1
|
Mkhitaryan M, Avetisyan T, Yeritsyan H, Harutyunyan H, Yenkoyan K. Unraveling the Genetic and Environmental Risk Factors of Autism Spectrum Disorder Through a Case-Control Study in Armenia. Health Sci Rep 2025; 8:e70801. [PMID: 40330750 PMCID: PMC12050264 DOI: 10.1002/hsr2.70801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Background and Aims Autism spectrum disorder (ASD) is influenced by genetic and environmental factors. This study investigates genetic mutations and prenatal factors associated with ASD, including their interplay, in a multicenter case-control study. Methods The study included 297 participants (149 ASD cases, 148 controls). Genetic data were analyzed using Multiplex Ligation-dependent Probe Amplification (MLPA) for ASD-associated chromosomal regions. Environmental data covered prenatal, perinatal, and neonatal factors. Logistic regression and stratified analyses were performed. Results Significant associations were found between ASD and mutations in 15q11-15q13, 16p11, and 11q13 regions, particularly in SHANK2 and SHANK3. Females with ASD showed higher mutation rates than males. Prenatal factors (e.g., MgB6 use, labor-inducing drugs, maternal stress, pregnancy complications, shorter interpregnancy intervals) exacerbated ASD risk when combined with specific genetic variations. Duphaston use during pregnancy, alongside certain mutations, may influence ASD risk, potentially offering protective effects. Conclusion This study highlights the importance of integrating genetic and environmental factors in ASD research. Findings underscore the need for tailored early interventions, prenatal care advice, and genetic counseling for at-risk families. Future studies should validate these findings in larger, diverse populations and explore underlying mechanisms.
Collapse
Affiliation(s)
| | - Tamara Avetisyan
- COBRAIN CenterYSMUYerevanArmenia
- Muratsan University Hospital Complex, YSMUYerevanArmenia
| | | | | | | |
Collapse
|
2
|
Budisteanu M, Papuc SM, Erbescu A, Glangher A, Andrei E, Rad F, Hinescu ME, Arghir A. Review of structural neuroimaging and genetic findings in autism spectrum disorder - a clinical perspective. Rev Neurosci 2025; 36:295-314. [PMID: 39566028 DOI: 10.1515/revneuro-2024-0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/03/2024] [Indexed: 11/22/2024]
Abstract
Autism spectrum disorders (ASDs) are neurodevelopmental conditions characterized by deficits in social relationships and communication and restrictive, repetitive behaviors and interests. ASDs form a heterogeneous group from a clinical and genetic perspective. Currently, ASDs diagnosis is based on the clinical observation of the individual's behavior. The subjective nature of behavioral diagnoses, in the context of ASDs heterogeneity, contributes to significant variation in the age at ASD diagnosis. Early detection has been proved to be critical in ASDs, as early start of appropriate therapeutic interventions greatly improve the outcome for some children. Structural magnetic resonance imaging (MRI) is widely used in the diagnostic work-up of neurodevelopmental conditions, including ASDs, mostly for brain malformations detection. Recently, the focus of brain imaging shifted towards quantitative MRI parameters, aiming to identify subtle changes that may establish early detection biomarkers. ASDs have a strong genetic component; deletions and duplications of several genomic loci have been strongly associated with ASDs risk. Consequently, a multitude of neuroimaging and genetic findings emerged in ASDs in the recent years. The association of gross or subtle changes in brain morphometry and volumes with different genetic defects has the potential to bring new insights regarding normal development and pathomechanisms of various disorders affecting the brain. Still, the clinical implications of these discoveries and the impact of genetic abnormalities on brain structure and function are unclear. Here we review the literature on brain imaging correlated with the most prevalent genomic imbalances in ASD, and discuss the potential clinical impact.
Collapse
Affiliation(s)
- Magdalena Budisteanu
- Alexandru Obregia Clinical Hospital of Psychiatry, 041914, Soseaua Berceni 10, Bucharest, Romania
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
- Faculty of Medicine, Titu Maiorescu University, 031593, Calea Vacaresti 187, Bucharest, Romania
| | - Sorina Mihaela Papuc
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
| | - Alina Erbescu
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
| | - Adelina Glangher
- Alexandru Obregia Clinical Hospital of Psychiatry, 041914, Soseaua Berceni 10, Bucharest, Romania
| | - Emanuela Andrei
- Alexandru Obregia Clinical Hospital of Psychiatry, 041914, Soseaua Berceni 10, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, 050474, Bulevardul Eroii Sanitari 8, Bucharest, Romania
| | - Florina Rad
- Alexandru Obregia Clinical Hospital of Psychiatry, 041914, Soseaua Berceni 10, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, 050474, Bulevardul Eroii Sanitari 8, Bucharest, Romania
| | - Mihail Eugen Hinescu
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, 050474, Bulevardul Eroii Sanitari 8, Bucharest, Romania
| | - Aurora Arghir
- Victor Babes National Institute of Pathology, 050096, Splaiul Independentei 99-101, Bucharest, Romania
| |
Collapse
|
3
|
Adamo M, Gayer M, Jacobs A, Raynaud Q, Sebbah R, di Domenicantonio G, Latypova A, Vionnet N, Kherif F, Lutti A, Pitteloud N, Draganski B. Enduring differential patterns of neuronal loss and myelination along 6-month pulsatile gonadotropin-releasing hormone therapy in individuals with Down syndrome. Brain Commun 2025; 7:fcaf117. [PMID: 40190351 PMCID: PMC11969670 DOI: 10.1093/braincomms/fcaf117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/08/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025] Open
Abstract
Despite major progress in understanding the impact of the triplicated chromosome 21 on the brain and behaviour in Down syndrome, our knowledge of the underlying neurobiology in humans is still limited. We sought to address some of the pertinent questions about the drivers of brain structure differences and their associations with cognitive function in Down syndrome. To this aim, in a pilot magnetic resonance imaging (MRI) study, we monitored brain anatomy in individuals with Down syndrome receiving pulsatile gonadotropin-releasing hormone (GnRH) therapy over 6 months in comparison with typically developed age- and sex-matched healthy controls. We analysed cross-sectional (Down syndrome/healthy controls n = 11/27; Down syndrome-2 females/9 males, age 26.7 ± 5.0 years old; healthy controls-8 females/19 males, age 24.1 ± 2.5 years old) and longitudinal (Down syndrome/healthy controls n = 8/13; Down syndrome-1 female/7 males, age 26.4 ± 5.3 years old; healthy controls-4 females/9 males, 24.7 ± 2.2 years old) relaxometry and diffusion-weighted MRI data alongside standard cognitive assessment. The statistical tests looked for cross-sectional baseline differences and for differential changes over time between Down syndrome and healthy controls. The post hoc analysis confined to the Down syndrome group, tested for potential time-dependent interactions between individuals' overall cognitive performance and associated brain anatomy changes. The brain MRI statistical analyses covered both grey and white matter regions across the whole brain allowing for investigation of regional volume, macromolecular/myelin and iron content, additionally to diffusion tensor and neurite orientation and dispersion density characterization across major white matter tracts. The cross-sectional analysis showed reduced frontal, temporal and cerebellar volumes in Down syndrome with only the cerebellar differences remaining significant after adjustment for the presence of microcephaly (P family-wise-corrected < 0.05). The volume reductions were paralleled by decreased cortical and subcortical macromolecular/myelin content confined to the cortical motor system, thalamus and basal ganglia (P family-wise-corrected < 0.05). All major white matter tracts showed a ubiquitous mean diffusivity and intracellular volume fraction reduction contrasted with no differences in magnetization transfer saturation metrics (P family-wise-corrected < 0.05). Compared with healthy controls over the same period, Down syndrome individuals under GnRH therapy showed cognitive improvement (Montreal Cognitive Assessment from 11.4 ± 5.5 to 15.1 ± 5.6; P < 0.01) on the background of stability of the observed differential neuroanatomical patterns. Despite the lack of adequate Down syndrome control group, we interpret the obtained cross-sectional and longitudinal findings in young adults as evidence for predominant neurodevelopmental neuronal loss due to dysfunctional neurogenesis without signs for short-term myelin loss.
Collapse
Affiliation(s)
- Michela Adamo
- Department of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Mihaly Gayer
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - An Jacobs
- Department of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Quentin Raynaud
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Raphael Sebbah
- Department of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Giulia di Domenicantonio
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Adeliya Latypova
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Nathalie Vionnet
- Department of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Ferath Kherif
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Antoine Lutti
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Nelly Pitteloud
- Department of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Bogdan Draganski
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
- Neurology Department, Max Planck Institute for Human Cognitive and Brain Sciences, D-04103 Leipzig, Germany
- Department of Neurology, Inselspital, University of Bern, CH-3010 Bern, Switzerland
- University Institute for Diagnostic and Interventional Neuroradiology, Inselspital, University of Bern, CH-3010 Bern, Switzerland
| |
Collapse
|
4
|
Elkhateeb N, Crookes R, Spiller M, Pavinato L, Palermo F, Brusco A, Parker M, Park SM, Mendes AC, Saraiva JM, Hammer TB, Nazaryan-Petersen L, Barakat TS, Wilke M, Bhoj E, Ahrens-Nicklas RC, Li D, Nomakuchi T, Brilstra EH, Hunt D, Johnson D, Mansour S, Oprych K, Mehta SG, Platzer K, Schnabel F, Kiep H, Faust H, Prinzing G, Wiltrout K, Radley JA, Serrano Russi AH, Atallah I, Campos-Xavier B, Amor DJ, Morgan AT, Fagerberg C, Andersen UA, Andersen CB, Bijlsma EK, Bird LM, Mullegama SV, Green A, Isidor B, Cogné B, Kenny J, Lynch SA, Quin S, Low K, Herget T, Kortüm F, Levy RJ, Morrison JL, Wheeler PG, Narumanch T, Peron K, Matthews N, Uhlman J, Bell L, Pang L, Scurr I, Belles RS, Salbert BA, Schaefer GB, Green S, Ros A, Rodríguez-Palmero A, Višnjar T, Writzl K, Vasudevan PC, Balasubramanian M. Expanding the phenotype and genotype spectrum of TAOK1 neurodevelopmental disorder and delineating TAOK2 neurodevelopmental disorder. Genet Med 2025; 27:101348. [PMID: 39737487 DOI: 10.1016/j.gim.2024.101348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025] Open
Abstract
PURPOSE The thousand and one kinase (TAOK) proteins are a group of serine/threonine-protein kinases involved in signaling pathways, cytoskeleton regulation, and neuronal development. TAOK1 variants are associated with a neurodevelopmental disorder (NDD) characterized by distinctive facial features, hypotonia, and feeding difficulties. TAOK2 variants have been reported to be associated with autism and early-onset obesity. However, a distinct TAOK2-NDD has not yet been delineated. METHODS We retrospectively studied the clinical and genetic data of individuals recruited from several centers with TAOK1 and TAOK2 variants that were detected through exome and genome sequencing. RESULTS We report 50 individuals with TAOK1 variants with associated phenotypes, including neurodevelopmental abnormalities (100%), macrocephaly (83%), and hypotonia (58%). We report male genital anomalies and hypoglycemia as novel phenotypes. Thirty-seven unique TAOK1 variants were identified. Most of the missense variants clustered in the protein kinase domain at residues that are intolerant to missense variation. We report 10 individuals with TAOK2 variants with associated phenotypes, including neurodevelopmental abnormalities (100%), macrocephaly (75%), autism (75%), and obesity (70%). CONCLUSION We describe the largest cohort of TAOK1-NDD to date, to our knowledge, expanding its phenotype and genotype spectrum with 30 novel variants. We delineated the phenotype of a novel TAOK2-NDD associated with neurodevelopmental abnormalities, autism, macrocephaly, and obesity.
Collapse
Affiliation(s)
- Nour Elkhateeb
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
| | - Renarta Crookes
- Sheffield Diagnostic Genetics Service, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, United Kingdom
| | - Michael Spiller
- Sheffield Diagnostic Genetics Service, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, United Kingdom
| | - Lisa Pavinato
- Department of Medical Sciences, University of Turin, Turin, Italy; Institute of Oncology Research (IOR), Bellinzona Institutes of Science (BIOS(+)), Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Flavia Palermo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alfredo Brusco
- Department of Neurosciences Rita Levi-Montalcini, University of Turin, Turin, Italy; Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Michael Parker
- Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom
| | - Soo-Mi Park
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ariana Costa Mendes
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Jorge M Saraiva
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal; University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, Portugal; Clinical Academic Center of Coimbra, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Trine Bjørg Hammer
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark; Department of Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Lusine Nazaryan-Petersen
- Department of Genetics, Rigshospitalet, Copenhagen, Denmark; Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands; ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Martina Wilke
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Elizabeth Bhoj
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Dong Li
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Tomoki Nomakuchi
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Eva H Brilstra
- Department of Genetics and Brain Center, University Medical Center Utrecht, The Netherlands
| | - David Hunt
- Wessex Clinical Genetics Service, Princess Anne Hospital, University Hospital Southampton NHS Trust, Southampton, United Kingdom
| | - Diana Johnson
- Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom
| | - Sahar Mansour
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, United Kingdom; School of Biological and Molecular Sciences, St George's University of London, London, United Kingdom
| | - Kathryn Oprych
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Sarju G Mehta
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | - Franziska Schnabel
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | - Henriette Kiep
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | - Helene Faust
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | | | | | - Jessica A Radley
- North West Thames Regional Genomics Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, United Kingdom
| | - Alvaro H Serrano Russi
- Division of Genetics, Department of Pediatrics, East Tennessee State University (ETSU), Quillen College of Medicine, TN
| | - Isis Atallah
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Belinda Campos-Xavier
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - David J Amor
- Speech and Language, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Angela T Morgan
- Speech and Language, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Christina Fagerberg
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark; Department of Clinical Genetics, Lillebaelt Hospital, location Vejle Hospital, Vejle, Denmark
| | - Ulla A Andersen
- Department of Mental Health, Odense University Hospital, Odense, Denmark
| | | | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Lynne M Bird
- Division of Genetics and Dysmorphology, Department of Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, CA
| | | | - Andrew Green
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland; University College Dublin School of Medicine and Medical Science, Dublin, Ireland
| | - Bertrand Isidor
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France; CHU Nantes, Service de Génétique Médicale, Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Benjamin Cogné
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Janna Kenny
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Sally A Lynch
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Shauna Quin
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Karen Low
- Department of Clinical Genetics, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom; Bristol Medical School, University of Bristol, United Kingdom
| | - Theresia Herget
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rebecca J Levy
- Neurology and Neurological Sciences, Division of Child Neurology, Stanford University and Lucile Packard Children's Hospital, Palo Alto, CA
| | | | | | - TaraChandra Narumanch
- Division of Genetics, Department of Pediatrics, West Virginia University, Morgantown, WV
| | - Kristina Peron
- Division of Genetics, Department of Pediatrics, West Virginia University, Morgantown, WV
| | - Nicole Matthews
- Division of Genetics, Department of Pediatrics, West Virginia University, Morgantown, WV
| | | | - Lauren Bell
- University of Illinois College of Medicine, Peoria, IL
| | - Lewis Pang
- Exeter Genomics Laboratory, RILD Building, Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Ingrid Scurr
- Department of Clinical Genetics, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | | | | | | | - Sarah Green
- University of Arkansas for Medical Sciences, Little Rock, AR
| | - Andrea Ros
- Department of Genetics, Hospital Universitari Germans Trias i Pujol, Catalonia, Spain
| | - Agustí Rodríguez-Palmero
- Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Catalonia, Spain; Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Tanja Višnjar
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Karin Writzl
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Pradeep C Vasudevan
- Department of Clinical Genetics, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Meena Balasubramanian
- Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom; Division of Clinical Medicine, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
5
|
Huguet G, Renne T, Poulain C, Dubuc A, Kumar K, Kazem S, Engchuan W, Shanta O, Douard E, Proulx C, Jean-Louis M, Saci Z, Mollon J, Schultz LM, Knowles EEM, Cox SR, Porteous D, Davies G, Redmond P, Harris SE, Schumann G, Dumas G, Labbe A, Pausova Z, Paus T, Scherer SW, Sebat J, Almasy L, Glahn DC, Jacquemont S. Effects of gene dosage on cognitive ability: A function-based association study across brain and non-brain processes. CELL GENOMICS 2024; 4:100721. [PMID: 39667348 PMCID: PMC11701252 DOI: 10.1016/j.xgen.2024.100721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/30/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024]
Abstract
Copy-number variants (CNVs) that increase the risk for neurodevelopmental disorders also affect cognitive ability. However, such CNVs remain challenging to study due to their scarcity, limiting our understanding of gene-dosage-sensitive biological processes linked to cognitive ability. We performed a genome-wide association study (GWAS) in 258,292 individuals, which identified-for the first time-a duplication at 2q12.3 associated with higher cognitive performance. We developed a functional-burden analysis, which tested the association between cognition and CNVs disrupting 6,502 gene sets biologically defined across tissues, cell types, and ontologies. Among those, 864 gene sets were associated with cognition, and effect sizes of deletion and duplication were negatively correlated. The latter suggested that functions across all biological processes were sensitive to either deletions (e.g., subcortical regions, postsynaptic) or duplications (e.g., cerebral cortex, presynaptic). Associations between non-brain tissues and cognition were driven partly by constrained genes, which may shed light on medical comorbidities in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Guillaume Huguet
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada.
