601
|
Exome Pool-Seq in neurodevelopmental disorders. Eur J Hum Genet 2017; 25:1364-1376. [PMID: 29158550 PMCID: PMC5865117 DOI: 10.1038/s41431-017-0022-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/20/2017] [Accepted: 09/22/2017] [Indexed: 12/22/2022] Open
Abstract
High throughput sequencing has greatly advanced disease gene identification, especially in heterogeneous entities. Despite falling costs this is still an expensive and laborious technique, particularly when studying large cohorts. To address this problem we applied Exome Pool-Seq as an economic and fast screening technology in neurodevelopmental disorders (NDDs). Sequencing of 96 individuals can be performed in eight pools of 12 samples on less than one Illumina sequencer lane. In a pilot study with 96 cases we identified 27 variants, likely or possibly affecting function. Twenty five of these were identified in 923 established NDD genes (based on SysID database, status November 2016) (ACTB, AHDC1, ANKRD11, ATP6V1B2, ATRX, CASK, CHD8, GNAS, IFIH1, KCNQ2, KMT2A, KRAS, MAOA, MED12, MED13L, RIT1, SETD5, SIN3A, TCF4, TRAPPC11, TUBA1A, WAC, ZBTB18, ZMYND11), two in 543 (SysID) candidate genes (ZNF292, BPTF), and additionally a de novo loss-of-function variant in LRRC7, not previously implicated in NDDs. Most of them were confirmed to be de novo, but we also identified X-linked or autosomal-dominantly or autosomal-recessively inherited variants. With a detection rate of 28%, Exome Pool-Seq achieves comparable results to individual exome analyses but reduces costs by >85%. Compared with other large scale approaches using Molecular Inversion Probes (MIP) or gene panels, it allows flexible re-analysis of data. Exome Pool-Seq is thus well suited for large-scale, cost-efficient and flexible screening in characterized but heterogeneous entities like NDDs.
Collapse
|
602
|
Jin ZB, Li Z, Liu Z, Jiang Y, Cai XB, Wu J. Identification of de novo germline mutations and causal genes for sporadic diseases using trio-based whole-exome/genome sequencing. Biol Rev Camb Philos Soc 2017; 93:1014-1031. [PMID: 29154454 DOI: 10.1111/brv.12383] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 09/28/2017] [Accepted: 10/10/2017] [Indexed: 12/14/2022]
Abstract
Whole-genome or whole-exome sequencing (WGS/WES) of the affected proband together with normal parents (trio) is commonly adopted to identify de novo germline mutations (DNMs) underlying sporadic cases of various genetic disorders. However, our current knowledge of the occurrence and functional effects of DNMs remains limited and accurately identifying the disease-causing DNM from a group of irrelevant DNMs is complicated. Herein, we provide a general-purpose discussion of important issues related to pathogenic gene identification based on trio-based WGS/WES data. Specifically, the relevance of DNMs to human sporadic diseases, current knowledge of DNM biogenesis mechanisms, and common strategies or software tools used for DNM detection are reviewed, followed by a discussion of pathogenic gene prioritization. In addition, several key factors that may affect DNM identification accuracy and causal gene prioritization are reviewed. Based on recent major advances, this review both sheds light on how trio-based WGS/WES technologies can play a significant role in the identification of DNMs and causal genes for sporadic diseases, and also discusses existing challenges.
Collapse
Affiliation(s)
- Zi-Bing Jin
- Division of Ophthalmic Genetics, The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China.,State Key Laboratory of Ophthalmology Optometry and Vision Science, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhongshan Li
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhenwei Liu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yi Jiang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xue-Bi Cai
- Division of Ophthalmic Genetics, The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China.,State Key Laboratory of Ophthalmology Optometry and Vision Science, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jinyu Wu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| |
Collapse
|
603
|
Spicer TP, Hubbs C, Vaissiere T, Collia D, Rojas C, Kilinc M, Vick K, Madoux F, Baillargeon P, Shumate J, Martemyanov KA, Page DT, Puthanveettil S, Hodder P, Davis R, Miller CA, Scampavia L, Rumbaugh G. Improved Scalability of Neuron-Based Phenotypic Screening Assays for Therapeutic Discovery in Neuropsychiatric Disorders. MOLECULAR NEUROPSYCHIATRY 2017; 3:141-150. [PMID: 29594133 DOI: 10.1159/000481731] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/08/2017] [Indexed: 02/05/2023]
Abstract
There is a pressing need to improve approaches for drug discovery related to neuropsychiatric disorders (NSDs). Therapeutic discovery in neuropsychiatric disorders would benefit from screening assays that can measure changes in complex phenotypes linked to disease mechanisms. However, traditional assays that track complex neuronal phenotypes, such as neuronal connectivity, exhibit poor scalability and are not compatible with high-throughput screening (HTS) procedures. Therefore, we created a neuronal phenotypic assay platform that focused on improving the scalability and affordability of neuron-based assays capable of tracking disease-relevant phenotypes. First, using inexpensive laboratory-level automation, we industrialized primary neuronal culture production, which enabled the creation of scalable assays within functioning neural networks. We then developed a panel of phenotypic assays based on culturing of primary neurons from genetically modified mice expressing HTS-compatible reporters that capture disease-relevant phenotypes. We demonstrated that a library of 1,280 compounds was quickly screened against both assays using only a few litters of mice in a typical academic laboratory setting. Finally, we implemented one assay in a fully automated high-throughput academic screening facility, illustrating the scalability of assays designed using this platform. These methodological improvements simplify the creation of highly scalable neuron-based phenotypic assays designed to improve drug discovery in CNS disorders.
Collapse
Affiliation(s)
| | - Christopher Hubbs
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Thomas Vaissiere
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | | | - Camilo Rojas
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Murat Kilinc
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Kyle Vick
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA.,Department of Aerie Pharmaceuticals, Durham, NC, USA
| | - Franck Madoux
- Department of Molecular Medicine, Jupiter, FL, USA.,Department of Amgen, Thousand Oaks, CA, USA
| | | | | | | | - Damon T Page
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | | | - Peter Hodder
- Department of Molecular Medicine, Jupiter, FL, USA.,Department of Amgen, Thousand Oaks, CA, USA
| | - Ronald Davis
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Courtney A Miller
- Department of Molecular Medicine, Jupiter, FL, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | | | - Gavin Rumbaugh
- Department of Molecular Medicine, Jupiter, FL, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| |
Collapse
|
604
|
Clinical whole-exome sequencing for the diagnosis of rare disorders with congenital anomalies and/or intellectual disability: substantial interest of prospective annual reanalysis. Genet Med 2017; 20:645-654. [DOI: 10.1038/gim.2017.162] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/25/2017] [Indexed: 12/11/2022] Open
|
605
|
De Novo Mutations in Protein Kinase Genes CAMK2A and CAMK2B Cause Intellectual Disability. Am J Hum Genet 2017; 101:768-788. [PMID: 29100089 DOI: 10.1016/j.ajhg.2017.10.003] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/09/2017] [Indexed: 02/08/2023] Open
Abstract
Calcium/calmodulin-dependent protein kinase II (CAMK2) is one of the first proteins shown to be essential for normal learning and synaptic plasticity in mice, but its requirement for human brain development has not yet been established. Through a multi-center collaborative study based on a whole-exome sequencing approach, we identified 19 exceedingly rare de novo CAMK2A or CAMK2B variants in 24 unrelated individuals with intellectual disability. Variants were assessed for their effect on CAMK2 function and on neuronal migration. For both CAMK2A and CAMK2B, we identified mutations that decreased or increased CAMK2 auto-phosphorylation at Thr286/Thr287. We further found that all mutations affecting auto-phosphorylation also affected neuronal migration, highlighting the importance of tightly regulated CAMK2 auto-phosphorylation in neuronal function and neurodevelopment. Our data establish the importance of CAMK2A and CAMK2B and their auto-phosphorylation in human brain function and expand the phenotypic spectrum of the disorders caused by variants in key players of the glutamatergic signaling pathway.
Collapse
|
606
|
De Novo Missense Mutations in DHX30 Impair Global Translation and Cause a Neurodevelopmental Disorder. Am J Hum Genet 2017; 101:716-724. [PMID: 29100085 DOI: 10.1016/j.ajhg.2017.09.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/19/2017] [Indexed: 11/21/2022] Open
Abstract
DHX30 is a member of the family of DExH-box helicases, which use ATP hydrolysis to unwind RNA secondary structures. Here we identified six different de novo missense mutations in DHX30 in twelve unrelated individuals affected by global developmental delay (GDD), intellectual disability (ID), severe speech impairment and gait abnormalities. While four mutations are recurrent, two are unique with one affecting the codon of one recurrent mutation. All amino acid changes are located within highly conserved helicase motifs and were found to either impair ATPase activity or RNA recognition in different in vitro assays. Moreover, protein variants exhibit an increased propensity to trigger stress granule (SG) formation resulting in global translation inhibition. Thus, our findings highlight the prominent role of translation control in development and function of the central nervous system and also provide molecular insight into how DHX30 dysfunction might cause a neurodevelopmental disorder.
Collapse
|
607
|
Hamdan FF, Myers CT, Cossette P, Lemay P, Spiegelman D, Laporte AD, Nassif C, Diallo O, Monlong J, Cadieux-Dion M, Dobrzeniecka S, Meloche C, Retterer K, Cho MT, Rosenfeld JA, Bi W, Massicotte C, Miguet M, Brunga L, Regan BM, Mo K, Tam C, Schneider A, Hollingsworth G, FitzPatrick DR, Donaldson A, Canham N, Blair E, Kerr B, Fry AE, Thomas RH, Shelagh J, Hurst JA, Brittain H, Blyth M, Lebel RR, Gerkes EH, Davis-Keppen L, Stein Q, Chung WK, Dorison SJ, Benke PJ, Fassi E, Corsten-Janssen N, Kamsteeg EJ, Mau-Them FT, Bruel AL, Verloes A, Õunap K, Wojcik MH, Albert DV, Venkateswaran S, Ware T, Jones D, Liu YC, Mohammad SS, Bizargity P, Bacino CA, Leuzzi V, Martinelli S, Dallapiccola B, Tartaglia M, Blumkin L, Wierenga KJ, Purcarin G, O’Byrne JJ, Stockler S, Lehman A, Keren B, Nougues MC, Mignot C, Auvin S, Nava C, Hiatt SM, Bebin M, Shao Y, Scaglia F, Lalani SR, Frye RE, Jarjour IT, Jacques S, Boucher RM, Riou E, Srour M, Carmant L, Lortie A, Major P, Diadori P, Dubeau F, D’Anjou G, Bourque G, Berkovic SF, Sadleir LG, Campeau PM, Kibar Z, Lafrenière RG, Girard SL, Mercimek-Mahmutoglu S, Boelman C, Rouleau GA, et alHamdan FF, Myers CT, Cossette P, Lemay P, Spiegelman D, Laporte AD, Nassif C, Diallo O, Monlong J, Cadieux-Dion M, Dobrzeniecka S, Meloche C, Retterer K, Cho MT, Rosenfeld JA, Bi W, Massicotte C, Miguet M, Brunga L, Regan BM, Mo K, Tam C, Schneider A, Hollingsworth G, FitzPatrick DR, Donaldson A, Canham N, Blair E, Kerr B, Fry AE, Thomas RH, Shelagh J, Hurst JA, Brittain H, Blyth M, Lebel RR, Gerkes EH, Davis-Keppen L, Stein Q, Chung WK, Dorison SJ, Benke PJ, Fassi E, Corsten-Janssen N, Kamsteeg EJ, Mau-Them FT, Bruel AL, Verloes A, Õunap K, Wojcik MH, Albert DV, Venkateswaran S, Ware T, Jones D, Liu YC, Mohammad SS, Bizargity P, Bacino CA, Leuzzi V, Martinelli S, Dallapiccola B, Tartaglia M, Blumkin L, Wierenga KJ, Purcarin G, O’Byrne JJ, Stockler S, Lehman A, Keren B, Nougues MC, Mignot C, Auvin S, Nava C, Hiatt SM, Bebin M, Shao Y, Scaglia F, Lalani SR, Frye RE, Jarjour IT, Jacques S, Boucher RM, Riou E, Srour M, Carmant L, Lortie A, Major P, Diadori P, Dubeau F, D’Anjou G, Bourque G, Berkovic SF, Sadleir LG, Campeau PM, Kibar Z, Lafrenière RG, Girard SL, Mercimek-Mahmutoglu S, Boelman C, Rouleau GA, Scheffer IE, Mefford HC, Andrade DM, Rossignol E, Minassian BA, Michaud JL, Michaud JL. High Rate of Recurrent De Novo Mutations in Developmental and Epileptic Encephalopathies. Am J Hum Genet 2017; 101:664-685. [PMID: 29100083 DOI: 10.1016/j.ajhg.2017.09.008] [Show More Authors] [Citation(s) in RCA: 336] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/11/2017] [Indexed: 12/30/2022] Open
Abstract
Developmental and epileptic encephalopathy (DEE) is a group of conditions characterized by the co-occurrence of epilepsy and intellectual disability (ID), typically with developmental plateauing or regression associated with frequent epileptiform activity. The cause of DEE remains unknown in the majority of cases. We performed whole-genome sequencing (WGS) in 197 individuals with unexplained DEE and pharmaco-resistant seizures and in their unaffected parents. We focused our attention on de novo mutations (DNMs) and identified candidate genes containing such variants. We sought to identify additional subjects with DNMs in these genes by performing targeted sequencing in another series of individuals with DEE and by mining various sequencing datasets. We also performed meta-analyses to document enrichment of DNMs in candidate genes by leveraging our WGS dataset with those of several DEE and ID series. By combining these strategies, we were able to provide a causal link between DEE and the following genes: NTRK2, GABRB2, CLTC, DHDDS, NUS1, RAB11A, GABBR2, and SNAP25. Overall, we established a molecular diagnosis in 63/197 (32%) individuals in our WGS series. The main cause of DEE in these individuals was de novo point mutations (53/63 solved cases), followed by inherited mutations (6/63 solved cases) and de novo CNVs (4/63 solved cases). De novo missense variants explained a larger proportion of individuals in our series than in other series that were primarily ascertained because of ID. Moreover, these DNMs were more frequently recurrent than those identified in ID series. These observations indicate that the genetic landscape of DEE might be different from that of ID without epilepsy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jacques L Michaud
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC H3T1C5, Canada; Department of Neurosciences, Université de Montréal, Montreal, QC H3T1J4, Canada; Department of Pediatrics, Université de Montréal, Montreal, QC H3T1C5, Canada.
| |
Collapse
|
608
|
Lamers IJC, Reijnders MRF, Venselaar H, Kraus A, Jansen S, de Vries BBA, Houge G, Gradek GA, Seo J, Choi M, Chae JH, van der Burgt I, Pfundt R, Letteboer SJF, van Beersum SEC, Dusseljee S, Brunner HG, Doherty D, Kleefstra T, Roepman R. Recurrent De Novo Mutations Disturbing the GTP/GDP Binding Pocket of RAB11B Cause Intellectual Disability and a Distinctive Brain Phenotype. Am J Hum Genet 2017; 101:824-832. [PMID: 29106825 DOI: 10.1016/j.ajhg.2017.09.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/19/2017] [Indexed: 12/20/2022] Open
Abstract
The Rab GTPase family comprises ∼70 GTP-binding proteins, functioning in vesicle formation, transport and fusion. They are activated by a conformational change induced by GTP-binding, allowing interactions with downstream effectors. Here, we report five individuals with two recurrent de novo missense mutations in RAB11B; c.64G>A; p.Val22Met in three individuals and c.202G>A; p.Ala68Thr in two individuals. An overlapping neurodevelopmental phenotype, including severe intellectual disability with absent speech, epilepsy, and hypotonia was observed in all affected individuals. Additionally, visual problems, musculoskeletal abnormalities, and microcephaly were present in the majority of cases. Re-evaluation of brain MRI images of four individuals showed a shared distinct brain phenotype, consisting of abnormal white matter (severely decreased volume and abnormal signal), thin corpus callosum, cerebellar vermis hypoplasia, optic nerve hypoplasia and mild ventriculomegaly. To compare the effects of both variants with known inactive GDP- and active GTP-bound RAB11B mutants, we modeled the variants on the three-dimensional protein structure and performed subcellular localization studies. We predicted that both variants alter the GTP/GDP binding pocket and show that they both have localization patterns similar to inactive RAB11B. Evaluation of their influence on the affinity of RAB11B to a series of binary interactors, both effectors and guanine nucleotide exchange factors (GEFs), showed induction of RAB11B binding to the GEF SH3BP5, again similar to inactive RAB11B. In conclusion, we report two recurrent dominant mutations in RAB11B leading to a neurodevelopmental syndrome, likely caused by altered GDP/GTP binding that inactivate the protein and induce GEF binding and protein mislocalization.
