1
|
Levitin MO, Rawlins LE, Sanchez-Andrade G, Arshad OA, Collins SC, Sawiak SJ, Iffland PH, Andersson MHL, Bupp C, Cambridge EL, Coomber EL, Ellis I, Herkert JC, Ironfield H, Jory L, Kretz PF, Kant SG, Neaverson A, Nibbeling E, Rowley C, Relton E, Sanderson M, Scott EM, Stewart H, Shuen AY, Schreiber J, Tuck L, Tonks J, Terkelsen T, van Ravenswaaij-Arts C, Vasudevan P, Wenger O, Wright M, Day A, Hunter A, Patel M, Lelliott CJ, Crino PB, Yalcin B, Crosby AH, Baple EL, Logan DW, Hurles ME, Gerety SS. Models of KPTN-related disorder implicate mTOR signalling in cognitive and overgrowth phenotypes. Brain 2023; 146:4766-4783. [PMID: 37437211 PMCID: PMC10629792 DOI: 10.1093/brain/awad231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/31/2023] [Accepted: 06/18/2023] [Indexed: 07/14/2023] Open
Abstract
KPTN-related disorder is an autosomal recessive disorder associated with germline variants in KPTN (previously known as kaptin), a component of the mTOR regulatory complex KICSTOR. To gain further insights into the pathogenesis of KPTN-related disorder, we analysed mouse knockout and human stem cell KPTN loss-of-function models. Kptn -/- mice display many of the key KPTN-related disorder phenotypes, including brain overgrowth, behavioural abnormalities, and cognitive deficits. By assessment of affected individuals, we have identified widespread cognitive deficits (n = 6) and postnatal onset of brain overgrowth (n = 19). By analysing head size data from their parents (n = 24), we have identified a previously unrecognized KPTN dosage-sensitivity, resulting in increased head circumference in heterozygous carriers of pathogenic KPTN variants. Molecular and structural analysis of Kptn-/- mice revealed pathological changes, including differences in brain size, shape and cell numbers primarily due to abnormal postnatal brain development. Both the mouse and differentiated induced pluripotent stem cell models of the disorder display transcriptional and biochemical evidence for altered mTOR pathway signalling, supporting the role of KPTN in regulating mTORC1. By treatment in our KPTN mouse model, we found that the increased mTOR signalling downstream of KPTN is rapamycin sensitive, highlighting possible therapeutic avenues with currently available mTOR inhibitors. These findings place KPTN-related disorder in the broader group of mTORC1-related disorders affecting brain structure, cognitive function and network integrity.
Collapse
Affiliation(s)
- Maria O Levitin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Evox Therapeutics Limited, Oxford OX4 4HG, UK
| | - Lettie E Rawlins
- RILD Wellcome Wolfson Medical Research Centre, University of Exeter, Exeter EX2 5DW, UK
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX1 2ED, UK
| | | | - Osama A Arshad
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Stephan C Collins
- INSERM Unit 1231, Université de Bourgogne Franche-Comté, Dijon 21078, France
| | - Stephen J Sawiak
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge CB2 3EB, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Phillip H Iffland
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Malin H L Andersson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Caleb Bupp
- Spectrum Health, Helen DeVos Children’s Hospital, Grand Rapids, MI 49503, USA
| | - Emma L Cambridge
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Eve L Coomber
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Ian Ellis
- Department of Clinical Genetics, Alder Hey Children’s Hospital, Liverpool L14 5AB, UK
| | - Johanna C Herkert
- Department of Genetics, University Medical Centre, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Holly Ironfield
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Logan Jory
- Haven Clinical Psychology Practice Ltd, Bude, Cornwall EX23 9HP, UK
| | | | - Sarina G Kant
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
- Department of Clinical Genetics, Leiden University Medical Center, Leiden 2300 RC, The Netherlands
| | - Alexandra Neaverson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Esther Nibbeling
- Laboratory for Diagnostic Genome Analysis, Department of Clinical Genetics, Leiden University Medical Center, Leiden 3015 GD, The Netherlands
| | - Christine Rowley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Institute of Metabolic Science, Cambridge University, Cambridge CB2 0QQ, UK
| | - Emily Relton
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7YH, UK
| | - Mark Sanderson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Ethan M Scott
- New Leaf Center, Clinic for Special Children, Mount Eaton, OH 44659, USA
| | - Helen Stewart
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Trust, Oxford OX3 7HE, UK
| | - Andrew Y Shuen
- London Health Sciences Centre, London, ON N6A 5W9, Canada
- Division of Medical Genetics, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5W9, Canada
| | - John Schreiber
- Department of Neurology, Children’s National Medical Center, Washington DC 20007, USA
| | - Liz Tuck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - James Tonks
- Haven Clinical Psychology Practice Ltd, Bude, Cornwall EX23 9HP, UK
| | - Thorkild Terkelsen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus DK-8200, Denmark
| | - Conny van Ravenswaaij-Arts
- Department of Genetics, University Medical Centre, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Pradeep Vasudevan
- Department of Clinical Genetics, University Hospitals of Leicester, Leicester Royal Infirmary, Leicester LE1 7RH, UK
| | - Olivia Wenger
- New Leaf Center, Clinic for Special Children, Mount Eaton, OH 44659, USA
| | - Michael Wright
- Institute of Human Genetics, International Centre for Life, Newcastle upon Tyne NE1 7RU, UK
| | - Andrew Day
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Qkine Ltd., Cambridge CB5 8HW, UK
| | - Adam Hunter
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Minal Patel
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Christopher J Lelliott
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Institute of Metabolic Science, Cambridge University, Cambridge CB2 0QQ, UK
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Binnaz Yalcin
- INSERM Unit 1231, Université de Bourgogne Franche-Comté, Dijon 21078, France
| | - Andrew H Crosby
- RILD Wellcome Wolfson Medical Research Centre, University of Exeter, Exeter EX2 5DW, UK
| | - Emma L Baple
- RILD Wellcome Wolfson Medical Research Centre, University of Exeter, Exeter EX2 5DW, UK
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX1 2ED, UK
| | - Darren W Logan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Waltham Petcare Science Institute, Waltham on the Wolds LE14 4RT, UK
| | - Matthew E Hurles
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Sebastian S Gerety
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| |
Collapse
|
2
|
Rojas RA, Kutateladze AA, Plummer L, Stamou M, Keefe DL, Salnikov KB, Delaney A, Hall JE, Sadreyev R, Ji F, Fliers E, Gambosova K, Quinton R, Merino PM, Mericq V, Seminara SB, Crowley WF, Balasubramanian R. Phenotypic continuum between Waardenburg syndrome and idiopathic hypogonadotropic hypogonadism in humans with SOX10 variants. Genet Med 2021; 23:629-636. [PMID: 33442024 PMCID: PMC8335791 DOI: 10.1038/s41436-020-01051-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
PURPOSE SOX10 variants previously implicated in Waardenburg syndrome (WS) have now been linked to Kallmann syndrome (KS), the anosmic form of idiopathic hypogonadotropic hypogonadism (IHH). We investigated whether SOX10-associated WS and IHH represent elements of a phenotypic continuum within a unifying disorder or if they represent phenotypically distinct allelic disorders. METHODS Exome sequencing from 1,309 IHH subjects (KS: 632; normosmic idiopathic hypogonadotropic hypogonadism [nIIHH]: 677) were reviewed for SOX10 rare sequence variants (RSVs). The genotypic and phenotypic spectrum of SOX10-related IHH (this study and literature) and SOX10-related WS cases (literature) were reviewed and compared with SOX10-RSV spectrum in gnomAD population. RESULTS Thirty-seven SOX10-associated IHH cases were identified as follows: current study: 16 KS; 4 nIHH; literature: 16 KS; 1 nIHH. Twenty-three IHH cases (62%; all KS), had ≥1 known WS-associated feature(s). Moreover, five previously reported SOX10-associated WS cases showed IHH-related features. Four SOX10 missense RSVs showed allelic overlap between IHH-ascertained and WS-ascertained cases. The SOX10-HMG domain showed an enrichment of RSVs in disease states versus gnomAD. CONCLUSION SOX10 variants contribute to both anosmic (KS) and normosmic (nIHH) forms of IHH. IHH and WS represent SOX10-associated developmental defects that lie along a unifying phenotypic continuum. The SOX10-HMG domain is critical for the pathogenesis of SOX10-related human disorders.