| | - Thomas Renne
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Cécile Poulain
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Alma Dubuc
- École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Kuldeep Kumar
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Sayeh Kazem
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Worrawat Engchuan
- The Hospital for Sick Children, Genetics and Genome Biology, Toronto, ON, Canada; The Hospital for Sick Children, The Centre for Applied Genomics, Toronto, ON, Canada
| | - Omar Shanta
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Elise Douard
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Catherine Proulx
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Martineau Jean-Louis
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Zohra Saci
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Josephine Mollon
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura M Schultz
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emma E M Knowles
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon R Cox
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - David Porteous
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK; Medical Genetics Section, Centre for Genomic & Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK; Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Gail Davies
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Paul Redmond
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Sarah E Harris
- Lothian Birth Cohorts, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Gunter Schumann
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Guillaume Dumas
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Mila - Quebec Artificial Intelligence Institute, Montréal, QC, Canada
| | - Aurélie Labbe
- Département de Sciences de la Décision, HEC Montreal, Montreal, QC, Canada
| | - Zdenka Pausova
- Research Institute of the Hospital for Sick Children, Toronto, ON, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada; ECOGENE-21, Chicoutimi, QC, Canada
| | - Tomas Paus
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Psychiatry and Addictology, Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Stephen W Scherer
- The Hospital for Sick Children, Genetics and Genome Biology, Toronto, ON, Canada; The Hospital for Sick Children, The Centre for Applied Genomics, Toronto, ON, Canada; McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Jonathan Sebat
- University of California, San Diego, Department of Psychiatry, Department of Cellular & Molecular Medicine, Beyster Center of Psychiatric Genomics, San Diego, CA, USA
| | - Laura Almasy
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David C Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Olin Neuropsychiatric Research Center, Institute of Living, Hartford Hospital, Hartford, CT, USA
| | - Sébastien Jacquemont
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada; Department of Pediatrics, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
6
|
Auwerx C, Kutalik Z, Reymond A. The pleiotropic spectrum of proximal 16p11.2 CNVs. Am J Hum Genet 2024; 111:2309-2346. [PMID: 39332410 PMCID: PMC11568765 DOI: 10.1016/j.ajhg.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/29/2024] Open
Abstract
Recurrent genomic rearrangements at 16p11.2 BP4-5 represent one of the most common causes of genomic disorders. Originally associated with increased risk for autism spectrum disorder, schizophrenia, and intellectual disability, as well as adiposity and head circumference, these CNVs have since been associated with a plethora of phenotypic alterations, albeit with high variability in expressivity and incomplete penetrance. Here, we comprehensively review the pleiotropy associated with 16p11.2 BP4-5 rearrangements to shine light on its full phenotypic spectrum. Illustrating this phenotypic heterogeneity, we expose many parallels between findings gathered from clinical versus population-based cohorts, which often point to the same physiological systems, and emphasize the role of the CNV beyond neuropsychiatric and anthropometric traits. Revealing the complex and variable clinical manifestations of this CNV is crucial for accurate diagnosis and personalized treatment strategies for carrier individuals. Furthermore, we discuss areas of research that will be key to identifying factors contributing to phenotypic heterogeneity and gaining mechanistic insights into the molecular pathways underlying observed associations, while demonstrating how diversity in affected individuals, cohorts, experimental models, and analytical approaches can catalyze discoveries.
Collapse
Affiliation(s)
- Chiara Auwerx
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland; Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Zoltán Kutalik
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
7
|
Verbesselt J, Breckpot J, Zink I, Swillen A. Language Profiles of School-Age Children With 16p11.2 Copy Number Variants in a Clinically Ascertained Cohort. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2024; 67:4487-4503. [PMID: 39418585 PMCID: PMC11567083 DOI: 10.1044/2024_jslhr-24-00257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/18/2024] [Accepted: 08/01/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Individuals with proximal 16p11.2 copy number variants (CNVs), either deletions (16p11.2DS) or duplications (16p11.2Dup), are predisposed to neurodevelopmental difficulties and disorders, such as language disorders, intellectual disability, and autism spectrum disorder. The purpose of the current study was to characterize language profiles of school-age children with proximal 16p11.2 CNVs, in relation to the normative sample and unaffected siblings of children with 16p11.2DS. METHOD Standardized language tests were conducted in 33 school-age children with BP4-BP5 16p11.2 CNVs and eight unaffected siblings of children with 16p11.2DS to evaluate language production and comprehension skills across various language domains. A standardized intelligence test was also administered, and parents completed a standardized questionnaire to assess autistic traits. Language profiles were compared across 16p11.2 CNVs and intrafamilial pairs. The influence of nonverbal intelligence and autistic traits on language outcomes was investigated. RESULTS No significant differences were found between children with 16p11.2DS and those with 16p11.2Dup, although both groups exhibited significantly poorer language skills compared to the normative sample and unaffected siblings of children with 16p11.2DS. Severe language deficits were identified in 70% of individuals with 16p11.2 CNVs across all language subdomains, with significantly better receptive vocabulary skills than overall receptive language abilities. In children with 16p11.2DS, expressive language deficits were more pronounced than receptive deficits. In contrast, only in children with 16p11.2Dup did nonverbal intelligence influence their language outcomes. CONCLUSIONS The current study contributes to the deeper understanding of language profiles in 16p11.2 CNVs in a clinically ascertained cohort, indicating generalized deficits across multiple language domains, rather than a syndrome-specific pattern targeting specific subdomains. The findings underscore the importance of early diagnosis, targeted therapy, and monitoring of language skills in children with 16p11.2 CNVs. SUPPLEMENTAL MATERIAL https://doi.org/10.23641/asha.27228702.
Collapse
Affiliation(s)
- Jente Verbesselt
- Department of Human Genetics, Catholic University Leuven, Belgium
- Research Group Experimental Oto-Rhino-Laryngology, Department of Neurosciences, Leuven Brain Institute, Catholic University Leuven, Belgium
| | - Jeroen Breckpot
- Department of Human Genetics, Catholic University Leuven, Belgium
- Centre for Human Genetics, University Hospitals Leuven, Belgium
| | - Inge Zink
- Research Group Experimental Oto-Rhino-Laryngology, Department of Neurosciences, Leuven Brain Institute, Catholic University Leuven, Belgium
- MUCLA, Department of Oto-Rhino-Laryngology, Head & Neck Surgery, University Hospitals Leuven, Belgium
| | - Ann Swillen
- Department of Human Genetics, Catholic University Leuven, Belgium
- Centre for Human Genetics, University Hospitals Leuven, Belgium
| |
Collapse
|
8
|
Mergener R, Nascimento LPC, Böttcher AK, Nunes MR, Zen PRG. What Can Really Be Considered a Syndrome? An Insight Based on 16p11.2 Microduplication. Cell Biochem Funct 2024; 42:e4121. [PMID: 39264303 DOI: 10.1002/cbf.4121] [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: 06/04/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
What is the definition of Syndrome? Since the beginning of studies in genetics, certain terminologies have been created and used to define groups of diseases or alterations. With the advancement of knowledge and the emergence of new technologies, the use of basic concepts is being done in a mistaken or often confusing way. Because of this, revisiting and readjusting the old terms becomes imminent. Here, we explore these concepts and their use, through a literature compilation of an already well-defined genetic alteration (16q11.2 microduplication). We bring comparisons in clinical and molecular scope of the alteration itself and its diagnostic methods, to improve the report of cases, rescuing terminologies and their applicability nowadays.
Collapse
Affiliation(s)
- Rafaella Mergener
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Lívia Polisseni Cotta Nascimento
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Kalise Böttcher
- Undergraduate Program in Biomedical Science, UFCSPA, Porto Alegre, Rio Grande do Sul, Brazil
| | - Marcela Rodrigues Nunes
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- Medical Genetics Resident, UFCSPA/Irmandade da Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Paulo Ricardo Gazzola Zen
- Department of Clinical Medicine, Medical Genetics, UFCSPA/ISCMPA, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
9
|
Nussinov R, Yavuz BR, Jang H. Single cell spatial biology over developmental time can decipher pediatric brain pathologies. Neurobiol Dis 2024; 199:106597. [PMID: 38992777 DOI: 10.1016/j.nbd.2024.106597] [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: 03/27/2024] [Revised: 06/18/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024] Open
Abstract
Pediatric low grade brain tumors and neurodevelopmental disorders share proteins, signaling pathways, and networks. They also share germline mutations and an impaired prenatal differentiation origin. They may differ in the timing of the events and proliferation. We suggest that their pivotal distinct, albeit partially overlapping, outcomes relate to the cell states, which depend on their spatial location, and timing of gene expression during brain development. These attributes are crucial as the brain develops sequentially, and single-cell spatial organization influences cell state, thus function. Our underlying premise is that the root cause in neurodevelopmental disorders and pediatric tumors is impaired prenatal differentiation. Data related to pediatric brain tumors, neurodevelopmental disorders, brain cell (sub)types, locations, and timing of expression in the developing brain are scant. However, emerging single cell technologies, including transcriptomic, spatial biology, spatial high-resolution imaging performed over the brain developmental time, could be transformational in deciphering brain pathologies thereby pharmacology.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
10
|
Kundu S, Sair H, Sherr EH, Mukherjee P, Rohde GK. Discovering the gene-brain-behavior link in autism via generative machine learning. SCIENCE ADVANCES 2024; 10:eadl5307. [PMID: 38865470 PMCID: PMC11168471 DOI: 10.1126/sciadv.adl5307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/08/2024] [Indexed: 06/14/2024]
Abstract
Autism is traditionally diagnosed behaviorally but has a strong genetic basis. A genetics-first approach could transform understanding and treatment of autism. However, isolating the gene-brain-behavior relationship from confounding sources of variability is a challenge. We demonstrate a novel technique, 3D transport-based morphometry (TBM), to extract the structural brain changes linked to genetic copy number variation (CNV) at the 16p11.2 region. We identified two distinct endophenotypes. In data from the Simons Variation in Individuals Project, detection of these endophenotypes enabled 89 to 95% test accuracy in predicting 16p11.2 CNV from brain images alone. Then, TBM enabled direct visualization of the endophenotypes driving accurate prediction, revealing dose-dependent brain changes among deletion and duplication carriers. These endophenotypes are sensitive to articulation disorders and explain a portion of the intelligence quotient variability. Genetic stratification combined with TBM could reveal new brain endophenotypes in many neurodevelopmental disorders, accelerating precision medicine, and understanding of human neurodiversity.
Collapse
Affiliation(s)
- Shinjini Kundu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Haris Sair
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Elliott H. Sherr
- Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Pratik Mukherjee
- Department of Radiology, University of California San Francisco, San Francisco, USA
| | - Gustavo K. Rohde
- Department of Biomedical Engineering, University of Virginia, Charlottesville, USA
- Department of Electrical and Computer Engineering, University of Virginia, Charlottesville, USA
| |
Collapse
|
11
|
Leone R, Zuglian C, Brambilla R, Morella I. Understanding copy number variations through their genes: a molecular view on 16p11.2 deletion and duplication syndromes. Front Pharmacol 2024; 15:1407865. [PMID: 38948459 PMCID: PMC11211608 DOI: 10.3389/fphar.2024.1407865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/16/2024] [Indexed: 07/02/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) include a broad spectrum of pathological conditions that affect >4% of children worldwide, share common features and present a variegated genetic origin. They include clinically defined diseases, such as autism spectrum disorders (ASD), attention-deficit/hyperactivity disorder (ADHD), motor disorders such as Tics and Tourette's syndromes, but also much more heterogeneous conditions like intellectual disability (ID) and epilepsy. Schizophrenia (SCZ) has also recently been proposed to belong to NDDs. Relatively common causes of NDDs are copy number variations (CNVs), characterised by the gain or the loss of a portion of a chromosome. In this review, we focus on deletions and duplications at the 16p11.2 chromosomal region, associated with NDDs, ID, ASD but also epilepsy and SCZ. Some of the core phenotypes presented by human carriers could be recapitulated in animal and cellular models, which also highlighted prominent neurophysiological and signalling alterations underpinning 16p11.2 CNVs-associated phenotypes. In this review, we also provide an overview of the genes within the 16p11.2 locus, including those with partially known or unknown function as well as non-coding RNAs. A particularly interesting interplay was observed between MVP and MAPK3 in modulating some of the pathological phenotypes associated with the 16p11.2 deletion. Elucidating their role in intracellular signalling and their functional links will be a key step to devise novel therapeutic strategies for 16p11.2 CNVs-related syndromes.