Collapse
Affiliation(s)
- Ideke J C Lamers
- Department of Human Genetics, and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Margot R F Reijnders
- Department of Human Genetics, and Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands.
| | - Hanka Venselaar
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Alison Kraus
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds, LS7 4SA, UK
| | - Sandra Jansen
- Department of Human Genetics, and Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Bert B A de Vries
- Department of Human Genetics, and Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Gunnar Houge
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, N-5021, Norway
| | - Gyri Aasland Gradek
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, N-5021, Norway
| | - Jieun Seo
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Murim Choi
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jong-Hee Chae
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Ineke van der Burgt
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Stef J F Letteboer
- Department of Human Genetics, and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Sylvia E C van Beersum
- Department of Human Genetics, and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Simone Dusseljee
- Department of Human Genetics, and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Han G Brunner
- Department of Human Genetics, and Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands; Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, 6229 ER, the Netherlands
| | - Dan Doherty
- Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, WA 98195, USA
| | - Tjitske Kleefstra
- Department of Human Genetics, and Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Ronald Roepman
- Department of Human Genetics, and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands.
| |
Collapse
|
609
|
Okamoto N, Nakao H, Niihori T, Aoki Y. Patient with a novel purine-rich element binding protein A mutation. Congenit Anom (Kyoto) 2017; 57:201-204. [PMID: 28164378 DOI: 10.1111/cga.12214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/17/2017] [Accepted: 01/30/2017] [Indexed: 12/30/2022]
Abstract
There have been several reports on 5q31.3 microdeletion syndrome. The overlapping deleted region includes purine-rich element binding protein A (PURA), which encodes transcriptional activator protein Pur-α. Patients with PURA mutations show moderate to severe neurodevelopmental delay and learning disability. Neonatal hypotonia, respiratory insufficiency, feeding difficulties, and seizures are often seen. Dysmorphic features including myopathic faces are helpful as clinical signs of the diagnosis. We report a patient with a novel PURA mutation detected by whole-exome sequencing. We suggest that PURA abnormality is a recognizable syndrome.
Collapse
Affiliation(s)
- Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - Hideto Nakao
- Department of Neonatology, Hyogo Prefectural Kobe Children's Hospital Perinatal Center, Kobe, Japan
| | - Tetsuya Niihori
- Department of Medical Genetics, Tohoku University School of Medicine, Sendai, Japan
| | - Yoko Aoki
- Department of Medical Genetics, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|
610
|
Gosalia N, Economides AN, Dewey FE, Balasubramanian S. MAPPIN: a method for annotating, predicting pathogenicity and mode of inheritance for nonsynonymous variants. Nucleic Acids Res 2017; 45:10393-10402. [PMID: 28977528 PMCID: PMC5737764 DOI: 10.1093/nar/gkx730] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/21/2017] [Indexed: 01/24/2023] Open
Abstract
Nonsynonymous single nucleotide variants (nsSNVs) constitute about 50% of known disease-causing mutations and understanding their functional impact is an area of active research. Existing algorithms predict pathogenicity of nsSNVs; however, they are unable to differentiate heterozygous, dominant disease-causing variants from heterozygous carrier variants that lead to disease only in the homozygous state. Here, we present MAPPIN (Method for Annotating, Predicting Pathogenicity, and mode of Inheritance for Nonsynonymous variants), a prediction method which utilizes a random forest algorithm to distinguish between nsSNVs with dominant, recessive, and benign effects. We apply MAPPIN to a set of Mendelian disease-causing mutations and accurately predict pathogenicity for all mutations. Furthermore, MAPPIN predicts mode of inheritance correctly for 70.3% of nsSNVs. MAPPIN also correctly predicts pathogenicity for 87.3% of mutations from the Deciphering Developmental Disorders Study with a 78.5% accuracy for mode of inheritance. When tested on a larger collection of mutations from the Human Gene Mutation Database, MAPPIN is able to significantly discriminate between mutations in known dominant and recessive genes. Finally, we demonstrate that MAPPIN outperforms CADD and Eigen in predicting disease inheritance modes for all validation datasets. To our knowledge, MAPPIN is the first nsSNV pathogenicity prediction algorithm that provides mode of inheritance predictions, adding another layer of information for variant prioritization.
Collapse
Affiliation(s)
- Nehal Gosalia
- Regeneron Genetics Center, Tarrytown, NY 10591, USA.,Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA
| | - Aris N Economides
- Regeneron Genetics Center, Tarrytown, NY 10591, USA.,Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA
| | | | | |
Collapse
|
611
|
SOX5-Null Heterozygous Mutation in a Family with Adult-Onset Hyperkinesia and Behavioral Abnormalities. Case Rep Genet 2017; 2017:2721615. [PMID: 29214085 PMCID: PMC5682053 DOI: 10.1155/2017/2721615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/13/2017] [Indexed: 12/15/2022] Open
Abstract
SOX5 encodes a conserved transcription factor implicated in cell-fate decisions of the neural lineage. SOX5 haploinsufficiency induced by larger genomic deletions has been linked to a recognizable pediatric syndrome combining developmental delay with intellectual disability, mild dysmorphism, inadequate behavior, and variable additional features including motor disturbances. In contrast to SOX5-involving deletions, examples of pathogenic SOX5 small coding variations are sparse in the literature and have been described only in singular cases with phenotypic abnormalities akin to those seen in the SOX5 microdeletion syndrome. Here a novel SOX5 loss-of-function point mutation, c.13C>T (p.Arg5X), is reported, identified in the course of exome sequencing applied to the diagnosis of an unexplained adult-onset motor disorder. Aged 43 years, our female index patient demonstrated abrupt onset of mixed generalized hyperkinesia, with dystonic and choreiform movements being the most salient features. The movement disorder was accompanied by behavioral problems such as anxiety and mood instability. The mutation was found to be inherited to the patient's son who manifested abnormal behavior including diminished social functioning, paranoid ideation, and anxiety since adolescence. Our results expand the compendium of SOX5 damaging single-nucleotide variation mutations and suggest that SOX5 haploinsufficiency might not be restrictively associated with childhood-onset syndromic disease.
Collapse
|
612
|
de Kovel CGF, Syrbe S, Brilstra EH, Verbeek N, Kerr B, Dubbs H, Bayat A, Desai S, Naidu S, Srivastava S, Cagaylan H, Yis U, Saunders C, Rook M, Plugge S, Muhle H, Afawi Z, Klein KM, Jayaraman V, Rajagopalan R, Goldberg E, Marsh E, Kessler S, Bergqvist C, Conlin LK, Krok BL, Thiffault I, Pendziwiat M, Helbig I, Polster T, Borggraefe I, Lemke JR, van den Boogaardt MJ, Møller RS, Koeleman BPC. Neurodevelopmental Disorders Caused by De Novo Variants in KCNB1 Genotypes and Phenotypes. JAMA Neurol 2017; 74:1228-1236. [PMID: 28806457 DOI: 10.1001/jamaneurol.2017.1714] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Importance Knowing the range of symptoms seen in patients with a missense or loss-of-function variant in KCNB1 and how these symptoms correlate with the type of variant will help clinicians with diagnosis and prognosis when treating new patients. Objectives To investigate the clinical spectrum associated with KCNB1 variants and the genotype-phenotype correlations. Design, Setting, and Participants This study summarized the clinical and genetic information of patients with a presumed pathogenic variant in KCNB1. Patients were identified in research projects or during clinical testing. Information on patients from previously published articles was collected and authors contacted if feasible. All patients were seen at a clinic at one of the participating institutes because of presumed genetic disorder. They were tested in a clinical setting or included in a research project. Main Outcomes and Measures The genetic variant and its inheritance and information on the patient's symptoms and characteristics in a predefined format. All variants were identified with massive parallel sequencing and confirmed with Sanger sequencing in the patient. Absence of the variant in the parents could be confirmed with Sanger sequencing in all families except one. Results Of 26 patients (10 female, 15 male, 1 unknown; mean age at inclusion, 9.8 years; age range, 2-32 years) with developmental delay, 20 (77%) carried a missense variant in the ion channel domain of KCNB1, with a concentration of variants in region S5 to S6. Three variants that led to premature stops were located in the C-terminal and 3 in the ion channel domain. Twenty-one of 25 patients (84%) had seizures, with 9 patients (36%) starting with epileptic spasms between 3 and 18 months of age. All patients had developmental delay, with 17 (65%) experiencing severe developmental delay; 14 (82%) with severe delay had behavioral problems. The developmental delay was milder in 4 of 6 patients with stop variants and in a patient with a variant in the S2 transmembrane element rather than the S4 to S6 region. Conclusions and Relevance De novo KCNB1 missense variants in the ion channel domain and loss-of-function variants in this domain and the C-terminal likely cause neurodevelopmental disorders with or without seizures. Patients with presumed pathogenic variants in KCNB1 have a variable phenotype. However, the type and position of the variants in the protein are (imperfectly) correlated with the severity of the disorder.
Collapse
Affiliation(s)
- Carolien G F de Kovel
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Language and Genetics, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Steffen Syrbe
- Division of Child Neurology and Inherited Metabolic Diseases, Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Eva H Brilstra
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Nienke Verbeek
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bronwyn Kerr
- Institute of Evolution, Systems and Genomics, Faculty of Medical and Human Sciences, University of Manchester, Manchester, England.,Manchester Centre For Genomic Medicine, Central Manchester University Hospitals National Health Service Foundation Trust, Manchester, England.,Manchester Academic Health Science Centre, Manchester, England
| | - Holly Dubbs
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Allan Bayat
- Department of Pediatrics, University Hospital of Hvidovre, Copenhagen, Denmark
| | - Sonal Desai
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, Maryland
| | - Sakkubai Naidu
- Department of Neurology and Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland.,Hugo Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland
| | | | - Hande Cagaylan
- Department of Molecular Biology and Genetics, Bogaziçi University, Istanbul, Turkey
| | - Uluc Yis
- Division of Child Neurology, Department of Pediatrics, School of Medicine, Dokuz Eylül University, İzmir, Turkey
| | - Carol Saunders
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, Missouri.,Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri.,Pediatric Pathology and Laboratory Medicine, University of Missouri-Kansas City School of Medicine, Kansas City
| | - Martin Rook
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Susanna Plugge
- Department of Biomedical Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hiltrud Muhle
- Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian Albrechts University, Kiel, Germany
| | - Zaid Afawi
- Department of Physiology and Pharmacology, Tel Aviv University Medical School, Ramat Aviv, Israel
| | - Karl-Martin Klein
- Department of Neurology, Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, University Hospital, Goethe-University Frankfurt, Frankfurt, Germany
| | - Vijayakumar Jayaraman
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ramakrishnan Rajagopalan
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ethan Goldberg
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Eric Marsh
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sudha Kessler
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Christina Bergqvist
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Laura K Conlin
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Bryan L Krok
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Isabelle Thiffault
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, Missouri.,Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Manuela Pendziwiat
- Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian Albrechts University, Kiel, Germany
| | - Ingo Helbig
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,University Medical Center Schleswig-Holstein, Christian Albrechts University, Kiel, Germany
| | - Tilman Polster
- Epilepsiezentrum Bethel, Krankenhaus Mara, Kinderepileptologie, Bielefeld, Germany
| | - Ingo Borggraefe
- Department of Pediatric Neurology, Developmental Medicine and Social Pediatrics Dr. von Hauner's Children's Hospital, University of Munich, Munich, Germany
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Leipzig, Germany
| | | | - Rikke S Møller
- Danish Epilepsy Centre, Dianalund, Denmark.,Institute for Regional Health Services, University of Southern Denmark, Odense, Denmark
| | - Bobby P C Koeleman
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
613
|
Miyamoto T, Akutsu SN, Matsuura S. Updated summary of genome editing technology in human cultured cells linked to human genetics studies. J Hum Genet 2017; 63:133-143. [PMID: 29167553 DOI: 10.1038/s10038-017-0349-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 12/22/2022]
Abstract
Current deep-sequencing technology provides a mass of nucleotide variations associated with human genetic disorders to accelerate the identification of causative mutations. To understand the etiology of genetic disorders, reverse genetics in human cultured cells is a useful approach for modeling a disease in vitro. However, gene targeting in human cultured cells is difficult because of their low activity of homologous recombination. Engineered endonucleases enable enhancement of the local activation of DNA repair pathways at the human genome target site to rewrite the desired sequence, thereby efficiently generating disease-modeling cultured cell clones. These edited cells can be used to explore the molecular functions of a causative gene product to uncover the etiological mechanisms. The correction of mutations in patient cells using genome editing technology could contribute to the development of unique gene therapies. This technology can also be applied to screening causative mutations. Rare genetic disorders and non-exonic mutation-caused diseases remain frontier in the field of human genetics as it is difficult to validate whether the extracted nucleotide variants are mutation or polymorphism. When isogenic human cultured cells with a candidate variant reproduce the pathogenic phenotypes, it is confirmed that the variant is a causative mutation.