Collapse
Affiliation(s)
- Rebecca A Rojas
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Anna A Kutateladze
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lacey Plummer
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Maria Stamou
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - David L Keefe
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kathyrn B Salnikov
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Angela Delaney
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Janet E Hall
- National Institute of Environmental Health Sciences, Research Triangle, NC, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Eric Fliers
- Amsterdam University Medical Center, location AMC, Department of Endocrinology and Metabolism, Amsterdam, The Netherlands
| | - Katarina Gambosova
- Stormont-Vail Health, Cotton O'Neil Diabetes and Endocrinology, Topeka, KS, USA
| | - Richard Quinton
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-tyne, UK
| | - Paulina M Merino
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile
| | - Veronica Mericq
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile
| | - Stephanie B Seminara
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - William F Crowley
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ravikumar Balasubramanian
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
3
|
Krick MV, Desmarais E, Samaras A, Guéret E, Dimitroglou A, Pavlidis M, Tsigenopoulos C, Guinand B. Family-effects in the epigenomic response of red blood cells to a challenge test in the European sea bass (Dicentrarchus labrax, L.). BMC Genomics 2021; 22:111. [PMID: 33563212 PMCID: PMC7871408 DOI: 10.1186/s12864-021-07420-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/31/2021] [Indexed: 12/13/2022] Open
Abstract
Abstract Background In fish, minimally invasive blood sampling is widely used to monitor physiological stress with blood plasma biomarkers. As fish blood cells are nucleated, they might be a source a potential new markers derived from ‘omics technologies. We modified the epiGBS (epiGenotyping By Sequencing) technique to explore changes in genome-wide cytosine methylation in the red blood cells (RBCs) of challenged European sea bass (Dicentrarchus labrax), a species widely studied in both natural and farmed environments. Results We retrieved 501,108,033 sequencing reads after trimming, with a mean mapping efficiency of 73.0% (unique best hits). Minor changes in RBC methylome appeared to manifest after the challenge test and a family-effect was detected. Only fifty-seven differentially methylated cytosines (DMCs) close to 51 distinct genes distributed on 17 of 24 linkage groups (LGs) were detected between RBCs of pre- and post-challenge individuals. Thirty-seven of these genes were previously reported as differentially expressed in the brain of zebrafish, most of them involved in stress coping differences. While further investigation remains necessary, few DMC-related genes associated to the Brain Derived Neurotrophic Factor, a protein that favors stress adaptation and fear memory, appear relevant to integrate a centrally produced stress response in RBCs. Conclusion Our modified epiGBS protocol was powerful to analyze patterns of cytosine methylation in RBCs of D. labrax and to evaluate the impact of a challenge using minimally invasive blood samples. This study is the first approximation to identify epigenetic biomarkers of exposure to stress in fish. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07420-9.
Collapse
Affiliation(s)
- Madoka Vera Krick
- UMR UM CNRS IRD EPHE ISEM- Institut des Sciences de l'Evolution de Montpellier, Montpellier, France
| | - Erick Desmarais
- UMR UM CNRS IRD EPHE ISEM- Institut des Sciences de l'Evolution de Montpellier, Montpellier, France
| | | | - Elise Guéret
- UMR UM CNRS IRD EPHE ISEM- Institut des Sciences de l'Evolution de Montpellier, Montpellier, France.,Univ. Montpellier, CNRS, INSERM, Montpellier, France.,Montpellier GenomiX, France Génomique, Montpellier, France
| | | | - Michalis Pavlidis
- Department of Biology, University of Crete, 70013, Heraklion, Greece
| | - Costas Tsigenopoulos
- Hellenic Centre for Marine Research (HCMR), Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), 715 00, Heraklion, Greece
| | - Bruno Guinand
- UMR UM CNRS IRD EPHE ISEM- Institut des Sciences de l'Evolution de Montpellier, Montpellier, France.
| |
Collapse
|
4
|
Fang ZH, Nosková A, Crysnanto D, Neuenschwander S, Vögeli P, Pausch H. A 63-bp insertion in exon 2 of the porcine KIF21A gene is associated with arthrogryposis multiplex congenita. Anim Genet 2020; 51:820-823. [PMID: 32686171 DOI: 10.1111/age.12984] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022]
Abstract
A recessive form of arthrogryposis multiplex congenita (AMC) was detected 20 years ago in the Swiss Large White (SLW) pig population. A diagnostic marker test enabled the identification of carrier animals, but the underlying causal mutation remains unknown. To identify the mutation underlying AMC, we collected SNP chip genotyping data for 11 affected piglets and 23 healthy pigs. Association testing using 47 829 SNPs confirmed that AMC maps to SSC5 (P = 9.4 × 10-13 ). Subsequent autozygosity mapping revealed a common 6.06 Mb region (from 66 757 970 to 72 815 151 bp) of extended homozygosity in 11 piglets affected by AMC. Using WGS data, we detected a 63-bp insertion compatible with the recessive inheritance of AMC in the second exon of KIF21A gene encoding Kinesin Family Member 21A. The 63-bp insertion is predicted to introduce a premature stop codon in KIF21A gene (p.Val41_Phe42insTer) that truncates 1614 amino acids (~97%) from the protein. We found that this deleterious allele still segregates at a frequency of 0.1% in the SLW pig population. Carrier animals can now be detected unambiguously and excluded from breeding.
Collapse
Affiliation(s)
- Z-H Fang
- Animal Genomics, Institute of Agricultural Science, D-USYS, ETH Zürich, Zürich, 8092, Switzerland
| | - A Nosková
- Animal Genomics, Institute of Agricultural Science, D-USYS, ETH Zürich, Zürich, 8092, Switzerland
| | - D Crysnanto
- Animal Genomics, Institute of Agricultural Science, D-USYS, ETH Zürich, Zürich, 8092, Switzerland
| | - S Neuenschwander
- Animal Genetics unit, Institute of Agricultural Science, D-USYS, ETH Zürich, Zürich, 8092, Switzerland
| | - P Vögeli
- Animal Genetics unit, Institute of Agricultural Science, D-USYS, ETH Zürich, Zürich, 8092, Switzerland
| | - H Pausch
- Animal Genomics, Institute of Agricultural Science, D-USYS, ETH Zürich, Zürich, 8092, Switzerland
| |
Collapse
|
5
|
LaMantia A. The strengths of the genetic approach to understanding neural systems development and function: Ray Guillery's synthesis. Eur J Neurosci 2019; 49:888-899. [PMID: 29883004 PMCID: PMC6369024 DOI: 10.1111/ejn.13985] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/04/2018] [Accepted: 05/23/2018] [Indexed: 12/29/2022]
Abstract
The organization and function of sensory systems, especially the mammalian visual system, has been the focus of philosophers and scientists for centuries-from Descartes and Newton onward. Nevertheless, the utility of understanding development and its genetic foundations for deeper insight into neural function has been debated: Do you need to know how something is assembled-a car, for example-to understand how it works or how to use it-to turn on the ignition and drive? This review addresses this issue for sensory pathways. The pioneering work of the late Rainer W. (Ray) Guillery provides an unequivocal answer to this central question: Using genetics for mechanistic exploration of sensory system development yields essential knowledge of organization and function. Ray truly built the foundation for this now accepted tenet of modern neuroscience. His work on the development and reorganization of visual pathways in albino mammals-all with primary genetic mutations in genes for pigmentation-defined the genetic approach to neural systems development, function and plasticity. The work that followed his lead in a variety of sensory systems, including my own work in the developing olfactory system, proceeds directly from Ray's fundamental contributions.