Collapse
Affiliation(s)
- Roberta Leone
- Università di Pavia, Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Pavia, Italy
| | - Cecilia Zuglian
- Università di Pavia, Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Pavia, Italy
| | - Riccardo Brambilla
- Università di Pavia, Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Pavia, Italy
- Cardiff University, School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff, United Kingdom
| | - Ilaria Morella
- Cardiff University, School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff, United Kingdom
| |
Collapse
|
12
|
Kim J, Vanrobaeys Y, Kelvington B, Peterson Z, Baldwin E, Gaine ME, Nickl-Jockschat T, Abel T. Dissecting 16p11.2 hemi-deletion to study sex-specific striatal phenotypes of neurodevelopmental disorders. Mol Psychiatry 2024; 29:1310-1321. [PMID: 38278994 PMCID: PMC11189748 DOI: 10.1038/s41380-024-02411-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/28/2024]
Abstract
Neurodevelopmental disorders (NDDs) are polygenic in nature and copy number variants (CNVs) are ideal candidates to study the nature of this polygenic risk. The disruption of striatal circuits is considered a central mechanism in NDDs. The 16p11.2 hemi-deletion (16p11.2 del/+) is one of the most common CNVs associated with NDD, and 16p11.2 del/+ mice show sex-specific striatum-related behavioral phenotypes. However, the critical genes among the 27 genes in the 16p11.2 region that underlie these phenotypes remain unknown. Previously, we applied a novel strategy to identify candidate genes associated with the sex-specific phenotypes of 16p11.2 del/+ mice and highlighted three genes within the deleted region: thousand and one amino acid protein kinase 2 (Taok2), seizure-related 6 homolog-like 2 (Sez6l2), and major vault protein (Mvp). Using CRISPR/Cas9, we generated mice carrying null mutations in Taok2, Sez6l2, and Mvp (3 gene hemi-deletion (3g del/+)). Hemi-deletion of these 3 genes recapitulates sex-specific behavioral alterations in striatum-dependent behavioral tasks observed in 16p11.2 del/+ mice, specifically male-specific hyperactivity and impaired motivation for reward seeking. Moreover, RNAseq analysis revealed that 3g del/+ mice exhibit gene expression changes in the striatum similar to 16p11.2 del/+ mice exclusively in males. Subsequent analysis identified translation dysregulation and/or extracellular signal-regulated kinase signaling as plausible molecular mechanisms underlying male-specific, striatum-dependent behavioral alterations. Interestingly, ribosomal profiling supported the notion of translation dysregulation in both 3g del/+ and 16p11.2 del/+ male mice. However, mice carrying a 4-gene deletion (with an additional deletion of Mapk3) exhibited fewer phenotypic similarities with 16p11.2 del/+ mice. Together, the mutation of 3 genes within the 16p11.2 region phenocopies striatal sex-specific phenotypes of 16p11.2 del/+ mice. These results support the importance of a polygenic approach to study NDDs and underscore that the effects of the large genetic deletions result from complex interactions between multiple candidate genes.
Collapse
Affiliation(s)
- Jaekyoon Kim
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
| | - Yann Vanrobaeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa, IA, USA
| | - Benjamin Kelvington
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
| | - Zeru Peterson
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA, USA
| | - Emily Baldwin
- The Iowa Medical Scientist Training Program, University of Iowa, Iowa, IA, USA
| | - Marie E Gaine
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa, IA, USA
| | - Thomas Nickl-Jockschat
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA.
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA.
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
| |
Collapse
|
13
|
Deng X, Lu S, Li Y, Fang X, Zhang R, Shen X, Du J, Xie S. Association between increased BMI and cognitive function in first-episode drug-naïve male schizophrenia. Front Psychiatry 2024; 15:1362674. [PMID: 38505798 PMCID: PMC10948420 DOI: 10.3389/fpsyt.2024.1362674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Objective Although the adverse effects of obesity in schizophrenia are documented, there is limited research exists on the implications for untreated initial schizophrenia. Our investigation aimed to explore the connections between BMI and cognitive function in first-episode drug-naïve (FEDN)schizophrenia. Methods We enrolled 143 FEDN schizophrenia patients, and collected data on their body mass index, fasting blood glucose and lipid levels. Cognitive function was measured with the MATRICS Consensus Cognitive Battery (MCCB). Using correlation and regression analysis to assess the relationship between BMI and cognitive performance. Results The prevalence rate of overweight plus obesity in FEDN schizophrenia patients was 33.57%. Patients with FEDN schizophrenia exhibited extensive cognitive impairment, and those who were overweight/obesity demonstrated more severe impairments in working memory and visual learning when compared to normal/under weight counterparts. Correlation analysis indicated a negative association between working memory and BMI and TG, as well as a link between visual learning and BMI and LDL-C. Multiple linear regression analysis revealed that a higher BMI predicted a decrease in working memory in FEDN schizophrenia patients. Conclusion Our results indicate that the rate of overweight plus obesity is high in FEDN schizophrenia patients, and there is an association between BMI and cognitive function in schizophrenia, particularly in relation to working memory.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinglun Du
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Shiping Xie
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Kim U, Jung YM, Oh S, Bae JH, Lee J, Park CW, Park JS, Jun JK, Lee SM. Chromosomal Microarray Analysis in Fetuses With Ultrasonographic Soft Markers: A Meta-Analysis of the Current Evidence. J Korean Med Sci 2024; 39:e70. [PMID: 38442716 PMCID: PMC10911939 DOI: 10.3346/jkms.2024.39.e70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/17/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Ultrasonographic soft markers are normal variants, rather than fetal abnormalities, and guidelines recommend a detailed survey of fetal anatomy to determine the necessity of antenatal karyotyping. Anecdotal reports have described cases with ultrasonographic soft markers in which chromosomal microarray analysis (CMA) revealed pathogenic copy number variants (CNVs) despite normal results on conventional karyotyping, but CMA for ultrasonographic soft markers remains a matter of debate. In this systematic review, we evaluated the clinical significance of CMA for pregnancies with isolated ultrasonographic soft markers and a normal fetal karyotype. METHODS An electronic search was conducted by an experienced librarian through the MEDLINE, Embase, and Cochrane CENTRAL databases. We reviewed 3,338 articles (3,325 identified by database searching and 13 by a hand search) about isolated ultrasonographic soft markers, and seven ultrasonographic markers (choroid plexus cysts, echogenic bowel, echogenic intracardiac focus, hypoplastic nasal bone, short femur [SF], single umbilical artery, and urinary tract dilatation) were included for this study. RESULTS Seven eligible articles were included in the final review. Pathogenic or likely pathogenic CNVs were found in fetuses with isolated ultrasonographic soft markers and a normal karyotype. The overall prevalence of pathogenic or likely pathogenic CNVs was 2.0% (41 of 2,048). The diagnostic yield of CMA was highest in fetuses with isolated SF (9 of 225, 3.9%). CONCLUSION CMA could aid in risk assessment and pregnancy counseling in pregnancies where the fetus has isolated ultrasonographic soft markers along with a normal karyotype.
Collapse
Affiliation(s)
- Uisuk Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Young Mi Jung
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
- Department of Obstetrics and Gynecology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Sohee Oh
- Department of Biostatistics, Seoul Metropolitan Government-Seoul National University Hospital Boramae Medical Center, Seoul, Korea
| | - Ji Hye Bae
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Jeesun Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Chan-Wook Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Joong Shin Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Jong Kwan Jun
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Mi Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
Maillard AM, Romascano D, Villalón-Reina JE, Moreau CA, Almeida Osório JM, Richetin S, Junod V, Yu P, Misic B, Thompson PM, Fornari E, Gygax MJ, Jacquemont S, Chabane N, Rodríguez-Herreros B. Pervasive alterations of intra-axonal volume and network organization in young children with a 16p11.2 deletion. Transl Psychiatry 2024; 14:95. [PMID: 38355713 PMCID: PMC10866898 DOI: 10.1038/s41398-024-02810-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/04/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
Reciprocal Copy Number Variants (CNVs) at the 16p11.2 locus confer high risk for autism spectrum disorder (ASD) and other neurodevelopmental disorders (NDDs). Morphometric MRI studies have revealed large and pervasive volumetric alterations in carriers of a 16p11.2 deletion. However, the specific neuroanatomical mechanisms underlying such alterations, as well as their developmental trajectory, are still poorly understood. Here we explored differences in microstructural brain connectivity between 24 children carrying a 16p11.2 deletion and 66 typically developing (TD) children between 2 and 8 years of age. We found a large pervasive increase of intra-axonal volume widespread over a high number of white matter tracts. Such microstructural alterations in 16p11.2 deletion children were already present at an early age, and led to significant changes in the global efficiency and integration of brain networks mainly associated to language, motricity and socio-emotional behavior, although the widespread pattern made it unlikely to represent direct functional correlates. Our results shed light on the neuroanatomical basis of the previously reported increase of white matter volume, and align well with analogous evidence of altered axonal diameter and synaptic function in 16p11.2 mice models. We provide evidence of a prevalent mechanistic deviation from typical maturation of brain structural connectivity associated with a specific biological risk to develop ASD. Future work is warranted to determine how this deviation contributes to the emergence of symptoms observed in young children diagnosed with ASD and other NDDs.
Collapse
Affiliation(s)
- Anne M Maillard
- Service des Troubles du Spectre de l'Autisme et apparentés, Département de psychiatrie, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - David Romascano
- Service des Troubles du Spectre de l'Autisme et apparentés, Département de psychiatrie, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Julio E Villalón-Reina
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California (USC), Marina del Rey, CA, USA
| | - Clara A Moreau
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California (USC), Marina del Rey, CA, USA
| | - Joana M Almeida Osório
- Service des Troubles du Spectre de l'Autisme et apparentés, Département de psychiatrie, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Sonia Richetin
- Service des Troubles du Spectre de l'Autisme et apparentés, Département de psychiatrie, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Vincent Junod
- Unité de Neurologie et neuroréhabilitation pédiatrique, Département femme-mère-enfant, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Paola Yu
- Service des Troubles du Spectre de l'Autisme et apparentés, Département de psychiatrie, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Bratislav Misic
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, Montréal, QC, H3A 2B4, Canada
- McConnell Brain Imaging Center, McGill University, Montréal, QC, H3A 2B4, Canada
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California (USC), Marina del Rey, CA, USA
| | - Eleonora Fornari
- Biomedical Imaging Center (CIBM), Department of Radiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Marine Jequier Gygax
- Service des Troubles du Spectre de l'Autisme et apparentés, Département de psychiatrie, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Sébastien Jacquemont
- Sainte Justine Hospital Research Center, Montréal, QC, Canada
- Department of Pediatrics, University of Montréal, Montreal, QC, Canada
| | - Nadia Chabane
- Service des Troubles du Spectre de l'Autisme et apparentés, Département de psychiatrie, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Borja Rodríguez-Herreros
- Service des Troubles du Spectre de l'Autisme et apparentés, Département de psychiatrie, Lausanne University Hospital (CHUV), Lausanne, Switzerland.
| |
Collapse
|
16
|
Liu L, Wang J, Liu X, Wang J, Chen L, Zhu H, Mai J, Hu T, Liu S. Prenatal prevalence and postnatal manifestations of 16p11.2 deletions: A new insights into neurodevelopmental disorders based on clinical investigations combined with multi-omics analysis. Clin Chim Acta 2024; 552:117671. [PMID: 37984529 DOI: 10.1016/j.cca.2023.117671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/17/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND The 16p11.2 deletion is one of the most common genetic aetiologies of neurodevelopmental disorders (NDDs). The prenatal phenotype of 16p11.2 deletion and the potential mechanism associated with postnatal clinical manifestations were largely unknow. We revealed the developmental trajectories of 16p11.2 deletion from the prenatal to postnatal periods and to identify key signaling pathways and candidate genes contributing to neurodevelopmental abnormalities. METHODS In this 5-y retrospective cohort study, women with singleton pregnancies who underwent amniocentesis for chromosomal abnormalities were included. Test of copy-number variations (CNVs) involved single nucleotide polymorphism-array and CNV-seq was performed to detected 16p11.2 deletion. For infants born carrying the 16p11.2 deletion, neurological and intellectual evaluations using the Chinese version of the Gesell Development Scale. For patients observed to have vertebral malformations, Sanger sequencing for T-C-A haplotype of TBX6 was performed. For those infants with clinical manifestations, whole-exome sequencing was consecutively performed in trios to rule out single-gene diseases, and transcriptomics combined with untargeted metabolomics were performed. RESULTS The prevalence of 16p11.2 deletion was 0.063% (55/86,035) in the prenatal period. Up to 80% (20/25) of the 16p11.2 deletions were proven de novo by parental confirmation. Approximately half of 16p11.2 deletions (28/55) were detected with prenatal abnormal ultrasound findings. Vertebral malformations were identified as the most distinctive structural malformations and were enriched in fetuses with 16p11.2 deletions compared with controls (90.9‰ [5/55] vs. 8.4‰ [72/85,980]; P < 0.001). All 5 fetuses with vertebral malformations were confirmed to have the TBX6 haplotype of T-C-A. Overall, 47.6% (10/21) infants birthed were diagnosed with NDDs of different degrees. Language impairment was the predominant manifestation (7/10; 70.0%), followed by motor delay (5/10; 50%). Multi-omics analysis indicated that MAPK3 was the central hub of the differentially expressed gene (DEG) network. We firstly reported that histidine-associated metabolism may be the core metabolic pathway related to the 16p11.2 deletion. CONCLUSION We demonstrated the prenatal presentation, incomplete penetrance and variable expressivity of the 16p11.2 deletion. We identified vertebral malformations were the most distinctive prenatal phenotypes, and language impairment was the predominant postnatal manifestation. Most of the 16p11.2 deletion was de novo. Meanwhile, we suggested that MAPK3 and histidine-associated metabolism may contribute to neurodevelopmental abnormalities of 16p11.2 deletion.
Collapse
Affiliation(s)
- Lan Liu
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Medical College, Tibet University, Lhasa, Tibet 850000, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China
| | - Jiamin Wang
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xijing Liu
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Wang
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Chen
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongmei Zhu
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingqun Mai
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ting Hu
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Shanling Liu
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
17
|
Rusu A, Chevalier C, de Chaumont F, Nalesso V, Brault V, Hérault Y, Ey E. Day-to-day spontaneous social behaviours is quantitatively and qualitatively affected in a 16p11.2 deletion mouse model. Front Behav Neurosci 2023; 17:1294558. [PMID: 38173978 PMCID: PMC10763239 DOI: 10.3389/fnbeh.2023.1294558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Background Autism spectrum disorders affect more than 1% of the population, impairing social communication and increasing stereotyped behaviours. A micro-deletion of the 16p11.2 BP4-BP5 chromosomic region has been identified in 1% of patients also displaying intellectual disabilities. In mouse models generated to understand the mechanisms of this deletion, learning and memory deficits were pervasive in most genetic backgrounds, while social communication deficits were only detected in some models. Methods To complement previous studies, we itemized the social deficits in the mouse model of 16p11.2 deletion on a hybrid C57BL/6N × C3H.Pde6b+ genetic background. We examined whether behavioural deficits were visible over long-term observation periods lasting several days and nights, to parallel everyday-life assessment of patients. We recorded the individual and social behaviours of mice carrying a heterozygous deletion of the homologous 16p11.2 chromosomic region (hereafter Del/+) and their wild-type littermates from both sexes over two or three consecutive nights during social interactions of familiar mixed-genotype quartets of males and of females, and of same-genotype unfamiliar female pairs. Results We observed that Del/+ mice of both sexes increased significantly their locomotor activity compared to wild-type littermates. In the social domain, Del/+ mice of both sexes displayed widespread deficits, even more so in males than in females in quartets of familiar individuals. In pairs, significant perturbations of the organisation of the social communication and behaviours appeared in Del/+ females. Discussion Altogether, this suggests that, over long recording periods, the phenotype of the 16p11.2 Del/+ mice was differently affected in the locomotor activity and the social domains and between the two sexes. These findings confirm the importance of testing models in long-term conditions to provide a comprehensive view of their phenotype that will refine the study of cellular and molecular mechanisms and complement pre-clinical targeted therapeutic trials.
