Missense variants in ANKRD11 cause KBG syndrome by impairment of stability or transcriptional activity of the encoded protein

Purpose: Although haploinsufficiency of ANKRD11 is among the most common genetic causes of neurodevelopmental disorders, the role of rare ANKRD11 missense variation remains unclear. We characterized the clinical, molecular and functional spectra of ANKRD11 missense variants. Methods: We collected clinical information of individuals with ANKRD11 missense variants and evaluated phenotypic fit to KBG syndrome. We assessed pathogenicity of variants by in silico analyses and cell-based experiments. Results: We identified 29 individuals with (mostly de novo) ANKRD11 missense variants, who presented with syndromic neurodevelopmental disorders and were phenotypically similar to individuals with KBG syndrome caused by ANKRD11 protein truncating variants or 16q24.3 microdeletions. Missense variants significantly clustered in Repression Domain 2. Cellularly, most variants caused reduced ANKRD11 stability. One variant resulted in decreased proteasome degradation and loss of ANKRD11 transcriptional activity. Conclusion: Our study indicates that pathogenic heterozygous missense variants in ANKRD11 cause the clinically recognizable KBG syndrome. Disrupted transrepression capacity and reduced protein stability each independently lead to ANKRD11 loss-of-function, consistent with haploinsufficiency. This highlights the diagnostic relevance of ANKRD11 missense variants, but also poses diagnostic challenges, as the KBG-associated phenotype may be mild and inherited pathogenic ANKRD11 (missense) variants are increasingly observed, warranting stringent variant classification and careful phenotyping.

[1]  Oriol Vinyals,et al.  Highly accurate protein structure prediction with AlphaFold , 2021, Nature.

[2]  Yun Yang,et al.  Two loss‐of‐function ANKRD11 variants in Chinese patients with short stature and a possible molecular pathway , 2020, American journal of medical genetics. Part A.

[3]  C. Hanly,et al.  Description of neurodevelopmental phenotypes associated with 10 genetic neurodevelopmental disorders: A scoping review , 2020, Clinical genetics.

[4]  Rebecca C. Spillmann,et al.  Mutation-specific pathophysiological mechanisms define different neurodevelopmental disorders associated with SATB1 dysfunction , 2020, bioRxiv.

[5]  Patrick J. Short,et al.  Evidence for 28 genetic disorders discovered by combining healthcare and research data , 2020, Nature.

[6]  B. Puisac,et al.  Pathogenic variants in EP300 and ANKRD11 in patients with phenotypes overlapping Cornelia de Lange syndrome , 2020, American journal of medical genetics. Part A.

[7]  M. Digilio,et al.  KBG syndrome: Common and uncommon clinical features based on 31 new patients , 2020, American journal of medical genetics. Part A.

[8]  Ryan L. Collins,et al.  The mutational constraint spectrum quantified from variation in 141,456 humans , 2020, Nature.

[9]  Michael F. Wangler,et al.  Biases in arginine codon usage correlate with genetic disease risk , 2019, bioRxiv.

[10]  F. Cremers,et al.  Identification of splice defects due to noncanonical splice site or deep‐intronic variants in ABCA4 , 2019, Human mutation.

[11]  David G. Knowles,et al.  Predicting Splicing from Primary Sequence with Deep Learning , 2019, Cell.

[12]  Tudor Groza,et al.  Expansion of the Human Phenotype Ontology (HPO) knowledge base and resources , 2018, Nucleic Acids Res..

[13]  Chunlei Liu,et al.  ClinVar: improving access to variant interpretations and supporting evidence , 2017, Nucleic Acids Res..

[14]  M. Tekin,et al.  KBG syndrome , 2017, Orphanet Journal of Rare Diseases.

[15]  G. Vriend,et al.  Spatial Clustering of de Novo Missense Mutations Identifies Candidate Neurodevelopmental Disorder-Associated Genes. , 2017, American journal of human genetics.