Collapse
Affiliation(s)
- Anthony‐Samuel LaMantia
- Institute for Neuroscience and Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of Columbia
| |
Collapse
|
6
|
Canonical TGF-β Signaling Negatively Regulates Neuronal Morphogenesis through TGIF/Smad Complex-Mediated CRMP2 Suppression. J Neurosci 2018; 38:4791-4810. [PMID: 29695415 DOI: 10.1523/jneurosci.2423-17.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 03/07/2018] [Accepted: 03/20/2018] [Indexed: 11/21/2022] Open
Abstract
Functional neuronal connectivity requires proper neuronal morphogenesis and its dysregulation causes neurodevelopmental diseases. Transforming growth factor-β (TGF-β) family cytokines play pivotal roles in development, but little is known about their contribution to morphological development of neurons. Here we show that the Smad-dependent canonical signaling of TGF-β family cytokines negatively regulates neuronal morphogenesis during brain development. Mechanistically, activated Smads form a complex with transcriptional repressor TG-interacting factor (TGIF), and downregulate the expression of a neuronal polarity regulator, collapsin response mediator protein 2. We also demonstrate that TGF-β family signaling inhibits neurite elongation of human induced pluripotent stem cell-derived neurons. Furthermore, the expression of TGF-β receptor 1, Smad4, or TGIF, which have mutations found in patients with neurodevelopmental disorders, disrupted neuronal morphogenesis in both mouse (male and female) and human (female) neurons. Together, these findings suggest that the regulation of neuronal morphogenesis by an evolutionarily conserved function of TGF-β signaling is involved in the pathogenesis of neurodevelopmental diseases.SIGNIFICANCE STATEMENT Canonical transforming growth factor-β (TGF-β) signaling plays a crucial role in multiple organ development, including brain, and mutations in components of the signaling pathway associated with several human developmental disorders. In this study, we found that Smads/TG-interacting factor-dependent canonical TGF-β signaling regulates neuronal morphogenesis through the suppression of collapsin response mediator protein-2 (CRMP2) expression during brain development, and that function of this signaling is evolutionarily conserved in the mammalian brain. Mutations in canonical TGF-β signaling factors identified in patients with neurodevelopmental disorders disrupt the morphological development of neurons. Thus, our results suggest that proper control of TGF-β/Smads/CRMP2 signaling pathways is critical for the precise execution of neuronal morphogenesis, whose impairment eventually results in neurodevelopmental disorders.
Collapse
|
7
|
Ma H, Yu H, Li T, Zhao Y, Hou M, Chen Z, Wang Y, Sun T. JIP3 regulates neuronal radial migration by mediating TrkB axonal anterograde transport in the developing cerebral cortex. Biochem Biophys Res Commun 2017; 485:790-795. [PMID: 28259553 DOI: 10.1016/j.bbrc.2017.02.132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 02/27/2017] [Indexed: 11/27/2022]
Abstract
Radial migration is essential for the precise lamination and the coordinated function of the cerebral cortex. However, the molecular mechanisms for neuronal radial migration are not clear. Here, we report that c-Jun NH2-terminal kinase (JNK)-interacting protein-3 (JIP3) is highly expressed in the brain of embryonic mice and essential for radial migration. Knocking down JIP3 by in utero electroporation specifically perturbs the radial migration of cortical neurons but has no effect on neurogenesis and neuronal differentiation. Furthermore, we illustrate that JIP3 knockdown delays but does not block the migration of cortical neurons by investigating the distribution of neurons with JIP3 knocked down in the embryo and postnatal mouse. Finally, we find that JIP3 regulates cortical neuronal migration by mediating TrkB axonal anterograde transport during brain development. These findings deepen our understanding of the regulation of neuronal development by JIP3 and provide us a novel view on the regulating mechanisms of neuronal radial migration.
Collapse
Affiliation(s)
- Huixian Ma
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Hui Yu
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Ting Li
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Yan Zhao
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Ming Hou
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Zheyu Chen
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Yue Wang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China.
| | - Tao Sun
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China; Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
8
|
Kinane TB, Lin AE, Lahoud-Rahme M, Westra SJ, Mark EJ. Case 4-2017. A 2-Month-Old Girl with Growth Retardation and Respiratory Failure. N Engl J Med 2017; 376:562-574. [PMID: 28177866 DOI: 10.1056/nejmcpc1613465] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- T Bernard Kinane
- From the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Massachusetts General Hospital, and the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Harvard Medical School - both in Boston
| | - Angela E Lin
- From the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Massachusetts General Hospital, and the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Harvard Medical School - both in Boston
| | - Manuella Lahoud-Rahme
- From the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Massachusetts General Hospital, and the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Harvard Medical School - both in Boston
| | - Sjirk J Westra
- From the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Massachusetts General Hospital, and the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Harvard Medical School - both in Boston
| | - Eugene J Mark
- From the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Massachusetts General Hospital, and the Departments of Pediatrics (T.B.K., A.E.L.), Cardiology (M.L.-R.), Radiology (S.J.W.), and Pathology (E.J.M.), Harvard Medical School - both in Boston
| |
Collapse
|
9
|
Chen T, Wu Q, Zhang Y, Lu T, Yue W, Zhang D. Tcf4 Controls Neuronal Migration of the Cerebral Cortex through Regulation of Bmp7. Front Mol Neurosci 2016; 9:94. [PMID: 27752241 PMCID: PMC5046712 DOI: 10.3389/fnmol.2016.00094] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/20/2016] [Indexed: 11/14/2022] Open
Abstract
Background: Transcription factor 4 (TCF4) is found to be associated with schizophrenia. TCF4 mutations also cause Pitt-Hopkins Syndrome, a neurodevelopmental disorder associated with severe mental retardation. However, the function of TCF4 during brain development remains unclear. Results: Here, we report that Tcf4 is expressed in the developing cerebral cortex. In utero suppression of Tcf4 arrested neuronal migration, leading to accumulation of ectopic neurons in the intermediate zone. Knockdown of Tcf4 impaired leading process formation. Furthermore, Bone Morphogenetic Protein 7 (Bmp7) is upregulated in Tcf4-deficient neurons. In vivo gain of function and rescue experiments demonstrated that Bmp7 is the major downstream effector of Tcf4 required for neuronal migration. Conclusion: Thus, we have uncovered a new Tcf4/Bmp7-dependent mechanism underlying neuronal migration, and provide insights into the pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Tianda Chen
- Institute of Mental Health, Peking University Sixth Hospital, BeijingChina; Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, BeijingChina
| | - Qinwei Wu
- Institute of Mental Health, Peking University Sixth Hospital, BeijingChina; Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijing, China; Peking-Tsinghua Center for Life Sciences, Peking UniversityBeijing, China
| | - Yang Zhang
- Institute of Mental Health, Peking University Sixth Hospital, BeijingChina; Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, BeijingChina
| | - Tianlan Lu
- Institute of Mental Health, Peking University Sixth Hospital, BeijingChina; Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, BeijingChina
| | - Weihua Yue
- Institute of Mental Health, Peking University Sixth Hospital, BeijingChina; Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, BeijingChina
| | - Dai Zhang
- Institute of Mental Health, Peking University Sixth Hospital, BeijingChina; Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, BeijingChina; Peking-Tsinghua Center for Life Sciences, Peking UniversityBeijing, China; PKU-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| |
Collapse
|
10
|
Park JG, Tischfield MA, Nugent AA, Cheng L, Di Gioia SA, Chan WM, Maconachie G, Bosley TM, Summers CG, Hunter DG, Robson CD, Gottlob I, Engle EC. Loss of MAFB Function in Humans and Mice Causes Duane Syndrome, Aberrant Extraocular Muscle Innervation, and Inner-Ear Defects. Am J Hum Genet 2016; 98:1220-1227. [PMID: 27181683 DOI: 10.1016/j.ajhg.2016.03.023] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/21/2016] [Indexed: 11/16/2022] Open
Abstract
Duane retraction syndrome (DRS) is a congenital eye-movement disorder defined by limited outward gaze and retraction of the eye on attempted inward gaze. Here, we report on three heterozygous loss-of-function MAFB mutations causing DRS and a dominant-negative MAFB mutation causing DRS and deafness. Using genotype-phenotype correlations in humans and Mafb-knockout mice, we propose a threshold model for variable loss of MAFB function. Postmortem studies of DRS have reported abducens nerve hypoplasia and aberrant innervation of the lateral rectus muscle by the oculomotor nerve. Our studies in mice now confirm this human DRS pathology. Moreover, we demonstrate that selectively disrupting abducens nerve development is sufficient to cause secondary innervation of the lateral rectus muscle by aberrant oculomotor nerve branches, which form at developmental decision regions close to target extraocular muscles. Thus, we present evidence that the primary cause of DRS is failure of the abducens nerve to fully innervate the lateral rectus muscle in early development.