Collapse
Affiliation(s)
- Anna Rusu
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire‑UMR 7104-UMR-S 1258, Illkirch, France
| | - Claire Chevalier
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire‑UMR 7104-UMR-S 1258, Illkirch, France
| | - Fabrice de Chaumont
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Valérie Nalesso
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire‑UMR 7104-UMR-S 1258, Illkirch, France
| | - Véronique Brault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire‑UMR 7104-UMR-S 1258, Illkirch, France
| | - Yann Hérault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire‑UMR 7104-UMR-S 1258, Illkirch, France
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Elodie Ey
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire‑UMR 7104-UMR-S 1258, Illkirch, France
| |
Collapse
|
18
|
Kretz PF, Wagner C, Mikhaleva A, Montillot C, Hugel S, Morella I, Kannan M, Fischer MC, Milhau M, Yalcin I, Brambilla R, Selloum M, Herault Y, Reymond A, Collins SC, Yalcin B. Dissecting the autism-associated 16p11.2 locus identifies multiple drivers in neuroanatomical phenotypes and unveils a male-specific role for the major vault protein. Genome Biol 2023; 24:261. [PMID: 37968726 PMCID: PMC10647150 DOI: 10.1186/s13059-023-03092-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 10/18/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Using mouse genetic studies and systematic assessments of brain neuroanatomical phenotypes, we set out to identify which of the 30 genes causes brain defects at the autism-associated 16p11.2 locus. RESULTS We show that multiple genes mapping to this region interact to regulate brain anatomy, with female mice exhibiting far fewer brain neuroanatomical phenotypes. In male mice, among the 13 genes associated with neuroanatomical defects (Mvp, Ppp4c, Zg16, Taok2, Slx1b, Maz, Fam57b, Bola2, Tbx6, Qprt, Spn, Hirip3, and Doc2a), Mvp is the top driver implicated in phenotypes pertaining to brain, cortex, hippocampus, ventricles, and corpus callosum sizes. The major vault protein (MVP), the main component of the vault organelle, is a conserved protein found in eukaryotic cells, yet its function is not understood. Here, we find MVP expression highly specific to the limbic system and show that Mvp regulates neuronal morphology, postnatally and specifically in males. We also recapitulate a previously reported genetic interaction and show that Mvp+/-;Mapk3+/- mice exhibit behavioral deficits, notably decreased anxiety-like traits detected in the elevated plus maze and open field paradigms. CONCLUSIONS Our study highlights multiple gene drivers in neuroanatomical phenotypes, interacting with each other through complex relationships. It also provides the first evidence for the involvement of the major vault protein in the regulation of brain size and neuroanatomy, specifically in male mice.
Collapse
Affiliation(s)
- Perrine F Kretz
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
| | - Christel Wagner
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
| | - Anna Mikhaleva
- Center for Integrative Genomics, University of Lausanne, CH-1015, Lausanne, Switzerland
| | | | - Sylvain Hugel
- Institute of Cellular and Integrative neuroscience, CNRS, UPR321267000, Strasbourg, France
| | - Ilaria Morella
- School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Meghna Kannan
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
| | - Marie-Christine Fischer
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
| | - Maxence Milhau
- Inserm UMR1231, Université de Bourgogne, 21000, Dijon, France
| | - Ipek Yalcin
- Institute of Cellular and Integrative neuroscience, CNRS, UPR321267000, Strasbourg, France
| | - Riccardo Brambilla
- School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, CF24 4HQ, UK
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università degli Studi di Pavia, Pavia, Italy
| | - Mohammed Selloum
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
- University of Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, ICS, 67400, Illkirch, France
| | - Yann Herault
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
- University of Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, ICS, 67400, Illkirch, France
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Stephan C Collins
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
- Current address: Université de Bourgogne, Inserm UMR1231, 21000, Dijon, France
| | - Binnaz Yalcin
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France.
- Current address: Université de Bourgogne, Inserm UMR1231, 21000, Dijon, France.
| |
Collapse
|
19
|
Wang H, Makowski C, Zhang Y, Qi A, Kaufmann T, Smeland OB, Fiecas M, Yang J, Visscher PM, Chen CH. Chromosomal inversion polymorphisms shape human brain morphology. Cell Rep 2023; 42:112896. [PMID: 37505983 PMCID: PMC10508191 DOI: 10.1016/j.celrep.2023.112896] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/27/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
The impact of chromosomal inversions on human brain morphology remains underexplored. We studied 35 common inversions classified from genotypes of 33,018 adults with European ancestry. The inversions at 2p22.3, 16p11.2, and 17q21.31 reach genome-wide significance, followed by 8p23.1 and 6p21.33, in their association with cortical and subcortical morphology. The 17q21.31, 8p23.1, and 16p11.2 regions comprise the LRRC37, OR7E, and NPIP duplicated gene families. We find the 17q21.31 MAPT inversion region, known for harboring neurological risk, to be the most salient locus among common variants for shaping and patterning the cortex. Overall, we observe the inverted orientations decreasing brain size, with the exception that the 2p22.3 inversion is associated with increased subcortical volume and the 8p23.1 inversion is associated with increased motor cortex. These significant inversions are in the genomic hotspots of neuropsychiatric loci. Our findings are generalizable to 3,472 children and demonstrate inversions as essential genetic variation to understand human brain phenotypes.
Collapse
Affiliation(s)
- Hao Wang
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA 92093, USA
| | - Carolina Makowski
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA 92093, USA
| | - Yanxiao Zhang
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA; School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| | - Anna Qi
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA 92093, USA
| | - Tobias Kaufmann
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, 72076 Tübingen, Germany; Norwegian Centre for Mental Disorders Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
| | - Olav B Smeland
- Norwegian Centre for Mental Disorders Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
| | - Mark Fiecas
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN 55455, USA
| | - Jian Yang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Chi-Hua Chen
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
20
|
Nascimento LPC, Mergener R, Nunes MR, Muniz VF, Catao JR, da Silveira AKB, Dorfman LE, Graziadio C, Zen PRG. 16p11.2 Microduplication Syndrome with Increased Fluid in the Cisterna: Coincidence or Phenotype Extension? Genes (Basel) 2023; 14:1583. [PMID: 37628634 PMCID: PMC10454344 DOI: 10.3390/genes14081583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
We report the first case of a child with 16p11.2 microduplication syndrome with increased fluid in the cisterna magna seen on magnetic resonance imaging (MRI). This finding may correspond to a Blake's Pouch Cyst (BPC) or a Mega Cisterna Magna (MCM), being impossible to differentiate through image examination. The molecular duplication was diagnosed using chromosomal microarray analysis with single nucleotide polymorphism (SNP). We review the clinical and neuroimaging features in published case reports in order to observe the findings described in the literature so far and present a skull three-dimensional model to contribute to a better understanding. Despite the variable expressivity of the syndrome being well known, there is no case described in the available literature that mentions the association of 16p11.2 microduplication and the presence of BPC or MCM seen in neuroimaging exams. This finding may represent an extension of the phenotype not yet reported or may present itself as a coincidence in a child with various malformations.
Collapse
Affiliation(s)
- Lívia Polisseni Cotta Nascimento
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil; (L.P.C.N.); (R.M.); (V.F.M.)
- Medical Residency Committee, Federal University of Health Sciences of Porto Alegre (UFCSPA)/Brotherhood of the Santa Casa de Misericórdia of Porto Alegre (ISCMPA), Porto Alegre 90050-170, RS, Brazil;
| | - Rafaella Mergener
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil; (L.P.C.N.); (R.M.); (V.F.M.)
| | - Marcela Rodrigues Nunes
- Medical Residency Committee, Federal University of Health Sciences of Porto Alegre (UFCSPA)/Brotherhood of the Santa Casa de Misericórdia of Porto Alegre (ISCMPA), Porto Alegre 90050-170, RS, Brazil;
| | - Victória Feitosa Muniz
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil; (L.P.C.N.); (R.M.); (V.F.M.)
| | - Juliana Rossi Catao
- Medicine Course, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil;
| | - Ana Kalise Böttcher da Silveira
- Undergraduate Program in Biomedical Science, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil;
| | - Luiza Emy Dorfman
- Health School, University of Vale do Rio dos Sinos (Unisinos), São Leopoldo 93022-750, RS, Brazil;
| | - Carla Graziadio
- Department of Clinical Medicine, Federal University of Health Sciences of Porto Alegre (UFCSPA)/Brotherhood of the Santa Casa de Misericórdia of Porto Alegre (ISCMPA), Porto Alegre 90050-170, RS, Brazil;
| | - Paulo Ricardo Gazzola Zen
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil; (L.P.C.N.); (R.M.); (V.F.M.)
- Department of Clinical Medicine, Federal University of Health Sciences of Porto Alegre (UFCSPA)/Brotherhood of the Santa Casa de Misericórdia of Porto Alegre (ISCMPA), Porto Alegre 90050-170, RS, Brazil;
| |
Collapse
|
21
|
Dhanasekara CS, Ancona D, Cortes L, Hu A, Rimu AH, Robohm-Leavitt C, Payne D, Wakefield SM, Mastergeorge AM, Kahathuduwa CN. Association Between Autism Spectrum Disorders and Cardiometabolic Diseases: A Systematic Review and Meta-analysis. JAMA Pediatr 2023; 177:248-257. [PMID: 36716018 PMCID: PMC9887535 DOI: 10.1001/jamapediatrics.2022.5629] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/11/2022] [Indexed: 01/31/2023]
Abstract
Importance Although the increased risk of obesity among individuals with autism has been well established, evidence on the association between autism, cardiometabolic disorders, and obesity remains inconclusive. Objective To examine the association between autism spectrum disorders and cardiometabolic diseases in a systematic review and meta-analysis. Data Sources PubMed, Scopus, Web of Science, ProQuest, Embase, and Ovid databases were searched from inception through July 31, 2022, without restrictions on date of publication or language. Study Selection Observational or baseline data of interventional studies reporting the prevalence of cardiometabolic risk factors (ie, diabetes, hypertension, dyslipidemia, atherosclerotic macrovascular disease) among children and/or adults with autism and matched with participants without autism were included. Data Extraction and Synthesis Screening, data extraction, and quality assessment were performed independently by at least 2 researchers. DerSimonian-Laird random-effects meta-analyses were performed using the meta package in R. Main Outcomes and Measures Relative risks (RRs) of diabetes, hypertension, dyslipidemia, and atherosclerotic macrovascular disease among individuals with autism were the primary outcomes. Secondary outcomes included the RR of type 1 and type 2 diabetes, heart disease, stroke, and peripheral vascular disease. Results A total of 34 studies were evaluated and included 276 173 participants with autism and 7 733 306 participants without autism (mean [range] age, 31.2 [3.8-72.8] years; pooled proportion [range] of female individuals, 47% [0-66%]). Autism was associated with greater risks of developing diabetes overall (RR, 1.57; 95% CI, 1.23-2.01; 20 studies), type 1 diabetes (RR, 1.64; 95% CI, 1.06-2.54; 6 studies), and type 2 diabetes (RR, 2.47; 95% CI, 1.30-4.70; 3 studies). Autism was also associated with increased risks of dyslipidemia (RR, 1.69; 95% CI, 1.20-2.40; 7 studies) and heart disease (RR, 1.46; 95% CI, 1.42-1.50; 3 studies). Yet, there was no significantly associated increased risk of hypertension and stroke with autism (RR, 1.22; 95% CI, 0.98-1.52; 12 studies; and RR, 1.19; 95% CI, 0.63-2.24; 4 studies, respectively). Meta-regression analyses revealed that children with autism were at a greater associated risk of developing diabetes and hypertension compared with adults. High between-study heterogeneity was a concern for several meta-analyses. Conclusions and Relevance Results suggest that the associated increased risk of cardiometabolic diseases should prompt clinicians to vigilantly monitor individuals with autism for potential contributors, signs of cardiometabolic disease, and their complications.
Collapse
Affiliation(s)
- Chathurika S. Dhanasekara
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| | - Dominic Ancona
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
| | - Leticia Cortes
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
| | - Amy Hu
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
| | - Afrina H. Rimu
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| | - Christina Robohm-Leavitt
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
| | - Drew Payne
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| | - Sarah M. Wakefield
- Department of Psychiatry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| | - Ann M. Mastergeorge
- Department of Human Development and Family Sciences, College of Human Sciences, Texas Tech University, Lubbock
| | - Chanaka N. Kahathuduwa
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
- Department of Psychiatry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
- Department of Neurology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| |
Collapse
|
22
|
Pérez-Cano L, Azidane Chenlo S, Sabido-Vera R, Sirci F, Durham L, Guney E. Translating precision medicine for autism spectrum disorder: A pressing need. Drug Discov Today 2023; 28:103486. [PMID: 36623795 DOI: 10.1016/j.drudis.2023.103486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/01/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Autism spectrum disorder (ASD) is a heterogenous group of neurodevelopmental disorders (NDDs) with a high unmet medical need. Currently, ASD is diagnosed according to behavior-based criteria that overlook clinical and genomic heterogeneity, thus repeatedly resulting in failed clinical trials. Here, we summarize the scientific evidence pointing to the pressing need to create a precision medicine framework for ASD and other NDDs. We discuss the role of omics and systems biology to characterize more homogeneous disease subtypes with different underlying pathophysiological mechanisms and to determine corresponding tailored treatments. Finally, we provide recent initiatives towards tackling the complexity in NDDs for precision medicine and cost-effective drug discovery.
