-TAD ,gene misregulation ,Genetics ,medicine ,Humans ,developmental disorders ,030304 developmental biology ,Genome, Human ,Human Genome ,Breakpoint ,Cytogenetics ,Chromatin Assembly and Disassembly ,Genomic ,Human genome ,030217 neurology & neurosurgery ,Comparative genomic hybridization - Abstract
Genome-wide analysis methods, such as array comparative genomic hybridization (CGH) and whole-genome sequencing (WGS), have greatly advanced the identification of structural variants (SVs) in the human genome. However, even with standard high-throughput sequencing techniques, complex rearrangements with multiple breakpoints are often difficult to resolve, and predicting their effects on gene expression and phenotype remains a challenge. Here, we address these problems by using high-throughput chromosome conformation capture (Hi-C) generated from cultured cells of nine individuals with developmental disorders (DDs). Three individuals had previously been identified as harboring duplications at the SOX9 locus and six had been identified with translocations. Hi-C resolved the positions of the duplications and was instructive in interpreting their distinct pathogenic effects, including the formation of new topologically associating domains (neo-TADs). Hi-C was very sensitive in detecting translocations, and it revealed previously unrecognized complex rearrangements at the breakpoints. In several cases, we observed the formation of fused-TADs promoting ectopic enhancer-promoter interactions that were likely to be involved in the disease pathology. In summary, we show that Hi-C is a sensible method for the detection of complex SVs in a clinical setting. The results help interpret the possible pathogenic effects of the SVs in individuals with DDs.
- Published
- 2020
- Full Text
- View/download PDF
15. Systematic analysis and prediction of genes associated with disorders on chromosome X
- Author
-
Elsa Leitão, Christopher Schröder, Ilaria Parenti, Carine Dalle, Agnès Rastetter, Theresa Kühnel, Alma Kuechler, Sabine Kaya, Bénédicte Gérard, Elise Schaefer, Caroline Nava, Nathalie Drouot, Camille Engel, Juliette Piard, Bénédicte Duban-Bedu, Laurent Villard, Alexander P.A. Stegmann, Els K. Vanhoutte, Job A.J Verdonshot, Frank J. Kaiser, Frédéric Tran Mau-Them, Marcello Scala, Pasquale Striano, Suzanna G.M. Frints, Emanuela Argilli, Elliott H. Sherr, Fikret Elder, Julien Buratti, Boris Keren, Cyril Mignot, Delphine Héron, Jean-Louis Mandel, Jozef Gecz, Vera M. Kalscheuer, Bernhard Horsthemke, Amélie Piton, and Christel Depienne
- Abstract
Disease gene discovery on chromosome (chr) X is challenging owing to its unique modes of inheritance. We undertook a systematic analysis of human chrX genes. We observe a higher proportion of disorder-associated genes and an enrichment of genes involved in cognition, language, and seizures on chrX compared to autosomes. We analyze gene constraints, exon and promoter conservation, expression and paralogues, and report 127 genes sharing one or more attributes with known chrX disorder genes. Using a neural network trained to distinguish disease-associated from dispensable genes, we classify 235 genes, including 121 of the 127, as having high probability of being disease-associated. We provide evidence of an excess of variants in predicted genes in existing databases. Finally, we report damaging variants in CDK16 and TRPC5 in patients with intellectual disability or autism spectrum disorders. This study predicts large-scale gene-disease associations that could be used for prioritization of X-linked pathogenic variants.
- Published
- 2022
- Full Text
- View/download PDF
16. Pathogenic variants in E3 ubiquitin ligase RLIM/RNF12 lead to a syndromic X-linked intellectual disability and behavior disorder
- Author
-
Kees E. P. van Roozendaal, Molka Kammoun, Michael Field, Andreas Dufke, Joris Vermeesch, Annick Toutain, Hao Hu, Theresa Mihalic Mosher, Joep P.M. Geraedts, Hans-Hilger Ropers, Peter White, Jan Liebelt, Sungjin Moon, Vera M. Kalscheuer, Joost Gribnau, Bas de Hoon, Germán Rodríguez Criado, Marie Shaw, Ute Grasshoff, Stefan A. Haas, Benjamin J. Kelly, Lynne Hobson, Marjan De Rademaeker, Christelle Golzio, Suzanna G.M. Frints, Olaf Riess, Claudia S. Bauer, Eric Haan, Nicholas Katsanis, Peter Bauer, Karen W. Gripp, Renee Carroll, Jozef Gecz, Jean Pierre Fryns, Cristina Gontan, Aysegul Ozanturk, Eveline Rentmeester, Martine Raynaud, Scott E. Hickey, Daniel C. Koboldt, Sylvie Manouvrier-Hanu, Lucinda Murray, Koen Devriendt, Christopher Schroeder, Kathryn Friend, Developmental Biology, Obstetrics & Gynecology, MUMC+: DA KG Bedrijfsbureau (9), RS: GROW - R4 - Reproductive and Perinatal Medicine, Klinische Genetica, MUMC+: DA KG Lab Centraal Lab (9), Maastricht University Medical Centre (MUMC), Maastricht University [Maastricht], Duke University [Durham], Hospital Universitario Virgen del Rocío [Sevilla], University of Tübingen, Erasmus University Medical Center [Rotterdam] (Erasmus MC), Hunter Genetics, Clinique de Génétique médicale Guy Fontaine [CHRU LIlle], Hôpital Jeanne de Flandres, Université de Lille, Droit et Santé-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Maladies RAres du DEveloppement embryonnaire et du MEtabolisme : du Phénotype au Génotype et à la Fonction - ULR 7364 (RADEME), Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Nationwide Children's Hospital, Ohio State University [Columbus] (OSU), University Hospitals Leuven [Leuven], Nemours/Alfred I. du Pont Hospital for Children, Hôpital Bretonneau, Centre Hospitalier Régional Universitaire de Tours (CHRU Tours), Imagerie et cerveau (iBrain - Inserm U1253 - UNIV Tours ), Université de Tours (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM), Max Planck Institute for Molecular Genetics (MPIMG), Max-Planck-Gesellschaft, University of Adelaide, Women’s and Children’s Hospital [Adelaide], SA Pathology [Adelaide, SA, Australia], Vrije Universiteit Brussel (VUB), South Australian Health and Medical Research Institute [ Adelaide] (SAHMRI), 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), univOAK, Archive ouverte, Reproduction and Genetics, Clinical sciences, and Medical Genetics
- Subjects
Male ,0301 basic medicine ,X-linked intellectual disability ,PROTEIN ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,FUNCTIONAL-ACTIVITY ,Mice ,0302 clinical medicine ,Genes, X-Linked ,X Chromosome Inactivation ,RNF12 ,Missense mutation ,TRANSCRIPTION ,Child ,Zebrafish ,Genetics ,Middle Aged ,Phenotype ,Pedigree ,Ubiquitin ligase ,Psychiatry and Mental health ,medicine.anatomical_structure ,Child, Preschool ,Female ,Adult ,Conduct Disorder ,Adolescent ,Ubiquitin-Protein Ligases ,NPAS3 ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Intellectual Disability ,medicine ,Ring finger ,Animals ,Humans ,Molecular Biology ,Transcription factor ,RLIM ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,CHROMOSOME INACTIVATION ,MUTATIONS ,Infant, Newborn ,Ubiquitination ,Wild type ,Zebrafish Proteins ,medicine.disease ,biology.organism_classification ,HEK293 Cells ,030104 developmental biology ,Mutation ,Mental Retardation, X-Linked ,biology.protein ,LIM COFACTORS ,030217 neurology & neurosurgery ,Transcription Factors ,GENE UBE2A CAUSE - Abstract
RLIM, also known as RNF12, is an X-linked E3 ubiquitin ligase acting as a negative regulator of LIM-domain containing transcription factors and participates in X-chromosome inactivation (XCI) in mice. We report the genetic and clinical findings of 84 individuals from nine unrelated families, eight of whom who have pathogenic variants in RLIM (RING finger LIM domain-interacting protein). A total of 40 affected males have X-linked intellectual disability (XLID) and variable behavioral anomalies with or without congenital malformations. In contrast, 44 heterozygous female carriers have normal cognition and behavior, but eight showed mild physical features. All RLIM variants identified are missense changes co-segregating with the phenotype and predicted to affect protein function. Eight of the nine altered amino acids are conserved and lie either within a domain essential for binding interacting proteins or in the C-terminal RING finger catalytic domain. In vitro experiments revealed that these amino acid changes in the RLIM RING finger impaired RLIM ubiquitin ligase activity. In vivo experiments in rlim mutant zebrafish showed that wild type RLIM rescued the zebrafish rlim phenotype, whereas the patient-specific missense RLIM variants failed to rescue the phenotype and thus represent likely severe loss-of-function mutations. In summary, we identified a spectrum of RLIM missense variants causing syndromic XLID and affecting the ubiquitin ligase activity of RLIM, suggesting that enzymatic activity of RLIM is required for normal development, cognition and behavior.
- Published
- 2019
- Full Text
- View/download PDF
17. Novel pathogenic EIF2S3 missense variants causing clinically variable MEHMO syndrome with impaired eIF2γ translational function, and literature review
- Author
-
Urania, Kotzaeridou, Sara K, Young-Baird, Vanessa, Suckow, Alexis G, Thornburg, Matias, Wagner, Inga, Harting, Stine, Christ, Tim, Strom, Thomas E, Dever, and Vera M, Kalscheuer
- Subjects
Male ,Epilepsy ,Adolescent ,Hypogonadism ,Eukaryotic Initiation Factor-2 ,Mutation, Missense ,Infant ,Article ,Child, Preschool ,Protein Biosynthesis ,Eif2gamma ,Intellectual Disability ,Mehmo ,X-linked ,Mutation ,Mental Retardation, X-Linked ,Microcephaly ,Humans ,Female ,Genetic Predisposition to Disease ,Genitalia ,Obesity ,Child - Abstract
Rare pathogenic EIF2S3 missense and terminal deletion variants cause the X-linked intellectual disability (ID) syndrome MEHMO, or a milder phenotype including pancreatic dysfunction and hypopituitarism. We present two unrelated male patients who carry novel EIF2S3 pathogenic missense variants (p.(Thr144Ile) and p.(Ile159Leu)) thereby broadening the limited genetic spectrum and underscoring clinically variable expressivity of MEHMO. While the affected male with p.(Thr144Ile) presented with severe cognitive and motor delay, severe microcephaly, epileptic seizures responsive to treatments, hypogenitalism, central obesity, facial dysmorphic features, and diabetes, the affected male with p.(Ile159Leu) presented with moderate ID, mild motor delay, microcephaly, epileptic seizures resistant to treatment, central obesity, and mild dysmorphic facial features. Both variants are located in the highly conserved guanine nucleotide-binding domain of the EIF2S3 encoded eIF2γ subunit of the heterotrimeric translation initiation factor 2 (eIF2) complex. Further, we investigated both variants in a structural model and in yeast. The reduced growth rates and lowered fidelity of translation with increased initiation at non-AUG codons observed for both mutants in these studies strongly support pathogenicity of the variants.
- Published
- 2020
18. Whole genome sequencing identifies a duplicated region encompassing Xq13.2q13.3 in a large Iranian family with intellectual disability
- Author
-
Hans-Hilger Ropers, Sepideh Mehvari, Zohreh Fattahi, Maryam Beheshtian, Daniel Auld, Hossein Najmabadi, Yasser Riazalhosseini, Kimia Kahrizi, Farzaneh Larti, Vera M. Kalscheuer, Sanaz Arzhangi, Seyedeh Sedigheh Abedini, and Hao Hu
- Subjects
0301 basic medicine ,Male ,Monocarboxylic Acid Transporters ,Candidate gene ,lcsh:QH426-470 ,Ubiquitin-Protein Ligases ,Nerve Tissue Proteins ,030105 genetics & heredity ,Biology ,Short stature ,Clinical Reports ,03 medical and health sciences ,Genetic linkage ,Intellectual disability ,Gene duplication ,Chromosome Duplication ,Genetics ,medicine ,Humans ,Copy-number variation ,Genetic Testing ,Molecular Biology ,Genetics (clinical) ,X chromosome ,Whole genome sequencing ,Chromosomes, Human, X ,whole genome sequencing ,Clinical Report ,Symporters ,Xq13.2q13.3 ,Genetic Diseases, X-Linked ,Xq duplication ,medicine.disease ,Pedigree ,lcsh:Genetics ,030104 developmental biology ,intellectual disability ,medicine.symptom - Abstract
Background The X chromosome has historically been one of the most thoroughly investigated chromosomes regarding intellectual disability (ID), whose etiology is attributed to many factors including copy number variations (CNVs). Duplications of the long arm of the X chromosome have been reported in patients with ID, short stature, facial anomalies, and in many cases hypoplastic genitalia and/or behavioral abnormalities. Methods Here, we report on a large Iranian family with X‐linked ID caused by a duplication on the X chromosome identified by whole genome sequencing in combination with linkage analysis. Results Seven affected males in different branches of the family presented with ID, short stature, seizures, facial anomalies, behavioral abnormalities (aggressiveness, self‐injury, anxiety, impaired social interactions, and shyness), speech impairment, and micropenis. The duplication of the region Xq13.2q13.3, which is ~1.8 Mb in size, includes seven protein‐coding OMIM genes. Three of these genes, namely SLC16A2, RLIM, and NEXMIF, if impaired, can lead to syndromes presenting with ID. Of note, this duplicated region was located within a linkage interval with a LOD score >3. Conclusion Our report indicates that CNVs should be considered in multi‐affected families where no candidate gene defect has been identified in sequencing data analysis., We report a duplication on the X chromosome encompassing Xq13.2q13.3 in a large Iranian family with X‐linked intellectual disability. There are seven affected males in ones and twos within two generations of the pedigree who presented with a similar phenotype, including ID, short stature, seizures, facial anomalies, behavioral abnormalities, speech impairment, and micropenis.
