Your browser doesn't support javascript.
loading
A clustering of heterozygous missense variants in the crucial chromatin modifier WDR5 defines a new neurodevelopmental disorder.
Snijders Blok, Lot; Verseput, Jolijn; Rots, Dmitrijs; Venselaar, Hanka; Innes, A Micheil; Stumpel, Connie; Õunap, Katrin; Reinson, Karit; Seaby, Eleanor G; McKee, Shane; Burton, Barbara; Kim, Katherine; van Hagen, Johanna M; Waisfisz, Quinten; Joset, Pascal; Steindl, Katharina; Rauch, Anita; Li, Dong; Zackai, Elaine H; Sheppard, Sarah E; Keena, Beth; Hakonarson, Hakon; Roos, Andreas; Kohlschmidt, Nicolai; Cereda, Anna; Iascone, Maria; Rebessi, Erika; Kernohan, Kristin D; Campeau, Philippe M; Millan, Francisca; Taylor, Jesse A; Lochmüller, Hanns; Higgs, Martin R; Goula, Amalia; Bernhard, Birgitta; Velasco, Danita J; Schmanski, Andrew A; Stark, Zornitza; Gallacher, Lyndon; Pais, Lynn; Marcogliese, Paul C; Yamamoto, Shinya; Raun, Nicholas; Jakub, Taryn E; Kramer, Jamie M; den Hoed, Joery; Fisher, Simon E; Brunner, Han G; Kleefstra, Tjitske.
Afiliación
  • Snijders Blok L; Human Genetics Department, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Verseput J; Language & Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands.
  • Rots D; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Venselaar H; Human Genetics Department, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Innes AM; Human Genetics Department, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Stumpel C; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Õunap K; Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands.
  • Reinson K; The Department of Medical Genetics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
  • Seaby EG; Department of Clinical Genetics and School for Oncology and Developmental Biology (GROW-School for Oncology and Reproduction), Maastricht UMC+, Maastricht, the Netherlands.
  • McKee S; Department of Clinical Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia.
  • Burton B; Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.
  • Kim K; Department of Clinical Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia.
  • van Hagen JM; Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.
  • Waisfisz Q; Translational Genomics Group, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
  • Joset P; Genomic Informatics Group, University Hospital Southampton, Southampton, UK.
  • Steindl K; Northern Ireland Regional Genetics Service, Belfast City Hospital, Belfast HSC Trust, Belfast BT9 7AB, UK.
  • Rauch A; Ann and Robert H. Lurie Children's Hospital and Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
  • Li D; Ann and Robert H. Lurie Children's Hospital and Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
  • Zackai EH; Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
  • Sheppard SE; Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
  • Keena B; Medical Genetics, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland.
  • Hakonarson H; Institute of Medical Genetics, University of Zuirch, Schlieren-Zurich, Switzerland.
  • Roos A; Institute of Medical Genetics, University of Zuirch, Schlieren-Zurich, Switzerland.
  • Kohlschmidt N; University Children's Hospital Zurich, Zurich, Switzerland.
  • Cereda A; Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Iascone M; Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Rebessi E; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
  • Kernohan KD; Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Campeau PM; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
  • Millan F; Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Taylor JA; Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Lochmüller H; Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Higgs MR; Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Goula A; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
  • Bernhard B; Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, Centre for Neuromuscular Disorders in Children, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
  • Velasco DJ; Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.
  • Schmanski AA; Department of Neurology, University Hospital Bergmannsheil, Heimer Institute for Muscle Research, 44789 Bochum, Germany.
  • Stark Z; Institute of Clinical Genetics and Tumor Genetics, Bonn, Germany.
  • Gallacher L; Department of Pediatrics, ASST Papa Giovanni XXIII, Bergamo, Italy.
  • Pais L; Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy.
  • Marcogliese PC; Pediatric Neurological Unit and Epilespy Center, Fatebenefratelli Hospital, Milan, Italy.
  • Yamamoto S; Newborn Screening Ontario, Children's Hospital of Eastern Ontario and Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.
  • Raun N; CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada.
  • Jakub TE; Sainte-Justine Hospital, University of Montreal, Montreal, QC H3T 1C5, Canada.
  • Kramer JM; GeneDx, Gaithersburg, MD 20877, USA.
  • den Hoed J; Division of Plastic Surgery, Department of Surgery, The Children's Hospital of Philadelpia, Philadelphia, PA, USA.
  • Fisher SE; Children's Hospital of Eastern Ontario Research Institute, Division of Neurology, Department of Medicine, the Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada.
  • Brunner HG; Lysine Methylation and DNA Damage Laboratory, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
  • Kleefstra T; Lysine Methylation and DNA Damage Laboratory, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
HGG Adv ; 4(1): 100157, 2023 01 12.
Article en En | MEDLINE | ID: mdl-36408368
WDR5 is a broadly studied, highly conserved key protein involved in a wide array of biological functions. Among these functions, WDR5 is a part of several protein complexes that affect gene regulation via post-translational modification of histones. We collected data from 11 unrelated individuals with six different rare de novo germline missense variants in WDR5; one identical variant was found in five individuals and another variant in two individuals. All individuals had neurodevelopmental disorders including speech/language delays (n = 11), intellectual disability (n = 9), epilepsy (n = 7), and autism spectrum disorder (n = 4). Additional phenotypic features included abnormal growth parameters (n = 7), heart anomalies (n = 2), and hearing loss (n = 2). Three-dimensional protein structures indicate that all the residues affected by these variants are located at the surface of one side of the WDR5 protein. It is predicted that five out of the six amino acid substitutions disrupt interactions of WDR5 with RbBP5 and/or KMT2A/C, as part of the COMPASS (complex proteins associated with Set1) family complexes. Our experimental approaches in Drosophila melanogaster and human cell lines show normal protein expression, localization, and protein-protein interactions for all tested variants. These results, together with the clustering of variants in a specific region of WDR5 and the absence of truncating variants so far, suggest that dominant-negative or gain-of-function mechanisms might be at play. All in all, we define a neurodevelopmental disorder associated with missense variants in WDR5 and a broad range of features. This finding highlights the important role of genes encoding COMPASS family proteins in neurodevelopmental disorders.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Proteínas de Drosophila / Trastornos del Neurodesarrollo / Trastorno del Espectro Autista / Trastornos del Desarrollo del Lenguaje Límite: Animals / Humans Idioma: En Revista: HGG Adv Año: 2023 Tipo del documento: Article País de afiliación: Países Bajos Pais de publicación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Proteínas de Drosophila / Trastornos del Neurodesarrollo / Trastorno del Espectro Autista / Trastornos del Desarrollo del Lenguaje Límite: Animals / Humans Idioma: En Revista: HGG Adv Año: 2023 Tipo del documento: Article País de afiliación: Países Bajos Pais de publicación: Estados Unidos