Collapse
Affiliation(s)
- Laura Pérez-Cano
- Discovery and Data Science (DDS) Unit, STALICLA SL, Moll de Barcelona, s/n, Edif Este, 08039 Barcelona, Spain
| | - Sara Azidane Chenlo
- Discovery and Data Science (DDS) Unit, STALICLA SL, Moll de Barcelona, s/n, Edif Este, 08039 Barcelona, Spain
| | - Rubén Sabido-Vera
- Discovery and Data Science (DDS) Unit, STALICLA SL, Moll de Barcelona, s/n, Edif Este, 08039 Barcelona, Spain
| | - Francesco Sirci
- Discovery and Data Science (DDS) Unit, STALICLA SL, Moll de Barcelona, s/n, Edif Este, 08039 Barcelona, Spain
| | - Lynn Durham
- Discovery and Data Science (DDS) Unit, STALICLA SL, Moll de Barcelona, s/n, Edif Este, 08039 Barcelona, Spain; Drug Development Unit (DDU), STALICLA SA, Avenue de Sécheron 15, 1202 Geneva, Switzerland.
| | - Emre Guney
- Discovery and Data Science (DDS) Unit, STALICLA SL, Moll de Barcelona, s/n, Edif Este, 08039 Barcelona, Spain.
| |
Collapse
|
23
|
Abel T, Kim J, Vanrobaeys Y, Peterson Z, Kelvington B, Gaine M, Nickl-Jockschat T. Dissecting 16p11.2 hemi-deletion to study sex-specific striatal phenotypes of neurodevelopmental disorders. RESEARCH SQUARE 2023:rs.3.rs-2565823. [PMID: 36824977 PMCID: PMC9949238 DOI: 10.21203/rs.3.rs-2565823/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Neurodevelopmental disorders (NDDs) are polygenic in nature and copy number variants (CNVs) are ideal candidates to study the nature of this polygenic risk. The disruption of striatal circuits is considered a central mechanism in NDDs. The 16p11.2 hemi-deletion (16p11.2 del) is one of the most common CNVs associated with NDD, and 16p11.2 del/+ mice show sex-specific striatum-related behavioral phenotypes. However, the critical genes among the 27 genes in the 16p11.2 region that underlie these phenotypes remain unknown. Previously, we applied a novel strategy to identify candidate genes associated with the sex-specific phenotypes of 16p11.2 del/+ mice and identified 3 genes of particular importance within the deleted region: thousand and one amino acid protein kinase 2 (Taok2), seizure-related 6 homolog-like 2 (Sez6l2), and major vault protein (Mvp). Using the CRISPR/Cas9 technique, we generated 3 gene hemi-deletion (3g del/+) mice carrying null mutations in Taok2, Sez6l2, and Mvp. We assessed striatum-dependent phenotypes of these 3g del/+ mice in behavioral, molecular, and imaging studies. Hemi-deletion of Taok2, Sez6l2, and Mvp induces sex-specific behavioral alterations in striatum-dependent behavioral tasks, specifically male-specific hyperactivity and impaired motivation for reward seeking, resembling behavioral phenotypes of 16p11.2 del/+ mice. Moreover, RNAseq analysis revealed that 3g del/+ mice exhibit gene expression changes in the striatum similar to 16p11.2 del/+ mice, but only in males. Pathway analysis identified ribosomal dysfunction and translation dysregulation as molecular mechanisms underlying male-specific, striatum-dependent behavioral alterations. Together, the mutation of 3 genes within the 16p11.2 region phenocopies striatal sex-specific phenotypes of 16p11.2 del/+ mice, unlike single gene mutation studies. These results support the importance of a polygenic approach to study NDDs and our novel strategy to identify genes of interest using gene expression patterns in brain regions, such as the striatum, which are impacted in these disorders.
Collapse
|
24
|
Forrest MP, Dos Santos M, Piguel NH, Wang YZ, Hawkins NA, Bagchi VA, Dionisio LE, Yoon S, Simkin D, Martin-de-Saavedra MD, Gao R, Horan KE, George AL, LeDoux MS, Kearney JA, Savas JN, Penzes P. Rescue of neuropsychiatric phenotypes in a mouse model of 16p11.2 duplication syndrome by genetic correction of an epilepsy network hub. Nat Commun 2023; 14:825. [PMID: 36808153 PMCID: PMC9938216 DOI: 10.1038/s41467-023-36087-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 01/16/2023] [Indexed: 02/19/2023] Open
Abstract
Neuropsychiatric disorders (NPDs) are frequently co-morbid with epilepsy, but the biological basis of shared risk remains poorly understood. The 16p11.2 duplication is a copy number variant that confers risk for diverse NPDs including autism spectrum disorder, schizophrenia, intellectual disability and epilepsy. We used a mouse model of the 16p11.2 duplication (16p11.2dup/+) to uncover molecular and circuit properties associated with this broad phenotypic spectrum, and examined genes within the locus capable of phenotype reversal. Quantitative proteomics revealed alterations to synaptic networks and products of NPD risk genes. We identified an epilepsy-associated subnetwork that was dysregulated in 16p11.2dup/+ mice and altered in brain tissue from individuals with NPDs. Cortical circuits from 16p11.2dup/+ mice exhibited hypersynchronous activity and enhanced network glutamate release, which increased susceptibility to seizures. Using gene co-expression and interactome analysis, we show that PRRT2 is a major hub in the epilepsy subnetwork. Remarkably, correcting Prrt2 copy number rescued aberrant circuit properties, seizure susceptibility and social deficits in 16p11.2dup/+ mice. We show that proteomics and network biology can identify important disease hubs in multigenic disorders, and reveal mechanisms relevant to the complex symptomatology of 16p11.2 duplication carriers.
Collapse
Affiliation(s)
- Marc P Forrest
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Marc Dos Santos
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Nicolas H Piguel
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yi-Zhi Wang
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Nicole A Hawkins
- Department of Pharmacology Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Vikram A Bagchi
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Leonardo E Dionisio
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sehyoun Yoon
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Dina Simkin
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Pharmacology Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Maria Dolores Martin-de-Saavedra
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - Ruoqi Gao
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Katherine E Horan
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Alfred L George
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Pharmacology Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Mark S LeDoux
- Department of Psychology, University of Memphis, Memphis, TN, 38152, USA
- Veracity Neuroscience LLC, Memphis, TN, 38157, USA
| | - Jennifer A Kearney
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Pharmacology Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Jeffrey N Savas
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Peter Penzes
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
25
|
Hampl SE, Hassink SG, Skinner AC, Armstrong SC, Barlow SE, Bolling CF, Avila Edwards KC, Eneli I, Hamre R, Joseph MM, Lunsford D, Mendonca E, Michalsky MP, Mirza N, Ochoa ER, Sharifi M, Staiano AE, Weedn AE, Flinn SK, Lindros J, Okechukwu K. Clinical Practice Guideline for the Evaluation and Treatment of Children and Adolescents With Obesity. Pediatrics 2023; 151:e2022060640. [PMID: 36622135 DOI: 10.1542/peds.2022-060640] [Citation(s) in RCA: 419] [Impact Index Per Article: 209.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 01/10/2023] Open
|
26
|
Ruthig VA, Lamb DJ. Modeling development of genitourinary birth defects to understand disruption due to changes in gene dosage. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2022; 10:412-424. [PMID: 36636694 PMCID: PMC9831917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 12/25/2022] [Indexed: 01/14/2023]
Abstract
Genitourinary development is a delicately orchestrated process that begins in the embryo. Once complete, the genitourinary system is a collection of functionally disparate organs spread throughout the abdominal and pelvic regions. These distinct organs are interconnected through an elaborate duct system which aggregates the organs' products to a common exit point. The complicated nature of the genitourinary system makes it highly susceptible to developmental disruptions that produce anomalies. In fact, genitourinary anomalies are among the most common class of human birth defects. Aside from congenital anomalies of the kidney and urinary tract (CAKUT), for males, these birth defects can also occur in the penis (hypospadias) and testis (cryptorchism), which impact male fertility and male mental health. As genetic technology has advanced, it has become clear that a subset of cases of genitourinary birth defects are due to gene variation causing dosage changes in critical regulatory genes. Here we first review the parallels between human and mouse genitourinary development. We then demonstrate how translational research leverages mouse models of human gene variation cases to advance mechanistic understanding of causation in genitourinary birth defects. We close with a view to the future highlighting upcoming technologies that will provide a deeper understanding of gene variation affecting regulation of genitourinary development, which should ultimately advance treatment options for patients.
Collapse
Affiliation(s)
- Victor A Ruthig
- Department of Urology, Weill Cornell MedicineNew York, NY, USA
- Sexual Medicine Laboratory, Weill Cornell MedicineNew York, NY, USA
| | - Dolores J Lamb
- Department of Urology, Weill Cornell MedicineNew York, NY, USA
- Center for Reproductive Genomics, Weill Cornell MedicineNew York, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell MedicineNew York, NY, USA
| |
Collapse
|
27
|
Jiménez-Romero MS, Fernández-Urquiza M, Benítez-Burraco A. Language and Communication Deficits in Chromosome 16p11.2 Deletion Syndrome. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2022; 65:4724-4740. [PMID: 36410413 DOI: 10.1044/2022_jslhr-22-00160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
PURPOSE Chromosome 16p11.2 deletion syndrome (OMIM #611913) is a rare genetic condition resulting from the partial deletion of approximately 35 genes located at Chromosome 16. Affected people exhibit a variable clinical profile, featuring mild dysmorphisms, motor problems, developmental delay, mild intellectual disability (ID), socialization deficits and/or autism spectrum disorder (ASD) traits, and problems with language. Specifically, a precise characterization of the speech, language, and communication (dis)abilities of people with this condition is still pending. METHOD We used standardized tests and samples of naturalistic speech to provide a longitudinal profile of the speech, language, and communication problems of a boy with Chromosome 16p11.2 deletion syndrome and without ID or ASD. RESULTS The proband shows impaired expressive abilities as well as problems with receptive language, dysprosody, and ASD-like communication deficits, such as impaired interactive skills, perseverative verbal behavior, overabundance of tangential responses, and lack of metapragmatic awareness and communicative use of gaze, meeting the criteria for social pragmatic communication disorder. CONCLUSIONS Our results support the view that language and communication impairment should be regarded as one core symptom of Chromosome 16p11.2 deletion syndrome, even without a diagnosis of ASD or ID. Clinical implications of our results, with a focus on therapeutic interventions for children with 16p11.2 deletion syndrome and no ASD or ID, are also discussed. SUPPLEMENTAL MATERIAL https://doi.org/10.23641/asha.21561714.
Collapse
Affiliation(s)
| | | | - Antonio Benítez-Burraco
- Department of Spanish, Linguistics, and Theory of Literature (Linguistics), Faculty of Philology, University of Seville, Spain
| |
Collapse
|
28
|
Duński E, Pękowska A. Keeping the balance: Trade-offs between human brain evolution, autism, and schizophrenia. Front Genet 2022; 13:1009390. [DOI: 10.3389/fgene.2022.1009390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/12/2022] [Indexed: 11/22/2022] Open
Abstract
The unique qualities of the human brain are a product of a complex evolutionary process. Evolution, famously described by François Jacob as a “tinkerer,” builds upon existing genetic elements by modifying and repurposing them for new functions. Genetic changes in DNA may lead to the emergence of new genes or cause altered gene expression patterns. Both gene and regulatory element mutations may lead to new functions. Yet, this process may lead to side-effects. An evolutionary trade-off occurs when an otherwise beneficial change, which is important for evolutionary success and is under strong positive selection, concurrently results in a detrimental change in another trait. Pleiotropy occurs when a gene affects multiple traits. Antagonistic pleiotropy is a phenomenon whereby a genetic variant leads to an increase in fitness at one life-stage or in a specific environment, but simultaneously decreases fitness in another respect. Therefore, it is conceivable that the molecular underpinnings of evolution of highly complex traits, including brain size or cognitive ability, under certain conditions could result in deleterious effects, which would increase the susceptibility to psychiatric or neurodevelopmental diseases. Here, we discuss possible trade-offs and antagonistic pleiotropies between evolutionary change in a gene sequence, dosage or activity and the susceptibility of individuals to autism spectrum disorders and schizophrenia. We present current knowledge about genes and alterations in gene regulatory landscapes, which have likely played a role in establishing human-specific traits and have been implicated in those diseases.
Collapse
|
29
|
Sun H, Wu M, Wang M, Zhang X, Zhu J. The regulatory role of endoplasmic reticulum chaperone proteins in neurodevelopment. Front Neurosci 2022; 16:1032607. [DOI: 10.3389/fnins.2022.1032607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
The endoplasmic reticulum (ER) is the largest tubular reticular organelle spanning the cell. As the main site of protein synthesis, Ca2+ homeostasis maintenance and lipid metabolism, the ER plays a variety of essential roles in eukaryotic cells, with ER molecular chaperones participate in all these processes. In recent years, it has been reported that the abnormal expression of ER chaperones often leads to a variety of neurodevelopmental disorders (NDDs), including abnormal neuronal migration, neuronal morphogenesis, and synaptic function. Neuronal development is a complex and precisely regulated process. Currently, the mechanism by which neural development is regulated at the ER level remains under investigation. Therefore, in this work, we reviewed the recent advances in the roles of ER chaperones in neural development and developmental disorders caused by the deficiency of these molecular chaperones.
Collapse
|
30
|
Scharrenberg R, Richter M, Johanns O, Meka DP, Rücker T, Murtaza N, Lindenmaier Z, Ellegood J, Naumann A, Zhao B, Schwanke B, Sedlacik J, Fiehler J, Hanganu-Opatz IL, Lerch JP, Singh KK, de Anda FC. TAOK2 rescues autism-linked developmental deficits in a 16p11.2 microdeletion mouse model. Mol Psychiatry 2022; 27:4707-4721. [PMID: 36123424 PMCID: PMC9734055 DOI: 10.1038/s41380-022-01785-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 12/14/2022]
Abstract
The precise development of the neocortex is a prerequisite for higher cognitive and associative functions. Despite numerous advances that have been made in understanding neuronal differentiation and cortex development, our knowledge regarding the impact of specific genes associated with neurodevelopmental disorders on these processes is still limited. Here, we show that Taok2, which is encoded in humans within the autism spectrum disorder (ASD) susceptibility locus 16p11.2, is essential for neuronal migration. Overexpression of de novo mutations or rare variants from ASD patients disrupts neuronal migration in an isoform-specific manner. The mutated TAOK2α variants but not the TAOK2β variants impaired neuronal migration. Moreover, the TAOK2α isoform colocalizes with microtubules. Consequently, neurons lacking Taok2 have unstable microtubules with reduced levels of acetylated tubulin and phosphorylated JNK1. Mice lacking Taok2 develop gross cortical and cortex layering abnormalities. Moreover, acute Taok2 downregulation or Taok2 knockout delayed the migration of upper-layer cortical neurons in mice, and the expression of a constitutively active form of JNK1 rescued these neuronal migration defects. Finally, we report that the brains of the Taok2 KO and 16p11.2 del Het mouse models show striking anatomical similarities and that the heterozygous 16p11.2 microdeletion mouse model displayed reduced levels of phosphorylated JNK1 and neuronal migration deficits, which were ameliorated upon the introduction of TAOK2α in cortical neurons and in the developing cortex of those mice. These results delineate the critical role of TAOK2 in cortical development and its contribution to neurodevelopmental disorders, including ASD.