- Published
- 2020
19. Author response for 'Comprehensive genotype-phenotype correlation in AP-4 deficiency syndrome; Adding data from a large cohort of Iranian patients'
- Author
-
Seyedeh Sedigheh Abedini, Vera M. Kalscheuer, Sanaz Arzhangi, Hossein Najmabadi, Zohreh Fattahi, Maryam Beheshtian, Reza Najafipour, Hao Hu, K. Kahrizi, Mahsa Fadaee, Hans-Hilger Ropers, Payman Jamali, Sepideh Mehvari, Tara Akhtarkhavari, and Marzieh Mohseni
- Subjects
Oncology ,Correlation ,medicine.medical_specialty ,Deficiency syndrome ,business.industry ,Internal medicine ,medicine ,business ,Genotype phenotype ,Large cohort - Published
- 2020
- Full Text
- View/download PDF
20. Author response for 'Novel pathogenic <scp> EIF2S3 </scp> missense variants causing clinically variable <scp>MEHMO</scp> syndrome with impaired <scp>eIF2γ</scp> translational function, and literature review'
- Author
-
Matias Wagner, Thomas E. Dever, Vera M. Kalscheuer, Stine Christ, Vanessa Suckow, Inga Harting, Alexis G. Thornburg, Urania Kotzaeridou, Tim M. Strom, and Sara K. Young-Baird
- Subjects
business.industry ,Missense mutation ,Medicine ,MEHMO syndrome ,business ,EIF2S3 ,Bioinformatics ,Function (biology) ,Variable (mathematics) - Published
- 2020
- Full Text
- View/download PDF
21. Comprehensive genotype-phenotype correlation in AP-4 deficiency syndrome; Adding data from a large cohort of Iranian patients
- Author
-
Seyedeh Sedigheh Abedini, Zohreh Fattahi, Kimia Kahrizi, Maryam Beheshtian, Hossein Najmabadi, Marzieh Mohseni, Sepideh Mehvari, Payman Jamali, Tara Akhtarkhavari, Hans-Hilger Ropers, Vera M. Kalscheuer, Hao Hu, Sanaz Arzhangi, Mahsa Fadaee, and Reza Najafipour
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Microcephaly ,Adolescent ,Adaptor Protein Complex 4 ,Consanguinity ,030105 genetics & heredity ,Iran ,Quadriplegia ,Corpus Callosum ,Cohort Studies ,03 medical and health sciences ,Internal medicine ,Intellectual Disability ,Intellectual disability ,Genetics ,medicine ,Humans ,Spasticity ,Global developmental delay ,Child ,Spastic tetraplegia ,Genetics (clinical) ,Genetic Association Studies ,business.industry ,Brain ,medicine.disease ,Phenotype ,Pedigree ,030104 developmental biology ,Child, Preschool ,Mutation ,Female ,medicine.symptom ,business ,Ventriculomegaly - Abstract
Mutations in adaptor protein complex‐4 (AP‐4) genes have first been identified in 2009, causing a phenotype termed as AP‐4 deficiency syndrome. Since then several patients with overlapping phenotypes, comprised of intellectual disability (ID) and spastic tetraplegia have been reported. To delineate the genotype‐phenotype correlation of the AP‐4 deficiency syndrome, we add the data from 30 affected individuals from 12 out of 640 Iranian families with ID in whom we detected disease‐causing variants in AP‐4 complex subunits, using next‐generation sequencing. Furthermore, by comparing genotype‐phenotype findings of those affected individuals with previously reported patients, we further refine the genotype‐phenotype correlation in this syndrome. The most frequent reported clinical findings in the 101 cases consist of ID and/or global developmental delay (97%), speech disorders (92.1%), inability to walk (90.1%), spasticity (77.2%), and microcephaly (75.2%). Spastic tetraplegia has been reported in 72.3% of the investigated patients. The major brain imaging findings are abnormal corpus callosum morphology (63.4%) followed by ventriculomegaly (44.5%). Our result might suggest the AP‐4 deficiency syndrome as a major differential diagnostic for unknown hereditary neurodegenerative disorders.
- Published
- 2020
22. A Point Mutation in the RNA Recognition Motif of CSTF2 Associated with Intellectual Disability in Humans Causes Defects in 3′ End Processing
- Author
-
Elahe Masoumzadeh, Pierre Billuart, Clinton C. MacDonald, Michael P. Latham, Vera M. Kalscheuer, Petar N. Grozdanov, Thierry Bienvenu, and Marie Ange Delrue
- Subjects
Genetics ,0303 health sciences ,Messenger RNA ,Polyadenylation ,RNA recognition motif ,MRNA cleavage ,Point mutation ,RNA ,RNA-binding protein ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
SUMMARYCSTF2 encodes an RNA-binding protein that is essential for mRNA cleavage and polyadenylation (C/P). No disease-associated mutations have been described for this gene. Here, we report a mutation in the RNA recognition motif (RRM) of CSTF2 that changes an aspartic acid at position 50 to alanine (p.D50A), resulting in intellectual disability in male patients. In mice, this mutation was sufficient to alter polyadenylation sites in over 1,000 genes critical for brain development. Using a reporter gene assay, we demonstrated that C/P efficiency of CSTF2D50A was lower than wild type. To account for this, we determined that p.D50A changed locations of amino acid side chains altering RNA binding sites in the RRM. The changes modified the electrostatic potential of the RRM leading to a greater affinity for RNA. These results highlight the importance of 3′ end mRNA processing in correct expression of genes important for brain plasticity and neuronal development.
- Published
- 2020
- Full Text
- View/download PDF
23. Effect of inbreeding on intellectual disability revisited by trio sequencing
- Author
-
Payman Jamali, Zhila Ghaderi, Hans-Hilger Ropers, Haleh Habibi, Fatemeh Pourfatemi, Farahnaz Sabbagh Kermani, Zohreh Mehrjoo, Kimia Kahrizi, Farnaz Sadeghinia, Hao Hu, Vera M. Kalscheuer, Bettina Lipkowitz, Reza Najafipour, Sanaz Arzhangi, Maryam Rahimi, Pooneh Nikuei, Atefeh Khoshaeen, Marzieh Mohseni, Masoumeh Hosseini, Hossein Najmabadi, Vanessa Suckow, Milad Falahat Chian, Faezeh Mojahedi, Sepideh Mehvari, Zohreh Fattahi, Maryam Beheshtian, Roshanak Jazayeri, Mohammad-Reza Khodaie-Ardakani, S. Hassan Tonekaboni, Tara Akhtarkhavari, and Thomas F. Wienker
- Subjects
Male ,0301 basic medicine ,Genes, Recessive ,Iran ,030105 genetics & heredity ,Biology ,Carrier testing ,Consanguinity ,Middle East ,03 medical and health sciences ,Intellectual Disability ,Exome Sequencing ,Intellectual disability ,Genetics ,medicine ,Humans ,Exome ,Family ,Inbreeding ,Genetics (clinical) ,De novo mutations ,Exome sequencing ,High rate ,Homozygote ,Disease gene identification ,medicine.disease ,Pedigree ,030104 developmental biology ,Parental consanguinity ,Mutation ,Female - Abstract
In outbred Western populations, most individuals with intellectual disability (ID) are sporadic cases, dominant de novo mutations (DNM) are frequent, and autosomal recessive ID (ARID) is very rare. Due to the high rate of parental consanguinity which raises the risk for ARID and other recessive disorders, the prevalence of ID is significantly higher in Near- and Middle-East countries. Indeed, homozygosity mapping and sequencing in consanguineous families have already identified a plethora of ARID genes, but due to the design of these studies, DNMs could not be systematically assessed, and the proportion of cases that are potentially preventable by avoiding consanguineous marriages or through carrier testing is hitherto unknown. This prompted us to perform whole exome sequencing in 100 sporadic ID patients from Iran and their healthy consanguineous parents. In 61 patients, we identified apparently causative changes in known ID genes. Of these, 44 were homozygous recessive and 17 dominant de novo mutations. Assuming that the DNM rate is stable, these results suggest that parental consanguinity raises the ID risk about 3.6-fold, and about 4.1-4.25-fold for children of first-cousin unions. These results do not rhyme with recent opinions that consanguinity-related health risks are generally small and have been 'overstated' in the past. This article is protected by copyright. All rights reserved.
- Published
- 2018
- Full Text
- View/download PDF
24. O-GlcNAc transferase missense mutations linked to X-linked intellectual disability deregulate genes involved in cell fate determination and signaling
- Author
-
Nithya Selvan, Shawn Levy, Charles E. Schwartz, Lynne Hobson, Marie Shaw, Juliet Taylor, Ahm M. Huq, Fatema J. Serajee, Nripesh Prasad, Jozef Gecz, Stephan N. George, Salim Aftimos, Lance Wells, and Vera M. Kalscheuer
- Subjects
0301 basic medicine ,Genetics ,education.field_of_study ,Mutation ,X-linked intellectual disability ,Cellular differentiation ,Population ,Glycobiology and Extracellular Matrices ,Cell Biology ,Biology ,medicine.disease ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,Tetratricopeptide ,030104 developmental biology ,medicine ,Missense mutation ,education ,Molecular Biology ,Gene ,Human embryonic stem cell line - Abstract
It is estimated that ∼1% of the world's population has intellectual disability, with males affected more often than females. OGT is an X-linked gene encoding for the enzyme O-GlcNAc transferase (OGT), which carries out the reversible addition of N-acetylglucosamine (GlcNAc) to Ser/Thr residues of its intracellular substrates. Three missense mutations in the tetratricopeptide (TPR) repeats of OGT have recently been reported to cause X-linked intellectual disability (XLID). Here, we report the discovery of two additional novel missense mutations (c.775 G>A, p.A259T, and c.1016 A>G, p.E339G) in the TPR domain of OGT that segregate with XLID in affected families. Characterization of all five of these XLID missense variants of OGT demonstrates modest declines in thermodynamic stability and/or activities of the variants. We engineered each of the mutations into a male human embryonic stem cell line using CRISPR/Cas9. Investigation of the global O-GlcNAc profile as well as OGT and O-GlcNAc hydrolase levels by Western blotting showed no gross changes in steady-state levels in the engineered lines. However, analyses of the differential transcriptomes of the OGT variant-expressing stem cells revealed shared deregulation of genes involved in cell fate determination and liver X receptor/retinoid X receptor signaling, which has been implicated in neuronal development. Thus, here we reveal two additional mutations encoding residues in the TPR regions of OGT that appear causal for XLID and provide evidence that the relatively stable and active TPR variants may share a common, unelucidated mechanism of altering gene expression profiles in human embryonic stem cells.