Collapse
Affiliation(s)
- Tatsuo Miyamoto
- Department of Genetics and Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| | - Silvia Natsuko Akutsu
- Department of Genetics and Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shinya Matsuura
- Department of Genetics and Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
614
|
Li X, Kim Y, Tsang EK, Davis JR, Damani FN, Chiang C, Hess GT, Zappala Z, Strober BJ, Scott AJ, Li A, Ganna A, Bassik MC, Merker JD, Hall IM, Battle A, Montgomery SB. The impact of rare variation on gene expression across tissues. Nature 2017; 550:239-243. [PMID: 29022581 PMCID: PMC5877409 DOI: 10.1038/nature24267] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 09/13/2017] [Indexed: 12/24/2022]
Abstract
Rare genetic variants are abundant in humans and are expected to contribute to individual disease risk. While genetic association studies have successfully identified common genetic variants associated with susceptibility, these studies are not practical for identifying rare variants. Efforts to distinguish pathogenic variants from benign rare variants have leveraged the genetic code to identify deleterious protein-coding alleles, but no analogous code exists for non-coding variants. Therefore, ascertaining which rare variants have phenotypic effects remains a major challenge. Rare non-coding variants have been associated with extreme gene expression in studies using single tissues, but their effects across tissues are unknown. Here we identify gene expression outliers, or individuals showing extreme expression levels for a particular gene, across 44 human tissues by using combined analyses of whole genomes and multi-tissue RNA-sequencing data from the Genotype-Tissue Expression (GTEx) project v6p release. We find that 58% of underexpression and 28% of overexpression outliers have nearby conserved rare variants compared to 8% of non-outliers. Additionally, we developed RIVER (RNA-informed variant effect on regulation), a Bayesian statistical model that incorporates expression data to predict a regulatory effect for rare variants with higher accuracy than models using genomic annotations alone. Overall, we demonstrate that rare variants contribute to large gene expression changes across tissues and provide an integrative method for interpretation of rare variants in individual genomes.
Collapse
Affiliation(s)
- Xin Li
- Department of Pathology, Stanford University, Stanford, California 94305, USA
| | - Yungil Kim
- Department of Computer Science, Johns Hopkins University, Baltimore 21218, Maryland, USA
| | - Emily K Tsang
- Department of Pathology, Stanford University, Stanford, California 94305, USA
- Biomedical Informatics Program, Stanford University, Stanford, California 94305, USA
| | - Joe R Davis
- Department of Pathology, Stanford University, Stanford, California 94305, USA
- Department of Genetics, Stanford University, Stanford, California 94305, USA
| | - Farhan N Damani
- Department of Computer Science, Johns Hopkins University, Baltimore 21218, Maryland, USA
| | - Colby Chiang
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri 63108, USA
| | - Gaelen T Hess
- Department of Genetics, Stanford University, Stanford, California 94305, USA
| | - Zachary Zappala
- Department of Pathology, Stanford University, Stanford, California 94305, USA
- Department of Genetics, Stanford University, Stanford, California 94305, USA
| | - Benjamin J Strober
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Alexandra J Scott
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri 63108, USA
| | - Amy Li
- Department of Genetics, Stanford University, Stanford, California 94305, USA
| | - Andrea Ganna
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University, Stanford, California 94305, USA
| | - Jason D Merker
- Department of Pathology, Stanford University, Stanford, California 94305, USA
| | - Ira M Hall
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri 63108, USA
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri 63110, USA
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Alexis Battle
- Department of Computer Science, Johns Hopkins University, Baltimore 21218, Maryland, USA
| | - Stephen B Montgomery
- Department of Pathology, Stanford University, Stanford, California 94305, USA
- Department of Genetics, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
615
|
Haploinsufficiency of the Chromatin Remodeler BPTF Causes Syndromic Developmental and Speech Delay, Postnatal Microcephaly, and Dysmorphic Features. Am J Hum Genet 2017; 101:503-515. [PMID: 28942966 DOI: 10.1016/j.ajhg.2017.08.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/10/2017] [Indexed: 12/13/2022] Open
Abstract
Bromodomain PHD finger transcription factor (BPTF) is the largest subunit of nucleosome remodeling factor (NURF), a member of the ISWI chromatin-remodeling complex. However, the clinical consequences of disruption of this complex remain largely uncharacterized. BPTF is required for anterior-posterior axis formation of the mouse embryo and was shown to promote posterior neuroectodermal fate by enhancing Smad2-activated wnt8 expression in zebrafish. Here, we report eight loss-of-function and two missense variants (eight de novo and two of unknown origin) in BPTF on 17q24.2. The BPTF variants were found in unrelated individuals aged between 2.1 and 13 years, who manifest variable degrees of developmental delay/intellectual disability (10/10), speech delay (10/10), postnatal microcephaly (7/9), and dysmorphic features (9/10). Using CRISPR-Cas9 genome editing of bptf in zebrafish to induce a loss of gene function, we observed a significant reduction in head size of F0 mutants compared to control larvae. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and phospho-histone H3 (PH3) staining to assess apoptosis and cell proliferation, respectively, showed a significant increase in cell death in F0 mutants compared to controls. Additionally, we observed a substantial increase of the ceratohyal angle of the craniofacial skeleton in bptf F0 mutants, indicating abnormal craniofacial patterning. Taken together, our data demonstrate the pathogenic role of BPTF haploinsufficiency in syndromic neurodevelopmental anomalies and extend the clinical spectrum of human disorders caused by ablation of chromatin remodeling complexes.
Collapse
|
616
|
Chérot E, Keren B, Dubourg C, Carré W, Fradin M, Lavillaureix A, Afenjar A, Burglen L, Whalen S, Charles P, Marey I, Heide S, Jacquette A, Heron D, Doummar D, Rodriguez D, Billette de Villemeur T, Moutard ML, Guët A, Xavier J, Périsse D, Cohen D, Demurger F, Quélin C, Depienne C, Odent S, Nava C, David V, Pasquier L, Mignot C. Using medical exome sequencing to identify the causes of neurodevelopmental disorders: Experience of 2 clinical units and 216 patients. Clin Genet 2017; 93:567-576. [DOI: 10.1111/cge.13102] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 12/19/2022]
Affiliation(s)
- E. Chérot
- Service de Génétique Médicale; CLAD Ouest CHU Hôpital Sud; Rennes France
- Laboratoire de Génétique moléculaire et Génomique; CHU Pontchaillou; Rennes France
| | - B. Keren
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
- Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme"; UPMC; Paris France
- Groupe de Recherche Clinique (GRC) "ConCer-LD"; UPMC; Paris France
| | - C. Dubourg
- Laboratoire de Génétique moléculaire et Génomique; CHU Pontchaillou; Rennes France
- CNRS UMR 6290 (IGDR); Université de Rennes 1; Rennes France
| | - W. Carré
- Laboratoire de Génétique moléculaire et Génomique; CHU Pontchaillou; Rennes France
| | - M. Fradin
- Service de Génétique Médicale; CLAD Ouest CHU Hôpital Sud; Rennes France
| | - A. Lavillaureix
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
| | - A. Afenjar
- Centre de Référence Déficiences Intellectuelles de Causes Rares Hôpital de la Pitié-Salpêtrière; Paris France
- APHP, GHUEP, Hôpital Armand Trousseau; Centre de Référence 'Malformations et maladies congénitales du cervelet'; Paris France
- Département de Génétique, APHP, GHUEP; Hôpital Armand-Trousseau; Paris France
| | - L. Burglen
- APHP, GHUEP, Hôpital Armand Trousseau; Centre de Référence 'Malformations et maladies congénitales du cervelet'; Paris France
- Département de Génétique, APHP, GHUEP; Hôpital Armand-Trousseau; Paris France
| | - S. Whalen
- APHP, GHUEP, Hôpital Armand Trousseau; Centre de Référence 'Malformations et maladies congénitales du cervelet'; Paris France
- Département de Génétique, APHP, GHUEP; Hôpital Armand-Trousseau; Paris France
| | - P. Charles
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
- Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme"; UPMC; Paris France
- Centre de Référence Déficiences Intellectuelles de Causes Rares Hôpital de la Pitié-Salpêtrière; Paris France
| | - I. Marey
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
- Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme"; UPMC; Paris France
- Centre de Référence Déficiences Intellectuelles de Causes Rares Hôpital de la Pitié-Salpêtrière; Paris France
| | - S. Heide
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
- Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme"; UPMC; Paris France
- Centre de Référence Déficiences Intellectuelles de Causes Rares Hôpital de la Pitié-Salpêtrière; Paris France
| | - A. Jacquette
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
- Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme"; UPMC; Paris France
- Centre de Référence Déficiences Intellectuelles de Causes Rares Hôpital de la Pitié-Salpêtrière; Paris France
| | - D. Heron
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
- Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme"; UPMC; Paris France
- Centre de Référence Déficiences Intellectuelles de Causes Rares Hôpital de la Pitié-Salpêtrière; Paris France
| | - D. Doummar
- Groupe de Recherche Clinique (GRC) "ConCer-LD"; UPMC; Paris France
- AP-HP, Service de Neuropédiatrie; Hôpital Armand Trousseau; Paris France
- AP-HP, Hôpital Armand Trousseau; Centre de Référence Neurogénétique de l'Enfant à l'adulte; Paris France
| | - D. Rodriguez
- Groupe de Recherche Clinique (GRC) "ConCer-LD"; UPMC; Paris France
- AP-HP, Service de Neuropédiatrie; Hôpital Armand Trousseau; Paris France
- AP-HP, Hôpital Armand Trousseau; Centre de Référence Neurogénétique de l'Enfant à l'adulte; Paris France
| | - T. Billette de Villemeur
- AP-HP, Service de Neuropédiatrie; Hôpital Armand Trousseau; Paris France
- AP-HP, Service de Pédiatrie; Hôpital Louis Mourier; Colombes France
| | - M.-L. Moutard
- Groupe de Recherche Clinique (GRC) "ConCer-LD"; UPMC; Paris France
- AP-HP, Service de Neuropédiatrie; Hôpital Armand Trousseau; Paris France
| | - A. Guët
- AP-HP, Service de Pédiatrie; Hôpital Louis Mourier; Colombes France
| | - J. Xavier
- AP-HP, Department of Child and Adolescent Psychiatry; Groupe Hospitalier Pitié-Salpêtrière et University Pierre and Marie Curie; Paris France
- Sorbonne Universités, UPMC, CNRS UMR 7222; Institut des Systèmes Intelligents et Robotiques; Paris France
| | - D. Périsse
- AP-HP, Department of Child and Adolescent Psychiatry; Groupe Hospitalier Pitié-Salpêtrière et University Pierre and Marie Curie; Paris France
| | - D. Cohen
- AP-HP, Department of Child and Adolescent Psychiatry; Groupe Hospitalier Pitié-Salpêtrière et University Pierre and Marie Curie; Paris France
- Sorbonne Universités, UPMC, CNRS UMR 7222; Institut des Systèmes Intelligents et Robotiques; Paris France
| | - F. Demurger
- Service de Génétique Médicale; CLAD Ouest CHU Hôpital Sud; Rennes France
| | - C. Quélin
- Service de Génétique Médicale; CLAD Ouest CHU Hôpital Sud; Rennes France
| | - C. Depienne
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127; Institut du Cerveau et de la Moelle épinière, ICM; Paris France
| | - S. Odent
- Service de Génétique Médicale; CLAD Ouest CHU Hôpital Sud; Rennes France
- CNRS UMR 6290 (IGDR); Université de Rennes 1; Rennes France
| | - C. Nava
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127; Institut du Cerveau et de la Moelle épinière, ICM; Paris France
| | - V. David
- Laboratoire de Génétique moléculaire et Génomique; CHU Pontchaillou; Rennes France
- CNRS UMR 6290 (IGDR); Université de Rennes 1; Rennes France
| | - L. Pasquier
- Service de Génétique Médicale; CLAD Ouest CHU Hôpital Sud; Rennes France
- INSERM CIC pédiatrique 1414; CHU Pontchaillou; Rennes France
| | - C. Mignot
- Département de Génétique, AP-HP; Hôpital de la Pitié-Salpêtrière; Paris France
- Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme"; UPMC; Paris France
- Groupe de Recherche Clinique (GRC) "ConCer-LD"; UPMC; Paris France
| |
Collapse
|
617
|
King DA, Sifrim A, Fitzgerald TW, Rahbari R, Hobson E, Homfray T, Mansour S, Mehta SG, Shehla M, Tomkins SE, Vasudevan PC, Hurles ME. Detection of structural mosaicism from targeted and whole-genome sequencing data. Genome Res 2017; 27:1704-1714. [PMID: 28855261 PMCID: PMC5630034 DOI: 10.1101/gr.212373.116] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/18/2017] [Indexed: 01/11/2023]
Abstract
Structural mosaic abnormalities are large post-zygotic mutations present in a subset of cells and have been implicated in developmental disorders and cancer. Such mutations have been conventionally assessed in clinical diagnostics using cytogenetic or microarray testing. Modern disease studies rely heavily on exome sequencing, yet an adequate method for the detection of structural mosaicism using targeted sequencing data is lacking. Here, we present a method, called MrMosaic, to detect structural mosaic abnormalities using deviations in allele fraction and read coverage from next-generation sequencing data. Whole-exome sequencing (WES) and whole-genome sequencing (WGS) simulations were used to calculate detection performance across a range of mosaic event sizes, types, clonalities, and sequencing depths. The tool was applied to 4911 patients with undiagnosed developmental disorders, and 11 events among nine patients were detected. For eight of these 11 events, mosaicism was observed in saliva but not blood, suggesting that assaying blood alone would miss a large fraction, possibly >50%, of mosaic diagnostic chromosomal rearrangements.
Collapse
Affiliation(s)
- Daniel A King
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Alejandro Sifrim
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Tomas W Fitzgerald
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Raheleh Rahbari
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Emma Hobson
- Department of Clinical Genetics, Chapel Allerton Hospital, Leeds LS7 4SA, United Kingdom
| | - Tessa Homfray
- Southwest Thames Regional Genetics Centre, St George's Healthcare NHS Trust, London SW17 0RE, United Kingdom
| | - Sahar Mansour
- Southwest Thames Regional Genetics Centre, St George's Healthcare NHS Trust, London SW17 0RE, United Kingdom
| | - Sarju G Mehta
- East Anglian Regional Genetics Service, Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Mohammed Shehla
- South East Thames Regional Genetics Centre, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Susan E Tomkins
- Department of Clinical Genetics, St Michael's Hospital, Bristol BS2 8EG, United Kingdom
| | | | - Matthew E Hurles
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| |
Collapse
|
618
|
Bishop DV, Snowling MJ, Thompson PA, Greenhalgh T, and the CATALISE‐2 consortium. Phase 2 of CATALISE: a multinational and multidisciplinary Delphi consensus study of problems with language development: Terminology. J Child Psychol Psychiatry 2017; 58:1068-1080. [PMID: 28369935 PMCID: PMC5638113 DOI: 10.1111/jcpp.12721] [Citation(s) in RCA: 737] [Impact Index Per Article: 92.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Lack of agreement about criteria and terminology for children's language problems affects access to services as well as hindering research and practice. We report the second phase of a study using an online Delphi method to address these issues. In the first phase, we focused on criteria for language disorder. Here we consider terminology. METHODS The Delphi method is an iterative process in which an initial set of statements is rated by a panel of experts, who then have the opportunity to view anonymised ratings from other panel members. On this basis they can either revise their views or make a case for their position. The statements are then revised based on panel feedback, and again rated by and commented on by the panel. In this study, feedback from a second round was used to prepare a final set of statements in narrative form. The panel included 57 individuals representing a range of professions and nationalities. RESULTS We achieved at least 78% agreement for 19 of 21 statements within two rounds of ratings. These were collapsed into 12 statements for the final consensus reported here. The term 'Language Disorder' is recommended to refer to a profile of difficulties that causes functional impairment in everyday life and is associated with poor prognosis. The term, 'Developmental Language Disorder' (DLD) was endorsed for use when the language disorder was not associated with a known biomedical aetiology. It was also agreed that (a) presence of risk factors (neurobiological or environmental) does not preclude a diagnosis of DLD, (b) DLD can co-occur with other neurodevelopmental disorders (e.g. ADHD) and (c) DLD does not require a mismatch between verbal and nonverbal ability. CONCLUSIONS This Delphi exercise highlights reasons for disagreements about terminology for language disorders and proposes standard definitions and nomenclature.