Collapse
Affiliation(s)
- Robin Scharrenberg
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Melanie Richter
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| | - Ole Johanns
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Durga Praveen Meka
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Tabitha Rücker
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Nadeem Murtaza
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, L8S 4A9, Canada
| | - Zsuzsa Lindenmaier
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, M5T 3H7, Canada
| | - Anne Naumann
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Bing Zhao
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Birgit Schwanke
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Jan Sedlacik
- Department of Neuroradiology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jens Fiehler
- Department of Neuroradiology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford, OX3 9DU, UK
| | - Karun K Singh
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
| | - Froylan Calderon de Anda
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
31
|
Tai DJC, Razaz P, Erdin S, Gao D, Wang J, Nuttle X, de Esch CE, Collins RL, Currall BB, O'Keefe K, Burt ND, Yadav R, Wang L, Mohajeri K, Aneichyk T, Ragavendran A, Stortchevoi A, Morini E, Ma W, Lucente D, Hastie A, Kelleher RJ, Perlis RH, Talkowski ME, Gusella JF. Tissue- and cell-type-specific molecular and functional signatures of 16p11.2 reciprocal genomic disorder across mouse brain and human neuronal models. Am J Hum Genet 2022; 109:1789-1813. [PMID: 36152629 PMCID: PMC9606388 DOI: 10.1016/j.ajhg.2022.08.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/23/2022] [Indexed: 01/29/2023] Open
Abstract
Chromosome 16p11.2 reciprocal genomic disorder, resulting from recurrent copy-number variants (CNVs), involves intellectual disability, autism spectrum disorder (ASD), and schizophrenia, but the responsible mechanisms are not known. To systemically dissect molecular effects, we performed transcriptome profiling of 350 libraries from six tissues (cortex, cerebellum, striatum, liver, brown fat, and white fat) in mouse models harboring CNVs of the syntenic 7qF3 region, as well as cellular, transcriptional, and single-cell analyses in 54 isogenic neural stem cell, induced neuron, and cerebral organoid models of CRISPR-engineered 16p11.2 CNVs. Transcriptome-wide differentially expressed genes were largely tissue-, cell-type-, and dosage-specific, although more effects were shared between deletion and duplication and across tissue than expected by chance. The broadest effects were observed in the cerebellum (2,163 differentially expressed genes), and the greatest enrichments were associated with synaptic pathways in mouse cerebellum and human induced neurons. Pathway and co-expression analyses identified energy and RNA metabolism as shared processes and enrichment for ASD-associated, loss-of-function constraint, and fragile X messenger ribonucleoprotein target gene sets. Intriguingly, reciprocal 16p11.2 dosage changes resulted in consistent decrements in neurite and electrophysiological features, and single-cell profiling of organoids showed reciprocal alterations to the proportions of excitatory and inhibitory GABAergic neurons. Changes both in neuronal ratios and in gene expression in our organoid analyses point most directly to calretinin GABAergic inhibitory neurons and the excitatory/inhibitory balance as targets of disruption that might contribute to changes in neurodevelopmental and cognitive function in 16p11.2 carriers. Collectively, our data indicate the genomic disorder involves disruption of multiple contributing biological processes and that this disruption has relative impacts that are context specific.
Collapse
Affiliation(s)
- Derek J C Tai
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Parisa Razaz
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Serkan Erdin
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Dadi Gao
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jennifer Wang
- Center for Quantitative Health, Division of Clinical Research, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Xander Nuttle
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Celine E de Esch
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ryan L Collins
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Benjamin B Currall
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kathryn O'Keefe
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nicholas D Burt
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rachita Yadav
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lily Wang
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kiana Mohajeri
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tatsiana Aneichyk
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ashok Ragavendran
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alexei Stortchevoi
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elisabetta Morini
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Weiyuan Ma
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Diane Lucente
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Raymond J Kelleher
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Roy H Perlis
- Center for Quantitative Health, Division of Clinical Research, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Michael E Talkowski
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - James F Gusella
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
32
|
Ahlberg R, Garcia-Argibay M, Hirvikoski T, Boman M, Chen Q, Taylor MJ, Frans E, Bölte S, Larsson H. Shared familial risk factors between autism spectrum disorder and obesity - a register-based familial coaggregation cohort study. J Child Psychol Psychiatry 2022; 63:890-899. [PMID: 34881437 DOI: 10.1111/jcpp.13538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Meta-analyses suggest an association between autism spectrum disorder (ASD) and obesity, but the factors underlying this association remain unclear. This study investigated the association between ASD and obesity stratified on intellectual disability (ID). In addition, in order to gain insight into possible shared etiological factors, the potential role of shared familial liability was examined. METHOD We studied a cohort of 3,141,696 individuals by linking several Swedish nationwide registers. We identified 35,461 individuals with ASD and 61,784 individuals with obesity. Logistic regression models were used to estimate the association between ASD and obesity separately by ID and sex and by adjusting for parental education, psychiatric comorbidity, and psychotropic medication. Potential shared familial etiologic factors were examined by comparing the risk of obesity in full siblings, maternal and paternal half-siblings, and full- and half-cousins of individuals with ASD to the risk of obesity in relatives of individuals without ASD. RESULTS Individuals with ASD + ID (OR = 3.76 [95% CI, 3.38-4.19]) and ASD-ID (OR = 3.40 [95% CI, 3.23-3.58]) had an increased risk for obesity compared with individuals without ASD. The associations remained statistically significant when adjusting for parental education, psychiatric comorbidity, and medication. Sex-stratified analyses indicated a higher relative risk for males compared with females, with statistically significant interaction effects for ASD-ID, but not for ASD+ID in the fully adjusted model. First-degree relatives of individuals with ASD+ID and ASD-ID had an increased risk of obesity compared with first-degree relatives of individuals without ASD. The obesity risk was similar in second-degree relatives of individuals with ASD+ID but was lower for and ASD-ID. Full cousins of individuals with ASD+ID had a higher risk compared with half-cousins of individuals with ASD+ID). A similar difference in the obesity risk between full cousins and half-cousins was observed for ASD-ID. CONCLUSIONS Individuals with ASD and their relatives are at increased risk for obesity. The risk might be somewhat higher for males than females. This warrants further studies examining potential common pleiotropic genetic factors and shared family-wide environmental factors for ASD and obesity. Such research might aid in identifying specific risks and underlying mechanisms in common between ASD and obesity.
Collapse
Affiliation(s)
- Richard Ahlberg
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | | | - Tatja Hirvikoski
- Division of Neuropsychiatry, Department of Women's and Children's Health, Center of Neurodevelopmental Disorders at Karolinska Institutet (KIND), Karolinska Institutet & Center for Psychiatry Research, Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.,Habilitation & Health, Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Marcus Boman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Qi Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mark J Taylor
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Emma Frans
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sven Bölte
- Division of Neuropsychiatry, Department of Women's and Children's Health, Center of Neurodevelopmental Disorders at Karolinska Institutet (KIND), Karolinska Institutet & Center for Psychiatry Research, Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.,Child and Adolescent Psychiatry, Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.,Curtin Autism Research Group, School of Occupational Therapy, Social Work and Speech Pathology, Curtin University, Perth, WA, Australia
| | - Henrik Larsson
- School of Medical Sciences, Örebro University, Örebro, Sweden.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
33
|
Possible association of 16p11.2 copy number variation with altered lymphocyte and neutrophil counts. NPJ Genom Med 2022; 7:38. [PMID: 35715439 PMCID: PMC9205872 DOI: 10.1038/s41525-022-00308-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 05/23/2022] [Indexed: 11/09/2022] Open
Abstract
Recurrent copy-number variations (CNVs) at chromosome 16p11.2 are associated with neurodevelopmental diseases, skeletal system abnormalities, anemia, and genitourinary defects. Among the 40 protein-coding genes encompassed within the rearrangement, some have roles in leukocyte biology and immunodeficiency, like SPN and CORO1A. We therefore investigated leukocyte differential counts and disease in 16p11.2 CNV carriers. In our clinically-recruited cohort, we identified three deletion carriers from two families (out of 32 families assessed) with neutropenia and lymphopenia. They had no deleterious single-nucleotide or indel variant in known cytopenia genes, suggesting a possible causative role of the deletion. Noticeably, all three individuals had the lowest copy number of the human-specific BOLA2 duplicon (copy-number range: 3–8). Consistent with the lymphopenia and in contrast with the neutropenia associations, adult deletion carriers from UK biobank (n = 74) showed lower lymphocyte (Padj = 0.04) and increased neutrophil (Padj = 8.31e-05) counts. Mendelian randomization studies pinpointed to reduced CORO1A, KIF22, and BOLA2-SMG1P6 expressions being causative for the lower lymphocyte counts. In conclusion, our data suggest that 16p11.2 deletion, and possibly also the lowest dosage of the BOLA2 duplicon, are associated with low lymphocyte counts. There is a trend between 16p11.2 deletion with lower copy-number of the BOLA2 duplicon and higher susceptibility to moderate neutropenia. Higher numbers of cases are warranted to confirm the association with neutropenia and to resolve the involvement of the deletion coupled with deleterious variants in other genes and/or with the structure and copy number of segments in the CNV breakpoint regions.
Collapse
|
34
|
Tomasello DL, Wlodkowic D. Noninvasive Electrophysiology: Emerging Prospects in Aquatic Neurotoxicity Testing. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:4788-4794. [PMID: 35196004 DOI: 10.1021/acs.est.1c08471] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The significance of neurotoxicological risks associated with anthropogenic pollution is gaining increasing recognition worldwide. In this regard, perturbations in behavioral traits upon exposure to environmentally relevant concentrations of neurotoxic and neuro-modulating contaminants have been linked to diminished ecological fitness of many aquatic species. Despite an increasing interest in behavioral testing in aquatic ecotoxicology there is, however, a notable gap in understanding of the neurophysiological foundations responsible for the altered behavioral phenotypes. One of the canonical approaches to explain the mechanisms of neuro-behavioral changes is functional analysis of neuronal transmission. In aquatic animals it requires, however, invasive, complex, and time-consuming electrophysiology techniques. In this perspective, we highlight emerging prospects of noninvasive, in situ electrophysiology based on multielectrode arrays (MEAs). This technology has only recently been pioneered for the detection and analysis of transient electrical signals in the central nervous system of small model organisms such as zebrafish. The analysis resembles electroencephalography (EEG) applications and provides an appealing strategy for mechanistic explorative studies as well as routine neurotoxicity risk assessment. We outline the prospective future applications and existing challenges of this emerging analytical strategy that is poised to bring new vistas for aquatic ecotoxicology such as greater mechanistic understanding of eco-neurotoxicity and thus more robust risk assessment protocols.
Collapse
Affiliation(s)
- Danielle L Tomasello
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, United States
| | - Donald Wlodkowic
- The Neurotox Lab, School of Science, RMIT University, Melbourne, Victoria 3083, Australia
| |
Collapse
|
35
|
Madeo SF, Stanghellini I, Predieri B, Ciancia S, Leo F, Bruzzi P, Calabrese O, Iughetti L. Copy Number Variation Analysis Increases the Number of Candidate Loci Associated with Pediatric Obesity. Horm Res Paediatr 2022; 94:251-262. [PMID: 34482307 DOI: 10.1159/000519299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/28/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Obesity is a multifactorial disease caused by the interaction of genetic, environmental, and behavioral factors. Currently, only a small number of obese children undergo genetic analysis, usually when obesity is associated with dysmorphic features. The aim of this study was to identify genomic rearrangement causing obesity. METHODS We analyzed the DNA of children and adolescents by single-nucleotide polymorphism-array (platform CytoScan HD, Affymetrix). Patients included in this study were obese with dysmorphic features and/or intellectual disabilities and/or neuropsychomotor signs. RESULTS Ninety-four children and adolescents with obesity (9.25 ± 4.04 years old, 60 males) were enrolled in the study. Dysmorphic features were found in 64 out of 94 subjects (68.1%), intellectual disability was found in 23 subjects (24.5%), and other neuropsychomotor signs in 31 (32.9%). Copy number variations (CNVs) were identified in 43 out of 94 patients (45.7%): among these 14 subjects showed at least 1 deletion, 22 duplication, whereas 7 patients showed both deletion and duplication. In 20 subjects (13 males), CNVs were linked or possibly related with obesity; in 23 subjects, this correlation cannot be inferred. CONCLUSION A genetic origin of obesity was detected in about half of our obese children and adolescents with associated dysmorphic features and/or intellectual disability and/or neuropsychomotor signs. In these children, array-CGH analysis can be useful to identify causative genetic mutations, with consequent advantage in therapeutic management and follow-up of these patients.
Collapse
Affiliation(s)
- Simona Filomena Madeo
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Ilaria Stanghellini
- Medical Genetics Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Barbara Predieri
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Ciancia
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Leo
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Patrizia Bruzzi
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Olga Calabrese
- Medical Genetics Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Lorenzo Iughetti
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
36
|
Zambrano-Zaragoza JF, Vázquez-Reyes A, Durán-Avelar MDJ, Gutiérrez-Franco J, Vibanco-Pérez N, Agraz-Cibrián JM, Pérez-Cambero H, Ayón-Pérez MF. Deleted genes associated with obesity in Mexican patients diagnosed with nonalcoholic fatty liver disease. Ann Hum Genet 2022; 86:237-244. [PMID: 35343586 DOI: 10.1111/ahg.12466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/18/2022] [Accepted: 03/10/2022] [Indexed: 11/29/2022]
Abstract
AIM Nonalcoholic fatty liver disease (NAFLD) is a complex metabolic condition in which both lifestyle and genetic factors have a pathogenic role. The LEP gene encodes leptin, which regulates appetite, body weight, and several metabolic functions. Proopiomelanocortin (POMC), regulates food intake and energy balance. The aim of the study was to determine partial or complete deletions of genes associated with obesity in patients diagnosed with NAFLD. MATERIAL AND METHODS Blood samples and DNA from 43 individuals diagnosed with NAFLD by ultrasonographic technique (Fibroscan) were obtained. The partial or complete deletions of genes were determined by MLPA (Multiplex Ligation-dependent Probe Amplification) using the SALSA probemix P220-B2 Obesity only on 43 individuals. Fifty blood samples from healthy individuals were included. RESULTS Eleven out of 43 individuals analyzed by MLPA presented some deletion of the genes analyzed: six were female and five were male. The partial or complete deletion of the LEPR and POMC genes was observed in eight patients (18.6%), SIM1 in six patients (13.9%), GRIK2 and SH2B1 in two patients (4.7%), SEZGL2 in four patients (9.3%), and MCR4 in one patient (2.3%). CONCLUSION Partial deletion was observed in LEPR, POMC, SIM1, GRIK2, SH2B1, SEZGL2, and MCR4 genes in 26% of the cases, and we suggest that these alterations probably has a potential relationship for the development of NAFLD.