- Published
- 2018
- Full Text
- View/download PDF
25. Variant in the X-chromosome spliceosomal gene GPKOW causes male-lethal microcephaly with intrauterine growth restriction
- Author
-
Mark A. Corbett, Vera M. Kalscheuer, Renee Carroll, Jennie Slee, Raman Kumar, Marie Shaw, and Jozef Gecz
- Subjects
0301 basic medicine ,Male ,Microcephaly ,Spliceosome ,RNA Splicing ,Short Report ,030105 genetics & heredity ,Biology ,medicine.disease_cause ,03 medical and health sciences ,Pregnancy ,Genetics ,medicine ,Humans ,Allele ,Gene ,Genetics (clinical) ,X chromosome ,Cells, Cultured ,Mutation ,Fetal Growth Retardation ,Wild type ,RNA-Binding Proteins ,Genetic Diseases, X-Linked ,Syndrome ,medicine.disease ,Molecular biology ,030104 developmental biology ,RNA splicing ,Female - Abstract
Congenital microcephaly, with or without additional developmental defects, is a heterogeneous disorder resulting from impaired brain development during early fetal life. The majority of causative genetic variants identified thus far are inherited in an autosomal recessive manner and impact key cellular pathways such as mitosis, DNA damage response and repair, apoptosis and splicing. Here, we report a novel donor splice site variant in the G-patch domain and KOW motifs (GPKOW) gene (NG_021310.2:g.6126G>A, NM_015698.4:c.331+5G>A) that segregates with affected and carrier status in a multigenerational family with an X-linked perinatal lethal condition characterized by severe microcephaly and intrauterine growth restriction (IUGR). GPKOW is a core member of the spliceosome that has been shown in numerous model organisms and in human cells to be essential for survival. By investigating GPKOW transcripts in lymphoblastoid cell lines (LCLs) of three carrier females, we show that the GPKOW c.331+5G>A variant disrupts normal splicing of its pre-mRNAs. In a clonal culture expressing only the c.331+5G>A allele isolated from one carrier female LCL, we observed an 80% reduction in wild type GPKOW mRNA, 70% reduction in the full length GPKOW protein and the presence of a truncated GPKOW protein with possible dominant negative effect. Based on our and published data we propose that the GPKOW gene is essential for fetal development and when disrupted, leads to a severe, male-lethal phenotype characterised by microcephaly and IUGR.
- Published
- 2017
26. Multigenic truncation of the semaphorin–plexin pathway by a germline chromothriptic rearrangement associated with Moebius syndrome
- Author
-
Juan M M Mendez, Niels Tommerup, Dustin C. Hancks, Iben Bache, Mads Bak, Vera M. Kalscheuer, Lusine Nazaryan-Petersen, Mana M. Mehrjouy, Inés Oliveira, and Merete Bugge
- Subjects
Male ,Candidate gene ,animal structures ,In silico ,Semaphorins ,Chromosomal rearrangement ,Biology ,Article ,Germline ,Chromosome Breakpoints ,03 medical and health sciences ,Fatal Outcome ,Semaphorin ,Genetics ,Humans ,Germ-Line Mutation ,Genetics (clinical) ,030304 developmental biology ,Gene Rearrangement ,Chromothripsis ,0303 health sciences ,Membrane Glycoproteins ,030305 genetics & heredity ,Breakpoint ,Plexin ,Intracellular Signaling Peptides and Proteins ,High-Throughput Nucleotide Sequencing ,Semaphorin-3A ,Middle Aged ,Mobius Syndrome ,biology.protein - Abstract
Moebius syndrome (MBS) is a congenital disorder caused by paralysis of the facial and abducens nerves. While a number of candidate genes have been suspected, so far only mutations in PLXND1 and REV3L are confirmed to cause MBS. Here, we fine mapped the breakpoints of a complex chromosomal rearrangement (CCR) 46,XY,t(7;8;11;13) in a patient with MBS, which revealed 41 clustered breakpoints with typical hallmarks of chromothripsis. Among 12 truncated protein-coding genes, SEMA3A is known to bind to the MBS-associated PLXND1. Intriguingly, the CCR also truncated PIK3CG, which in silico interacts with REVL3 encoded by the other known MBS-gene REV3L, and with the SEMA3A/PLXND1 complex via FLT1. Additional studies of other complex rearrangements may reveal whether the multiple breakpoints in germline chromothripsis may predispose to complex multigenic disorders. This article is protected by copyright. All rights reserved.
- Published
- 2019
- Full Text
- View/download PDF
27. Author response for 'Identification of disease causing variants in the EXOSC gene family underlying autosomal recessive intellectual disability in Iranian families'
- Author
-
Raheleh Vazehan, Hans-Hilger Ropers, Elham Parsimehr, Zahra Kalhor, Seyedeh Sedighe Abedini, Mahboubeh Kamgar, Faezeh Mojahedi, K. Kahrizi, Ariana Kariminejad, Farahnaz Sabbagh Kermani, Mahsa Fadaee, Zohreh Fattahi, Vera M. Kalscheuer, Mehrshid Faraji Zonooz, Maryam Beheshtian, Sanaz Arzhangi, Shokouh Sadat Mahdavi, Payman Jamali, and Hossein Najmabadi
- Subjects
Genetics ,Intellectual disability ,medicine ,Gene family ,Identification (biology) ,Disease ,Biology ,medicine.disease - Published
- 2019
- Full Text
- View/download PDF
28. Identification of disease-causing variants in the EXOSC gene family underlying autosomal recessive intellectual disability in Iranian families
- Author
-
Shokouh Sadat Mahdavi, Ariana Kariminejad, Hossein Najmabadi, Raheleh Vazehan, Kimia Kahrizi, Payman Jamali, Seyedeh Sedigheh Abedini, Faezeh Mojahedi, Mehrshid Faraji Zonooz, Hans-Hilger Ropers, Vera M. Kalscheuer, Sanaz Arzhangi, Mahsa Fadaee, Zohreh Fattahi, Maryam Beheshtian, Zahra Kalhor, Mahboubeh Kamgar, Farahnaz Sabbagh Kermani, and Elham Parsimehr
- Subjects
0301 basic medicine ,Male ,Genes, Recessive ,Disease ,030105 genetics & heredity ,Biology ,Iran ,Cohort Studies ,03 medical and health sciences ,Consanguinity ,Intellectual Disability ,Clinical information ,Intellectual disability ,Exome Sequencing ,Genetics ,medicine ,Gene family ,Humans ,Family ,Child ,Genetics (clinical) ,Rna processing ,Exosome Multienzyme Ribonuclease Complex ,fungi ,Infant ,medicine.disease ,Phenotype ,humanities ,Pedigree ,030104 developmental biology ,Child, Preschool ,Mutation ,Neurodevelopmental delay ,Identification (biology) ,Female ,geographic locations - Abstract
Neurodevelopmental delay and intellectual disability (ID) can arise from numerous genetic defects. To date, variants in the EXOSC gene family have been associated with such disorders. Using next-generation sequencing (NGS), known and novel variants in this gene family causing autosomal recessive ID (ARID) have been identified in five Iranian families. By collecting clinical information on these families and comparing their phenotypes with previously reported patients, we further describe the clinical variability of ARID resulting from alterations in the EXOSC gene family, and emphasize the role of RNA processing dysregulation in ARID.
- Published
- 2019
29. A recurrent missense variant in SLC9A7 causes nonsyndromic X-linked intellectual disability with alteration of Golgi acidification and aberrant glycosylation
- Author
-
Anna Hackett, Jozef Gecz, Alina Ilie, Marie Shaw, Wujood Khayat, Tracy Dudding-Byth, Michael Field, Louise Christie, Brian Kirmse, John Orlowski, Mark A. Corbett, Jane Juusola, Vera M. Kalscheuer, and Kathryn Friend
- Subjects
Glycosylation ,Sodium-Hydrogen Exchangers ,X-linked intellectual disability ,Mutation, Missense ,Golgi Apparatus ,CHO Cells ,medicine.disease_cause ,Transfection ,03 medical and health sciences ,chemistry.chemical_compound ,symbols.namesake ,Cricetulus ,Viral Envelope Proteins ,Cricetinae ,Intellectual Disability ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Genetics (clinical) ,chemistry.chemical_classification ,0303 health sciences ,Mutation ,Membrane Glycoproteins ,biology ,Chinese hamster ovary cell ,030305 genetics & heredity ,Cell Membrane ,Genetic Diseases, X-Linked ,General Medicine ,Golgi apparatus ,medicine.disease ,biology.organism_classification ,Cell biology ,Transport protein ,Protein Transport ,chemistry ,Gene Expression Regulation ,Vesicular stomatitis virus ,symbols ,General Article ,Glycoprotein ,Acids ,trans-Golgi Network - Abstract
We report two unrelated families with multigenerational nonsyndromic intellectual disability segregating with a recurrent de novo missense variant (c.1543C>T:p.Leu515Phe) in the alkali cation/proton exchanger gene SLC9A7 (also commonly referred to as NHE7). SLC9A7 is located on human X chromosome at Xp11.3 and has not yet been associated with a human phenotype. The gene is widely transcribed, but especially abundant in brain, skeletal muscle and various secretory tissues. Within cells, SLC9A7 resides in the Golgi apparatus, with prominent enrichment in the trans-Golgi network (TGN) and post-Golgi vesicles. In transfected Chinese hamster ovary AP-1 cells, the Leu515Phe mutant protein was correctly targeted to the TGN/post-Golgi vesicles, but its N-linked oligosaccharide maturation as well as that of a co-transfected secretory membrane glycoprotein, vesicular stomatitis virus G (VSVG) glycoprotein, was reduced compared to cells co-expressing SLC9A7 wild-type and VSVG. This correlated with alkalinization of the TGN/post-Golgi compartments, suggestive of a gain-of-function. Membrane trafficking of glycosylation-deficient Leu515Phe and co-transfected VSVG to the cell surface, however, was relatively unaffected. Mass spectrometry analysis of patient sera also revealed an abnormal N-glycosylation profile for transferrin, a clinical diagnostic marker for congenital disorders of glycosylation. These data implicate a crucial role for SLC9A7 in the regulation of TGN/post-Golgi pH homeostasis and glycosylation of exported cargo which may underlie the cellular pathophysiology and neurodevelopmental deficits associated with this particular nonsyndromic form of X-linked intellectual
- Published
- 2019
30. Genetics of intellectual disability in consanguineous families
- Author
-
Sarah Azimi, Leila Nouri Vahid, Krystyna Keleman, Pooneh Nikuei, Tara Akhtarkhavari, Thomas F. Wienker, Beate Albrecht, Hossein Khodaei, Mohammad Reza Ebrahimpour, Mohammad Javad Soltani Banavandi, Marzieh Mohseni, Vanessa Suckow, Aria Jankhah, Milad Bastami, Behzad Davarnia, Vera M. Kalscheuer, Farzaneh Larti, Saeide Akbari, Kimia Kahrizi, Jamileh Rezazadeh Varaghchi, Bettina Lipkowitz, Sanaz Arzhangi, Morteza Oladnabi, Monika Cohen, Sabine Otto, Zohreh Fattahi, Luciana Musante, Payman Jamali, Maryam Beheshtian, Masoumeh Hosseini, Maryam Taghdiri, Wei Chen, Seyedeh Sedigheh Abedini, Bernd Timmermann, Hans-Hilger Ropers, Andreas Tzschach, Gholamreza Bahrami, Birgit Zirn, Hossein Najmabadi, Dagmar Wieczorek, Ingrid Bader, Gabriele Gillessen-Kaesbach, Cornelia Oppitz, Elaheh Papari, Hao Hu, Ralf Herwig, Fatemeh Pourfatemi, Jutta Gärtner, Faezeh Mojahedi, Hossein Dehghani, Sepideh Mehvari, and Seyed Hassan Tonekaboni
- Subjects
Adult ,Male ,0301 basic medicine ,Medizin ,Genes, Recessive ,Consanguinity ,Iran ,Biology ,DNA sequencing ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Intellectual Disability ,Exome Sequencing ,Intellectual disability ,medicine ,Humans ,Exome ,Family ,Protein Interaction Maps ,Molecular Biology ,Gene ,De novo mutations ,Affected offspring ,Whole genome sequencing ,Genetics ,Whole Genome Sequencing ,Homozygote ,High-Throughput Nucleotide Sequencing ,Middle Aged ,medicine.disease ,Pedigree ,Psychiatry and Mental health ,030104 developmental biology ,Mutation ,Female ,030217 neurology & neurosurgery - Abstract
Autosomal recessive (AR) gene defects are the leading genetic cause of intellectual disability (ID) in countries with frequent parental consanguinity, which account for about 1/7th of the world population. Yet, compared to autosomal dominant de novo mutations, which are the predominant cause of ID in Western countries, the identification of AR-ID genes has lagged behind. Here, we report on whole exome and whole genome sequencing in 404 consanguineous predominantly Iranian families with two or more affected offspring. In 219 of these, we found likely causative variants, involving 77 known and 77 novel AR-ID (candidate) genes, 21 X-linked genes, as well as 9 genes previously implicated in diseases other than ID. This study, the largest of its kind published to date, illustrates that high-throughput DNA sequencing in consanguineous families is a superior strategy for elucidating the thousands of hitherto unknown gene defects underlying AR-ID, and it sheds light on their prevalence.