Collapse
Affiliation(s)
- Jong G Park
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurology, Harvard Medical School, Boston, MA 02115, USA; Duke University School of Medicine, Durham, NC 27710, USA
| | - Max A Tischfield
- Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Alicia A Nugent
- Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Long Cheng
- Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Silvio Alessandro Di Gioia
- Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Wai-Man Chan
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Gail Maconachie
- Ulverscroft Eye Unit, University of Leicester, Leicester LE2 7LX, UK; Department of Neuroscience, Psychology, and Behavior, University of Leicester, Leicester LE2 7LX, UK
| | - Thomas M Bosley
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD 21287, USA
| | - C Gail Summers
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - David G Hunter
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Caroline D Robson
- Department of Radiology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Radiology, Harvard Medical School, Boston, MA 02115, USA
| | - Irene Gottlob
- Ulverscroft Eye Unit, University of Leicester, Leicester LE2 7LX, UK; Department of Neuroscience, Psychology, and Behavior, University of Leicester, Leicester LE2 7LX, UK
| | - Elizabeth C Engle
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurology, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA; Department of Ophthalmology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
11
|
Avedisova AS. [The relationship between schizophrenia and epilepsy: the history and current state of the problem]. Zh Nevrol Psikhiatr Im S S Korsakova 2016. [PMID: 28635730 DOI: 10.17116/jnevro201611691126-132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The review deals with the background and current state of the relationship between schizophrenia and epilepsy. The author presents results of recent epidemiological, neuropsychological, neurophysiological and genetic studies, indicating that these diseases may be different clinical variants of a common etiologic process (neurodevelopmental brain abnormalities), while epilepsy with chronic psychotic symptoms can be considered as an experimental model of schizophrenia pathogenesis.
Collapse
Affiliation(s)
- A S Avedisova
- Serbsky Federal Medical Research Center for Psychiatry and Narcology, Moscow, Russia, Soloviev Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| |
Collapse
|
12
|
Homberg JR, Kyzar EJ, Stewart AM, Nguyen M, Poudel MK, Echevarria DJ, Collier AD, Gaikwad S, Klimenko VM, Norton W, Pittman J, Nakamura S, Koshiba M, Yamanouchi H, Apryatin SA, Scattoni ML, Diamond DM, Ullmann JFP, Parker MO, Brown RE, Song C, Kalueff AV. Improving treatment of neurodevelopmental disorders: recommendations based on preclinical studies. Expert Opin Drug Discov 2015; 11:11-25. [DOI: 10.1517/17460441.2016.1115834] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Evan J Kyzar
- Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- The International Stress and Behavior Society (ISBS), Kiev, Ukraine
| | | | | | | | - David J Echevarria
- The International Stress and Behavior Society (ISBS), Kiev, Ukraine
- Department of Psychology, University of Southern Mississippi, Hattiesburg, MS, USA
| | - Adam D Collier
- Department of Psychology, University of Southern Mississippi, Hattiesburg, MS, USA
| | - Siddharth Gaikwad
- The International Stress and Behavior Society (ISBS), Kiev, Ukraine
- Research Institute of Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Neuroscience Graduate Hospital, China Medical University Hospital, Taichung, Taiwan
| | - Viktor M Klimenko
- The International Stress and Behavior Society (ISBS), Kiev, Ukraine
- Pavlov Physiology Department, Institute of Experimental Medicine, St. Petersburg, Russia
| | - William Norton
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Julian Pittman
- Department of Biological and Environmental Sciences, Troy University, Troy, AL, USA
| | - Shun Nakamura
- The International Stress and Behavior Society (ISBS), Kiev, Ukraine
- Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Mamiko Koshiba
- The International Stress and Behavior Society (ISBS), Kiev, Ukraine
- Departments of Pediatrics and Biochemistry, Saitama University Medical School, Saitama, Japan
| | - Hideo Yamanouchi
- Departments of Pediatrics and Biochemistry, Saitama University Medical School, Saitama, Japan
| | | | - Maria Luisa Scattoni
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanita, Rome, Italy
| | - David M Diamond
- Department of Psychology, University of South Florida, Tampa, FL, USA
- Research and Development Service, J.A. Haley Veterans Hospital, Tampa, FL, USA
| | - Jeremy FP Ullmann
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Matthew O Parker
- School of Health Sciences and Social Work, University of Portsmouth, Portsmouth, UK
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Cai Song
- Research Institute of Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Neuroscience Graduate Hospital, China Medical University Hospital, Taichung, Taiwan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Allan V Kalueff
- Research Institute of Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Institute for Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Institute of Chemical Technology and Institute of Natural Sciences, Ural Federal University, Ekaterinburg, Russia
| |
Collapse
|
13
|
NDUFV2 regulates neuronal migration in the developing cerebral cortex through modulation of the multipolar-bipolar transition. Brain Res 2015; 1625:102-10. [PMID: 26327164 DOI: 10.1016/j.brainres.2015.08.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 08/20/2015] [Accepted: 08/23/2015] [Indexed: 11/23/2022]
Abstract
Abnormalities during brain development are tightly linked several psychiatric disorders. Mutations in NADH dehydrogenase ubiquinone flavoprotein 2 (NDUFV2) are responsible for schizophrenia, bipolar disorder and Parkinson׳s disease. However, the function of NDUFV2 during brain development remains unclear. Here we reported that ndufv2 is expressed in the developing cerebral cortex. In utero suppression of ndufv2 arrested neuronal migration, leading to accumulation of ectopic neurons in the intermediate zone. ndufv2 inhibition did not affect radial glia scaffold, progenitor cells or neurons survival. However, the loss of ndufv2 impairs neuronal multipolar-bipolar transition in vivo and polarization in vitro. Moreover, ndufv2 affected actin cytoskeleton and tubulin stabilization in cortical neurons. Overall, our findings establish a new NDUFV2 dependent mechanism underlying neuronal migration and psychiatric disorders.
Collapse
|
14
|
Waltereit R, Banaschewski T, Meyer-Lindenberg A, Poustka L. Interaction of neurodevelopmental pathways and synaptic plasticity in mental retardation, autism spectrum disorder and schizophrenia: implications for psychiatry. World J Biol Psychiatry 2014; 15:507-16. [PMID: 24079538 DOI: 10.3109/15622975.2013.838641] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Schizophrenia (SCZ), autism spectrum disorder (ASD) and mental retardation (MR) are psychiatric disorders with high heritability. They differ in their clinical presentation and in their time course of major symptoms, which predominantly occurs for MR and ASD during childhood and for SCZ during young adult age. Recent findings with focus on the developmental neurobiology of these disorders emphasize shared mechanisms of common origin. These findings propose a continuum of genetic risk factors impacting on synaptic plasticity with MR causing impairments in global cognitive abilities, ASD in social cognition and SCZ in both global and social cognition. METHODS We assess here the historical developments that led to the current disease concepts of the three disorders. We then analyse, based on the functions of genes mutated in two or three of the disorders, selected mechanisms shared in neurodevelopmental pathways and synaptic plasticity. RESULTS The analysis of the psychopathological constructs supports the existence of three distinct clinical entities but also elaborates important associations. Similarly, there are common mechanisms especially in global and social cognition. CONCLUSIONS We discuss implications from this integrated view on MR, ASD and SCZ for child & adolescent and adult psychiatry in pathophysiology and research perspectives.