Collapse
Affiliation(s)
| | | | - Paul A. Thompson
- Department of Experimental PsychologyUniversity of OxfordOxfordUK
| | - Trisha Greenhalgh
- Nuffield Department of Primary Care Health SciencesUniversity of OxfordOxfordUK
| | | |
Collapse
|
619
|
Soblet J, Dimov I, Graf von Kalckreuth C, Cano-Chervel J, Baijot S, Pelc K, Sottiaux M, Vilain C, Smits G, Deconinck N. BCL11A frameshift mutation associated with dyspraxia and hypotonia affecting the fine, gross, oral, and speech motor systems. Am J Med Genet A 2017; 176:201-208. [PMID: 28960836 PMCID: PMC5765401 DOI: 10.1002/ajmg.a.38479] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 12/08/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022]
Abstract
We report the case of a 7‐year‐old male of Western European origin presenting with moderate intellectual disability, severe childhood apraxia of speech in the presence of oral and manual dyspraxia, and hypotonia across motor systems including the oral and speech motor systems. Exome sequencing revealed a de novo frameshift protein truncating mutation in the fourth exon of BCL11A, a gene recently demonstrated as being involved in cognition and language development. Making parallels with a previously described patient with a 200 kb 2p15p16.1 deletion encompassing the entire BCL11A gene and displaying a similar phenotype, we characterize in depth how BCL11A is involved in clinical aspects of language development and oral praxis.
Collapse
Affiliation(s)
- Julie Soblet
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles, Brussels, Belgium
| | - Ivan Dimov
- Faculté de Médecine ULB, Université Libre de Bruxelles, Brussels, Belgium
| | - Clemens Graf von Kalckreuth
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Julie Cano-Chervel
- Department of Child and Adolescent Psychiatry, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Simon Baijot
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium.,Department of Child and Adolescent Psychiatry, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Karin Pelc
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Martine Sottiaux
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Catheline Vilain
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles, Brussels, Belgium
| | - Guillaume Smits
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Deconinck
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
620
|
Yoo Y, Jung J, Lee YN, Lee Y, Cho H, Na E, Hong J, Kim E, Lee JS, Lee JS, Hong C, Park SY, Wie J, Miller K, Shur N, Clow C, Ebel RS, DeBrosse SD, Henderson LB, Willaert R, Castaldi C, Tikhonova I, Bilgüvar K, Mane S, Kim KJ, Hwang YS, Lee SG, So I, Lim BC, Choi HJ, Seong JY, Shin YB, Jung H, Chae JH, Choi M. GABBR2mutations determine phenotype in rett syndrome and epileptic encephalopathy. Ann Neurol 2017; 82:466-478. [DOI: 10.1002/ana.25032] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Yongjin Yoo
- Department of Biomedical Sciences; Seoul National University College of Medicine; Seoul Republic of Korea
| | - Jane Jung
- Department of Anatomy, Brain Research Institute, and Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine; Seoul Republic of Korea
| | - Yoo-Na Lee
- Graduate School of Medicine; Korea University; Seoul Republic of Korea
| | - Youngha Lee
- Department of Biomedical Sciences; Seoul National University College of Medicine; Seoul Republic of Korea
| | - Hyosuk Cho
- Department of Biomedical Sciences; Seoul National University College of Medicine; Seoul Republic of Korea
| | - Eunjung Na
- Department of Anatomy, Brain Research Institute, and Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine; Seoul Republic of Korea
| | - JeaYeok Hong
- Department of Anatomy, Brain Research Institute, and Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine; Seoul Republic of Korea
| | - Eunjin Kim
- Department of Anatomy, Brain Research Institute, and Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine; Seoul Republic of Korea
| | - Jin Sook Lee
- Department of Pediatrics, Department of Genome Medicine and Science; Gachon University Gil Medical Center; Incheon Republic of Korea
| | - Je Sang Lee
- Department of Rehabilitation Medicine; Pusan National University College of Medicine; Pusan Republic of Korea
| | - Chansik Hong
- Department of Physiology; Chosun University School of Medicine; Kwangju Republic of Korea
| | - Sang-Yoon Park
- Department of Science in Korean Medicine, Cancer Preventive Material Developmental Research Center, College of Korean Medicine; Kyung Hee University; Seoul Republic of Korea
| | - Jinhong Wie
- Department of Biomedical Sciences; Seoul National University College of Medicine; Seoul Republic of Korea
- Department of Physiology; Seoul National University College of Medicine; Seoul Republic of Korea
| | | | | | | | - Roseànne S. Ebel
- UH Cleveland Medical Center; Center for Human Genetics; Cleveland OH
| | | | | | | | | | | | - Kaya Bilgüvar
- Yale Center for Genome Analysis; West Haven CT
- Department of Genetics; Yale University School of Medicine; New Haven CT
| | - Shrikant Mane
- Yale Center for Genome Analysis; West Haven CT
- Department of Genetics; Yale University School of Medicine; New Haven CT
| | - Ki Joong Kim
- Department of Pediatrics, Seoul National University College of Medicine; Seoul National University Children's Hospital; Seoul Republic of Korea
| | - Yong Seung Hwang
- Department of Pediatrics, Seoul National University College of Medicine; Seoul National University Children's Hospital; Seoul Republic of Korea
| | - Seok-Geun Lee
- Department of Science in Korean Medicine, Cancer Preventive Material Developmental Research Center, College of Korean Medicine; Kyung Hee University; Seoul Republic of Korea
| | - Insuk So
- Department of Biomedical Sciences; Seoul National University College of Medicine; Seoul Republic of Korea
- Department of Physiology; Seoul National University College of Medicine; Seoul Republic of Korea
| | - Byung Chan Lim
- Department of Pediatrics, Seoul National University College of Medicine; Seoul National University Children's Hospital; Seoul Republic of Korea
| | - Hee-Jung Choi
- Department of Biological Sciences; Seoul National University College of Natural Sciences; Seoul Republic of Korea
| | - Jae Young Seong
- Graduate School of Medicine; Korea University; Seoul Republic of Korea
| | - Yong Beom Shin
- Department of Rehabilitation Medicine; Pusan National University College of Medicine; Pusan Republic of Korea
| | - Hosung Jung
- Department of Anatomy, Brain Research Institute, and Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine; Seoul Republic of Korea
| | - Jong-Hee Chae
- Department of Pediatrics, Seoul National University College of Medicine; Seoul National University Children's Hospital; Seoul Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences; Seoul National University College of Medicine; Seoul Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine; Seoul National University Children's Hospital; Seoul Republic of Korea
| |
Collapse
|
621
|
Anazi S, Maddirevula S, Salpietro V, Asi YT, Alsahli S, Alhashem A, Shamseldin HE, AlZahrani F, Patel N, Ibrahim N, Abdulwahab FM, Hashem M, Alhashmi N, Al Murshedi F, Al Kindy A, Alshaer A, Rumayyan A, Al Tala S, Kurdi W, Alsaman A, Alasmari A, Banu S, Sultan T, Saleh MM, Alkuraya H, Salih MA, Aldhalaan H, Ben-Omran T, Al Musafri F, Ali R, Suleiman J, Tabarki B, El-Hattab AW, Bupp C, Alfadhel M, Al Tassan N, Monies D, Arold ST, Abouelhoda M, Lashley T, Houlden H, Faqeih E, Alkuraya FS. Expanding the genetic heterogeneity of intellectual disability. Hum Genet 2017; 136:1419-1429. [PMID: 28940097 DOI: 10.1007/s00439-017-1843-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/15/2017] [Indexed: 11/30/2022]
Abstract
Intellectual disability (ID) is a common morbid condition with a wide range of etiologies. The list of monogenic forms of ID has increased rapidly in recent years thanks to the implementation of genomic sequencing techniques. In this study, we describe the phenotypic and genetic findings of 68 families (105 patients) all with novel ID-related variants. In addition to established ID genes, including ones for which we describe unusual mutational mechanism, some of these variants represent the first confirmatory disease-gene links following previous reports (TRAK1, GTF3C3, SPTBN4 and NKX6-2), some of which were based on single families. Furthermore, we describe novel variants in 14 genes that we propose as novel candidates (ANKHD1, ASTN2, ATP13A1, FMO4, MADD, MFSD11, NCKAP1, NFASC, PCDHGA10, PPP1R21, SLC12A2, SLK, STK32C and ZFAT). We highlight MADD and PCDHGA10 as particularly compelling candidates in which we identified biallelic likely deleterious variants in two independent ID families each. We also highlight NCKAP1 as another compelling candidate in a large family with autosomal dominant mild intellectual disability that fully segregates with a heterozygous truncating variant. The candidacy of NCKAP1 is further supported by its biological function, and our demonstration of relevant expression in human brain. Our study expands the locus and allelic heterogeneity of ID and demonstrates the power of positional mapping to reveal unusual mutational mechanisms.
Collapse
Affiliation(s)
- Shams Anazi
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Sateesh Maddirevula
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Vincenzo Salpietro
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Yasmine T Asi
- Queen Square Brain Bank for Neurological Disorders, Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK
| | - Saud Alsahli
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Amal Alhashem
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hanan E Shamseldin
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fatema AlZahrani
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nisha Patel
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Niema Ibrahim
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Firdous M Abdulwahab
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mais Hashem
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nadia Alhashmi
- Department of Genetics, College of Medicine, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Fathiya Al Murshedi
- Department of Genetics, College of Medicine, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Adila Al Kindy
- Department of Genetics, College of Medicine, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Ahmad Alshaer
- Pediatric Neurology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Ahmed Rumayyan
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,Neurology Division, Department of Pediatrics, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Saeed Al Tala
- Department of Pediatrics and Genetic Unit, Armed Forces Hospital, Khamis Mushayt, Saudi Arabia
| | - Wesam Kurdi
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital, Riyadh, Saudi Arabia
| | - Abdulaziz Alsaman
- Department of Pediatric Subspecialties, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ali Alasmari
- Department of Pediatric Subspecialties, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Selina Banu
- Department of Pediatric Neurology, ICH and SSF Hospital Mirpur, Dhaka, 1216, Bangladesh
| | - Tipu Sultan
- Department of Pediatric Neurology, Institute of Child Health and The Children's Hospital Lahore, 381-D/2, Lahore, Pakistan
| | - Mohammed M Saleh
- Department of Pediatric Subspecialties, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Hisham Alkuraya
- Department of Ophthalmology, Specialized Medical Center Hospital, Riyadh, Saudi Arabia
| | - Mustafa A Salih
- Division of Pediatric Neurology, Department of Pediatrics, King Khalid University Hospital and College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hesham Aldhalaan
- Pediatric Neurology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Tawfeg Ben-Omran
- Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
| | - Fatima Al Musafri
- Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
| | - Rehab Ali
- Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
| | - Jehan Suleiman
- Division of Neurology, Department of Pediatrics, Tawam Hospital, Al Ain, United Arab Emirates
| | - Brahim Tabarki
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Ayman W El-Hattab
- Division of Clinical Genetics and Metabolic Disorders, Department of Pediatrics, Tawam Hospital, Al-Ain, United Arab Emirates
| | - Caleb Bupp
- Spectrum Health Genetics, Grand Rapids, MI, USA
| | - Majid Alfadhel
- Genetics Division, Department of Pediatrics, King Abdullah International Medical Research Centre, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Nada Al Tassan
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Dorota Monies
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Stefan T Arold
- Division of Biological and Environmental Sciences and Engineering (BESE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Mohamed Abouelhoda
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK
| | - Henry Houlden
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Eissa Faqeih
- Department of Pediatric Subspecialties, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia. .,Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia. .,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia. .,Spectrum Health Genetics, Grand Rapids, MI, USA. .,Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
622
|
Clinical and genetic analysis of a rare syndrome associated with neoteny. Genet Med 2017; 20:495-502. [DOI: 10.1038/gim.2017.140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 06/20/2017] [Indexed: 01/23/2023] Open
|
623
|
Peng J, Li Q, Shang X. Investigations on factors influencing HPO-based semantic similarity calculation. J Biomed Semantics 2017; 8:34. [PMID: 29297376 PMCID: PMC5763495 DOI: 10.1186/s13326-017-0144-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Although disease diagnosis has greatly benefited from next generation sequencing technologies, it is still difficult to make the right diagnosis purely based on sequencing technologies for many diseases with complex phenotypes and high genetic heterogeneity. Recently, calculating Human Phenotype Ontology (HPO)-based phenotype semantic similarity has contributed a lot for completing disease diagnosis. However, factors which affect the accuracy of HPO-based semantic similarity have not been evaluated systematically. Results In this study, we proposed a new framework called HPOFactor to evaluate these factors. Our model includes four components: (1) the size of annotation set, (2) the evidence code of annotations, (3) the quality of annotations and (4) the coverage of annotations respectively. Conclusions HPOFactor analyzes the four factors systematically based on two kinds of experiments: causative gene prediction and disease prediction. Furthermore, semantic similarity measurement could be designed based on the characteristic of these factors.
Collapse
Affiliation(s)
- Jiajie Peng
- Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Qianqian Li
- Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Xuequn Shang
- Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China.
| |
Collapse
|
624
|
An autism spectrum disorder-related de novo mutation hotspot discovered in the GEF1 domain of Trio. Nat Commun 2017; 8:601. [PMID: 28928363 PMCID: PMC5605661 DOI: 10.1038/s41467-017-00472-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/30/2017] [Indexed: 12/24/2022] Open
Abstract
The Rho guanine nucleotide exchange factor (RhoGEF) Trio promotes actin polymerization by directly activating the small GTPase Rac1. Recent studies suggest that autism spectrum disorder (ASD)-related behavioral phenotypes in animal models of ASD can be produced by dysregulation of Rac1’s control of actin polymerization at glutamatergic synapses. Here, in humans, we discover a large cluster of ASD-related de novo mutations in Trio’s Rac1 activating domain, GEF1. Our study reveals that these mutations produce either hypofunctional or hyperfunctional forms of Trio in rodent neurons in vitro. In accordance with pathological increases or decreases in glutamatergic neurotransmission observed in animal models of ASD, we find that these mutations result in either reduced synaptic AMPA receptor expression or enhanced glutamatergic synaptogenesis. Together, our findings implicate both excessive and reduced Trio activity and the resulting synaptic dysfunction in ASD-related pathogenesis, and point to the Trio-Rac1 pathway at glutamatergic synapses as a possible key point of convergence of many ASD-related genes. Trio is a RhoGEF protein that promotes actin polymerization and is implicated in the regulation of glutamatergic synapses in autism spectrum disorder (ASD). Here the authors identify a large cluster of de novo mutations in the GEF1 domain of Trio in whole-exome sequencing data from individuals with ASD, and confirm that some of these mutations lead to glutamatergic dysregulation in vitro.