- Published
- 2019
31. Deleterious de novo variants of X-linked ZC4H2 in females cause a variable phenotype with neurogenic arthrogryposis multiplex congenita
- Author
-
Merryn V. E. Macville, David Hunt, Richard Webster, Suzanna G.M. Frints, Alberto Fernández-Jaén, Shelagh Joss, Andrew G. L. Douglas, Margje Sinnema, Lesley M McGregor, Vera M. Kalscheuer, Abhijit Dixit, Paulien A. Terhal, Arthur Lee, Sébastien Jacquemont, Omar A. Abdul-Rahman, Peter Wieacker, Koen L.I. van Gassen, Norbert Utzig, Marcus Lee, Vanessa Suckow, Gunnar Houge, Danita Velasco, Cheryl Longman, Holly H. Zimmerman, Elizabeth C. Engle, Bryce A. Mendelsohn, Salwan Al-Nasiry, Suzanne M. Koudijs, Saskia M. Maas, Diana Baralle, Hiromi Hirata, Kees E. P. van Roozendaal, Servi J. C. Stevens, Raoul C.M. Hennekam, Roberto Colombo, Ulrike Kordaß, Gyri Aasland Gradek, Friederike Hennig, APH - Quality of Care, Human Genetics, Amsterdam Neuroscience - Complex Trait Genetics, MUMC+: DA KG Bedrijfsbureau (9), Klinische Genetica, MUMC+: DA KG Polikliniek (9), MUMC+: DA KG Lab Centraal Lab (9), Obstetrie & Gynaecologie, MUMC+: MA Medische Staf Obstetrie Gynaecologie (9), RS: GROW - R4 - Reproductive and Perinatal Medicine, and MUMC+: MA Med Staf Spec Neurologie (9)
- Subjects
Male ,INTELLECTUAL DISABILITY ,Enfermedad del sistema nervioso ,CONTRACTURES ,Pie zambo ,Genes, X-Linked ,Missense mutation ,Genetics(clinical) ,Frameshift Mutation ,EXCHANGE ,Zebrafish ,Genetics (clinical) ,Sequence Deletion ,media_common ,Arthrogryposis ,Genetics ,Sex Characteristics ,Paraplejía ,medicine.diagnostic_test ,2 microdeletion ,Intracellular Signaling Peptides and Proteins ,complicated spastic paraplegia/ spasticity ,METHYLATION ,Espasticidad muscular ,Nuclear Proteins ,spasticity ,Phenotype ,Pedigree ,Codon, Nonsense ,akinesia ,Female ,Neurogenic arthrogryposis multiplex congenita ,media_common.quotation_subject ,fetal hypo ,Nonsense ,Mutation, Missense ,Biology ,DIAGNOSIS ,Article ,Frameshift mutation ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Gene ,Genetic testing ,ZC4H2-Associated Rare Disorders (ZARD) ,SPECTRUM ,Arthrogryposis multiplex congenita ,MUTATIONS ,fetal hypo-/akinesia ,feet ,club foot/-feet ,ZC4H2 ,GENE ,Genética ,DELETIONS ,Disease Models, Animal ,complicated spastic paraplegia ,Mutation ,Xq11.2 microdeletion ,Xq11 ,MENTAL-RETARDATION ,club foot - Abstract
Pathogenic variants in the X-linked gene ZC4H2, which encodes a zinc-finger protein, cause an infrequently described syndromic form of arthrogryposis multiplex congenita (AMC) with central and peripheral nervous system involvement. We present genetic and detailed phenotypic information on 23 newly identified families and simplex cases that include 19 affected females from 18 families and 14 affected males from nine families. Of note, the 15 females with deleterious de novo ZC4H2 variants presented with phenotypes ranging from mild to severe, and their clinical features overlapped with those seen in affected males. By contrast, of the nine carrier females with inherited ZC4H2 missense variants that were deleterious in affected male relatives, four were symptomatic. We also compared clinical phenotypes with previously published cases of both sexes and provide an overview on 48 males and 57 females from 42 families. The spectrum of ZC4H2 defects comprises novel and recurrent mostly inherited missense variants in affected males, and de novo splicing, frameshift, nonsense, and partial ZC4H2 deletions in affected females. Pathogenicity of two newly identified missense variants was further supported by studies in zebrafish. We propose ZC4H2 as a good candidate for early genetic testing of males and females with a clinical suspicion of fetal hypo-/akinesia and/or (neurogenic) AMC. Sin financiación 4.124 JCR (2019) Q1, 45/178 Genetics & Heredity 2.410 SJR (2019) Q1, 43/356 Genetics No data IDR 2019 UEM
- Published
- 2019
32. TAF1, associated with intellectual disability in humans, is essential for embryogenesis and regulates neurodevelopmental processes in zebrafish
- Author
-
Sanna Gudmundsson, Maria Wilbe, Beata Filipek-Górniok, Anna-Maja Molin, Sara Ekvall, Josefin Johansson, Amin Allalou, Hans Gylje, Vera M. Kalscheuer, Johan Ledin, Göran Annerén, and Marie-Louise Bondeson
- Subjects
Adult ,Male ,Adolescent ,lcsh:Medicine ,taf1 ,Nervous System ,Article ,Intellectual Disability ,Genetics research ,Genetics ,Animals ,Humans ,Genetik ,lcsh:Science ,Child ,Zebrafish ,Histone Acetyltransferases ,TATA-Binding Protein Associated Factors ,Gene Expression Profiling ,lcsh:R ,Neurodevelopmental disorders ,RNA sequencing ,Zebrafish Proteins ,zebrafish ,Pedigree ,intellectual disability ,Child, Preschool ,Gene Knockdown Techniques ,Embryogenesis ,Mental Retardation, X-Linked ,lcsh:Q ,Female ,Transcription Factor TFIID ,Gene expression - Abstract
The TATA-box binding protein associated factor 1 (TAF1) protein is a key unit of the transcription factor II D complex that serves a vital function during transcription initiation. Variants of TAF1 have been associated with neurodevelopmental disorders, but TAF1's molecular functions remain elusive. In this study, we present a five-generation family affected with X-linked intellectual disability that co-segregated with a TAF1 c. 3568C>T, p.(Arg1190Cys) variant. All affected males presented with intellectual disability and dysmorphic features, while heterozygous females were asymptomatic and had completely skewed X-chromosome inactivation. We investigated the role of TAF1 and its association to neurodevelopment by creating the first complete knockout model of the TAF1 orthologue in zebrafish. A crucial function of human TAF1 during embryogenesis can be inferred from the model, demonstrating that intact taf1 is essential for embryonic development. Transcriptome analysis of taf1 zebrafish knockout revealed enrichment for genes associated with neurodevelopmental processes. In conclusion, we propose that functional TAF1 is essential for embryonic development and specifically neurodevelopmental processes. Title in Thesis list of papers: TAF1, associated with intellectual disability in humans, is essential for life and regulates neurodevelopmental processes in zebrafish
- Published
- 2019
33. De novo and inherited mutations in the X-linked gene CLCN4 are associated with syndromic intellectual disability and behavior and seizure disorders in males and females
- Author
-
Pawel Stankiewicz, Claude Moraine, Astrid Grimme, Martine Raynaud, Jillian Nicholl, D. Hamlin, Mauricio R. Delgado, Linda Manwaring, H Van Bokhoven, Zhiyv Niu, Stefanie Weinert, J. Wynn, Jozef Gecz, Thomas J. Jentsch, Vanessa Suckow, Vera M. Kalscheuer, Hossein Najmabadi, Jackie Boyle, A. Sommer, Maureen Holvoet, J. M. Goehringer, Eric Haan, M. P. Pietryga, Luis Rohena, John Tolmie, Luciana Musante, Utz Fischer, Floor A. M. Duijkers, Wendy K. Chung, Friederike Hennig, Jan Maarten Cobben, Elizabeth E. Palmer, Tjitske Kleefstra, H Van Esch, B. M. Faux, Michael Field, Kimia Kahrizi, Deepa Sirsi, Melanie Leffler, T. Stuhlmann, Dorothy K. Grange, Jill A. Rosenfeld, Hans-Hilger Ropers, S. P. Lodh, Marie Shaw, Sailaja Golla, E. Bernardo, Shelagh Joss, Thomas D. Challman, General Paediatrics, ANS - Cellular & Molecular Mechanisms, Paediatric Genetics, Other Research, and Human Genetics
- Subjects
Male ,0301 basic medicine ,Movement disorders ,Xenopus laevis ,Epilepsy ,0302 clinical medicine ,Genes, X-Linked ,Intellectual disability ,Missense mutation ,Child ,Genetics ,Genetic Diseases, X-Linked ,Syndrome ,Middle Aged ,White Matter ,Pedigree ,3. Good health ,Psychiatry and Mental health ,Phenotype ,Schizophrenia ,Child, Preschool ,Female ,Original Article ,medicine.symptom ,Function and Dysfunction of the Nervous System ,Adult ,medicine.medical_specialty ,Adolescent ,Frameshift mutation ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,All institutes and research themes of the Radboud University Medical Center ,Chloride Channels ,Intellectual Disability ,medicine ,Animals ,Humans ,Family ,Psychiatry ,Molecular Biology ,Germ-Line Mutation ,Aged ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,business.industry ,medicine.disease ,030104 developmental biology ,Mood disorders ,Cardiovascular and Metabolic Diseases ,Mutation ,Behavioral medicine ,Oocytes ,business ,Epileptic Syndromes ,030217 neurology & neurosurgery - Abstract
Variants in CLCN4, which encodes the chloride/hydrogen ion exchanger CIC-4 prominently expressed in brain, were recently described to cause X-linked intellectual disability and epilepsy. We present detailed phenotypic information on 52 individuals from 16 families with CLCN4-related disorder: 5 affected females and 2 affected males with a de novo variant in CLCN4 (6 individuals previously unreported) and 27 affected males, 3 affected females and 15 asymptomatic female carriers from 9 families with inherited CLCN4 variants (4 families previously unreported). Intellectual disability ranged from borderline to profound. Behavioral and psychiatric disorders were common in both child- and adulthood, and included autistic features, mood disorders, obsessive-compulsive behaviors and hetero- and autoaggression. Epilepsy was common, with severity ranging from epileptic encephalopathy to well-controlled seizures. Several affected individuals showed white matter changes on cerebral neuroimaging and progressive neurological symptoms, including movement disorders and spasticity. Heterozygous females can be as severely affected as males. The variability of symptoms in females is not correlated with the X inactivation pattern studied in their blood. The mutation spectrum includes frameshift, missense and splice site variants and one single-exon deletion. All missense variants were predicted to affect CLCN4's function based on in silico tools and either segregated with the phenotype in the family or were de novo. Pathogenicity of all previously unreported missense variants was further supported by electrophysiological studies in Xenopus laevis oocytes. We compare CLCN4-related disorder with conditions related to dysfunction of other members of the CLC family.Molecular Psychiatry advance online publication, 23 August 2016; doi:10.1038/mp.2016.135. ispartof: Molecular Psychiatry vol:23 issue:2 pages:222-230 ispartof: location:England status: published
- Published
- 2016
- Full Text
- View/download PDF
34. Intracellular CLC Transporters - From Kidney Stones to Intellectual Disability
- Author
-
Vera M. Kalscheuer, Michael Pusch, Alessandra Picollo, Sara Bertelli, Elizabeth E. Palmer, and Giovanni Zifarelli
- Subjects
business.industry ,Intellectual disability ,Biophysics ,Medicine ,Kidney stones ,Transporter ,Pharmacology ,business ,medicine.disease ,Intracellular - Published
- 2020
- Full Text
- View/download PDF
35. Author response for 'Effect of inbreeding on intellectual disability revisited by Trio sequencing'
- Author
-
Mohammad-Reza Khodaie-Ardakani, Farahnaz Sabbagh Kermani, K. Kahrizi, Roshanak Jazayeri, Pooneh Nikuei, Payman Jamali, Masoumeh Hosseini, Faezeh Mojahedi, Tara Akhtarkhavari, Zhila Ghaderi, Sepideh Mehvari, Atefeh Khoshaeen, Zohreh Fattahi, Marzieh Mohseni, Maryam Beheshtian, Zohreh Mehrjoo, Haleh Habibi, Vera M. Kalscheuer, Hao Hu, Bettina Lipkowitz, Hossein Najmabadi, Sanaz Arzhangi, Fatemeh Pourfatemi, Thomas F. Wienker, Vanessa Suckow, Seyed Hassan Tonekaboni, Hans-Hilger Ropers, Farnaz Sadeghinia, Reza Najafipour, Maryam Rahimi, and Milad Falahat Chian
- Subjects
Intellectual disability ,medicine ,Psychology ,medicine.disease ,Inbreeding ,Genealogy - Published
- 2018
- Full Text
- View/download PDF
36. The power of the Mediator complex-Expanding the genetic architecture and phenotypic spectrum of MED12-related disorders
- Author
-
Andreas Tzschach, W. Hachmann, C Jensen, Magdalena Nawara, Jarosław Poznański, K. Kahrizi, Jerzy Bal, Vera M. Kalscheuer, M Bienek, Andreas Dufke, H. Enders, Johannes R. Lemke, Monika Gos, T. Chilarska, Dorota Hoffman-Zacharska, Agnieszka Charzewska, Hossein Najmabadi, Barbara Oehl-Jaschkowitz, R. Maiwald, and Ewa Obersztyn
- Subjects
0301 basic medicine ,Male ,Models, Molecular ,Genotype ,FG syndrome ,X-linked intellectual disability ,Protein Conformation ,Mutation, Missense ,Biology ,MED12 ,03 medical and health sciences ,Structure-Activity Relationship ,Lujan–Fryns syndrome ,Genes, X-Linked ,X Chromosome Inactivation ,Exome Sequencing ,Genetics ,medicine ,Missense mutation ,Humans ,Genetic Predisposition to Disease ,Genetics (clinical) ,X chromosome ,Alleles ,Genetic Association Studies ,Mediator Complex ,Facies ,Genetic Variation ,medicine.disease ,Genetic architecture ,Pedigree ,030104 developmental biology ,Phenotype ,Amino Acid Substitution ,Female ,Single Palmar Crease - Abstract
MED12 is a member of the large Mediator complex that controls cell growth, development, and differentiation. Mutations in MED12 disrupt neuronal gene expression and lead to at least three distinct X-linked intellectual disability syndromes (FG, Lujan-Fryns, and Ohdo). Here, we describe six families with missense variants in MED12 (p.(Arg815Gln), p.(Val954Gly), p.(Glu1091Lys), p.(Arg1295Cys), p.(Pro1371Ser), and p.(Arg1148His), the latter being first reported in affected females) associated with a continuum of symptoms rather than distinct syndromes. The variants expanded the genetic architecture and phenotypic spectrum of MED12-related disorders. New clinical symptoms included brachycephaly, anteverted nares, bulbous nasal tip, prognathism, deep set eyes, and single palmar crease. We showed that MED12 variants, initially implicated in X-linked recessive disorders in males, may predict a potential risk for phenotypic expression in females, with no correlation of the X chromosome inactivation pattern in blood cells. Molecular modeling (Yasara Structure) performed to model the functional effects of the variants strongly supported the pathogenic character of the variants examined. We showed that molecular modeling is a useful method for in silico testing of the potential functional effects of MED12 variants and thus can be a valuable addition to the interpretation of the clinical and genetic findings.