Collapse
Affiliation(s)
- Robert Waltereit
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health and University of Heidelberg, Mannheim Medical Faculty , Mannheim , Germany
| | | | | | | |
Collapse
|
15
|
McCarthy SE, Gillis J, Kramer M, Lihm J, Yoon S, Berstein Y, Mistry M, Pavlidis P, Solomon R, Ghiban E, Antoniou E, Kelleher E, O’Brien C, Donohoe G, Gill M, Morris DW, McCombie WR, Corvin A. De novo mutations in schizophrenia implicate chromatin remodeling and support a genetic overlap with autism and intellectual disability. Mol Psychiatry 2014; 19:652-8. [PMID: 24776741 PMCID: PMC4031262 DOI: 10.1038/mp.2014.29] [Citation(s) in RCA: 270] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 01/21/2014] [Accepted: 02/24/2014] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a serious psychiatric disorder with a broadly undiscovered genetic etiology. Recent studies of de novo mutations (DNMs) in schizophrenia and autism have reinforced the hypothesis that rare genetic variation contributes to risk. We carried out exome sequencing on 57 trios with sporadic or familial schizophrenia. In sporadic trios, we observed a ~3.5-fold increase in the proportion of nonsense DNMs (0.101 vs 0.031, empirical P=0.01, Benjamini-Hochberg-corrected P=0.044). These mutations were significantly more likely to occur in genes with highly ranked probabilities of haploinsufficiency (P=0.0029, corrected P=0.006). DNMs of potential functional consequence were also found to occur in genes predicted to be less tolerant to rare variation (P=2.01 × 10(-)(5), corrected P=2.1 × 10(-)(3)). Genes with DNMs overlapped with genes implicated in autism (for example, AUTS2, CHD8 and MECP2) and intellectual disability (for example, HUWE1 and TRAPPC9), supporting a shared genetic etiology between these disorders. Functionally CHD8, MECP2 and HUWE1 converge on epigenetic regulation of transcription suggesting that this may be an important risk mechanism. Our results were consistent in an analysis of additional exome-based sequencing studies of other neurodevelopmental disorders. These findings suggest that perturbations in genes, which function in the epigenetic regulation of brain development and cognition, could have a central role in the susceptibility to, pathogenesis and treatment of mental disorders.
Collapse
Affiliation(s)
- Shane E. McCarthy
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Jesse Gillis
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Melissa Kramer
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Jayon Lihm
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Seungtai Yoon
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Yael Berstein
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Meeta Mistry
- Department of Psychiatry and Centre for High-throughput Biology, The University of British Columbia, Vancouver, Canada
| | - Paul Pavlidis
- Department of Psychiatry and Centre for High-throughput Biology, The University of British Columbia, Vancouver, Canada
| | - Rebecca Solomon
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Elena Ghiban
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Eric Antoniou
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Eric Kelleher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Carol O’Brien
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Gary Donohoe
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Michael Gill
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Derek W. Morris
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - W. Richard. McCombie
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
- The Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, USA
| | - Aiden Corvin
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
16
|
Roles of heat shock factor 1 in neuronal response to fetal environmental risks and its relevance to brain disorders. Neuron 2014; 82:560-72. [PMID: 24726381 DOI: 10.1016/j.neuron.2014.03.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2014] [Indexed: 12/25/2022]
Abstract
Prenatal exposure of the developing brain to various environmental challenges increases susceptibility to late onset of neuropsychiatric dysfunction; still, the underlying mechanisms remain obscure. Here we show that exposure of embryos to a variety of environmental factors such as alcohol, methylmercury, and maternal seizure activates HSF1 in cerebral cortical cells. Furthermore, Hsf1 deficiency in the mouse cortex exposed in utero to subthreshold levels of these challenges causes structural abnormalities and increases seizure susceptibility after birth. In addition, we found that human neural progenitor cells differentiated from induced pluripotent stem cells derived from schizophrenia patients show higher variability in the levels of HSF1 activation induced by environmental challenges compared to controls. We propose that HSF1 plays a crucial role in the response of brain cells to prenatal environmental insults and may be a key component in the pathogenesis of late-onset neuropsychiatric disorders.
Collapse
|
17
|
Abstract
Advances in genetic tools and sequencing technology in the past few years have vastly expanded our understanding of the genetics of neurodevelopmental disorders. Recent high-throughput sequencing analyses of structural brain malformations, cognitive and neuropsychiatric disorders, and localized cortical dysplasias have uncovered a diverse genetic landscape beyond classic Mendelian patterns of inheritance. The underlying genetic causes of neurodevelopmental disorders implicate numerous cell biological pathways critical for normal brain development.
Collapse
Affiliation(s)
- Wen F Hu
- Division of Genetics and Genomics, Department of Medicine; Manton Center for Orphan Disease Research; and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts 02115; , ,
| | | | | |
Collapse
|
18
|
Bosley TM, Abu-Amero KK, Oystreck DT. Congenital cranial dysinnervation disorders. Curr Opin Ophthalmol 2013; 24:398-406. [DOI: 10.1097/icu.0b013e3283645ad6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
19
|
Abstract
Mislocalization of axonal proteins can result in misassembly and/or miswiring of neural circuits, causing disease. To date, only a handful of genes that control polarized localization of axonal membrane proteins have been identified. Here we report that Drosophila X11/Mint proteins are required for targeting several proteins, including human amyloid precursor protein (APP) and Drosophila APP-like protein (APPL), to axonal membranes and for their exclusion from dendrites of the mushroom body in Drosophila, a brain structure involved in learning and memory. Axonal localization of APP is mediated by an endocytic motif, and loss of X11/Mint results in a dramatic increase in cell-surface levels of APPL, especially on dendrites. Mutations in genes required for endocytosis show similar mislocalization of these proteins to dendrites, and strongly enhance defects seen in X11/Mint mutants. These results suggest that X11/Mint-dependent endocytosis in dendrites may serve to promote the axonal localization of membrane proteins. Since X11/Mint binds to APP, and abnormal trafficking of APP contributes to Alzheimer's disease, deregulation of X11/Mint may be important for Alzheimer's disease pathogenesis.
Collapse
|
20
|
Norton WHJ. Toward developmental models of psychiatric disorders in zebrafish. Front Neural Circuits 2013; 7:79. [PMID: 23637652 PMCID: PMC3636468 DOI: 10.3389/fncir.2013.00079] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 04/09/2013] [Indexed: 12/20/2022] Open
Abstract
Psychiatric disorders are a diverse set of diseases that affect all aspects of mental function including social interaction, thinking, feeling, and mood. Although psychiatric disorders place a large economic burden on society, the drugs available to treat them are often palliative with variable efficacy and intolerable side-effects. The development of novel drugs has been hindered by a lack of knowledge about the etiology of these diseases. It is thus necessary to further investigate psychiatric disorders using a combination of human molecular genetics, gene-by-environment studies, in vitro pharmacological and biochemistry experiments, animal models, and investigation of the non-biological basis of these diseases, such as environmental effects. Many psychiatric disorders, including autism spectrum disorder, attention-deficit/hyperactivity disorder, mental retardation, and schizophrenia can be triggered by alterations to neural development. The zebrafish is a popular model for developmental biology that is increasingly used to study human disease. Recent work has extended this approach to examine psychiatric disorders as well. However, since psychiatric disorders affect complex mental functions that might be human specific, it is not possible to fully model them in fish. In this review, I will propose that the suitability of zebrafish for developmental studies, and the genetic tools available to manipulate them, provide a powerful model to study the roles of genes that are linked to psychiatric disorders during neural development. The relative speed and ease of conducting experiments in zebrafish can be used to address two areas of future research: the contribution of environmental factors to disease onset, and screening for novel therapeutic compounds.