Collapse
|
625
|
Braun SMG, Kirkland JG, Chory EJ, Husmann D, Calarco JP, Crabtree GR. Rapid and reversible epigenome editing by endogenous chromatin regulators. Nat Commun 2017; 8:560. [PMID: 28916764 PMCID: PMC5601922 DOI: 10.1038/s41467-017-00644-y] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/17/2017] [Indexed: 01/10/2023] Open
Abstract
Understanding the causal link between epigenetic marks and gene regulation remains a central question in chromatin biology. To edit the epigenome we developed the FIRE-Cas9 system for rapid and reversible recruitment of endogenous chromatin regulators to specific genomic loci. We enhanced the dCas9–MS2 anchor for genome targeting with Fkbp/Frb dimerizing fusion proteins to allow chemical-induced proximity of a desired chromatin regulator. We find that mSWI/SNF (BAF) complex recruitment is sufficient to oppose Polycomb within minutes, leading to activation of bivalent gene transcription in mouse embryonic stem cells. Furthermore, Hp1/Suv39h1 heterochromatin complex recruitment to active promoters deposits H3K9me3 domains, resulting in gene silencing that can be reversed upon washout of the chemical dimerizer. This inducible recruitment strategy provides precise kinetic information to model epigenetic memory and plasticity. It is broadly applicable to mechanistic studies of chromatin in mammalian cells and is particularly suited to the analysis of endogenous multi-subunit chromatin regulator complexes. Understanding the link between epigenetic marks and gene regulation requires the development of new tools to directly manipulate chromatin. Here the authors demonstrate a Cas9-based system to recruit chromatin remodelers to loci of interest, allowing rapid, reversible manipulation of epigenetic states.
Collapse
Affiliation(s)
- Simon M G Braun
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jacob G Kirkland
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Emma J Chory
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Dylan Husmann
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Joseph P Calarco
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Gerald R Crabtree
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA. .,Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
626
|
Shimojima K, Okamoto N, Goel H, Ondo Y, Yamamoto T. Familial 9q33q34 microduplication in siblings with developmental disorders and macrocephaly. Eur J Med Genet 2017; 60:650-654. [PMID: 28899818 DOI: 10.1016/j.ejmg.2017.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 10/18/2022]
Abstract
Because several genes responsible for epileptic encephalopathy are located on the 9q33q34 region, patients with chromosomal deletions of this region often show intractable epilepsy and neurodevelopmental disability. Contrary to these findings, chromosomal duplications of this region have never been reported previously. We identified a first case of 9q33q34 microduplications in siblings associated with developmental disorders and macrocephaly. Their mother was a mosaic carrier of this duplication. Duplicated regions involved STXBP1; the gene related to epileptic encephalopathy. Neurological features including developmental delay and macrocephaly observed in the present siblings may be derived from the extra-copy of STXBP1.
Collapse
Affiliation(s)
- Keiko Shimojima
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Himanshu Goel
- School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Yumiko Ondo
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshiyuki Yamamoto
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
627
|
Hara K, Taharazako S, Ikeda M, Fujita H, Mikami Y, Kikuchi S, Hishiki A, Yokoyama H, Ishikawa Y, Kanno SI, Tanaka K, Hashimoto H. Dynamic feature of mitotic arrest deficient 2-like protein 2 (MAD2L2) and structural basis for its interaction with chromosome alignment-maintaining phosphoprotein (CAMP). J Biol Chem 2017; 292:17658-17667. [PMID: 28887307 DOI: 10.1074/jbc.m117.804237] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/04/2017] [Indexed: 12/30/2022] Open
Abstract
Mitotic arrest deficient 2-like protein 2 (MAD2L2), also termed MAD2B or REV7, is involved in multiple cellular functions including translesion DNA synthesis (TLS), signal transduction, transcription, and mitotic events. MAD2L2 interacts with chromosome alignment-maintaining phosphoprotein (CAMP), a kinetochore-microtubule attachment protein in mitotic cells, presumably through a novel "WK" motif in CAMP. Structures of MAD2L2 in complex with binding regions of the TLS proteins REV3 and REV1 have revealed that MAD2L2 has two faces for protein-protein interactions that are regulated by its C-terminal region; however, the mechanisms underlying the MAD2L2-CAMP interaction and the mitotic role of MAD2L2 remain unknown. Here we have determined the structures of human MAD2L2 in complex with a CAMP fragment in two crystal forms. The overall structure of the MAD2L2-CAMP complex in both crystal forms was essentially similar to that of the MAD2L2-REV3 complex. However, the residue interactions between MAD2L2 and CAMP were strikingly different from those in the MAD2L2-REV3 complex. Furthermore, structure-based interaction analyses revealed an unprecedented mechanism involving CAMP's WK motif. Surprisingly, in one of the crystal forms, the MAD2L2-CAMP complex formed a dimeric structure in which the C-terminal region of MAD2L2 was swapped and adopted an immature structure. The structure provides direct evidence for the dynamic nature of MAD2L2 structure, which in turn may have implications for the protein-protein interaction mechanism and the multiple functions of this protein. This work is the first structural study of MAD2L2 aside from its role in TLS and might pave the way to clarify MAD2L2's function in mitosis.
Collapse
Affiliation(s)
- Kodai Hara
- From the School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan
| | - Shota Taharazako
- From the School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan
| | - Masanori Ikeda
- Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan, and
| | - Hiroki Fujita
- Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan, and
| | - Yoshiko Mikami
- Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan, and
| | - Sotaro Kikuchi
- From the School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan
| | - Asami Hishiki
- From the School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan
| | - Hideshi Yokoyama
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Yoshinobu Ishikawa
- From the School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan
| | - Shin-Ichiro Kanno
- Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan, and
| | - Kozo Tanaka
- Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan, and
| | - Hiroshi Hashimoto
- From the School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan,
| |
Collapse
|
628
|
Lelieveld SH, Wiel L, Venselaar H, Pfundt R, Vriend G, Veltman JA, Brunner HG, Vissers LE, Gilissen C. Spatial Clustering of de Novo Missense Mutations Identifies Candidate Neurodevelopmental Disorder-Associated Genes. Am J Hum Genet 2017; 101:478-484. [PMID: 28867141 DOI: 10.1016/j.ajhg.2017.08.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 08/04/2017] [Indexed: 10/19/2022] Open
Abstract
Haploinsufficiency (HI) is the best characterized mechanism through which dominant mutations exert their effect and cause disease. Non-haploinsufficiency (NHI) mechanisms, such as gain-of-function and dominant-negative mechanisms, are often characterized by the spatial clustering of mutations, thereby affecting only particular regions or base pairs of a gene. Variants leading to haploinsufficency might occasionally cluster as well, for example in critical domains, but such clustering is on the whole less pronounced with mutations often spread throughout the gene. Here we exploit this property and develop a method to specifically identify genes with significant spatial clustering patterns of de novo mutations in large cohorts. We apply our method to a dataset of 4,061 de novo missense mutations from published exome studies of trios with intellectual disability and developmental disorders (ID/DD) and successfully identify 15 genes with clustering mutations, including 12 genes for which mutations are known to cause neurodevelopmental disorders. For 11 out of these 12, NHI mutation mechanisms have been reported. Additionally, we identify three candidate ID/DD-associated genes of which two have an established role in neuronal processes. We further observe a higher intolerance to normal genetic variation of the identified genes compared to known genes for which mutations lead to HI. Finally, 3D modeling of these mutations on their protein structures shows that 81% of the observed mutations are unlikely to affect the overall structural integrity and that they therefore most likely act through a mechanism other than HI.
Collapse
|
629
|
Abstract
An international collaboration has assembled DNA samples and detailed phenotypic information from >13,000 individuals with a clinical diagnosis of either autism spectrum disorder (ASD) or intellectual disability/developmental delay (ID/DD). The application of molecular inversion probe technologies to sequence 208 candidate genes at scale in this impressive resource has identified a large set of plausibly causative mutations for these disorders.
Collapse
|
630
|
Reijnders MR, Ansor NM, Kousi M, Yue WW, Tan PL, Clarkson K, Clayton-Smith J, Corning K, Jones JR, Lam WW, Mancini GM, Marcelis C, Mohammed S, Pfundt R, Roifman M, Cohn R, Chitayat D, Millard TH, Katsanis N, Brunner HG, Banka S, Banka S. RAC1 Missense Mutations in Developmental Disorders with Diverse Phenotypes. Am J Hum Genet 2017; 101:466-477. [PMID: 28886345 DOI: 10.1016/j.ajhg.2017.08.007] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/01/2017] [Indexed: 01/01/2023] Open
Abstract
RAC1 is a widely studied Rho GTPase, a class of molecules that modulate numerous cellular functions essential for normal development. RAC1 is highly conserved across species and is under strict mutational constraint. We report seven individuals with distinct de novo missense RAC1 mutations and varying degrees of developmental delay, brain malformations, and additional phenotypes. Four individuals, each harboring one of c.53G>A (p.Cys18Tyr), c.116A>G (p.Asn39Ser), c.218C>T (p.Pro73Leu), and c.470G>A (p.Cys157Tyr) variants, were microcephalic, with head circumferences between -2.5 to -5 SD. In contrast, two individuals with c.151G>A (p.Val51Met) and c.151G>C (p.Val51Leu) alleles were macrocephalic with head circumferences of +4.16 and +4.5 SD. One individual harboring a c.190T>G (p.Tyr64Asp) allele had head circumference in the normal range. Collectively, we observed an extraordinary spread of ∼10 SD of head circumferences orchestrated by distinct mutations in the same gene. In silico modeling, mouse fibroblasts spreading assays, and in vivo overexpression assays using zebrafish as a surrogate model demonstrated that the p.Cys18Tyr and p.Asn39Ser RAC1 variants function as dominant-negative alleles and result in microcephaly, reduced neuronal proliferation, and cerebellar abnormalities in vivo. Conversely, the p.Tyr64Asp substitution is constitutively active. The remaining mutations are probably weakly dominant negative or their effects are context dependent. These findings highlight the importance of RAC1 in neuronal development. Along with TRIO and HACE1, a sub-category of rare developmental disorders is emerging with RAC1 as the central player. We show that ultra-rare disorders caused by private, non-recurrent missense mutations that result in varying phenotypes are challenging to dissect, but can be delineated through focused international collaboration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Siddharth Banka
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, Central Manchester NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester M13 9WL, UK; Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK.
| |
Collapse
|
631
|
Wangler MF, Yamamoto S, Chao HT, Posey JE, Westerfield M, Postlethwait J, Hieter P, Boycott KM, Campeau PM, Bellen HJ. Model Organisms Facilitate Rare Disease Diagnosis and Therapeutic Research. Genetics 2017; 207:9-27. [PMID: 28874452 PMCID: PMC5586389 DOI: 10.1534/genetics.117.203067] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/06/2017] [Indexed: 12/29/2022] Open
Abstract
Efforts to identify the genetic underpinnings of rare undiagnosed diseases increasingly involve the use of next-generation sequencing and comparative genomic hybridization methods. These efforts are limited by a lack of knowledge regarding gene function, and an inability to predict the impact of genetic variation on the encoded protein function. Diagnostic challenges posed by undiagnosed diseases have solutions in model organism research, which provides a wealth of detailed biological information. Model organism geneticists are by necessity experts in particular genes, gene families, specific organs, and biological functions. Here, we review the current state of research into undiagnosed diseases, highlighting large efforts in North America and internationally, including the Undiagnosed Diseases Network (UDN) (Supplemental Material, File S1) and UDN International (UDNI), the Centers for Mendelian Genomics (CMG), and the Canadian Rare Diseases Models and Mechanisms Network (RDMM). We discuss how merging human genetics with model organism research guides experimental studies to solve these medical mysteries, gain new insights into disease pathogenesis, and uncover new therapeutic strategies.
Collapse
Affiliation(s)
- Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine (BCM), Houston, Texas 77030
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine (BCM), Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine (BCM), Houston, Texas 77030
| | - Hsiao-Tuan Chao
- Department of Pediatrics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Department of Pediatrics, Section of Child Neurology, Baylor College of Medicine (BCM), Houston, Texas 77030
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
| | - Monte Westerfield
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - John Postlethwait
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - Philip Hieter
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4C, Canada
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ontario K1H 8L1, Canada
| | - Philippe M Campeau
- Department of Pediatrics, University of Montreal, Quebec H3T 1C5, Canada
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine (BCM), Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine (BCM), Houston, Texas 77030
- Howard Hughes Medical Institute, Baylor College of Medicine (BCM), Houston, Texas 77030
| |
Collapse
|
632
|
Chen C, Qi H, Shen Y, Pickrell J, Przeworski M. Contrasting Determinants of Mutation Rates in Germline and Soma. Genetics 2017; 207:255-267. [PMID: 28733365 PMCID: PMC5586376 DOI: 10.1534/genetics.117.1114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/01/2017] [Indexed: 12/13/2022] Open
Abstract
Recent studies of somatic and germline mutations have led to the identification of a number of factors that influence point mutation rates, including CpG methylation, expression levels, replication timing, and GC content. Intriguingly, some of the effects appear to differ between soma and germline: in particular, whereas mutation rates have been reported to decrease with expression levels in tumors, no clear effect has been detected in the germline. Distinct approaches were taken to analyze the data, however, so it is hard to know whether these apparent differences are real. To enable a cleaner comparison, we considered a statistical model in which the mutation rate of a coding region is predicted by GC content, expression levels, replication timing, and two histone repressive marks. We applied this model to both a set of germline mutations identified in exomes and to exonic somatic mutations in four types of tumors. Most determinants of mutations are shared: notably, we detected an effect of expression levels on both germline and somatic mutation rates. Moreover, in all tissues considered, higher expression levels are associated with greater strand asymmetry of mutations. However, mutation rates increase with expression levels in testis (and, more tentatively, in ovary), whereas they decrease with expression levels in somatic tissues. This contrast points to differences in damage or repair rates during transcription in soma and germline.