- Published
- 2018
37. MED13L-related intellectual disability: involvement of missense variants and delineation of the phenotype
- Author
-
Bertrand Isidor, Christine Francannet, D. Li, Samuel W. Baker, Gaëlle Vieville, Martine Doco-Fenzy, David Geneviève, I. Giurgea, Anna Gréen, Emily Fassi, Caroline Nava, Roseline Caumes, C. Fournier, Alexandra Afenjar, Françoise Devillard, Yuri A. Zarate, Damien Sanlaville, Michael Field, Elisabetta Lapi, Sandra Whalen, Emma Bedoukian, Alice Goldenberg, S. Steinwall Larsen, Jamal Ghoumid, Marjolaine Willems, M. Wenzel, Isabelle Marey, Sylvie Picker-Minh, Thomas Smol, Anne-Marie Guerrot, Dominique Bonneau, Gaetan Lesca, Delphine Héron, Elizabeth J. Bhoj, Véronique Satre, Sylvie Manouvrier-Hanu, Christine Coubes, Alain Verloes, Margarita Stefanova, Gaël Nicolas, Amélie Piton, Odile Boute-Benejean, Laurence Faivre, Caroline Thuillier, Bénédicte Gérard, Nicolas Chatron, Florence Petit, Beth Keena, Elise Boudry-Labis, C. Colson, Sonia Bouquillon, Avni Santani, Boris Keren, Lisa Ewans, Tony Roscioli, N. Le Meur, Paul Kuentz, Bryan L. Krock, Catherine Roche-Lestienne, Anne Dieux-Coeslier, Alban Ziegler, Pascale Saugier-Veber, Cyril Mignot, Vera M. Kalscheuer, Addie I. Nesbitt, Charles Coutton, Service de Génétique Médicale [Lille], Institut de génétique médicale-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Maladies RAres du DEveloppement embryonnaire et du MEtabolisme : du Phénotype au Génotype et à la Fonction - ULR 7364 (RADEME), Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Pôle de Biologie Pathologie Génétique [CHU Lille], Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Laboratoire de Diagnostic Génétique [CHU Strasbourg], Université de Strasbourg (UNISTRA)-CHU Strasbourg, CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Laboratoire de Cytogénétique Constitutionnelle [Hospices civils de Lyon], Hospices Civils de Lyon (HCL), CHU Trousseau [APHP], Children’s Hospital of Philadelphia (CHOP ), Service de génétique [Angers], Université d'Angers (UA)-Centre Hospitalier Universitaire d'Angers (CHU Angers), PRES Université Nantes Angers Le Mans (UNAM)-PRES Université Nantes Angers Le Mans (UNAM), MitoVasc - Physiopathologie Cardiovasculaire et Mitochondriale (MITOVASC), Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Département de génétique médicale, maladies rares et médecine personnalisée [CHRU Montpellier], Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Institute for Advanced Biosciences / Institut pour l'Avancée des Biosciences (Grenoble) (IAB), Centre Hospitalier Universitaire [Grenoble] (CHU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement français du sang - Auvergne-Rhône-Alpes (EFS)-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes [2016-2019] (UGA [2016-2019]), Laboratoire de Génétique Chromosomique [CHU de Grenoble], CHU Grenoble, University of New South Wales [Canberra Campus] (UNSW), Centre de génétique - Centre de référence des maladies rares, anomalies du développement et syndromes malformatifs (CHU de Dijon), Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand (CHU Dijon), Lipides - Nutrition - Cancer [Dijon - U1231] (LNC), Université de Bourgogne (UB)-Institut National de la Santé et de la Recherche Médicale (INSERM)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement, Washington University School of Medicine in St. Louis, Washington University in Saint Louis (WUSTL), Service de Génétique Médicale [CHU Clermont-Ferrand], CHU Estaing [Clermont-Ferrand], CHU Clermont-Ferrand-CHU Clermont-Ferrand, Maladies génétiques d'expression pédiatrique (U933), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Trousseau [APHP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Génétique du cancer et des maladies neuropsychiatriques (GMFC), Université de Rouen Normandie (UNIROUEN), Normandie Université (NU)-Normandie Université (NU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Linköping university hospital, Service de génétique médicale - Unité de génétique clinique [Nantes], Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes), Azienda Ospedaliero Universitaria A. Meyer [Firenze, Italy], Charité - UniversitätsMedizin = Charité - University Hospital [Berlin], Unité fonctionnelle de génétique clinique, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Robert Debré-Université Paris Diderot - Paris 7 (UPD7), Département de génétique [Robert Debré], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-AP-HP Hôpital universitaire Robert-Debré [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Service de génétique médicale [Montpellier], Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier)-Hôpital Arnaud de Villeneuve, University of Arkansas for Medical Sciences (UAMS), Max Planck Institute for Molecular Genetics (MPIMG), Max-Planck-Gesellschaft, Service de Génétique Cytogénétique et Embryologie [CHU Pitié-Salpêtrière], Service de génétique et embryologie médicales [CHU Trousseau], Physiopathologie Cardiovasculaire et Mitochondriale (MITOVASC), Centre Hospitalier Universitaire [Grenoble] (CHU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Etablissement français du sang - Auvergne-Rhône-Alpes (EFS)-Université Grenoble Alpes [2016-2019] (UGA [2016-2019]), Université Paris Diderot - Paris 7 (UPD7)-Hôpital Robert Debré-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), CHU Montpellier, Equipe GAD (LNC - U1231), Université de Bourgogne (UB)-Institut National de la Santé et de la Recherche Médicale (INSERM)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Université de Bourgogne (UB)-Institut National de la Santé et de la Recherche Médicale (INSERM)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement, Cellules Souches, Plasticité Cellulaire, Médecine Régénératrice et Immunothérapies (IRMB), Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Génomique et Médecine Personnalisée du Cancer et des Maladies Neuropsychiatriques (GPMCND), Institut de génétique médicale-Centre Hospitalier Régional Universitaire [Lille] ( CHRU Lille ), Maladies RAres du DEveloppement embryonnaire et du MEtabolisme : du Phénotype au Génotype et à la Fonction ( RADEME ), Hôpital Jeanne de Flandre [Lille]-Université de Lille-Centre Hospitalier Régional Universitaire de Lille ( CHRU de Lille ) -Clinique de Génétique médicale Guy Fontaine [CHRU LIlle]-Centre de référence maladies rares Anomalies du développement [CHRU Lille], Lipides - Nutrition - Cancer [Dijon - U1231] ( LNC ), Université de Bourgogne ( UB ) -AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale ( INSERM ), Université de Bourgogne ( UB ) -AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Université de Bourgogne ( UB ) -AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale ( INSERM ), Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand ( CHU Dijon ), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA), Laboratoire de Probabilités et Modèles Aléatoires (LPMA), Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Laboratoire de recherche en Hydrodynamique, Énergétique et Environnement Atmosphérique (LHEEA), École Centrale de Nantes (ECN)-Centre National de la Recherche Scientifique (CNRS), Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-CHU Pitié-Salpêtrière [APHP], Service de Génétique Clinique, Hôpital Femme Mère Enfant, Centre Hospitalier Universitaire de Lyon, Laboratoire Hippolyte Fizeau (FIZEAU), Université Nice Sophia Antipolis (... - 2019) (UNS), Université Côte d'Azur (UCA)-Université Côte d'Azur (UCA)-Institut national des sciences de l'Univers (INSU - CNRS)-Observatoire de la Côte d'Azur, Université Côte d'Azur (UCA)-Centre National de la Recherche Scientifique (CNRS), Systèmes de Référence Temps Espace (SYRTE), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut national des sciences de l'Univers (INSU - CNRS)-Observatoire de Paris, PSL Research University (PSL)-PSL Research University (PSL)-Centre National de la Recherche Scientifique (CNRS), Centre de recherche en neurosciences de Lyon (CRNL), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet [Saint-Étienne] (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Service de Génétique [CHU Caen], Université de Caen Normandie (UNICAEN), Normandie Université (NU)-Normandie Université (NU)-CHU Caen, Normandie Université (NU)-Tumorothèque de Caen Basse-Normandie (TCBN)-Tumorothèque de Caen Basse-Normandie (TCBN), Biologie, génétique et thérapies ostéoarticulaires et respiratoires (BIOTARGEN), Normandie Université (NU)-Normandie Université (NU), Département de génétique médicale, maladies rares et médecine personnalisée [CHRU de Montpellier], Centre Hospitalier Universitaire [Grenoble] (CHU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement français du sang - Auvergne-Rhône-Alpes (EFS)-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes (UGA), APERAM, Inconnu, CHU Clermont-Ferrand, Service de génétique [Rouen], CHU Rouen, Normandie Université (NU)-Normandie Université (NU)-Université de Rouen Normandie (UNIROUEN), Normandie Université (NU), FHU TRANSLAD, Département de génétique [CHU Rouen] (Centre Normandie de Génomique et de Médecine Personnalisée), CHU Pitié-Salpêtrière [APHP], Fossil Fuel Chemistry, University of Sofia, Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-Hôpital Robert Debré-Université Paris Diderot - Paris 7 (UPD7), Génétique et épigénétique des maladies métaboliques, neurosensorielles et du développement (Inserm U781), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de génétique clinique, Hôpital Jeanne de Flandre [Lille]-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), and International Livestock Research Institute
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Pediatrics ,Ataxia ,Mutation, Missense ,Intellectual disability ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,MESH: Phenotype ,MESH: Intellectual Disability ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Epilepsy ,MESH: Mediator Complex ,MESH: Child ,Genetics ,medicine ,Missense mutation ,Humans ,Mediator complex ,Child ,[ SDV.GEN.GH ] Life Sciences [q-bio]/Genetics/Human genetics ,Genetics (clinical) ,MESH: Mutation, Missense ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,MESH: Humans ,business.industry ,MESH: Child, Preschool ,medicine.disease ,Human genetics ,Hypotonia ,MESH: Male ,3. Good health ,MED13L ,Cardiopathy ,030104 developmental biology ,Phenotype ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Child, Preschool ,Medical genetics ,Female ,medicine.symptom ,Haploinsufficiency ,business ,MESH: Female ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
IF 3.269; International audience; Molecular anomalies in MED13L, leading to haploinsufficiency, have been reported in patients with moderate to severe intellectual disability (ID) and distinct facial features, with or without congenital heart defects. Phenotype of the patients was referred to "MED13L haploinsufficiency syndrome." Missense variants in MED13L were already previously described to cause the MED13L-related syndrome, but only in a limited number of patients. Here we report 36 patients with MED13L molecular anomaly, recruited through an international collaboration between centers of expertise for developmental anomalies. All patients presented with intellectual disability and severe language impairment. Hypotonia, ataxia, and recognizable facial gestalt were frequent findings, but not congenital heart defects. We identified seven de novo missense variations, in addition to protein-truncating variants and intragenic deletions. Missense variants clustered in two mutation hot-spots, i.e., exons 15-17 and 25-31. We found that patients carrying missense mutations had more frequently epilepsy and showed a more severe phenotype. This study ascertains missense variations in MED13L as a cause for MED13L-related intellectual disability and improves the clinical delineation of the condition.