[16]  M. Daly,et al.  Regional missense constraint improves variant deleteriousness prediction , 2017, bioRxiv.

[17]  R. Pfundt,et al.  Clinical and molecular findings in 39 patients with KBG syndrome caused by deletion or mutation of ANKRD11 , 2016, American journal of medical genetics. Part A.

[18]  J. Clayton-Smith,et al.  Clinical and genetic aspects of KBG syndrome , 2016, American Journal of Medical Genetics. Part A.

[19]  F. Cunningham,et al.  The Ensembl Variant Effect Predictor , 2016, Genome Biology.

[20]  Desmond G. Higgins,et al.  ProViz—a web-based visualization tool to investigate the functional and evolutionary features of protein sequences , 2016, Nucleic Acids Res..

[21]  D. Valle,et al.  GeneMatcher: A Matching Tool for Connecting Investigators with an Interest in the Same Gene , 2015, Human mutation.

[22]  Bale,et al.  Standards and Guidelines for the Interpretation of Sequence Variants: A Joint Consensus Recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology , 2015, Genetics in Medicine.

[23]  Lin Gao,et al.  HPOSim: An R Package for Phenotypic Similarity Measure and Enrichment Analysis Based on the Human Phenotype Ontology , 2015, PloS one.

[24]  S. Scherer,et al.  Ankrd11 is a chromatin regulator involved in autism that is essential for neural development. , 2015, Developmental cell.

[25]  Tomas W. Fitzgerald,et al.  Large-scale discovery of novel genetic causes of developmental disorders , 2014, Nature.

[26]  B. V. van Bon,et al.  Further delineation of the KBG syndrome phenotype caused by ANKRD11 aberrations , 2014, European Journal of Human Genetics.

[27]  Juan I. Young,et al.  Characterization of ANKRD11 mutations in humans and mice related to KBG syndrome , 2015, Human Genetics.

[28]  Mustafa Tekin,et al.  Mutations in ANKRD11 cause KBG syndrome, characterized by intellectual disability, skeletal malformations, and macrodontia. , 2011, American journal of human genetics.

[29]  Manuel Corpas,et al.  DECIPHER: Database of Chromosomal Imbalance and Phenotype in Humans Using Ensembl Resources. , 2009, American journal of human genetics.

[30]  Steve D. M. Brown,et al.  An ENU-induced mutation in the Ankrd11 gene results in an osteopenia-like phenotype in the mouse mutant Yoda. , 2008, Physiological genomics.

[31]  Chia-Wei Li,et al.  Characterization of transcriptional regulatory domains of ankyrin repeat cofactor-1. , 2007, Biochemical and biophysical research communications.

[32]  Philip S. Yu,et al.  A new method to measure the semantic similarity of GO terms , 2007, Bioinform..

[33]  A. Slavotinek,et al.  KBG Syndrome: Report of Twins, Neurological Characteristics, and Delineation of Diagnostic Criteria , 2007 .

[34]  Sofia Khan,et al.  Spectrum of disease-causing mutations in protein secondary structures , 2015 .

[35]  A. Zhang,et al.  Identification of a Novel Family of Ankyrin Repeats Containing Cofactors for p160 Nuclear Receptor Coactivators* , 2004, Journal of Biological Chemistry.

[36]  M. Walsh,et al.  CCAAT displacement protein/cut homolog recruits G9a histone lysine methyltransferase to repress transcription. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[37]  M. Nachman,et al.  Estimate of the mutation rate per nucleotide in humans. , 2000, Genetics.

[38]  J. Trent,et al.  WAF1, a potential mediator of p53 tumor suppression , 1993, Cell.

[39]  Peter J. Rousseeuw,et al.  Clustering by means of medoids , 1987 .

[40]  J. Opitz,et al.  The KBG syndrome-a syndrome of short stature, characteristic facies, mental retardation, macrodontia and skeletal anomalies. , 1975, Birth defects original article series.