Collapse
Affiliation(s)
- José Francisco Zambrano-Zaragoza
- Laboratorio de Inmunología. Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Alejandro Vázquez-Reyes
- Laboratorios de Investigación en Biología Molecular e Inmunología. Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Ma de Jesús Durán-Avelar
- Laboratorios de Investigación en Biología Molecular e Inmunología. Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Jorge Gutiérrez-Franco
- Laboratorio de Inmunología. Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Norberto Vibanco-Pérez
- Laboratorios de Investigación en Biología Molecular e Inmunología. Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Juan Manuel Agraz-Cibrián
- Laboratorio de Inmunología. Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Horacio Pérez-Cambero
- Comisión Estatal para la Protección Contra Riesgo Sanitario de Nayarit, Servicios de Salud de Nayarit, Tepic, Nayarit, México
| | - Miriam Fabiola Ayón-Pérez
- Laboratorios de Investigación en Biología Molecular e Inmunología. Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| |
Collapse
|
37
|
Tomasello DL, Kim JL, Khodour Y, McCammon JM, Mitalipova M, Jaenisch R, Futerman AH, Sive H. 16pdel lipid changes in iPSC-derived neurons and function of FAM57B in lipid metabolism and synaptogenesis. iScience 2022; 25:103551. [PMID: 34984324 PMCID: PMC8693007 DOI: 10.1016/j.isci.2021.103551] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/23/2021] [Accepted: 11/26/2021] [Indexed: 01/01/2023] Open
Abstract
The complex 16p11.2 deletion syndrome (16pdel) is accompanied by neurological disorders, including epilepsy, autism spectrum disorder, and intellectual disability. We demonstrated that 16pdel iPSC differentiated neurons from affected people show augmented local field potential activity and altered ceramide-related lipid species relative to unaffected. FAM57B, a poorly characterized gene in the 16p11.2 interval, has emerged as a candidate tied to symptomatology. We found that FAM57B modulates ceramide synthase (CerS) activity, but is not a CerS per se. In FAM57B mutant human neuronal cells and zebrafish brain, composition and levels of sphingolipids and glycerolipids associated with cellular membranes are disrupted. Consistently, we observed aberrant plasma membrane architecture and synaptic protein mislocalization, which were accompanied by depressed brain and behavioral activity. Together, these results suggest that haploinsufficiency of FAM57B contributes to changes in neuronal activity and function in 16pdel syndrome through a crucial role for the gene in lipid metabolism.
Collapse
Affiliation(s)
| | - Jiyoon L. Kim
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yara Khodour
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Maya Mitalipova
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anthony H. Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hazel Sive
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
38
|
Sønderby IE, Ching CRK, Thomopoulos SI, van der Meer D, Sun D, Villalon‐Reina JE, Agartz I, Amunts K, Arango C, Armstrong NJ, Ayesa‐Arriola R, Bakker G, Bassett AS, Boomsma DI, Bülow R, Butcher NJ, Calhoun VD, Caspers S, Chow EWC, Cichon S, Ciufolini S, Craig MC, Crespo‐Facorro B, Cunningham AC, Dale AM, Dazzan P, de Zubicaray GI, Djurovic S, Doherty JL, Donohoe G, Draganski B, Durdle CA, Ehrlich S, Emanuel BS, Espeseth T, Fisher SE, Ge T, Glahn DC, Grabe HJ, Gur RE, Gutman BA, Haavik J, Håberg AK, Hansen LA, Hashimoto R, Hibar DP, Holmes AJ, Hottenga J, Hulshoff Pol HE, Jalbrzikowski M, Knowles EEM, Kushan L, Linden DEJ, Liu J, Lundervold AJ, Martin‐Brevet S, Martínez K, Mather KA, Mathias SR, McDonald‐McGinn DM, McRae AF, Medland SE, Moberget T, Modenato C, Monereo Sánchez J, Moreau CA, Mühleisen TW, Paus T, Pausova Z, Prieto C, Ragothaman A, Reinbold CS, Reis Marques T, Repetto GM, Reymond A, Roalf DR, Rodriguez‐Herreros B, Rucker JJ, Sachdev PS, Schmitt JE, Schofield PR, Silva AI, Stefansson H, Stein DJ, Tamnes CK, Tordesillas‐Gutiérrez D, Ulfarsson MO, Vajdi A, van 't Ent D, van den Bree MBM, Vassos E, Vázquez‐Bourgon J, Vila‐Rodriguez F, Walters GB, Wen W, Westlye LT, Wittfeld K, Zackai EH, Stefánsson K, Jacquemont S, et alSønderby IE, Ching CRK, Thomopoulos SI, van der Meer D, Sun D, Villalon‐Reina JE, Agartz I, Amunts K, Arango C, Armstrong NJ, Ayesa‐Arriola R, Bakker G, Bassett AS, Boomsma DI, Bülow R, Butcher NJ, Calhoun VD, Caspers S, Chow EWC, Cichon S, Ciufolini S, Craig MC, Crespo‐Facorro B, Cunningham AC, Dale AM, Dazzan P, de Zubicaray GI, Djurovic S, Doherty JL, Donohoe G, Draganski B, Durdle CA, Ehrlich S, Emanuel BS, Espeseth T, Fisher SE, Ge T, Glahn DC, Grabe HJ, Gur RE, Gutman BA, Haavik J, Håberg AK, Hansen LA, Hashimoto R, Hibar DP, Holmes AJ, Hottenga J, Hulshoff Pol HE, Jalbrzikowski M, Knowles EEM, Kushan L, Linden DEJ, Liu J, Lundervold AJ, Martin‐Brevet S, Martínez K, Mather KA, Mathias SR, McDonald‐McGinn DM, McRae AF, Medland SE, Moberget T, Modenato C, Monereo Sánchez J, Moreau CA, Mühleisen TW, Paus T, Pausova Z, Prieto C, Ragothaman A, Reinbold CS, Reis Marques T, Repetto GM, Reymond A, Roalf DR, Rodriguez‐Herreros B, Rucker JJ, Sachdev PS, Schmitt JE, Schofield PR, Silva AI, Stefansson H, Stein DJ, Tamnes CK, Tordesillas‐Gutiérrez D, Ulfarsson MO, Vajdi A, van 't Ent D, van den Bree MBM, Vassos E, Vázquez‐Bourgon J, Vila‐Rodriguez F, Walters GB, Wen W, Westlye LT, Wittfeld K, Zackai EH, Stefánsson K, Jacquemont S, Thompson PM, Bearden CE, Andreassen OA. Effects of copy number variations on brain structure and risk for psychiatric illness: Large-scale studies from the ENIGMA working groups on CNVs. Hum Brain Mapp 2022; 43:300-328. [PMID: 33615640 PMCID: PMC8675420 DOI: 10.1002/hbm.25354] [Show More Authors] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/07/2021] [Accepted: 01/13/2021] [Indexed: 01/21/2023] Open
Abstract
The Enhancing NeuroImaging Genetics through Meta-Analysis copy number variant (ENIGMA-CNV) and 22q11.2 Deletion Syndrome Working Groups (22q-ENIGMA WGs) were created to gain insight into the involvement of genetic factors in human brain development and related cognitive, psychiatric and behavioral manifestations. To that end, the ENIGMA-CNV WG has collated CNV and magnetic resonance imaging (MRI) data from ~49,000 individuals across 38 global research sites, yielding one of the largest studies to date on the effects of CNVs on brain structures in the general population. The 22q-ENIGMA WG includes 12 international research centers that assessed over 533 individuals with a confirmed 22q11.2 deletion syndrome, 40 with 22q11.2 duplications, and 333 typically developing controls, creating the largest-ever 22q11.2 CNV neuroimaging data set. In this review, we outline the ENIGMA infrastructure and procedures for multi-site analysis of CNVs and MRI data. So far, ENIGMA has identified effects of the 22q11.2, 16p11.2 distal, 15q11.2, and 1q21.1 distal CNVs on subcortical and cortical brain structures. Each CNV is associated with differences in cognitive, neurodevelopmental and neuropsychiatric traits, with characteristic patterns of brain structural abnormalities. Evidence of gene-dosage effects on distinct brain regions also emerged, providing further insight into genotype-phenotype relationships. Taken together, these results offer a more comprehensive picture of molecular mechanisms involved in typical and atypical brain development. This "genotype-first" approach also contributes to our understanding of the etiopathogenesis of brain disorders. Finally, we outline future directions to better understand effects of CNVs on brain structure and behavior.
Collapse
Affiliation(s)
- Ida E. Sønderby
- Department of Medical GeneticsOslo University HospitalOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
| | - Christopher R. K. Ching
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Sophia I. Thomopoulos
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Dennis van der Meer
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Daqiang Sun
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and PsychologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
- Department of Mental HealthVeterans Affairs Greater Los Angeles Healthcare System, Los AngelesCaliforniaUSA
| | - Julio E. Villalon‐Reina
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Ingrid Agartz
- NORMENT, Institute of Clinical PsychiatryUniversity of OsloOsloNorway
- Department of Psychiatric ResearchDiakonhjemmet HospitalOsloNorway
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Katrin Amunts
- Institute of Neuroscience and Medicine (INM‐1)Research Centre JülichJülichGermany
- Cecile and Oskar Vogt Institute for Brain Research, Medical FacultyUniversity Hospital Düsseldorf, Heinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Celso Arango
- Department of Child and Adolescent PsychiatryInstitute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañon, IsSGM, Universidad Complutense, School of MedicineMadridSpain
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
| | | | - Rosa Ayesa‐Arriola
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
- Department of PsychiatryMarqués de Valdecilla University Hospital, Valdecilla Biomedical Research Institute (IDIVAL)SantanderSpain
| | - Geor Bakker
- Department of Psychiatry and NeuropsychologyMaastricht UniversityMaastrichtThe Netherlands
- Department of Radiology and Nuclear MedicineVU University Medical CenterAmsterdamThe Netherlands
| | - Anne S. Bassett
- Clinical Genetics Research ProgramCentre for Addiction and Mental HealthTorontoOntarioCanada
- Dalglish Family 22q Clinic for Adults with 22q11.2 Deletion Syndrome, Toronto General HospitalUniversity Health NetworkTorontoOntarioCanada
- Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
| | - Dorret I. Boomsma
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam Public Health (APH) Research InstituteAmsterdam UMCAmsterdamThe Netherlands
| | - Robin Bülow
- Institute of Diagnostic Radiology and NeuroradiologyUniversity Medicine GreifswaldGreifswaldGermany
| | - Nancy J. Butcher
- Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
- Child Health Evaluative SciencesThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
| | - Vince D. Calhoun
- Tri‐institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS)Georgia State, Georgia Tech, EmoryAtlantaGeorgiaUSA
| | - Svenja Caspers
- Institute of Neuroscience and Medicine (INM‐1)Research Centre JülichJülichGermany
- Institute for Anatomy IMedical Faculty & University Hospital Düsseldorf, University of DüsseldorfDüsseldorfGermany
| | - Eva W. C. Chow
- Clinical Genetics Research ProgramCentre for Addiction and Mental HealthTorontoOntarioCanada
- Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
| | - Sven Cichon
- Institute of Neuroscience and Medicine (INM‐1)Research Centre JülichJülichGermany
- Institute of Medical Genetics and PathologyUniversity Hospital BaselBaselSwitzerland
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Simone Ciufolini
- Department of Psychosis StudiesInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUnited Kingdom
| | - Michael C. Craig
- Department of Forensic and Neurodevelopmental SciencesThe Sackler Institute for Translational Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's CollegeLondonUnited Kingdom
| | | | - Adam C. Cunningham
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUnited Kingdom
| | - Anders M. Dale
- Center for Multimodal Imaging and GeneticsUniversity of California San DiegoLa JollaCaliforniaUSA
- Department RadiologyUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Paola Dazzan
- Department of Psychological MedicineInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUnited Kingdom
| | - Greig I. de Zubicaray
- Faculty of HealthQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
| | - Srdjan Djurovic
- Department of Medical GeneticsOslo University HospitalOsloNorway
- NORMENT, Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Joanne L. Doherty
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUnited Kingdom
- Cardiff University Brain Research Imaging Centre (CUBRIC)CardiffUnited Kingdom
| | - Gary Donohoe
- Center for Neuroimaging, Genetics and GenomicsSchool of Psychology, NUI GalwayGalwayIreland
| | - Bogdan Draganski
- LREN, Centre for Research in Neuroscience, Department of NeuroscienceUniversity Hospital Lausanne and University LausanneLausanneSwitzerland
- Neurology DepartmentMax‐Planck Institute for Human Brain and Cognitive SciencesLeipzigGermany
| | - Courtney A. Durdle
- MIND Institute and Department of Psychiatry and Behavioral SciencesUniversity of California DavisDavisCaliforniaUSA
| | - Stefan Ehrlich
- Division of Psychological and Social Medicine and Developmental NeurosciencesFaculty of Medicine, TU DresdenDresdenGermany
| | - Beverly S. Emanuel
- Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Thomas Espeseth
- Department of PsychologyUniversity of OsloOsloNorway
- Department of PsychologyBjørknes CollegeOsloNorway
| | - Simon E. Fisher
- Language and Genetics DepartmentMax Planck Institute for PsycholinguisticsNijmegenThe Netherlands
- Donders Institute for Brain, Cognition and BehaviourRadboud UniversityNijmegenThe Netherlands
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics UnitCenter for Genomic Medicine, Massachusetts General HospitalBostonMassachusettsUSA
- Department of Psychiatry, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - David C. Glahn
- Tommy Fuss Center for Neuropsychiatric Disease ResearchBoston Children's HospitalBostonMassachusettsUSA
- Department of PsychiatryHarvard Medical SchoolBostonMassachusettsUSA
| | - Hans J. Grabe
- German Center for Neurodegenerative Diseases (DZNE)Site Rostock/GreifswaldGreifswaldGermany
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldGermany
| | - Raquel E. Gur
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Youth Suicide Prevention, Intervention and Research CenterChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Boris A. Gutman
- Medical Imaging Research Center, Department of Biomedical EngineeringIllinois Institute of TechnologyChicagoIllinoisUSA
| | - Jan Haavik
- Department of BiomedicineUniversity of BergenBergenNorway
- Division of PsychiatryHaukeland University HospitalBergenNorway
| | - Asta K. Håberg
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health SciencesNorwegian University of Science and TechnologyTrondheimNorway
- Department of Radiology and Nuclear MedicineSt. Olavs HospitalTrondheimNorway
| | - Laura A. Hansen
- Department of Psychiatry and Biobehavioral SciencesUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Ryota Hashimoto
- Department of Pathology of Mental DiseasesNational Institute of Mental Health, National Center of Neurology and PsychiatryTokyoJapan
- Department of PsychiatryOsaka University Graduate School of MedicineOsakaJapan
| | - Derrek P. Hibar
- Personalized Healthcare AnalyticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Avram J. Holmes
- Department of PsychologyYale UniversityNew HavenConnecticutUSA
- Department of PsychiatryYale UniversityNew HavenConnecticutUSA
| | - Jouke‐Jan Hottenga
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Hilleke E. Hulshoff Pol
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | | | - Emma E. M. Knowles
- Department of Psychiatry, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of PsychiatryBoston Children's HospitalBostonMassachusettsUSA
| | - Leila Kushan
- Semel Institute for Neuroscience and Human BehaviorUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - David E. J. Linden
- School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUnited Kingdom
| | - Jingyu Liu
- Tri‐institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS)Georgia State, Georgia Tech, EmoryAtlantaGeorgiaUSA
- Computer ScienceGeorgia State UniversityAtlantaGeorgiaUSA
| | - Astri J. Lundervold
- Department of Biological and Medical PsychologyUniversity of BergenBergenNorway
| | - Sandra Martin‐Brevet
- LREN, Centre for Research in Neuroscience, Department of NeuroscienceUniversity Hospital Lausanne and University LausanneLausanneSwitzerland
| | - Kenia Martínez
- Department of Child and Adolescent PsychiatryInstitute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañon, IsSGM, Universidad Complutense, School of MedicineMadridSpain
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
- Facultad de PsicologíaUniversidad Autónoma de MadridMadridSpain
| | - Karen A. Mather
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of MedicineUniversity of New South WalesSydneyNew South WalesAustralia
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
| | - Samuel R. Mathias
- Department of PsychiatryHarvard Medical SchoolBostonMassachusettsUSA
- Department of PsychiatryBoston Children's HospitalBostonMassachusettsUSA