- Published
- 2018
- Full Text
- View/download PDF
38. X chromosome dosage and presence of SRY shape sex-specific differences in DNA methylation at an autosomal region in human cells
- Author
-
Natália D. Linhares, Francisco Martínez, Eugênia Ribeiro Valadares, Keelin M Greenlaw, Celia M. T. Greenwood, Germán Rodríguez Criado, Bianca Ho, Alfredo Brusco, Giovanni Battista Ferrero, Catherine Laprise, Abeer Al Tuwaijri, Elisa Giorgio, Anna K. Naumova, Sanny Moussette, Marta Svartman, and Vera M. Kalscheuer
- Subjects
0301 basic medicine ,Male ,lcsh:Medicine ,Locus (genetics) ,Biology ,Y chromosome ,lcsh:Physiology ,Cell Line ,Gender Studies ,DNA methylation ,Sex ,X chromosome ,03 medical and health sciences ,Endocrinology ,Humans ,Epigenetics ,Genes, sry ,ATRX ,Genetics ,Chromosomes, Human, X ,Sex Characteristics ,Chromosomes, Human, Y ,lcsh:QP1-981 ,Research ,lcsh:R ,Egg Proteins ,Membrane Proteins ,Methylation ,030104 developmental biology ,Testis determining factor ,Female - Abstract
Background Sexual dimorphism in DNA methylation levels is a recurrent epigenetic feature in different human cell types and has been implicated in predisposition to disease, such as psychiatric and autoimmune disorders. To elucidate the genetic origins of sex-specific DNA methylation, we examined DNA methylation levels in fibroblast cell lines and blood cells from individuals with different combinations of sex chromosome complements and sex phenotypes focusing on a single autosomal region––the differentially methylated region (DMR) in the promoter of the zona pellucida binding protein 2 (ZPBP2) as a reporter. Results Our data show that the presence of the sex determining region Y (SRY) was associated with lower methylation levels, whereas higher X chromosome dosage in the absence of SRY led to an increase in DNA methylation levels at the ZPBP2 DMR. We mapped the X-linked modifier of DNA methylation to the long arm of chromosome X (Xq13-q21) and tested the impact of mutations in the ATRX and RLIM genes, located in this region, on methylation levels. Neither ATRX nor RLIM mutations influenced ZPBP2 methylation in female carriers. Conclusions We conclude that sex-specific methylation differences at the autosomal locus result from interaction between a Y-linked factor SRY and at least one X-linked factor that acts in a dose-dependent manner. Electronic supplementary material The online version of this article (10.1186/s13293-018-0169-7) contains supplementary material, which is available to authorized users.
- Published
- 2018
39. De novo mutations in MSL3 cause an X-linked syndrome marked by impaired histone H4 lysine 16 acetylation
- Author
-
Olivier Vanakker, Maria Kirchhoff, Christian Gilissen, Jenny Morton, Ineke van der Burgt, Kelly Radtke, Tugce Aktas, Sarah Vergult, Diana Johnson, Tobias Rumpf, Tony Roscioli, André Reis, Alan Fryer, Salima El Chehadeh, Christel Thauvin-Robinet, Ange Line Bruel, Rolph Pfundt, Sander Pajusalu, David Francis, Asifa Akhtar, Iben Bache, Tiong Yang Tan, Vera M. Kalscheuer, Meredith Wilson, Björn Menten, Giuseppe Semplicio, Julien Thevenon, Richard Fisher, Yannis Duffourd, Gerhard Mittler, Witold G. Szymanski, M. Felicia Basilicata, Victoria McKay, Kristin Lindstrom, Han G. Brunner, Katrin Õunap, Jaya Ganesh, Claudia Isabelle Keller Valsecchi, Megan T. Cho, Laurence Faivre, MUMC+: DA Klinische Genetica (5), Klinische Genetica, RS: GROW - R4 - Reproductive and Perinatal Medicine, Max Planck Institute of Immunobiology and Epigenetics (MPI-IE), Max-Planck-Gesellschaft, Equipe GAD (LNC - U1231), Lipides - Nutrition - Cancer [Dijon - U1231] (LNC), Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de génétique - Centre de référence des maladies rares, anomalies du développement et syndromes malformatifs (CHU de Dijon), Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand (CHU Dijon), FHU TRANSLAD (CHU de Dijon), Université Bourgogne Franche-Comté [COMUE] (UBFC), West Midlands Regional Genetics Laboratory and Clinical Genetics Unit, Birmingham Women's Hospital, Copenhagen University Hospitals, Radboud University Medical Center [Nijmegen], Center for Medical Genetics [Ghent], Ghent University Hospital, Donders Institute for Brain, Cognition and Behaviour, Radboud university [Nijmegen], GeneDx [Gaithersburg, MD, USA], Murdoch Children's Research Institute (MCRI), Department of Clinical Genetics [Copenhagen], Rigshospitalet [Copenhagen], Copenhagen University Hospital-Copenhagen University Hospital, Institute of Human Genetics [Erlangen, Allemagne], Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Department of Clinical Genetics (Sheffield Children’s NHS Foundation Trust), Sheffield Children's NHS Foundation Trust, Liverpool Women's NHS Foundation Trust, Institute for Advanced Biosciences / Institut pour l'Avancée des Biosciences (Grenoble) (IAB), and Centre Hospitalier Universitaire [Grenoble] (CHU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement français du sang - Auvergne-Rhône-Alpes (EFS)-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes [2016-2019] (UGA [2016-2019])
- Subjects
Male ,0301 basic medicine ,CHROMATIN ,INTELLECTUAL DISABILITY ,Chromosomal Proteins, Non-Histone ,CHROMOSOME ,Cohort Studies ,Histones ,Mice ,Genes, X-Linked ,MSL complex ,Child ,Cells, Cultured ,Histone Acetyltransferases ,Epigenomics ,Acetylation ,Genetic Diseases, X-Linked ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Syndrome ,Chromatin ,Cell biology ,DNA-Binding Proteins ,DROSOPHILA ,Histone ,ACETYLTRANSFERASE ,Child, Preschool ,Female ,Adolescent ,Mice, Transgenic ,Biology ,Article ,Histone H4 ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,Genetics ,Animals ,Humans ,CELL-CYCLE ,Epigenetics ,DEACETYLASE ,MOF ,MSL3 ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,DOSAGE COMPENSATION COMPLEX ,Infant ,Dosage compensation complex ,PROTEIN INTERACTIONS ,HEK293 Cells ,030104 developmental biology ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Neurodevelopmental Disorders ,Case-Control Studies ,Mutation ,biology.protein ,Histone deacetylase ,Protein Processing, Post-Translational ,Transcription Factors ,Ultra-rare developmental disorders - Abstract
International audience; The etiological spectrum of ultra-rare developmental disorders remains to be fully defined. Chromatin regulatory mechanisms maintain cellular identity and function, where misregulation may lead to developmental defects. Here, we report pathogenic variations in MSL3, which encodes a member of the chromatin-associated male-specific lethal (MSL) complex responsible for bulk histone H4 lysine 16 acetylation (H4K16ac) in flies and mammals. These variants cause an X-linked syndrome affecting both sexes. Clinical features of the syndrome include global developmental delay, progressive gait disturbance, and recognizable facial dysmorphism. MSL3 mutations affect MSL complex assembly and activity, accompanied by a pronounced loss of H4K16ac levels in vivo. Patient-derived cells display global transcriptome alterations of pathways involved in morphogenesis and cell migration. Finally, we use histone deacetylase inhibitors to rebalance acetylation levels, alleviating some of the molecular and cellular phenotypes of patient cells. Taken together, we characterize a syndrome that allowed us to decipher the developmental importance of MSL3 in humans.
- Published
- 2018
- Full Text
- View/download PDF
40. FRMPD4 mutations cause X-linked intellectual disability and disrupt dendritic spine morphogenesis
- Author
-
Jamel Chelly, Joanna Kosińska, Jia-Hua Hu, Philippe M. Campeau, Cindy Skinner, Piotr Stawiński, Jennifer Castaneda, Elsa Rossignol, Sylwia Rzonca, Ruihua Wang, Hilde Van Esch, Lionel Van Maldergem, Tao Wang, Paul F. Worley, Charles E. Schwartz, Emmanuelle Lemyre, Elizabeth Vincent, Jerzy Bal, Vera M. Kalscheuer, Dax A. Hoffman, Juliette Piard, Mei Han, Centre de génétique humaine [CHRU Besançon], Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon), Johns Hopkins University School of Medicine [Baltimore], Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Université du Québec à Montréal = University of Québec in Montréal (UQAM), Institute of Mother and Child, University Hospitals Leuven [Leuven], 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), The Greenwood Genetic Center, Max Planck Institute for Molecular Genetics (MPIMG), Max-Planck-Gesellschaft, Centre d'Investigation Clinique de Besançon (Inserm CIC 1431), Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement français du sang [Bourgogne-Franche-Comté] (EFS [Bourgogne-Franche-Comté])-Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC), and univOAK, Archive ouverte
- Subjects
Adult ,Male ,0301 basic medicine ,Dendritic spine ,Adolescent ,X-linked intellectual disability ,Dendritic Spines ,Neurogenesis ,Nonsense mutation ,HOMER1 ,Dendritic spine morphogenesis ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,Biology ,medicine.disease_cause ,Young Adult ,03 medical and health sciences ,Intellectual Disability ,Morphogenesis ,Genetics ,medicine ,Humans ,Missense mutation ,Frameshift Mutation ,Molecular Biology ,Genetics (clinical) ,X-linked recessive inheritance ,Aged ,Mutation ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Intracellular Signaling Peptides and Proteins ,Articles ,Exons ,General Medicine ,Middle Aged ,medicine.disease ,Pedigree ,030104 developmental biology ,Female - Abstract
FRMPD4 (FERM and PDZ Domain Containing 4) is a neural scaffolding protein that interacts with PSD-95 to positively regulate dendritic spine morphogenesis, and with mGluR1/5 and Homer to regulate mGluR1/5 signaling. We report the genetic and functional characterization of 4 FRMPD4 deleterious mutations that cause a new X-linked intellectual disability (ID) syndrome. These mutations were found to be associated with ID in ten affected male patients from four unrelated families, following an apparent X-linked mode of inheritance. Mutations include deletion of an entire coding exon, a nonsense mutation, a frame-shift mutation resulting in premature termination of translation, and a missense mutation involving a highly conserved amino acid residue neighboring FRMPD4-FERM domain. Clinical features of these patients consisted of moderate to severe ID, language delay and seizures alongside with behavioral and/or psychiatric disturbances. In-depth functional studies showed that a frame-shift mutation, FRMPD4p.Cys618ValfsX, result in a disruption of FRMPD4 binding with PSD-95 and HOMER1, and a failure to increase spine density in transfected hippocampal neurons. Behavioral studies of frmpd4-KO mice identified hippocampus-dependent spatial learning and memory deficits in Morris Water Maze test. These findings point to an important role of FRMPD4 in normal cognitive development and function in humans and mice, and support the hypothesis that FRMPD4 mutations cause ID by disrupting dendritic spine morphogenesis in glutamatergic neurons. ispartof: Human Molecular Genetics vol:27 issue:4 pages:589-600 ispartof: location:England status: published
- Published
- 2018
- Full Text
- View/download PDF
41. IncreasedSTAG2dosage defines a novel cohesinopathy with intellectual disability and behavioral problems
- Author
-
Martine Raynaud, Raman Kumar, Hilde Van Esch, C Jensen, Bartlomiej Budny, Magdalena Badura-Stronka, Michael I. Love, Guy Froyen, Lachlan A. Jolly, Bregje W.M. van Bon, Jill A. Rosenfeld, Lina Basel-Vanagaite, M Bienek, Alison Gardner, Jillian Nicholl, Sau Wai Cheung, Elizabeth Thompson, Jozef Gecz, Anne Baxendale, Anna Latos-Bielenska, C Tan, Joshua A. Woenig, Stefan A. Haas, Mark A. Corbett, Marzena Wisniewska, Maureen Holvoet, Evelyn Douglas, Eric Haan, Michael Field, Kathryn Friend, Melanie Leffler, Jacqueline R. Batanian, Hao Hu, Pawel Stankiewicz, Reinhard Ullmann, and Vera M. Kalscheuer
- Subjects
Male ,DNA Copy Number Variations ,Cohesin complex ,Gene regulatory network ,Cell Cycle Proteins ,Biology ,Genome ,Intellectual Disability ,Intellectual disability ,Genetics ,medicine ,Humans ,Copy-number variation ,Molecular Biology ,Gene ,Genetics (clinical) ,X chromosome ,Problem Behavior ,Chromosomes, Human, X ,Reverse Transcriptase Polymerase Chain Reaction ,Antigens, Nuclear ,General Medicine ,medicine.disease ,Genetic architecture ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] - Abstract
Next generation genomic technologies have made a significant contribution to the understanding of the genetic architecture of human neurodevelopmental disorders. Copy number variants (CNVs) play an important role in the genetics of intellectual disability (ID). For many CNVs, and copy number gains in particular, the responsible dosage-sensitive gene(s) have been hard to identify. We have collected 18 different interstitial microduplications and one microtriplication of Xq25. There were 15 affected individuals from 6 different families and 13 singleton cases, 28 affected males in total. The critical overlapping region involved the STAG2 gene, which codes for a subunit of the cohesin complex that regulates cohesion of sister chromatids and gene transcription. We demonstrate that STAG2 is the dosage-sensitive gene within these CNVs, as gains of STAG2 mRNA and protein dysregulate disease-relevant neuronal gene networks in cells derived from affected individuals. We also show that STAG2 gains result in increased expression of OPHN1, a known X-chromosome ID gene. Overall we define a novel cohesinopathy due to copy number gain of Xq25 and STAG2 in particular. ispartof: Human Molecular Genetics vol:24 issue:25 pages:7171-81 ispartof: location:England status: published
- Published
- 2015
- Full Text
- View/download PDF
42. A Novel Mutation inRPL10(Ribosomal Protein L10) Causes X-Linked Intellectual Disability, Cerebellar Hypoplasia, and Spondylo-Epiphyseal Dysplasia
- Author
-
Lore Breitenbach-Koller, Emanuele Bellacchio, Andreas Friedrich, Matteo Di Capua, Francesco Emma, Sabine M. Klauck, Stefan A. Haas, Paolo Alfieri, Giorgia Piccini, Ginevra Zanni, Thomas Karl, Sabina Barresi, Marco Cappa, Vera M. Kalscheuer, and Enrico Bertini
- Subjects
Male ,Ribosomal Proteins ,Heterozygote ,Ribosomal Protein L10 ,X-linked intellectual disability ,Developmental Disabilities ,Population ,Ribosome biogenesis ,Neuroimaging ,Biology ,Nervous System Malformations ,Osteochondrodysplasias ,Ribosome ,Genes, X-Linked ,X Chromosome Inactivation ,Ribosomal protein ,Cerebellum ,Intellectual Disability ,Large ribosomal subunit ,Genetics ,medicine ,Humans ,Missense mutation ,education ,Genetic Association Studies ,Genetics (clinical) ,education.field_of_study ,Sequence Analysis, DNA ,medicine.disease ,Magnetic Resonance Imaging ,Phenotype ,Child, Preschool ,Mutation ,Mutation (genetic algorithm) ,Female - Abstract
RPL10 encodes ribosomal protein L10 (uL16), a highly conserved multifunctional component of the large ribosomal subunit, involved in ribosome biogenesis and function. Using X-exome resequencing, we identified a novel missense mutation (c.191C>T; p.(A64V)) in the N-terminal domain of the protein, in a family with two affected cousins presenting with X-linked intellectual disability, cerebellar hypoplasia, and spondylo-epiphyseal dysplasia (SED). We assessed the impact of the mutation on the translational capacity of the cell using yeast as model system. The mutation generates a functional ribosomal protein, able to complement the translational defects of a conditional lethal mutation of yeast rpl10. However, unlike previously reported mutations, this novel RPL10 missense mutation results in an increase in the actively translating ribosome population. Our results expand the mutational and clinical spectrum of RPL10 identifying a new genetic cause of SED and highlight the emerging role of ribosomal proteins in the pathogenesis of neurodevelopmental disorders.