Collapse
Affiliation(s)
- Chen Chen
- Department of Biological Sciences, Columbia University, New York, New York 10025
- New York Genome Center, New York, New York 10013
| | - Hongjian Qi
- Department of Systems Biology, Columbia University Medical Center, New York, New York 10032
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, New York 10025
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Medical Center, New York, New York 10032
- Department of Biomedical Informatics, Columbia University, New York, New York 10025
| | - Joseph Pickrell
- Department of Biological Sciences, Columbia University, New York, New York 10025
- New York Genome Center, New York, New York 10013
| | - Molly Przeworski
- Department of Biological Sciences, Columbia University, New York, New York 10025
- Department of Systems Biology, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
633
|
DDX3 Modulates Neurite Development via Translationally Activating an RNA Regulon Involved in Rac1 Activation. J Neurosci 2017; 36:9792-804. [PMID: 27656019 DOI: 10.1523/jneurosci.4603-15.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 08/01/2016] [Indexed: 01/23/2023] Open
Abstract
UNLABELLED The RNA helicase DDX3 is a component of neuronal granules, and its gene mutations are linked to intellectual disability (ID). Here we demonstrate that DDX3 depletion in neurons impairs neurite development by downregulating Rac1 level and activation. Moreover, DDX3 activates the translation of functionally coherent mRNAs involved in Rac1 activation including Rac1 Among the DDX3 regulon, Prkaca encodes the catalytic subunit of PKA, a potential activator of Rac1 in neurons. DDX3-modulated PKAcα and Rac1 expression tunes the strength of PKA-Rac1 signaling and thereby contributes to neurite outgrowth and dendritic spine formation. Inhibition of DDX3 activity or expression in neonatal mice impaired dendritic outgrowth and spine formation of hippocampal neurons, echoing neuronal deficits underling DDX3 mutation-associated ID. Finally, we provide evidence that DDX3 activates local protein synthesis through a 5' UTR-dependent mechanism in neurons. The novel DDX3 regulon may conduct a spatial and temporal control of Rac1 signaling to regulate neurite development. SIGNIFICANCE STATEMENT DDX3X mutations are linked to intellectual disability (ID). We provide first evidence that DDX3 is required for neurite outgrowth and dendritic spine formation in vitro and in vivo We identified a DDX3 regulon constituting functionally cohesive mRNAs involved in Rac1 signaling, which contributes to DDX3-modulated neurite development. Inhibition or ablation of DDX3 in vivo shortened neurite lengths and impaired dendritic spine formation in hippocampal neurons, reflecting the prevalence of ID-associated DDX3X mutations in the helicase domain. Mechanistically, DDX3 activates local protein synthesis of mRNAs sharing similar 5' UTR structures and therefore controls Rac1 signaling strength in neurites.
Collapse
|
634
|
Krier JB, Kalia SS, Green RC. Genomic sequencing in clinical practice: applications, challenges, and opportunities. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 27757064 PMCID: PMC5067147 DOI: 10.31887/dcns.2016.18.3/jkrier] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The development of massively parallel sequencing (or next-generation sequencing) has facilitated a rapid implementation of genomic sequencing in clinical medicine. Genomic sequencing (GS) is now an essential tool for evaluating rare disorders, identifying therapeutic targets in neoplasms, and screening for prenatal aneuploidy. Emerging applications, such as GS for preconception carrier screening and predisposition screening in healthy individuals, are being explored in research settings and utilized by members of the public eager to incorporate genomic information into their health management. The rapid pace of adoption has created challenges for all stakeholders in clinical GS, from standardizing variant interpretation approaches in clinical molecular laboratories to ensuring that nongeneticist clinicians are prepared for new types of clinical information. Clinical GS faces a pivotal moment, as the vast potential of new quantities and types of data enable further clinical innovation and complicated implementation questions continue to be resolved.
Collapse
Affiliation(s)
- Joel B Krier
- Genomes2People Research Program, Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | | | - Robert C Green
- Genomes2People Research Program, Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
635
|
Need AC, Goldstein DB. Neuropsychiatric genomics in precision medicine: diagnostics, gene discovery, and translation. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 27757059 PMCID: PMC5067142 DOI: 10.31887/dcns.2016.18.3/aneed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Only a few years after its development, next-generation sequencing is rapidly becoming an essential part of clinical care for patients with serious neurological conditions, especially in the diagnosis of early-onset and severe presentations. Beyond this diagnostic role, there has been an explosion in definitive gene discovery in a range of neuropsychiatric diseases. This is providing new pointers to underlying disease biology and is beginning to outline a new framework for genetic stratification of neuropsychiatric disease, with clear relevance to both individual treatment optimization and clinical trial design. Here, we outline these developments and chart the expected impact on the treatment of neurological, neurodevelopmental, and psychiatric disease.
Collapse
Affiliation(s)
- Anna C Need
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, W12 ONN, UK
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
636
|
Sollis E, Deriziotis P, Saitsu H, Miyake N, Matsumoto N, Hoffer MJV, Ruivenkamp CAL, Alders M, Okamoto N, Bijlsma EK, Plomp AS, Fisher SE. Equivalent missense variant in the FOXP2 and FOXP1 transcription factors causes distinct neurodevelopmental disorders. Hum Mutat 2017; 38:1542-1554. [PMID: 28741757 DOI: 10.1002/humu.23303] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 07/11/2017] [Accepted: 07/22/2017] [Indexed: 12/28/2022]
Abstract
The closely related paralogues FOXP2 and FOXP1 encode transcription factors with shared functions in the development of many tissues, including the brain. However, while mutations in FOXP2 lead to a speech/language disorder characterized by childhood apraxia of speech (CAS), the clinical profile of FOXP1 variants includes a broader neurodevelopmental phenotype with global developmental delay, intellectual disability, and speech/language impairment. Using clinical whole-exome sequencing, we report an identical de novo missense FOXP1 variant identified in three unrelated patients. The variant, p.R514H, is located in the forkhead-box DNA-binding domain and is equivalent to the well-studied p.R553H FOXP2 variant that cosegregates with CAS in a large UK family. We present here for the first time a direct comparison of the molecular and clinical consequences of the same mutation affecting the equivalent residue in FOXP1 and FOXP2. Detailed functional characterization of the two variants in cell model systems revealed very similar molecular consequences, including aberrant subcellular localization, disruption of transcription factor activity, and deleterious effects on protein interactions. Nonetheless, clinical manifestations were broader and more severe in the three cases carrying the p.R514H FOXP1 variant than in individuals with the p.R553H variant related to CAS, highlighting divergent roles of FOXP2 and FOXP1 in neurodevelopment.
Collapse
Affiliation(s)
- Elliot Sollis
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Pelagia Deriziotis
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mariëtte J V Hoffer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Claudia A L Ruivenkamp
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariëlle Alders
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Astrid S Plomp
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| |
Collapse
|
637
|
Behavioral Comorbidities and Drug Treatments in a Zebrafish scn1lab Model of Dravet Syndrome. eNeuro 2017; 4:eN-NWR-0066-17. [PMID: 28812061 PMCID: PMC5555352 DOI: 10.1523/eneuro.0066-17.2017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/11/2017] [Accepted: 07/30/2017] [Indexed: 12/15/2022] Open
Abstract
Loss-of-function mutations in SCN1A cause Dravet syndrome (DS), a catastrophic childhood epilepsy in which patients experience comorbid behavioral conditions, including movement disorders, sleep abnormalities, anxiety, and intellectual disability. To study the functional consequences of voltage-gated sodium channel mutations, we use zebrafish with a loss-of-function mutation in scn1lab, a zebrafish homolog of human SCN1A. Homozygous scn1labs552/s552 mutants exhibit early-life seizures, metabolic deficits, and early death. Here, we developed in vivo assays using scn1labs552 mutants between 3 and 6 d postfertilization (dpf). To evaluate sleep disturbances, we monitored larvae for 24 h with locomotion tracking software. Locomotor activity during dark (night phase) was significantly higher in mutants than in controls. Among anticonvulsant drugs, clemizole and diazepam, but not trazodone or valproic acid, decreased distance moved at night for scn1labs552 mutant larvae. To monitor exploratory behavior in an open field, we tracked larvae in a novel arena. Mutant larvae exhibited impaired exploratory behavior, with increased time spent near the edge of the arena and decreased mobility, suggesting greater anxiety. Both clemizole and diazepam, but not trazodone or valproic acid, decreased distance moved and increased time spent in the center of the arena. Counting inhibitory neurons in vivo revealed no differences between scn1labs552 mutants and siblings. Taken together, our results demonstrate conserved features of sleep, anxiety, and movement disorders in scn1lab mutant zebrafish, and provide evidence that a zebrafish model allows effective tests of treatments for behavioral comorbidities associated with DS.
Collapse
|
638
|
Bourchany A, Thauvin-Robinet C, Lehalle D, Bruel AL, Masurel-Paulet A, Jean N, Nambot S, Willems M, Lambert L, El Chehadeh-Djebbar S, Schaefer E, Jaquette A, St-Onge J, Poe C, Jouan T, Chevarin M, Callier P, Mosca-Boidron AL, Laurent N, Lefebvre M, Huet F, Houcinat N, Moutton S, Philippe C, Tran-Mau-Them F, Vitobello A, Kuentz P, Duffourd Y, Rivière JB, Thevenon J, Faivre L. Reducing diagnostic turnaround times of exome sequencing for families requiring timely diagnoses. Eur J Med Genet 2017; 60:595-604. [PMID: 28807864 DOI: 10.1016/j.ejmg.2017.08.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/27/2017] [Accepted: 08/10/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Whole-exome sequencing (WES) has now entered medical practice with powerful applications in the diagnosis of rare Mendelian disorders. Although the usefulness and cost-effectiveness of WES have been widely demonstrated, it is essential to reduce the diagnostic turnaround time to make WES a first-line procedure. Since 2011, the automation of laboratory procedures and advances in sequencing chemistry have made it possible to carry out diagnostic whole genome sequencing from the blood sample to molecular diagnosis of suspected genetic disorders within 50 h. Taking advantage of these advances, the main objective of the study was to improve turnaround times for sequencing results. METHODS WES was proposed to 29 patients with severe undiagnosed disorders with developmental abnormalities and faced with medical situations requiring rapid diagnosis. Each family gave consent. The extracted DNA was sequenced on a NextSeq500 (Illumina) instrument. Data were analyzed following standard procedures. Variants were interpreted using in-house software. Each rare variant affecting protein sequences with clinical relevance was tested for familial segregation. RESULTS The diagnostic rate was 45% (13/29), with a mean turnaround time of 40 days from reception of the specimen to delivery of results to the referring physician. Besides permitting genetic counseling, the rapid diagnosis for positive families led to two pre-natal diagnoses and two inclusions in clinical trials. CONCLUSIONS This pilot study demonstrated the feasibility of rapid diagnostic WES in our primary genetics center. It reduced the diagnostic odyssey and helped provide support to families.
Collapse
Affiliation(s)
- Aurélie Bourchany
- Département de Pédiatrie 1, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France; Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Christel Thauvin-Robinet
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France
| | - Daphné Lehalle
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France
| | - Ange-Line Bruel
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Alice Masurel-Paulet
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France
| | - Nolwenn Jean
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France
| | - Sophie Nambot
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France
| | - Marjorie Willems
- Département de Génétique Clinique, CHRU de Montpellier, Hôpital Arnaud de Villeneuve, Montpellier, France
| | - Laetitia Lambert
- Unité Fonctionnelle de Génétique Clinique, Service de Médecine Néonatale, Maternité Régionale Universitaire, Nancy, France
| | | | - Elise Schaefer
- Service de Génétique Médicale, Hôpital de Hautepierre, Strasbourg, France
| | - Aurélia Jaquette
- Centre de Génétique, Hôpital de la Pitié-Salpétrière, Paris, France
| | - Judith St-Onge
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Charlotte Poe
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Thibaud Jouan
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Martin Chevarin
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Patrick Callier
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Laboratoire de Génétique chromosomique moléculaire, Plateau technique de Biologie, CHU, Dijon, France
| | - Anne-Laure Mosca-Boidron
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Laboratoire de Génétique chromosomique moléculaire, Plateau technique de Biologie, CHU, Dijon, France
| | - Nicole Laurent
- Laboratoire d'anatomopathologie, Plateau technique de Biologie, CHU, Dijon, France
| | - Mathilde Lefebvre
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Frédéric Huet
- Département de Pédiatrie 1, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France; Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Nada Houcinat
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France
| | - Sébastien Moutton
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France
| | - Christophe Philippe
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Laboratoire de Génétique chromosomique moléculaire, Plateau technique de Biologie, CHU, Dijon, France
| | - Frédéric Tran-Mau-Them
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Laboratoire de Génétique chromosomique moléculaire, Plateau technique de Biologie, CHU, Dijon, France
| | - Antonio Vitobello
- Laboratoire de Génétique chromosomique moléculaire, Plateau technique de Biologie, CHU, Dijon, France
| | - Paul Kuentz
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Yannis Duffourd
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France
| | - Jean-Baptiste Rivière
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Laboratoire de Génétique chromosomique moléculaire, Plateau technique de Biologie, CHU, Dijon, France
| | - Julien Thevenon
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France.
| | - Laurence Faivre
- Equipe Génétique des Anomalies du Développement, INSERM UMR1231, Université de Bourgogne-Franche Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon et Université de Bourgogne, Dijon, France.
| |
Collapse
|
639
|
Guella I, McKenzie MB, Evans DM, Buerki SE, Toyota EB, Van Allen MI, Suri M, Elmslie F, Simon ME, van Gassen KL, Héron D, Keren B, Nava C, Connolly MB, Demos M, Farrer MJ, Adam S, Boelman C, Bolbocean C, Candido T, Eydoux P, Horvath G, Huh L, Nelson TN, Sinclair G, van Karnebeek C, Vercauteren S. De Novo Mutations in YWHAG Cause Early-Onset Epilepsy. Am J Hum Genet 2017; 101:300-310. [PMID: 28777935 DOI: 10.1016/j.ajhg.2017.07.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/11/2017] [Indexed: 12/31/2022] Open
Abstract
Massively parallel sequencing has revealed many de novo mutations in the etiology of developmental and epileptic encephalopathies (EEs), highlighting their genetic heterogeneity. Additional candidate genes have been prioritized in silico by their co-expression in the brain. Here, we evaluate rare coding variability in 20 candidates nominated with the use of a reference gene set of 51 established EE-associated genes. Variants within the 20 candidate genes were extracted from exome-sequencing data of 42 subjects with EE and no previous genetic diagnosis. We identified 7 rare non-synonymous variants in 7 of 20 genes and performed Sanger sequence validation in affected probands and parental samples. De novo variants were found only in SLC1A2 (aka EAAT2 or GLT1) (c.244G>A [p.Gly82Arg]) and YWHAG (aka 14-3-3γ) (c.394C>T [p.Arg132Cys]), highlighting the potential cause of EE in 5% (2/42) of subjects. Seven additional subjects with de novo variants in SLC1A2 (n = 1) and YWHAG (n = 6) were subsequently identified through online tools. We identified a highly significant enrichment of de novo variants in YWHAG, establishing their role in early-onset epilepsy, and we provide additional support for the prior assignment of SLC1A2. Hence, in silico modeling of brain co-expression is an efficient method for nominating EE-associated genes to further elucidate the disorder's etiology and genotype-phenotype correlations.