Collapse
Affiliation(s)
- William H J Norton
- Department of Biology, College of Medicine, Biological Sciences and Psychiatry, University of Leicester Leicester, UK
| |
Collapse
|
21
|
Yu T, Chahrour M, Coulter M, Jiralerspong S, Okamura-Ikeda K, Ataman B, Schmitz-Abe K, Harmin D, Adli M, Malik A, D’Gama A, Lim E, Sanders S, Mochida G, Partlow J, Sunu C, Felie J, Rodriguez J, Nasir R, Ware J, Joseph R, Hill R, Kwan B, Al-Saffar M, Mukaddes N, Hashmi A, Balkhy S, Gascon G, Hisama F, LeClair E, Poduri A, Oner O, Al-Saad S, Al-Awadi S, Bastaki L, Ben-Omran T, Teebi A, Al-Gazali L, Eapen V, Stevens C, Rappaport L, Gabriel S, Markianos K, State M, Greenberg M, Taniguchi H, Braverman N, Morrow E, Walsh C. Using whole-exome sequencing to identify inherited causes of autism. Neuron 2013; 77:259-73. [PMID: 23352163 PMCID: PMC3694430 DOI: 10.1016/j.neuron.2012.11.002] [Citation(s) in RCA: 320] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2012] [Indexed: 01/01/2023]
Abstract
Despite significant heritability of autism spectrum disorders (ASDs), their extreme genetic heterogeneity has proven challenging for gene discovery. Studies of primarily simplex families have implicated de novo copy number changes and point mutations, but are not optimally designed to identify inherited risk alleles. We apply whole-exome sequencing (WES) to ASD families enriched for inherited causes due to consanguinity and find familial ASD associated with biallelic mutations in disease genes (AMT, PEX7, SYNE1, VPS13B, PAH, and POMGNT1). At least some of these genes show biallelic mutations in nonconsanguineous families as well. These mutations are often only partially disabling or present atypically, with patients lacking diagnostic features of the Mendelian disorders with which these genes are classically associated. Our study shows the utility of WES for identifying specific genetic conditions not clinically suspected and the importance of partial loss of gene function in ASDs.
Collapse
Affiliation(s)
- T.W. Yu
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- The Autism Consortium, Boston, Massachusetts, USA, 02115
- Harvard Medical School, Boston, Massachusetts, USA, 02115
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA, 02114
| | - M.H. Chahrour
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- The Autism Consortium, Boston, Massachusetts, USA, 02115
- Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - M.E. Coulter
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - S. Jiralerspong
- Department of Human Genetics and Pediatrics, McGill University, Montreal Children’s Hospital Research Institute, Montreal, Quebec, Canada, H3H1P3
| | - K. Okamura-Ikeda
- Institute for Enzyme Research, The University of Tokushima, Tokushima, Japan
| | - B. Ataman
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - K. Schmitz-Abe
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - D.A. Harmin
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - M. Adli
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, Virginia, USA, 22908
| | - A.N. Malik
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - A.M. D’Gama
- Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - E.T. Lim
- Analytic and Translational Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA, 02114
| | - S.J. Sanders
- Department of Genetics, Center for Human Genetics and Genomics and Program on Neurogenetics, Yale University School of Medicine, New Haven, Connecticut, USA, 06510
| | - G.H. Mochida
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Harvard Medical School, Boston, Massachusetts, USA, 02115
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA, 02114
| | - J.N. Partlow
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
| | - C.M. Sunu
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
| | - J.M. Felie
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
| | - J. Rodriguez
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
| | - R.H. Nasir
- Harvard Medical School, Boston, Massachusetts, USA, 02115
- Division of Developmental Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
| | - J. Ware
- Harvard Medical School, Boston, Massachusetts, USA, 02115
- Division of Developmental Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
| | - R.M. Joseph
- The Autism Consortium, Boston, Massachusetts, USA, 02115
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, USA, 02118
| | - R.S. Hill
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - B.Y. Kwan
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada, N6A 5C1
| | - M. Al-Saffar
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Department of Paediatrics, Faculty of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - N.M. Mukaddes
- Istanbul Faculty of Medicine, Department of Child Psychiatry, Istanbul University, Istanbul, Turkey
| | - A. Hashmi
- Armed Forces Hospital, King Abdulaziz Naval Base, Jubail, Kingdom of Saudi Arabia
| | - S. Balkhy
- Department of Neurosciences and Pediatrics, King Faisal Specialist Hospital and Research Center, Jeddah, Kingdom of Saudi Arabia
| | - G.G. Gascon
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA, 02114
- Istanbul Faculty of Medicine, Department of Child Psychiatry, Istanbul University, Istanbul, Turkey
- Clinical Neurosciences and Pediatrics, Brown University School of Medicine, Providence, Rhode Island, 02912
| | - F.M. Hisama
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA, 98195
| | - E. LeClair
- Harvard Medical School, Boston, Massachusetts, USA, 02115
- Division of Developmental Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
| | - A. Poduri
- Harvard Medical School, Boston, Massachusetts, USA, 02115
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts, USA,02115
| | - O. Oner
- Department of Child and Adolescent Psychiatry, Dr Sami Ulus Childrens’ Hospital, Telsizler, Ankara, Turkey
| | - S. Al-Saad
- Kuwait Center for Autism, Kuwait City, Kuwait
| | | | - L. Bastaki
- Kuwait Medical Genetics Center, Kuwait City, Kuwait
| | - T. Ben-Omran
- Section of Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
- Departments of Pediatrics and Genetic Medicine, Weil-Cornell Medical College, New York and Doha, Qatar
| | - A. Teebi
- Section of Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
- Departments of Pediatrics and Genetic Medicine, Weil-Cornell Medical College, New York and Doha, Qatar
| | - L. Al-Gazali
- Department of Paediatrics, Faculty of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - V. Eapen
- Academic Unit of Child Psychiatry South West Sydney (AUCS), University of New South Wales, Sydney, New South Wales, Australia
| | - C.R. Stevens
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA, 02142
| | - L. Rappaport
- The Autism Consortium, Boston, Massachusetts, USA, 02115
- Harvard Medical School, Boston, Massachusetts, USA, 02115
- Division of Developmental Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
| | - S.B. Gabriel
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA, 02142
| | - K. Markianos
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - M.W. State
- Department of Genetics, Center for Human Genetics and Genomics and Program on Neurogenetics, Yale University School of Medicine, New Haven, Connecticut, USA, 06510
| | - M.E. Greenberg
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - H. Taniguchi
- Institute for Enzyme Research, The University of Tokushima, Tokushima, Japan
| | - N.E. Braverman
- Department of Human Genetics and Pediatrics, McGill University, Montreal Children’s Hospital Research Institute, Montreal, Quebec, Canada, H3H1P3
| | - E.M. Morrow
- The Autism Consortium, Boston, Massachusetts, USA, 02115
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, 02912
- Department of Psychiatry and Human Behavior, Brown University, Providence, Rhode Island, 02912
| | - C.A. Walsh
- Division of Genetics, Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts, USA, 02115
- The Autism Consortium, Boston, Massachusetts, USA, 02115
- Harvard Medical School, Boston, Massachusetts, USA, 02115
| |
Collapse
|
22
|
Connecting signaling pathways underlying communication to ASD vulnerability. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2013; 113:97-133. [PMID: 24290384 DOI: 10.1016/b978-0-12-418700-9.00004-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Language is a human-specific trait that likely facilitated the rapid increase in higher cognitive function in our species. A consequence of the selective pressures that have permitted language and cognition to flourish in humans is the unique vulnerability of humans to developing cognitive disorders such as autism. Therefore, progress in understanding the genetic and molecular mechanisms of language evolution should provide insight into such disorders. Here, we discuss the few genes that have been identified in both autism-related pathways and language. We also detail the use of animal models to uncover the function of these genes at a mechanistic and circuit level. Finally, we present the use of comparative genomics to identify novel genes and gene networks involved in autism. Together, all of these approaches will allow for a broader and deeper view of the molecular brain mechanisms involved in the evolution of language and the gene disruptions associated with autism.