- Published
- 2015
- Full Text
- View/download PDF
43. Tentative clinical diagnosis of Lujan-Fryns syndrome-A conglomeration of different genetic entities?
- Author
-
Karl Hackmann, Sigrid Tinschert, Albrecht Kobelt, Evelin Schröck, Alma Kuechler, Andreas Tzschach, Tim Ripperger, Stefan A. Haas, Dagmar Wieczorek, Johannes R. Lemke, Andreas Rump, Jean-Pierre Fryns, Beate Albrecht, Vera M. Kalscheuer, Konrad Oexle, and Nataliya Di Donato
- Subjects
Male ,0301 basic medicine ,X-linked intellectual disability ,Medizin ,Marfan Syndrome ,MED12 ,Craniofacial Abnormalities ,03 medical and health sciences ,Genes, X-Linked ,Lujan–Fryns syndrome ,Intellectual Disability ,Gene duplication ,Intellectual disability ,Genetics ,Humans ,Medicine ,Abnormalities, Multiple ,Genetics (clinical) ,Mediator Complex ,business.industry ,High-Throughput Nucleotide Sequencing ,RNA-Binding Proteins ,Karyotype ,medicine.disease ,Pedigree ,030104 developmental biology ,Mutation ,Mutation (genetic algorithm) ,Mental Retardation, X-Linked ,Autism ,Female ,business ,Acyltransferases - Abstract
The clinical diagnosis of Lujan-Fryns syndrome (LFS) comprises X-linked intellectual disability (XLID) with marfanoid habitus, distinct combination of minor facial anomalies and nasal speech. However the definition of syndrome was significantly broadened since the original report and implies ID with marfanoid habitus. Mutations of three genes (MED12, UPF3B, and ZDHHC9) have been reported in "broadly defined" LFS. We examined these genes in 28 individuals with a tentative clinical diagnosis of LFS but we did not identify any causative mutation. By molecular karyotyping we detected other disorders, i.e., Phelan-McDermid syndrome and 16p11.2 microduplication, each in one patient. One affected individual was carrier of a different recurrent duplication on 16p11.2 that has been reported several times to the DECIPHER and ISCA databases in individuals with autism, intellectual disability (ID), and developmental delay. It may represent a new duplication syndrome. We also identified previously unreported de novo duplication on chromosome 12p13.31 which we considered to be disease-causing. X-exome sequencing of four individuals revealed private or non-recurrent mutations in NKAP and LAS1L in one patient each. While LFS is defined as a form of XLID, there seem to be various conditions that have rather similar phenotypes. Therefore, the combination of ID and marfanoid habitus in a male patient is not sufficient for the diagnosis of LFS. We suggest that the diagnosis of LFS in patients with ID and marfanoid habitus should be made only in presence of specific facial features, nasal speech and obvious X-linked segregation of the disorder or an unambiguously pathogenic mutation in the MED12. (c) 2015 Wiley Periodicals, Inc.
- Published
- 2015
- Full Text
- View/download PDF
44. THOC2 Mutations Implicate mRNA-Export Pathway in X-Linked Intellectual Disability
- Author
-
Michael Field, Alison Gardner, Hilde Van Esch, Vera M. Kalscheuer, Jackie Boyle, Matthew F. Hunter, Evelyn Douglas, Marie Shaw, Jozef Gecz, Melanie Leffler, Lloyd Weir, C Jensen, Elizabeth E. Palmer, Martine Raynaud, Bregje W.M. van Bon, C Tan, Carolyn Rogers, Griet Van Buggenhout, Lachlan A. Jolly, Stefan A. Haas, Eric Haan, Kathryn Friend, Katrin Hoffmann, Mark A. Corbett, Raman Kumar, M Bienek, Joshua A. Woenig, Hao Hu, Huiying Zhao, Robin Reed, and Anna Hackett
- Subjects
Models, Molecular ,X-linked intellectual disability ,Protein subunit ,Molecular Sequence Data ,Active Transport, Cell Nucleus ,Mutation, Missense ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Report ,Genetics ,medicine ,Protein biosynthesis ,Humans ,Missense mutation ,Genetics(clinical) ,Amino Acid Sequence ,RNA, Messenger ,Genetics (clinical) ,X chromosome ,Exome sequencing ,030304 developmental biology ,Chromosomes, Human, X ,0303 health sciences ,Messenger RNA ,Base Sequence ,RNA-Binding Proteins ,Sequence Analysis, DNA ,Syndrome ,medicine.disease ,Pedigree ,Cell nucleus ,medicine.anatomical_structure ,Mental Retardation, X-Linked ,030217 neurology & neurosurgery ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] - Abstract
Item does not contain fulltext Export of mRNA from the cell nucleus to the cytoplasm is essential for protein synthesis, a process vital to all living eukaryotic cells. mRNA export is highly conserved and ubiquitous. Mutations affecting mRNA and mRNA processing or export factors, which cause aberrant retention of mRNAs in the nucleus, are thus emerging as contributors to an important class of human genetic disorders. Here, we report that variants in THOC2, which encodes a subunit of the highly conserved TREX mRNA-export complex, cause syndromic intellectual disability (ID). Affected individuals presented with variable degrees of ID and commonly observed features included speech delay, elevated BMI, short stature, seizure disorders, gait disturbance, and tremors. X chromosome exome sequencing revealed four missense variants in THOC2 in four families, including family MRX12, first ascertained in 1971. We show that two variants lead to decreased stability of THOC2 and its TREX-complex partners in cells derived from the affected individuals. Protein structural modeling showed that the altered amino acids are located in the RNA-binding domains of two complex THOC2 structures, potentially representing two different intermediate RNA-binding states of THOC2 during RNA transport. Our results show that disturbance of the canonical molecular pathway of mRNA export is compatible with life but results in altered neuronal development with other comorbidities.
- Published
- 2015
- Full Text
- View/download PDF
45. New insights into Brunner syndrome and potential for targeted therapy
- Author
-
Melanie Leffler, J. Earl, N.W. Cheung, Carolyn Rogers, Jozef Gecz, Hao Hu, Renee Carroll, Stefan A. Haas, Vera M. Kalscheuer, Bernard Champion, Michael Field, Elizabeth E. Palmer, and Marie Shaw
- Subjects
Pediatrics ,medicine.medical_specialty ,biology ,Monoamine oxidase ,business.industry ,Brunner syndrome ,Serotonin reuptake inhibitor ,medicine.disease ,Serotonergic ,030227 psychiatry ,03 medical and health sciences ,0302 clinical medicine ,Autism spectrum disorder ,Genetics ,medicine ,biology.protein ,Serotonin ,Monoamine oxidase A ,business ,030217 neurology & neurosurgery ,Genetics (clinical) ,Serotonin Measurement - Abstract
We report two families with Brunner syndrome living in one state of Australia. The first family had a predicted protein-truncating variant of monoamine oxidase A (MAOA) (p.S251KfsX2). Affected males had mild intellectual disability (ID), obsessive behaviour, limited friendships and were introverted and placid during clinical interview. The family disclosed episodic explosive aggression after a diagnosis was made. The second family had a missense variant in MAOA (p.R45W). Affected males had borderline-mild ID, attention deficit disorder and limited friendships. One had a history of explosive aggression in childhood and episodic symptoms of flushing, headaches and diarrhoea. Their carrier mother had normal intelligence but similar episodic symptoms. Characteristic biochemical abnormalities included high serum serotonin and urinary metanephrines and low urinary 5-hydroxyindoleacetic acid (5-HIAA) and vanillylmandelic acid (VMA). Symptomatic individuals in the second family had particularly high serotonin levels, and treatment with a serotonin reuptake inhibitor and dietary modification resulted in reversal of biochemical abnormalities, reduction of 'serotonergic' symptoms and behavioural improvement. Brunner syndrome should be considered as a cause of mild ID with paroxysmal behavioural symptoms. It can be screened for with serum/urine metanephrine and serotonin measurement. Cautious treatment with a serotonin reuptake inhibitor, dietary modifications and avoidance of medications contraindicated in patients on monoamine oxidase inhibitors can improve symptoms.