Collapse
|
640
|
Singh T, Walters JTR, Johnstone M, Curtis D, Suvisaari J, Torniainen M, Rees E, Iyegbe C, Blackwood D, McIntosh AM, Kirov G, Geschwind D, Murray RM, Di Forti M, Bramon E, Gandal M, Hultman CM, Sklar P, INTERVAL Study, UK10K Consortium, Palotie A, Sullivan PF, O'Donovan MC, Owen MJ, Barrett JC. The contribution of rare variants to risk of schizophrenia in individuals with and without intellectual disability. Nat Genet 2017; 49:1167-1173. [PMID: 28650482 PMCID: PMC5533219 DOI: 10.1038/ng.3903] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 06/01/2017] [Indexed: 12/15/2022]
Abstract
By performing a meta-analysis of rare coding variants in whole-exome sequences from 4,133 schizophrenia cases and 9,274 controls, de novo mutations in 1,077 family trios, and copy number variants from 6,882 cases and 11,255 controls, we show that individuals with schizophrenia carry a significant burden of rare, damaging variants in 3,488 genes previously identified as having a near-complete depletion of loss-of-function variants. In patients with schizophrenia who also have intellectual disability, this burden is concentrated in risk genes associated with neurodevelopmental disorders. After excluding known risk genes for neurodevelopmental disorders, a significant rare variant burden persists in other genes intolerant of loss-of-function variants; although this effect is notably stronger in patients with both schizophrenia and intellectual disability, it is also seen in patients with schizophrenia who do not have intellectual disability. Together, our results show that rare, damaging variants contribute to the risk of schizophrenia both with and without intellectual disability and support an overlap of genetic risk between schizophrenia and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Tarjinder Singh
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1HH, Cambridge, UK
| | - James T. R. Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Mandy Johnstone
- Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK
| | - David Curtis
- University College London (UCL) Genetics Institute, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK
- Centre for Psychiatry, Barts and the London School of Medicine and Dentistry, London, UK
| | - Jaana Suvisaari
- National Institute for Health and Welfare (THL), Helsinki FI-00271, Finland
| | - Minna Torniainen
- National Institute for Health and Welfare (THL), Helsinki FI-00271, Finland
| | - Elliott Rees
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Conrad Iyegbe
- Institute of Psychiatry, King's College London, 16 De Crespigny Park, London SE5 8AF, UK
| | - Douglas Blackwood
- Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK
| | - Andrew M. McIntosh
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, 7 George Square, Edinburgh EH8 9JZ, UK
| | - Georg Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Daniel Geschwind
- UCLA David Geffen School of Medicine, Los Angeles, California 90095, USA
| | - Robin M. Murray
- Institute of Psychiatry, King's College London, 16 De Crespigny Park, London SE5 8AF, UK
| | - Marta Di Forti
- Institute of Psychiatry, King's College London, 16 De Crespigny Park, London SE5 8AF, UK
| | - Elvira Bramon
- Division of Psychiatry, University College London, Charles Bell House, Riding House Street, London W1W 7EJ, UK
| | - Michael Gandal
- UCLA David Geffen School of Medicine, Los Angeles, California 90095, USA
| | - Christina M. Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Pamela Sklar
- Division of Psychiatric Genomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | | | | | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki FI-00014 Finland
- Program in Medical and Population Genetics and Genetic Analysis Platform, The Broad Institute of MIT and Harvard, Cambridge MA 02132, USA
| | - Patrick F. Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, 27599-7264, USA
| | - Michael C. O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Michael J. Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Jeffrey C. Barrett
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1HH, Cambridge, UK
| |
Collapse
|
641
|
Corbett MA, Turner SJ, Gardner A, Silver J, Stankovich J, Leventer RJ, Derry CP, Carroll R, Ha T, Scheffer IE, Bahlo M, Jackson GD, Mackey DA, Berkovic SF, Gecz J. Familial epilepsy with anterior polymicrogyria as a presentation of COL18A1 mutations. Eur J Med Genet 2017; 60:437-443. [DOI: 10.1016/j.ejmg.2017.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/13/2017] [Accepted: 06/06/2017] [Indexed: 12/20/2022]
|
642
|
Kardon G, Ackerman KG, McCulley DJ, Shen Y, Wynn J, Shang L, Bogenschutz E, Sun X, Chung WK. Congenital diaphragmatic hernias: from genes to mechanisms to therapies. Dis Model Mech 2017; 10:955-970. [PMID: 28768736 PMCID: PMC5560060 DOI: 10.1242/dmm.028365] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Congenital diaphragmatic hernias (CDHs) and structural anomalies of the diaphragm are a common class of congenital birth defects that are associated with significant morbidity and mortality due to associated pulmonary hypoplasia, pulmonary hypertension and heart failure. In ∼30% of CDH patients, genomic analyses have identified a range of genetic defects, including chromosomal anomalies, copy number variants and sequence variants. The affected genes identified in CDH patients include transcription factors, such as GATA4, ZFPM2, NR2F2 and WT1, and signaling pathway components, including members of the retinoic acid pathway. Mutations in these genes affect diaphragm development and can have pleiotropic effects on pulmonary and cardiac development. New therapies, including fetal endoscopic tracheal occlusion and prenatal transplacental fetal treatments, aim to normalize lung development and pulmonary vascular tone to prevent and treat lung hypoplasia and pulmonary hypertension, respectively. Studies of the association between particular genetic mutations and clinical outcomes should allow us to better understand the origin of this birth defect and to improve our ability to predict and identify patients most likely to benefit from specialized treatment strategies.
Collapse
Affiliation(s)
- Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Kate G Ackerman
- Departments of Pediatrics (Critical Care) and Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David J McCulley
- Department of Pediatrics, University of Wisconsin, Madison, WI 53792, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Julia Wynn
- Departments of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Linshan Shang
- Departments of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Eric Bogenschutz
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wendy K Chung
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
643
|
Vamathevan J, Birney E. A Review of Recent Advances in Translational Bioinformatics: Bridges from Biology to Medicine. Yearb Med Inform 2017; 26:178-187. [PMID: 29063562 PMCID: PMC6239226 DOI: 10.15265/iy-2017-017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Indexed: 11/24/2022] Open
Abstract
Objectives: To highlight and provide insights into key developments in translational bioinformatics between 2014 and 2016. Methods: This review describes some of the most influential bioinformatics papers and resources that have been published between 2014 and 2016 as well as the national genome sequencing initiatives that utilize these resources to routinely embed genomic medicine into healthcare. Also discussed are some applications of the secondary use of patient data followed by a comprehensive view of the open challenges and emergent technologies. Results: Although data generation can be performed routinely, analyses and data integration methods still require active research and standardization to improve streamlining of clinical interpretation. The secondary use of patient data has resulted in the development of novel algorithms and has enabled a refined understanding of cellular and phenotypic mechanisms. New data storage and data sharing approaches are required to enable diverse biomedical communities to contribute to genomic discovery. Conclusion: The translation of genomics data into actionable knowledge for use in healthcare is transforming the clinical landscape in an unprecedented way. Exciting and innovative models that bridge the gap between clinical and academic research are set to open up the field of translational bioinformatics for rapid growth in a digital era.
Collapse
Affiliation(s)
- J. Vamathevan
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - E. Birney
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| |
Collapse
|
644
|
Ben-Shalom R, Keeshen CM, Berrios KN, An JY, Sanders SJ, Bender KJ. Opposing Effects on Na V1.2 Function Underlie Differences Between SCN2A Variants Observed in Individuals With Autism Spectrum Disorder or Infantile Seizures. Biol Psychiatry 2017; 82:224-232. [PMID: 28256214 PMCID: PMC5796785 DOI: 10.1016/j.biopsych.2017.01.009] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/14/2016] [Accepted: 01/10/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Variants in the SCN2A gene that disrupt the encoded neuronal sodium channel NaV1.2 are important risk factors for autism spectrum disorder (ASD), developmental delay, and infantile seizures. Variants observed in infantile seizures are predominantly missense, leading to a gain of function and increased neuronal excitability. How variants associated with ASD affect NaV1.2 function and neuronal excitability are unclear. METHODS We examined the properties of 11 ASD-associated SCN2A variants in heterologous expression systems using whole-cell voltage-clamp electrophysiology and immunohistochemistry. Resultant data were incorporated into computational models of developing and mature cortical pyramidal cells that express NaV1.2. RESULTS In contrast to gain of function variants that contribute to seizure, we found that all ASD-associated variants dampened or eliminated channel function. Incorporating these electrophysiological results into a compartmental model of developing excitatory neurons demonstrated that all ASD variants, regardless of their mechanism of action, resulted in deficits in neuronal excitability. Corresponding analysis of mature neurons predicted minimal change in neuronal excitability. CONCLUSIONS This functional characterization thus identifies SCN2A mutation and NaV1.2 dysfunction as the most frequently observed ASD risk factor detectable by exome sequencing and suggests that associated changes in neuronal excitability, particularly in developing neurons, may contribute to ASD etiology.
Collapse
Affiliation(s)
- Roy Ben-Shalom
- Center for Integrative Neuroscience, Kavli Institute for Fundamental Neuroscience, Department of Neurology, San Francisco, San Francisco; Computational Research Division , Lawrence Berkeley National Laboratory, Berkeley, California
| | - Caroline M Keeshen
- Center for Integrative Neuroscience, Kavli Institute for Fundamental Neuroscience, Department of Neurology, San Francisco, San Francisco
| | - Kiara N Berrios
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, Puerto Rico
| | - Joon Y An
- Department of Psychiatry, San Francisco, San Francisco; UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| | - Stephan J Sanders
- Department of Psychiatry, San Francisco, San Francisco; UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| | - Kevin J Bender
- Center for Integrative Neuroscience, Kavli Institute for Fundamental Neuroscience, Department of Neurology, San Francisco, San Francisco; UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco; Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, Puerto Rico.
| |
Collapse
|
645
|
Geisheker MR, Heymann G, Wang T, Coe BP, Turner TN, Stessman HA, Hoekzema K, Kvarnung M, Shaw M, Friend K, Liebelt J, Barnett C, Thompson EM, Haan E, Guo H, Anderlid BM, Nordgren A, Lindstrand A, Vandeweyer G, Alberti A, Avola E, Vinci M, Giusto S, Pramparo T, Pierce K, Nalabolu S, Michaelson JJ, Sedlacek Z, Santen GW, Peeters H, Hakonarson H, Courchesne E, Romano C, Kooy RF, Bernier RA, Nordenskjöld M, Gecz J, Xia K, Zweifel LS, Eichler EE. Hotspots of missense mutation identify neurodevelopmental disorder genes and functional domains. Nat Neurosci 2017; 20:1043-1051. [PMID: 28628100 PMCID: PMC5539915 DOI: 10.1038/nn.4589] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/19/2017] [Indexed: 12/17/2022]
Abstract
Although de novo missense mutations have been predicted to account for more cases of autism than gene-truncating mutations, most research has focused on the latter. We identified the properties of de novo missense mutations in patients with neurodevelopmental disorders (NDDs) and highlight 35 genes with excess missense mutations. Additionally, 40 amino acid sites were recurrently mutated in 36 genes, and targeted sequencing of 20 sites in 17,688 patients with NDD identified 21 new patients with identical missense mutations. One recurrent site substitution (p.A636T) occurs in a glutamate receptor subunit, GRIA1. This same amino acid substitution in the homologous but distinct mouse glutamate receptor subunit Grid2 is associated with Lurcher ataxia. Phenotypic follow-up in five individuals with GRIA1 mutations shows evidence of specific learning disabilities and autism. Overall, we find significant clustering of de novo mutations in 200 genes, highlighting specific functional domains and synaptic candidate genes important in NDD pathology.
Collapse
Affiliation(s)
| | - Gabriel Heymann
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Tianyun Wang
- The State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Bradley P. Coe
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Tychele N. Turner
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Holly A.F. Stessman
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Kendra Hoekzema
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Malin Kvarnung
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Marie Shaw
- Robinson Research Institute and the University of Adelaide at the Women’s and Children’s Hospital, North Adelaide, South Australia, Australia
| | - Kathryn Friend
- Robinson Research Institute and the University of Adelaide at the Women’s and Children’s Hospital, North Adelaide, South Australia, Australia
- SA Pathology, Adelaide, South Australia, Australia
| | - Jan Liebelt
- South Australian Clinical Genetics Service, SA Pathology (at Women’s and Children’s Hospital), Adelaide, South Australia, Australia
| | - Christopher Barnett
- South Australian Clinical Genetics Service, SA Pathology (at Women’s and Children’s Hospital), Adelaide, South Australia, Australia
- School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Elizabeth M. Thompson
- South Australian Clinical Genetics Service, SA Pathology (at Women’s and Children’s Hospital), Adelaide, South Australia, Australia
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Eric Haan
- South Australian Clinical Genetics Service, SA Pathology (at Women’s and Children’s Hospital), Adelaide, South Australia, Australia
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Hui Guo
- The State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Britt-Marie Anderlid
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Ann Nordgren
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Geert Vandeweyer
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Antonino Alberti
- Unit of Pediatrics & Medical Genetics, IRCCS Associazione Oasi Maria Santissima, Troina, Italy
| | - Emanuela Avola
- Unit of Pediatrics & Medical Genetics, IRCCS Associazione Oasi Maria Santissima, Troina, Italy
| | - Mirella Vinci
- Laboratory of Medical Genetics, IRCCS Associazione Oasi Maria Santissima, Troina, Italy
| | - Stefania Giusto
- Unit of Neurology, IRCCS Associazione Oasi Maria Santissima, Troina, Italy
| | - Tiziano Pramparo
- University of California, San Diego, Autism Center of Excellence, La Jolla, California, USA
| | - Karen Pierce
- University of California, San Diego, Autism Center of Excellence, La Jolla, California, USA
| | - Srinivasa Nalabolu
- University of California, San Diego, Autism Center of Excellence, La Jolla, California, USA
| | | | - Zdenek Sedlacek
- Department of Biology and Medical Genetics, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Gijs W.E. Santen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hilde Peeters
- Centre for Human Genetics, KU Leuven and Leuven Autism Research, Leuven, Belgium
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Division of Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eric Courchesne
- University of California, San Diego, Autism Center of Excellence, La Jolla, California, USA
| | - Corrado Romano
- Unit of Pediatrics & Medical Genetics, IRCCS Associazione Oasi Maria Santissima, Troina, Italy
| | - R. Frank Kooy
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Raphael A. Bernier
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Magnus Nordenskjöld
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Jozef Gecz
- Robinson Research Institute and the University of Adelaide at the Women’s and Children’s Hospital, North Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Kun Xia
- The State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Larry S. Zweifel
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Evan E. Eichler
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Howard Hughes Medical Institute, Seattle, Washington, USA
| |
Collapse
|
646
|
Benke D, Möhler H. Impact on GABA systems in monogenetic developmental CNS disorders: Clues to symptomatic treatment. Neuropharmacology 2017; 136:46-55. [PMID: 28764992 DOI: 10.1016/j.neuropharm.2017.07.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/26/2017] [Accepted: 07/28/2017] [Indexed: 12/26/2022]
Abstract
Animal studies of several single-gene disorders demonstrate that reversing the molecular signaling deficits can result in substantial symptomatic improvements in function. Focusing on the ratio of excitation to inhibition as a potential pathophysiological hallmark, seven single-gene developmental CNS disorders are reviewed which are characterized by a striking dysregulation of neuronal inhibition. Deficits in inhibition and excessive inhibition are found. The examples of developmental disorders encompass Neurofibromatosis type 1, Fragile X syndrome, Rett syndrome, Dravet syndrome including autism-like behavior, NONO-mutation-induced intellectual disability, Succinic semialdehyde dehydrogenase deficiency and Congenital nystagmus due to FRMD7 mutations. The phenotype/genotype correlations observed in animal models point to potential treatment options and will continue to inspire clinical research. Three drugs are presently in clinical trials: acamprosate and ganoxolon for Fragile X syndrome and SGS-742 for SSADH deficiency. This article is part of the "Special Issue Dedicated to Norman G. Bowery".