Collapse
|
23
|
Breuss M, Heng JIT, Poirier K, Tian G, Jaglin XH, Qu Z, Braun A, Gstrein T, Ngo L, Haas M, Bahi-Buisson N, Moutard ML, Passemard S, Verloes A, Gressens P, Xie Y, Robson KJH, Rani DS, Thangaraj K, Clausen T, Chelly J, Cowan NJ, Keays DA. Mutations in the β-tubulin gene TUBB5 cause microcephaly with structural brain abnormalities. Cell Rep 2012; 2:1554-62. [PMID: 23246003 PMCID: PMC3595605 DOI: 10.1016/j.celrep.2012.11.017] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 07/18/2012] [Accepted: 11/21/2012] [Indexed: 01/11/2023] Open
Abstract
The formation of the mammalian cortex requires the generation, migration, and differentiation of neurons. The vital role that the microtubule cytoskeleton plays in these cellular processes is reflected by the discovery that mutations in various tubulin isotypes cause different neurodevelopmental diseases, including lissencephaly (TUBA1A), polymicrogyria (TUBA1A, TUBB2B, TUBB3), and an ocular motility disorder (TUBB3). Here, we show that Tubb5 is expressed in neurogenic progenitors in the mouse and that its depletion in vivo perturbs the cell cycle of progenitors and alters the position of migrating neurons. We report the occurrence of three microcephalic patients with structural brain abnormalities harboring de novo mutations in TUBB5 (M299V, V353I, and E401K). These mutant proteins, which affect the chaperone-dependent assembly of tubulin heterodimers in different ways, disrupt neurogenic division and/or migration in vivo. Our results provide insight into the functional repertoire of the tubulin gene family, specifically implicating TUBB5 in embryonic neurogenesis and microcephaly.
Collapse
Affiliation(s)
- Martin Breuss
- Institute of Molecular Pathology, Dr Bohr-Gasse, Vienna 1030, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wu QF, Yang L, Li S, Wang Q, Yuan XB, Gao X, Bao L, Zhang X. Fibroblast Growth Factor 13 Is a Microtubule-Stabilizing Protein Regulating Neuronal Polarization and Migration. Cell 2012; 149:1549-64. [DOI: 10.1016/j.cell.2012.04.046] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/08/2012] [Accepted: 04/11/2012] [Indexed: 01/24/2023]
|
25
|
Clarke MC, Tanskanen A, Huttunen MO, Clancy M, Cotter DR, Cannon M. Evidence for shared susceptibility to epilepsy and psychosis: a population-based family study. Biol Psychiatry 2012; 71:836-9. [PMID: 22365727 DOI: 10.1016/j.biopsych.2012.01.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/09/2012] [Accepted: 01/09/2012] [Indexed: 12/25/2022]
Abstract
BACKGROUND There is emerging evidence of an etiological overlap between a range of neurodevelopmental disorders, including schizophrenia and epilepsy. Here we investigate shared familial vulnerability to psychotic illness and epilepsy in a family-based study. METHODS The study population consisted of parents and their children born in Helsinki between 1947 and 1990. The Finnish Hospital Discharge Register was used to determine psychiatric and neurological outcomes in adulthood for all offspring. Parental history of psychosis and epilepsy was determined by linking the Hospital Discharge Register and the Finnish Population Register. RESULTS Our total sample comprised 9653 families and 23,404 offspring. Individuals with epilepsy had a 5.5-fold increase in the risk of having a broadly defined psychotic disorder, an almost 8.5-fold increase in the risk of having schizophrenia, and a 6.3-fold increase in the risk of having bipolar disorder. There was strong evidence of clustering of the association between epilepsy and psychosis within families. Individuals with a parental history of epilepsy had a 2-fold increase in the risk of developing psychosis, compared with individuals without a parental history of epilepsy. Individuals with a parental history of psychosis had, reciprocally, a 2.7-fold increase in the risk of having a diagnosis of generalized epilepsy, compared with individuals without a parental history of psychosis. Post hoc analyses showed that these associations were not driven by the comorbidity of epilepsy and psychosis in the parents. CONCLUSIONS These findings support recent evidence of overlapping etiological factors between epilepsy and schizophrenia, especially recent evidence of a genetic overlap between these disorders.
Collapse
Affiliation(s)
- Mary C Clarke
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | | | | | | | | | | |
Collapse
|
26
|
Networks of neuronal genes affected by common and rare variants in autism spectrum disorders. PLoS Genet 2012; 8:e1002556. [PMID: 22412387 PMCID: PMC3297570 DOI: 10.1371/journal.pgen.1002556] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 01/11/2012] [Indexed: 11/19/2022] Open
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental disorders with phenotypic and genetic heterogeneity. Recent studies have reported rare and de novo mutations in ASD, but the allelic architecture of ASD remains unclear. To assess the role of common and rare variations in ASD, we constructed a gene co-expression network based on a widespread survey of gene expression in the human brain. We identified modules associated with specific cell types and processes. By integrating known rare mutations and the results of an ASD genome-wide association study (GWAS), we identified two neuronal modules that are perturbed by both rare and common variations. These modules contain highly connected genes that are involved in synaptic and neuronal plasticity and that are expressed in areas associated with learning and memory and sensory perception. The enrichment of common risk variants was replicated in two additional samples which include both simplex and multiplex families. An analysis of the combined contribution of common variants in the neuronal modules revealed a polygenic component to the risk of ASD. The results of this study point toward contribution of minor and major perturbations in the two sub-networks of neuronal genes to ASD risk.
Collapse
|
27
|
Konopka G, Wexler E, Rosen E, Mukamel Z, Osborn GE, Chen L, Lu D, Gao F, Gao K, Lowe JK, Geschwind DH. Modeling the functional genomics of autism using human neurons. Mol Psychiatry 2012; 17:202-14. [PMID: 21647150 PMCID: PMC3170664 DOI: 10.1038/mp.2011.60] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human neural progenitors from a variety of sources present new opportunities to model aspects of human neuropsychiatric disease in vitro. Such in vitro models provide the advantages of a human genetic background combined with rapid and easy manipulation, making them highly useful adjuncts to animal models. Here, we examined whether a human neuronal culture system could be utilized to assess the transcriptional program involved in human neural differentiation and to model some of the molecular features of a neurodevelopmental disorder, such as autism. Primary normal human neuronal progenitors (NHNPs) were differentiated into a post-mitotic neuronal state through addition of specific growth factors and whole-genome gene expression was examined throughout a time course of neuronal differentiation. After 4 weeks of differentiation, a significant number of genes associated with autism spectrum disorders (ASDs) are either induced or repressed. This includes the ASD susceptibility gene neurexin 1, which showed a distinct pattern from neurexin 3 in vitro, and which we validated in vivo in fetal human brain. Using weighted gene co-expression network analysis, we visualized the network structure of transcriptional regulation, demonstrating via this unbiased analysis that a significant number of ASD candidate genes are coordinately regulated during the differentiation process. As NHNPs are genetically tractable and manipulable, they can be used to study both the effects of mutations in multiple ASD candidate genes on neuronal differentiation and gene expression in combination with the effects of potential therapeutic molecules. These data also provide a step towards better understanding of the signaling pathways disrupted in ASD.
Collapse
|
28
|
Abstract
Rather than being polygenic, complex disorders probably represent umbrella terms for collections of conditions caused by rare, recent mutations in any of a large number of different genes.
Collapse
Affiliation(s)
- Kevin J Mitchell
- Smurfit Institute of Genetics and Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
29
|
Clapham KR, Yu TW, Ganesh VS, Barry B, Chan Y, Mei D, Parrini E, Funalot B, Dupuis L, Nezarati MM, du Souich C, van Karnebeek C, Guerrini R, Walsh CA. FLNA genomic rearrangements cause periventricular nodular heterotopia. Neurology 2012; 78:269-78. [PMID: 22238415 DOI: 10.1212/wnl.0b013e31824365e4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To identify copy number variant (CNV) causes of periventricular nodular heterotopia (PNH) in patients for whom FLNA sequencing is negative. METHODS Screening of 35 patients from 33 pedigrees on an Affymetrix 6.0 microarray led to the identification of one individual bearing a CNV that disrupted FLNA. FLNA-disrupting CNVs were also isolated in 2 other individuals by multiplex ligation probe amplification. These 3 cases were further characterized by high-resolution oligo array comparative genomic hybridization (CGH), and the precise junctional breakpoints of the rearrangements were identified by PCR amplification and sequencing. RESULTS We report 3 cases of PNH caused by nonrecurrent genomic rearrangements that disrupt one copy of FLNA. The first individual carried a 113-kb deletion that removes all but the first exon of FLNA. A second patient harbored a complex rearrangement including a deletion of the 3' end of FLNA accompanied by a partial duplication event. A third patient bore a 39-kb deletion encompassing all of FLNA and the neighboring gene EMD. High-resolution oligo array CGH of the FLNA locus suggests distinct molecular mechanisms for each of these rearrangements, and implicates nearby low copy repeats in their pathogenesis. CONCLUSIONS These results demonstrate that FLNA is prone to pathogenic rearrangements, and highlight the importance of screening for CNVs in individuals with PNH lacking FLNA point mutations.