- Published
- 2015
- Full Text
- View/download PDF
46. Redefining the MED13L syndrome
- Author
-
Barbara Delle Chiaie, Filip Roelens, Gabriela Soares, Abidemi A. Adegbola, Sylvie Picker-Minh, Angela M. Kaindl, Bert Callewaert, Thomas F. Wienker, Bertrand Isidor, Gerald F. Cox, Hossein Najmabadi, Annelies Dheedene, Vera M. Kalscheuer, Steven P. Angus, Nele Bockaert, Hao Hu, Fátima Lopes, Olivier Vanakker, Luciana Musante, Andrew Chess, Björn Menten, João Silva, Patrícia Maciel, Hans-Hilger Ropers, John F. Staropoli, Kimia Kahrizi, Karin Decaestecker, Vanessa Suckow, Oliver Bartsch, Cédric Le Caignec, Christoph Hübner, Ute Fischer, and Universidade do Minho
- Subjects
Male ,Adolescent ,Heart malformation ,Transposition of Great Vessels ,RNA polymerase II ,Bioinformatics ,Article ,Mediator ,Intellectual Disability ,Intellectual disability ,Genetics ,medicine ,Transcriptional regulation ,Humans ,Abnormalities, Multiple ,Child ,Transcription factor ,Genetics (clinical) ,Science & Technology ,Mediator Complex ,biology ,Muscular hypotonia ,Syndrome ,medicine.disease ,Phenotype ,Child, Preschool ,Mutation ,biology.protein ,Muscle Hypotonia ,Female ,Neurocognitive - Abstract
Congenital cardiac and neurodevelopmental deficits have been recently linked to the mediator complex subunit 13-like protein MED13L, a subunit of the CDK8-associated mediator complex that functions in transcriptional regulation through DNA-binding transcription factors and RNA polymerase II. Heterozygous MED13L variants cause transposition of the great arteries and intellectual disability (ID). Here, we report eight patients with predominantly novel MED13L variants who lack such complex congenital heart malformations. Rather, they depict a syndromic form of ID characterized by facial dysmorphism, ID, speech impairment, motor developmental delay with muscular hypotonia and behavioral difficulties. We thereby define a novel syndrome and significantly broaden the clinical spectrum associated with MED13L variants. A prominent feature of the MED13L neurocognitive presentation is profound language impairment, often in combination with articulatory deficits., We thank all families for participation in this study, Bettina Lipkowitz and Susanne Freier for excellent technical assistance. This work was supported by the Deutsches Humangenom-Programm (DHGP, grant number 01KW9908), the Nationales Genomforschungsnetzwerk (NGFN, project number 01GR0105), the German Research Foundation (SFB665), the Brain and Behavior Foundation (AA), the Berlin Institute of Health (BIH), the Sonnenfeld Stiftung, the Senate of Berlin by funds to the Berlin Institute for Medical Systems Biology (BIMSB), the Iranian National Science foundation, FEDER funds through the COMPETE program, Portuguese national funds through FCT - Fundacao para a Ciencia e Tecnologia (project PIC/IC/83026/2007, scholarship to FL SFRH/BD/84650/2010), the Max Planck Society and the EU FP 7 project GENCODYS (grant number 241995).
- Published
- 2015
- Full Text
- View/download PDF
47. De Novo Mutations in SLC25A24 Cause a Craniosynostosis Syndrome with Hypertrichosis, Progeroid Appearance, and Mitochondrial Dysfunction
- Author
-
Denise Horn, Penelope E. Bonnen, Anna Floriane Hennig, Marten Jäger, Fernando Scaglia, Bernd Wollnik, Stefan Mundlos, Christian Netzer, Markus Schuelke, Uwe Kornak, Beatrix Fauler, Luitgard Graul-Neumann, Namrata Saha, Holger Thiele, Peter Krawitz, Lara Segebrecht, Jochen Hecht, Nadja Ehmke, Thorsten Mielke, Gökhan Yigit, Rainer Koenig, Carlos A. Bacino, Friederike Hennig, Nicolai Adolphs, Janine Altmüller, Pilar L. Magoulas, Lukasz Smorag, Vera M. Kalscheuer, Peter Nürnberg, Ulrike Krüger, Björn Fischer-Zirnsak, and Esra Kılıç
- Subjects
0301 basic medicine ,Hypertrichosis ,Mitochondrion ,Microphthalmia ,Antiporters ,Cutis Laxa ,Craniofacial Abnormalities ,0302 clinical medicine ,Adenosine Triphosphate ,Progeria ,Exome ,Inner mitochondrial membrane ,Child ,Ductus Arteriosus, Patent ,Genetics (clinical) ,Growth Disorders ,Membrane Potential, Mitochondrial ,Fetal Growth Retardation ,3. Good health ,Mitochondria ,Premature aging ,Child, Preschool ,Female ,Gorlin-chaudhry-moss syndrome ,medicine.medical_specialty ,Mitochondrial DNA ,Adolescent ,Biology ,DNA, Mitochondrial ,Mitochondrial Proteins ,03 medical and health sciences ,Craniosynostoses ,Craniosynostosis ,Internal medicine ,Report ,Genetics ,medicine ,Humans ,Abnormalities, Multiple ,Cutis laxa ,SLC25A24 ,Calcium-Binding Proteins ,Infant ,Hydrogen Peroxide ,Fibroblasts ,medicine.disease ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,Oxidative stress ,Mitochondrial swelling ,Mutation ,Lipoatrophy ,030217 neurology & neurosurgery - Abstract
Gorlin-Chaudhry-Moss syndrome (GCMS) is a dysmorphic syndrome characterized by coronal craniosynostosis and severe midface hypoplasia, body and facial hypertrichosis, microphthalmia, short stature, and short distal phalanges. Variable lipoatrophy and cutis laxa are the basis for a progeroid appearance. Using exome and genome sequencing, we identified the recurrent de novo mutations c.650G>A (p.Arg217His) and c.649C>T (p.Arg217Cys) in SLC25A24 in five unrelated girls diagnosed with GCMS. Two of the girls had pronounced neonatal progeroid features and were initially diagnosed with Wiedemann-Rautenstrauch syndrome. SLC25A24 encodes a mitochondrial inner membrane ATP-Mg/Pi carrier. In fibroblasts from affected individuals, the mutated SLC25A24 showed normal stability. In contrast to control cells, the probands' cells showed mitochondrial swelling, which was exacerbated upon treatment with hydrogen peroxide (H2O2). The same effect was observed after overexpression of the mutant cDNA. Under normal culture conditions, the mitochondrial membrane potential of the probands' fibroblasts was intact, whereas ATP content in the mitochondrial matrix was lower than that in control cells. However, upon H2O2 exposure, the membrane potential was significantly elevated in cells harboring the mutated SLC25A24. No reduction of mitochondrial DNA copy number was observed. These findings demonstrate that mitochondrial dysfunction with increased sensitivity to oxidative stress is due to the SLC25A24 mutations. Our results suggest that the SLC25A24 mutations induce a gain of pathological function and link mitochondrial ATP-Mg/Pi transport to the development of skeletal and connective tissue. N.E. is a participant in the Berlin Institute of Health Charité Clinician Scientist Program, funded by the Charité - Universitätsmedizin Berlin and the Berlin Institute of Health. S.M. was supported by grants from the Deutsche Forschungsgemeinschaft (DFG) and the Max Planck Foundation, B.W. was supported by grants from the DFG SFB1002 project D02, and B.F.-Z. was supported by a grant from the DFG (FI 2240/1-1). U.K. received funding from FP7-EU grant agreement no. 602300 (SYBIL) and the DFG Research Unit FOR 2165 (249509554). Research reported in this publication was supported by National Institute of Neurological Disorders and Stroke of the National Institutes of Health under award number R01NS08372 to P.E.B.
- Published
- 2017
48. Epilepsy and intellectual disability linked protein Shrm4 interaction with GABA B Rs shapes inhibitory neurotransmission
- Author
-
Bernhard Bettler, Luisa Ponzoni, Luca Murru, Trevor G. Smart, Edoardo Moretto, Pamela Valnegri, Francesca Fanelli, Saad Hannan, Silvia Bassani, Daniela Braida, Matteo Fossati, Jeffrey D. Hildebrand, Carlo Sala, Maura Francolini, Jonathan Zapata, Christopher Heise, Lorena Benedetti, Maria Passafaro, Davide Mazza, Mariaelvina Sala, Vera M. Kalscheuer, Anna Longatti, Zapata, J, Moretto, E, Hannan, S, Murru, L, Longatti, A, Mazza, D, Benedetti, L, Fossati, M, Heise, C, Ponzoni, L, Valnegri, P, Braida, D, Sala, M, Francolini, M, Hildebrand, J, Kalscheuer, V, Fanelli, F, Sala, C, Bettler, B, Bassani, S, Smart, Tg, and Passafaro, M
- Subjects
0301 basic medicine ,Multidisciplinary ,Dendritic spine ,Dentate gyrus ,Science ,HEK 293 cells ,Synaptogenesis ,General Physics and Astronomy ,General Chemistry ,GABAB receptor ,Neurotransmission ,Biology ,Hippocampal formation ,Bioinformatics ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Chemistry (all) ,Biochemistry, Genetics and Molecular Biology (all) ,03 medical and health sciences ,Epilepsy ,030104 developmental biology ,medicine ,Neuroscience - Abstract
Shrm4, a protein expressed only in polarized tissues, is encoded by the KIAA1202 gene, whose mutations have been linked to epilepsy and intellectual disability. However, a physiological role for Shrm4 in the brain is yet to be established. Here, we report that Shrm4 is localized to synapses where it regulates dendritic spine morphology and interacts with the C terminus of GABAB receptors (GABABRs) to control their cell surface expression and intracellular trafficking via a dynein-dependent mechanism. Knockdown of Shrm4 in rat severely impairs GABABR activity causing increased anxiety-like behaviour and susceptibility to seizures. Moreover, Shrm4 influences hippocampal excitability by modulating tonic inhibition in dentate gyrus granule cells, in a process involving crosstalk between GABABRs and extrasynaptic δ-subunit-containing GABAARs. Our data highlights a role for Shrm4 in synaptogenesis and in maintaining GABABR-mediated inhibition, perturbation of which may be responsible for the involvement of Shrm4 in cognitive disorders and epilepsy.
- Published
- 2017
49. Mutations in two large pedigrees highlight the role of ZNF711 in X-linked intellectual disability
- Author
-
Arjan P.M. de Brouwer, Céline Helsmoortel, Ajay Anand Kumar, Geert Mortier, Edwin Reyniers, Tjitske Kleefstra, Geeit Vandeweyer, Sandra Janssens, Hans van Bokhoven, Ilse M. van der Werf, Anke Van Dijck, R. Frank Kooy, and Vera M. Kalscheuer
- Subjects
Adult ,Male ,0301 basic medicine ,Adolescent ,Autism Spectrum Disorder ,X-linked intellectual disability ,Population ,Gene Expression ,Pedigree chart ,Disease ,030105 genetics & heredity ,Biology ,medicine.disease_cause ,Severity of Illness Index ,03 medical and health sciences ,Genes, X-Linked ,Intellectual Disability ,Intellectual disability ,Genetics ,medicine ,Humans ,Articulation Disorders ,Exome ,Genetic Predisposition to Disease ,Child ,education ,Mutation ,education.field_of_study ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Base Sequence ,Genetic heterogeneity ,Sequence Analysis, DNA ,General Medicine ,Middle Aged ,medicine.disease ,Pedigree ,DNA-Binding Proteins ,Phenotype ,030104 developmental biology ,Autism ,Female ,Human medicine ,Genome-Wide Association Study - Abstract
Item does not contain fulltext Intellectual disability (ID) affects approximately 1-2% of the general population and is characterized by impaired cognitive abilities. ID is both clinically as well as genetically heterogeneous, up to 2000 genes are estimated to be involved in the emergence of the disease with various clinical presentations. For many genes, only a few patients have been reported and causality of some genes has been questioned upon the discovery of apparent loss-of-function mutations in healthy controls. Description of additional patients strengthens the evidence for the involvement of a gene in the disease and can clarify the clinical phenotype associated with mutations in a particular gene. Here, we present two large four-generation families with a total of 11 males affected with ID caused by mutations in ZNF711, thereby expanding the total number of families with ID and a ZNF711 mutation to four. Patients with mutations in ZNF711 all present with mild to moderate ID and poor speech accompanied by additional features in some patients, including autistic features and mild facial dysmorphisms, suggesting that ZNF711 mutations cause non-syndromic ID.
- Published
- 2017
50. Absent CNKSR2 causes seizures and intellectual, attention, and language deficits
- Author
-
Tove B. Haaland, Mary Lou Smith, Sarah Bowdin, Katja S. Brocke-Holmefjord, Gunnar Houge, Stephen W. Scherer, Katia J. Sinopoli, Brigitte Gilbert-Dussardier, Emmanuelle Lagrue, Christian R. Marshall, Catherine Vincent-Delorme, Susan Walker, Berge A. Minassian, Vera M. Kalscheuer, Andrea K. Vaags, Cindy Gilles, and Radu Harbuz
- Subjects
Disease ,medicine.disease ,Synapse ,Epilepsy ,Synaptic function ,Neurology ,Intellectual disability ,medicine ,CNKSR2 ,Neurology (clinical) ,Early childhood ,Psychology ,Neuroscience ,Brain function - Abstract
Synaptic function is central to brain function. Understanding the synapse is aided by studies of patients lacking individual synaptic proteins. Common neurological diseases are genetically complex. Their understanding is likewise simplified by studies of less common monogenic forms. We detail the disease caused by absence of the synaptic protein CNKSR2 in 8 patients ranging from 6 to 62 years old. The disease is characterized by intellectual disability, attention problems, and abrupt lifelong language loss following a brief early childhood epilepsy with continuous spike-waves in sleep. This study describes the phenotype of CNKSR2 deficiency and its involvement in systems underlying common neurological disorders. Ann Neurol 2014;76:758–764
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.