Collapse
Affiliation(s)
- Dietmar Benke
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Drug Discovery Network Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Hanns Möhler
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Vladimir-Prelog-Weg 10, 8023 Zurich, Switzerland.
| |
Collapse
|
647
|
Di Gregorio E, Riberi E, Belligni EF, Biamino E, Spielmann M, Ala U, Calcia A, Bagnasco I, Carli D, Gai G, Giordano M, Guala A, Keller R, Mandrile G, Arduino C, Maffè A, Naretto VG, Sirchia F, Sorasio L, Ungari S, Zonta A, Zacchetti G, Talarico F, Pappi P, Cavalieri S, Giorgio E, Mancini C, Ferrero M, Brussino A, Savin E, Gandione M, Pelle A, Giachino DF, De Marchi M, Restagno G, Provero P, Cirillo Silengo M, Grosso E, Buxbaum JD, Pasini B, De Rubeis S, Brusco A, Ferrero GB. Copy number variants analysis in a cohort of isolated and syndromic developmental delay/intellectual disability reveals novel genomic disorders, position effects and candidate disease genes. Clin Genet 2017; 92:415-422. [PMID: 28295210 DOI: 10.1111/cge.13009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Array-comparative genomic hybridization (array-CGH) is a widely used technique to detect copy number variants (CNVs) associated with developmental delay/intellectual disability (DD/ID). AIMS Identification of genomic disorders in DD/ID. MATERIALS AND METHODS We performed a comprehensive array-CGH investigation of 1,015 consecutive cases with DD/ID and combined literature mining, genetic evidence, evolutionary constraint scores, and functional information in order to assess the pathogenicity of the CNVs. RESULTS We identified non-benign CNVs in 29% of patients. Amongst the pathogenic variants (11%), detected with a yield consistent with the literature, we found rare genomic disorders and CNVs spanning known disease genes. We further identified and discussed 51 cases with likely pathogenic CNVs spanning novel candidate genes, including genes encoding synaptic components and/or proteins involved in corticogenesis. Additionally, we identified two deletions spanning potential Topological Associated Domain (TAD) boundaries probably affecting the regulatory landscape. DISCUSSION AND CONCLUSION We show how phenotypic and genetic analyses of array-CGH data allow unraveling complex cases, identifying rare disease genes, and revealing unexpected position effects.
Collapse
Affiliation(s)
- E Di Gregorio
- University of Torino, Department of Medical Sciences, Turin, Italy.,Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - E Riberi
- Department of Public Health and Pediatrics, University of Torino, Turin, Italy
| | - E F Belligni
- Department of Public Health and Pediatrics, University of Torino, Turin, Italy
| | - E Biamino
- Department of Public Health and Pediatrics, University of Torino, Turin, Italy
| | - M Spielmann
- Research Group Mundlos, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - U Ala
- Computational Biology Unit, Molecular Biotechnology Center (MBC), Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - A Calcia
- University of Torino, Department of Medical Sciences, Turin, Italy
| | - I Bagnasco
- Neuropsichiatria Infantile, Martini Hospital, ASL TO1, Turin, Italy
| | - D Carli
- University of Torino, Department of Medical Sciences, Turin, Italy
| | - G Gai
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - M Giordano
- Department of Health Sciences, Laboratory of Genetics, University of Eastern Piedmont and Interdisciplinary Research Center of Autoimmune Diseases, Novara, Italy
| | - A Guala
- SOC Pediatria, Castelli Hospital, Verbania, Italy
| | - R Keller
- Mental Health Department, ASL TO2, Adult Autism Center, Turin, Italy
| | - G Mandrile
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy.,Medical Genetics, San Luigi Gonzaga University Hospital, Orbassano (TO), Italy
| | - C Arduino
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - A Maffè
- Molecular Biology and Genetics Unit, Santa Croce e Carle Hospital, Cuneo, Italy
| | - V G Naretto
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - F Sirchia
- Molecular Biology and Genetics Unit, Santa Croce e Carle Hospital, Cuneo, Italy
| | - L Sorasio
- Pediatrics, Santa Croce e Carle Hospital, Cuneo, Italy
| | - S Ungari
- Molecular Biology and Genetics Unit, Santa Croce e Carle Hospital, Cuneo, Italy
| | - A Zonta
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - G Zacchetti
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy.,Department of Health Sciences, Laboratory of Genetics, University of Eastern Piedmont and Interdisciplinary Research Center of Autoimmune Diseases, Novara, Italy
| | - F Talarico
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - P Pappi
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - S Cavalieri
- University of Torino, Department of Medical Sciences, Turin, Italy
| | - E Giorgio
- University of Torino, Department of Medical Sciences, Turin, Italy
| | - C Mancini
- University of Torino, Department of Medical Sciences, Turin, Italy
| | - M Ferrero
- University of Torino, Department of Medical Sciences, Turin, Italy
| | - A Brussino
- University of Torino, Department of Medical Sciences, Turin, Italy
| | - E Savin
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - M Gandione
- Department of Neuropsychiatry, University of Torino, Turin, Italy
| | - A Pelle
- Medical Genetics, San Luigi Gonzaga University Hospital, Orbassano (TO), Italy.,Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - D F Giachino
- Medical Genetics, San Luigi Gonzaga University Hospital, Orbassano (TO), Italy.,Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - M De Marchi
- Medical Genetics, San Luigi Gonzaga University Hospital, Orbassano (TO), Italy.,Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - G Restagno
- Laboratory of Molecular Genetics, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - P Provero
- Computational Biology Unit, Molecular Biotechnology Center (MBC), Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - M Cirillo Silengo
- Department of Public Health and Pediatrics, University of Torino, Turin, Italy
| | - E Grosso
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - J D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - B Pasini
- Molecular Biology and Genetics Unit, Santa Croce e Carle Hospital, Cuneo, Italy
| | - S De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - A Brusco
- University of Torino, Department of Medical Sciences, Turin, Italy.,Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - G B Ferrero
- Department of Public Health and Pediatrics, University of Torino, Turin, Italy
| |
Collapse
|
648
|
Nozawa RS, Boteva L, Soares DC, Naughton C, Dun AR, Buckle A, Ramsahoye B, Bruton PC, Saleeb RS, Arnedo M, Hill B, Duncan RR, Maciver SK, Gilbert N. SAF-A Regulates Interphase Chromosome Structure through Oligomerization with Chromatin-Associated RNAs. Cell 2017. [PMID: 28622508 PMCID: PMC5473940 DOI: 10.1016/j.cell.2017.05.029] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Higher eukaryotic chromosomes are organized into topologically constrained functional domains; however, the molecular mechanisms required to sustain these complex interphase chromatin structures are unknown. A stable matrix underpinning nuclear organization was hypothesized, but the idea was abandoned as more dynamic models of chromatin behavior became prevalent. Here, we report that scaffold attachment factor A (SAF-A), originally identified as a structural nuclear protein, interacts with chromatin-associated RNAs (caRNAs) via its RGG domain to regulate human interphase chromatin structures in a transcription-dependent manner. Mechanistically, this is dependent on SAF-A's AAA+ ATPase domain, which mediates cycles of protein oligomerization with caRNAs, in response to ATP binding and hydrolysis. SAF-A oligomerization decompacts large-scale chromatin structure while SAF-A loss or monomerization promotes aberrant chromosome folding and accumulation of genome damage. Our results show that SAF-A and caRNAs form a dynamic, transcriptionally responsive chromatin mesh that organizes large-scale chromosome structures and protects the genome from instability.
Collapse
Affiliation(s)
- Ryu-Suke Nozawa
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Lora Boteva
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Dinesh C Soares
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Catherine Naughton
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Alison R Dun
- Edinburgh Super-Resolution Imaging Consortium, Institute of Biological Chemistry, Biophysics, and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, UK
| | - Adam Buckle
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Bernard Ramsahoye
- Centre for Genomics and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Peter C Bruton
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Rebecca S Saleeb
- Edinburgh Super-Resolution Imaging Consortium, Institute of Biological Chemistry, Biophysics, and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, UK
| | - Maria Arnedo
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Bill Hill
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Rory R Duncan
- Edinburgh Super-Resolution Imaging Consortium, Institute of Biological Chemistry, Biophysics, and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, UK
| | - Sutherland K Maciver
- Centre for Integrative Physiology, Edinburgh Medical School, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - Nick Gilbert
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
649
|
Celen C, Chuang JC, Luo X, Nijem N, Walker AK, Chen F, Zhang S, Chung AS, Nguyen LH, Nassour I, Budhipramono A, Sun X, Bok LA, McEntagart M, Gevers EF, Birnbaum SG, Eisch AJ, Powell CM, Ge WP, Santen GWE, Chahrour M, Zhu H. Arid1b haploinsufficient mice reveal neuropsychiatric phenotypes and reversible causes of growth impairment. eLife 2017; 6:e25730. [PMID: 28695822 PMCID: PMC5515576 DOI: 10.7554/elife.25730] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/24/2017] [Indexed: 12/17/2022] Open
Abstract
Sequencing studies have implicated haploinsufficiency of ARID1B, a SWI/SNF chromatin-remodeling subunit, in short stature (Yu et al., 2015), autism spectrum disorder (O'Roak et al., 2012), intellectual disability (Deciphering Developmental Disorders Study, 2015), and corpus callosum agenesis (Halgren et al., 2012). In addition, ARID1B is the most common cause of Coffin-Siris syndrome, a developmental delay syndrome characterized by some of the above abnormalities (Santen et al., 2012; Tsurusaki et al., 2012; Wieczorek et al., 2013). We generated Arid1b heterozygous mice, which showed social behavior impairment, altered vocalization, anxiety-like behavior, neuroanatomical abnormalities, and growth impairment. In the brain, Arid1b haploinsufficiency resulted in changes in the expression of SWI/SNF-regulated genes implicated in neuropsychiatric disorders. A focus on reversible mechanisms identified Insulin-like growth factor (IGF1) deficiency with inadequate compensation by Growth hormone-releasing hormone (GHRH) and Growth hormone (GH), underappreciated findings in ARID1B patients. Therapeutically, GH supplementation was able to correct growth retardation and muscle weakness. This model functionally validates the involvement of ARID1B in human disorders, and allows mechanistic dissection of neurodevelopmental diseases linked to chromatin-remodeling.
Collapse
Affiliation(s)
- Cemre Celen
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jen-Chieh Chuang
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Xin Luo
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Nadine Nijem
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, United States
| | - Angela K Walker
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, United States
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Fei Chen
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Shuyuan Zhang
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Andrew S Chung
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Liem H Nguyen
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ibrahim Nassour
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Albert Budhipramono
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Xuxu Sun
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Levinus A Bok
- Department of Pediatrics, Máxima Medical Center, Veldhoven, The Netherlands
| | - Meriel McEntagart
- Medical Genetics, St George's University Hospitals, NHS Foundation Trust, United Kingdom Caroline Brain, Endocrinology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Evelien F Gevers
- William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, United Kingdom
| | - Shari G Birnbaum
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Amelia J Eisch
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, and Mahoney Institute of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Craig M Powell
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, United States
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Woo-Ping Ge
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Gijs WE Santen
- Department of Clinical genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Chahrour
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, United States
| | - Hao Zhu
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
650
|
Weiner DJ, Wigdor EM, Ripke S, Walters RK, Kosmicki JA, Grove J, Samocha KE, Goldstein J, Okbay A, Bybjerg-Grauholm J, Werge T, Hougaard DM, Taylor J, iPSYCH-Broad Autism Group, Psychiatric Genomics Consortium Autism Group, Skuse D, Devlin B, Anney R, Sanders SJ, Bishop S, Mortensen PB, Børglum AD, Smith GD, Daly MJ, Robinson EB. Polygenic transmission disequilibrium confirms that common and rare variation act additively to create risk for autism spectrum disorders. Nat Genet 2017; 49:978-985. [PMID: 28504703 PMCID: PMC5552240 DOI: 10.1038/ng.3863] [Citation(s) in RCA: 326] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/13/2017] [Indexed: 12/16/2022]
Abstract
Autism spectrum disorder (ASD) risk is influenced by common polygenic and de novo variation. We aimed to clarify the influence of polygenic risk for ASD and to identify subgroups of ASD cases, including those with strongly acting de novo variants, in which polygenic risk is relevant. Using a novel approach called the polygenic transmission disequilibrium test and data from 6,454 families with a child with ASD, we show that polygenic risk for ASD, schizophrenia, and greater educational attainment is over-transmitted to children with ASD. These findings hold independent of proband IQ. We find that polygenic variation contributes additively to risk in ASD cases who carry a strongly acting de novo variant. Lastly, we show that elements of polygenic risk are independent and differ in their relationship with phenotype. These results confirm that the genetic influences on ASD are additive and suggest that they create risk through at least partially distinct etiologic pathways.
Collapse
Affiliation(s)
- Daniel J. Weiner
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Emilie M. Wigdor
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Psychiatry and Psychotherapy, Charité, Campus Mitte, Berlin, Germany
| | - Raymond K. Walters
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jack A. Kosmicki
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Genetics and Genomics, Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Jakob Grove
- Department of Biomedicine (Human Genetics), Aarhus University, Aarhus, Denmark
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Kaitlin E. Samocha
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jacqueline Goldstein
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Aysu Okbay
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Jonas Bybjerg-Grauholm
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
- Danish Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Thomas Werge
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Mental Health Services Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - David M. Hougaard
- Danish Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Jacob Taylor
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Psychiatry, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | | | - David Skuse
- Behavioural Sciences Unit, Institute of Child Health, University College London, London, UK
| | - Bernie Devlin
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Richard Anney
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, Wales, UK
| | - Stephan J. Sanders
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, 94158, USA
| | - Somer Bishop
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, 94158, USA
| | - Preben Bo Mortensen
- Department of Biomedicine (Human Genetics), Aarhus University, Aarhus, Denmark
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
- National Centre for Register-based Research, University of Aarhus, Aarhus, Denmark
| | - Anders D. Børglum
- Department of Biomedicine (Human Genetics), Aarhus University, Aarhus, Denmark
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Mark J. Daly
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Elise B. Robinson
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|