Collapse
Affiliation(s)
- K R Clapham
- Harvard-MIT Division of Health Sciences and Technology, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Corvin AP. Two patients walk into a clinic...a genomics perspective on the future of schizophrenia. BMC Biol 2011; 9:77. [PMID: 22078159 PMCID: PMC3214150 DOI: 10.1186/1741-7007-9-77] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 11/07/2011] [Indexed: 12/17/2022] Open
Abstract
Progress is being made in schizophrenia genomics, suggesting that this complex brain disorder involves rare, moderate to high-risk mutations and the cumulative impact of small genetic effects, coupled with environmental factors. The genetic heterogeneity underlying schizophrenia and the overlap with other neurodevelopmental disorders suggest that it will not continue to be viewed as a single disease. This has radical implications for clinical practice, as diagnosis and treatment will be guided by molecular etiology rather than clinical diagnostic criteria.
Collapse
Affiliation(s)
- Aiden P Corvin
- Department of Psychiatry, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
31
|
Mitchell KJ, Huang ZJ, Moghaddam B, Sawa A. Following the genes: a framework for animal modeling of psychiatric disorders. BMC Biol 2011; 9:76. [PMID: 22078115 PMCID: PMC3214139 DOI: 10.1186/1741-7007-9-76] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 11/07/2011] [Indexed: 01/19/2023] Open
Abstract
The number of individual cases of psychiatric disorders that can be ascribed to identified, rare, single mutations is increasing with great rapidity. Such mutations can be recapitulated in mice to generate animal models with direct etiological validity. Defining the underlying pathogenic mechanisms will require an experimental and theoretical framework to make the links from mutation to altered behavior in an animal or psychopathology in a human. Here, we discuss key elements of such a framework, including cell type-based phenotyping, developmental trajectories, linking circuit properties at micro and macro scales and definition of neurobiological phenotypes that are directly translatable to humans.
Collapse
Affiliation(s)
- Kevin J Mitchell
- Smurfit Institute of Genetics and Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Z Josh Huang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Bita Moghaddam
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
32
|
Moore S, Kelleher E, Corvin A. The shock of the new: progress in schizophrenia genomics. Curr Genomics 2011; 12:516-24. [PMID: 22547958 PMCID: PMC3219846 DOI: 10.2174/138920211797904089] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 09/20/2011] [Accepted: 09/29/2011] [Indexed: 12/31/2022] Open
Abstract
A growing list of common and rare genetic risk variants are being implicated in schizophrenia susceptibility. As with other complex genetic disorders most of the variance in genetic risk is still to be attributed. What can be learned from progress to date? The available data challenges how we conceptualize schizophrenia and suggests strong aetiological links with other psychiatric and developmental disorders. With the identification of rare copy number risk variants implicating specific genes (e.g. VIPR2 and NRXN1) it is increasingly possible to investigate molecular aetiology in patient subgroups to establish whether schizophrenia represents one or many different disease processes. This review summarizes recent research progress and suggests how the tools of modern genomics and neuroscience can be applied to best understand this devastating disorder.
Collapse
Affiliation(s)
- Susan Moore
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Institute of Molecular Medicine, Trinity College, Dublin, Ireland
| | - Eric Kelleher
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Institute of Molecular Medicine, Trinity College, Dublin, Ireland
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Institute of Molecular Medicine, Trinity College, Dublin, Ireland
| |
Collapse
|
33
|
Mitchell KJ. The miswired brain: making connections from neurodevelopment to psychopathology. BMC Biol 2011; 9:23. [PMID: 21489316 PMCID: PMC3076292 DOI: 10.1186/1741-7007-9-23] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 04/12/2011] [Indexed: 01/20/2023] Open
Affiliation(s)
- Kevin J Mitchell
- Smurfit Institute of Genetics and Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
34
|
Tischfield MA, Cederquist GY, Gupta ML, Engle EC. Phenotypic spectrum of the tubulin-related disorders and functional implications of disease-causing mutations. Curr Opin Genet Dev 2011; 21:286-94. [PMID: 21292473 DOI: 10.1016/j.gde.2011.01.003] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 12/23/2010] [Accepted: 01/03/2011] [Indexed: 12/17/2022]
Abstract
A spectrum of neurological disorders characterized by abnormal neuronal migration, differentiation, and axon guidance and maintenance have recently been attributed to missense and splice-site mutations in the genes that encode α-tubulin and β-tubulin isotypes TUBA1A, TUBA8, TUBB2B, and TUBB3, all of which putatively coassemble into neuronal microtubules. The resulting nervous system malformations can include different types of cortical malformations, defects in commissural fiber tracts, and degeneration of motor and sensory axons. Many clinical phenotypes and brain malformations are shared among the various mutations regardless of structural location and/or isotype, while others segregate with distinct amino acids or functional domains within tubulin. Collectively, these disorders provide novel paradigms for understanding the biological functions of microtubules and their core components in normal health and disease.
Collapse
Affiliation(s)
- Max A Tischfield
- Department of Molecular Biology and Genetics, Johns Hopkins Medical School, 725 North Wolfe St., PCTB 804, Baltimore, MD 21205, United States
| | | | | | | |
Collapse
|
35
|
What disorders of cortical development tell us about the cortex: one plus one does not always make two. Curr Opin Genet Dev 2011; 21:333-9. [PMID: 21288712 PMCID: PMC3139684 DOI: 10.1016/j.gde.2011.01.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 12/29/2010] [Accepted: 01/04/2011] [Indexed: 12/31/2022]
Abstract
The unique size and complexity of the human cerebral cortex are achieved via a long and precisely regulated developmental process controlling neurogenesis, neuronal migration and differentiation. Traditionally, disorders of cortical development have been classified on the basis of the most obvious defects in one of these developmental steps. However, the more we learn about the cellular biological roles of genes that are essential for cortical development, the more we realize that these functions map onto molecular processes, but not so cleanly onto anatomical processes. Essential genes might be involved in both proliferation and migration as well as differentiation, reflecting roles for underlying molecular mechanisms in different phases of development and causing a stunning variety of cortical defects.
Collapse
|
36
|
Corvin A, Donohoe G, Hargreaves A, Gallagher L, Gill M. The cognitive genetics of neuropsychiatric disorders. Curr Top Behav Neurosci 2011; 12:579-613. [PMID: 22367920 DOI: 10.1007/7854_2011_188] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Classification in psychiatry is heavily dependent on clinical symptoms and illness course. This ignores the critical role that cognitive problems play in neuropsychiatric disorders affecting different domains across the lifespan, from ADHD and autism to schizophrenia and Alzheimers disease. At this point, it is unclear whether cognitive mechanisms are specific to disorders, whether multiple processes can contribute to the same disorder, or whether aberrant neural processing can result in many different phenotypic outcomes. Understanding this would allow us to better grasp normal as well as pathological brain function. This could inform diagnostics based on understanding of neurophysiological processes and the consequent development of new therapeutics. Genetics, and the development of genomic research, offers real opportunities to understand the molecular mechanisms relevant to cognition. This chapter defines and describes the main cognitive phenotypes, which are investigated in psychiatric disorders. We review evidence for their heritability and early progress in the field using cytogenetic, linkage and candidate gene-based research methodologies. With high-throughput genomics it is now possible to explore novel common and rare risk variants for psychiatric disorders and their role in cognitive function at a genome-wide level. We review the results of early genomic studies and discuss the novel insights that they are starting to provide. Finally, we review the analysis of whole-genome DNA sequence data and the challenges that this will bring for cognitive genomics research.
Collapse
Affiliation(s)
- A Corvin
- Department of Psychiatry, Trinity College Dublin, Dublin, Ireland,
| | | | | | | | | |
Collapse
|