| | - Donna M. McDonald‐McGinn
- Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Division of Human Genetics and 22q and You CenterChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Allan F. McRae
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQueenslandAustralia
| | - Sarah E. Medland
- Psychiatric GeneticsQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Torgeir Moberget
- Department of Psychology, Faculty of Social SciencesUniversity of OsloOsloNorway
| | - Claudia Modenato
- LREN, Centre for Research in Neuroscience, Department of NeuroscienceUniversity Hospital Lausanne and University LausanneLausanneSwitzerland
- University of LausanneLausanneSwitzerland
| | - Jennifer Monereo Sánchez
- School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Faculty of Health, Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
- Department of Radiology and Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Clara A. Moreau
- Sainte Justine Hospital Research CenterUniversity of Montreal, MontrealQCCanada
| | - Thomas W. Mühleisen
- Institute of Neuroscience and Medicine (INM‐1)Research Centre JülichJülichGermany
- Cecile and Oskar Vogt Institute for Brain Research, Medical FacultyUniversity Hospital Düsseldorf, Heinrich‐Heine‐University DüsseldorfDüsseldorfGermany
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Tomas Paus
- Bloorview Research InstituteHolland Bloorview Kids Rehabilitation HospitalTorontoOntarioCanada
- Departments of Psychology and PsychiatryUniversity of TorontoTorontoOntarioCanada
| | - Zdenka Pausova
- Translational Medicine, The Hospital for Sick ChildrenTorontoOntarioCanada
| | - Carlos Prieto
- Bioinformatics Service, NucleusUniversity of SalamancaSalamancaSpain
| | | | - Céline S. Reinbold
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Centre for Lifespan Changes in Brain and Cognition, Department of PsychologyUniversity of OsloOsloNorway
| | - Tiago Reis Marques
- Department of Psychosis StudiesInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUnited Kingdom
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Hammersmith HospitalImperial College LondonLondonUnited Kingdom
| | - Gabriela M. Repetto
- Center for Genetics and GenomicsFacultad de Medicina, Clinica Alemana Universidad del DesarrolloSantiagoChile
| | - Alexandre Reymond
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - David R. Roalf
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - James J. Rucker
- Department of Psychological MedicineInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUnited Kingdom
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of MedicineUniversity of New South WalesSydneyNew South WalesAustralia
- Neuropsychiatric InstituteThe Prince of Wales HospitalSydneyNew South WalesAustralia
| | - James E. Schmitt
- Department of Radiology and PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Peter R. Schofield
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Medical SciencesUNSW SydneySydneyNew South WalesAustralia
| | - Ana I. Silva
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUnited Kingdom
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
| | | | - Dan J. Stein
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
| | - Christian K. Tamnes
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- Department of Psychiatric ResearchDiakonhjemmet HospitalOsloNorway
- PROMENTA Research Center, Department of PsychologyUniversity of OsloOsloNorway
| | - Diana Tordesillas‐Gutiérrez
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
- Neuroimaging Unit, Technological FacilitiesValdecilla Biomedical Research Institute (IDIVAL), SantanderSpain
| | - Magnus O. Ulfarsson
- Population Genomics, deCODE genetics/AmgenReykjavikIceland
- Faculty of Electrical and Computer EngineeringUniversity of Iceland, ReykjavikIceland
| | - Ariana Vajdi
- Semel Institute for Neuroscience and Human BehaviorUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Dennis van 't Ent
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Marianne B. M. van den Bree
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUnited Kingdom
| | - Evangelos Vassos
- Social, Genetic and Developmental Psychiatry CentreInstitute of Psychiatry, Psychology & Neuroscience, King's College LondonLondonUnited Kingdom
| | - Javier Vázquez‐Bourgon
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
- Department of PsychiatryMarqués de Valdecilla University Hospital, Valdecilla Biomedical Research Institute (IDIVAL)SantanderSpain
- School of MedicineUniversity of CantabriaSantanderSpain
| | - Fidel Vila‐Rodriguez
- Department of PsychiatryThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - G. Bragi Walters
- Population Genomics, deCODE genetics/AmgenReykjavikIceland
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Lars T. Westlye
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
- Department of PsychologyUniversity of OsloOsloNorway
- NORMENT, Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Katharina Wittfeld
- German Center for Neurodegenerative Diseases (DZNE)Site Rostock/GreifswaldGreifswaldGermany
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldGermany
| | - Elaine H. Zackai
- Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Kári Stefánsson
- Population Genomics, deCODE genetics/AmgenReykjavikIceland
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| | - Sebastien Jacquemont
- Sainte Justine Hospital Research CenterUniversity of Montreal, MontrealQCCanada
- Department of PediatricsUniversity of Montreal, MontrealQCCanada
| | - Paul M. Thompson
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Carrie E. Bearden
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and PsychologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
- Center for Neurobehavioral GeneticsUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
| |
Collapse
|
39
|
Modenato C, Martin-Brevet S, Moreau CA, Rodriguez-Herreros B, Kumar K, Draganski B, Sønderby IE, Jacquemont S. Lessons Learned From Neuroimaging Studies of Copy Number Variants: A Systematic Review. Biol Psychiatry 2021; 90:596-610. [PMID: 34509290 DOI: 10.1016/j.biopsych.2021.05.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 01/06/2023]
Abstract
Pathogenic copy number variants (CNVs) and aneuploidies alter gene dosage and are associated with neurodevelopmental psychiatric disorders such as autism spectrum disorder and schizophrenia. Brain mechanisms mediating genetic risk for neurodevelopmental psychiatric disorders remain largely unknown, but there is a rapid increase in morphometry studies of CNVs using T1-weighted structural magnetic resonance imaging. Studies have been conducted one mutation at a time, leaving the field with a complex catalog of brain alterations linked to different genomic loci. Our aim was to provide a systematic review of neuroimaging phenotypes across CNVs associated with developmental psychiatric disorders including autism and schizophrenia. We included 76 structural magnetic resonance imaging studies on 20 CNVs at the 15q11.2, 22q11.2, 1q21.1 distal, 16p11.2 distal and proximal, 7q11.23, 15q11-q13, and 22q13.33 (SHANK3) genomic loci as well as aneuploidies of chromosomes X, Y, and 21. Moderate to large effect sizes on global and regional brain morphometry are observed across all genomic loci, which is in line with levels of symptom severity reported for these variants. This is in stark contrast with the much milder neuroimaging effects observed in idiopathic psychiatric disorders. Data also suggest that CNVs have independent effects on global versus regional measures as well as on cortical surface versus thickness. Findings highlight a broad diversity of regional morphometry patterns across genomic loci. This heterogeneity of brain patterns provides insight into the weak effects reported in magnetic resonance imaging studies of cognitive dimension and psychiatric conditions. Neuroimaging studies across many more variants will be required to understand links between gene function and brain morphometry.
Collapse
Affiliation(s)
- Claudia Modenato
- Laboratory for Research in Neuroimaging, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Sandra Martin-Brevet
- Laboratory for Research in Neuroimaging, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Clara A Moreau
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada; Human Genetics and Cognitive Functions, Centre National de la Recherche Scientifique UMR 3571, Department of Neuroscience, Université de Paris, Institut Pasteur, Paris, France
| | - Borja Rodriguez-Herreros
- Service des Troubles du Spectre de l'Autisme et Apparentés, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Kuldeep Kumar
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada; Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Bogdan Draganski
- Laboratory for Research in Neuroimaging, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland; Neurology Department, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Ida E Sønderby
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Sébastien Jacquemont
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada; Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
40
|
Agrawal N, Lawler K, Davidson CM, Keogh JM, Legg R, Barroso I, Farooqi IS, Brand AH. Predicting novel candidate human obesity genes and their site of action by systematic functional screening in Drosophila. PLoS Biol 2021; 19:e3001255. [PMID: 34748544 PMCID: PMC8575313 DOI: 10.1371/journal.pbio.3001255] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 09/29/2021] [Indexed: 11/18/2022] Open
Abstract
The discovery of human obesity-associated genes can reveal new mechanisms to target for weight loss therapy. Genetic studies of obese individuals and the analysis of rare genetic variants can identify novel obesity-associated genes. However, establishing a functional relationship between these candidate genes and adiposity remains a significant challenge. We uncovered a large number of rare homozygous gene variants by exome sequencing of severely obese children, including those from consanguineous families. By assessing the function of these genes in vivo in Drosophila, we identified 4 genes, not previously linked to human obesity, that regulate adiposity (itpr, dachsous, calpA, and sdk). Dachsous is a transmembrane protein upstream of the Hippo signalling pathway. We found that 3 further members of the Hippo pathway, fat, four-jointed, and hippo, also regulate adiposity and that they act in neurons, rather than in adipose tissue (fat body). Screening Hippo pathway genes in larger human cohorts revealed rare variants in TAOK2 associated with human obesity. Knockdown of Drosophila tao increased adiposity in vivo demonstrating the strength of our approach in predicting novel human obesity genes and signalling pathways and their site of action.
Collapse
Affiliation(s)
- Neha Agrawal
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Katherine Lawler
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Catherine M. Davidson
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Julia M. Keogh
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Robert Legg
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | | | - Inês Barroso
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - I. Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Andrea H. Brand
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
41
|
Characterizing Sleep Problems in 16p11.2 Deletion and Duplication. J Autism Dev Disord 2021; 53:1462-1475. [PMID: 34633643 DOI: 10.1007/s10803-021-05311-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
Studies of 16p11.2 copy number variants (CNVs) provide an avenue to identify mechanisms of impairment and develop targeted treatments for individuals with neurodevelopmental disorders. 16p11.2 deletion and duplication phenotypes are currently being ascertained; however, sleep disturbances are minimally described. In this study, we examine sleep disturbance in a well-characterized national sample of 16p11.2 CNVs, the Simons Foundation Autism Research Initiative (SFARI) database of youth and adults (n = 692). Factor analyses and multilevel models of derived sleep questionnaires for youth (n = 345) and adults (n = 347) indicate that 16p11.2 carriers show elevated sleep disturbance relative to community controls. Non-carrier family members also show elevated sleep disturbance. However, sleep duration does not differ between carriers and controls. Further studies of sleep in 16p11.2 are needed.
Collapse
|
42
|
Moreau CA, Raznahan A, Bellec P, Chakravarty M, Thompson PM, Jacquemont S. Dissecting autism and schizophrenia through neuroimaging genomics. Brain 2021; 144:1943-1957. [PMID: 33704401 PMCID: PMC8370419 DOI: 10.1093/brain/awab096] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/23/2022] Open
Abstract
Neuroimaging genomic studies of autism spectrum disorder and schizophrenia have mainly adopted a 'top-down' approach, beginning with the behavioural diagnosis, and moving down to intermediate brain phenotypes and underlying genetic factors. Advances in imaging and genomics have been successfully applied to increasingly large case-control studies. As opposed to diagnostic-first approaches, the bottom-up strategy begins at the level of molecular factors enabling the study of mechanisms related to biological risk, irrespective of diagnoses or clinical manifestations. The latter strategy has emerged from questions raised by top-down studies: why are mutations and brain phenotypes over-represented in individuals with a psychiatric diagnosis? Are they related to core symptoms of the disease or to comorbidities? Why are mutations and brain phenotypes associated with several psychiatric diagnoses? Do they impact a single dimension contributing to all diagnoses? In this review, we aimed at summarizing imaging genomic findings in autism and schizophrenia as well as neuropsychiatric variants associated with these conditions. Top-down studies of autism and schizophrenia identified patterns of neuroimaging alterations with small effect-sizes and an extreme polygenic architecture. Genomic variants and neuroimaging patterns are shared across diagnostic categories suggesting pleiotropic mechanisms at the molecular and brain network levels. Although the field is gaining traction; characterizing increasingly reproducible results, it is unlikely that top-down approaches alone will be able to disentangle mechanisms involved in autism or schizophrenia. In stark contrast with top-down approaches, bottom-up studies showed that the effect-sizes of high-risk neuropsychiatric mutations are equally large for neuroimaging and behavioural traits. Low specificity has been perplexing with studies showing that broad classes of genomic variants affect a similar range of behavioural and cognitive dimensions, which may be consistent with the highly polygenic architecture of psychiatric conditions. The surprisingly discordant effect sizes observed between genetic and diagnostic first approaches underscore the necessity to decompose the heterogeneity hindering case-control studies in idiopathic conditions. We propose a systematic investigation across a broad spectrum of neuropsychiatric variants to identify putative latent dimensions underlying idiopathic conditions. Gene expression data on temporal, spatial and cell type organization in the brain have also considerable potential for parsing the mechanisms contributing to these dimensions' phenotypes. While large neuroimaging genomic datasets are now available in unselected populations, there is an urgent need for data on individuals with a range of psychiatric symptoms and high-risk genomic variants. Such efforts together with more standardized methods will improve mechanistically informed predictive modelling for diagnosis and clinical outcomes.
Collapse
Affiliation(s)
- Clara A Moreau
- Sainte Justine Research Center, University of Montréal, Montréal, Québec H3T 1C5, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montreal, Québec H3W 1W5, Canada
- Human Genetics and Cognitive Functions, CNRS UMR 3571, Université de Paris, Institut Pasteur, Paris, France
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, MD 20892, USA
| | - Pierre Bellec
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montreal, Québec H3W 1W5, Canada
| | - Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Hospital Mental Health University Institute, Verdun, Québec H4H 1R3, Canada
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Marina del Rey, CA 90033, USA
| | - Sebastien Jacquemont
- Sainte Justine Research Center, University of Montréal, Montréal, Québec H3T 1C5, Canada
| |
Collapse
|
43
|
Modenato C, Kumar K, Moreau C, Martin-Brevet S, Huguet G, Schramm C, Jean-Louis M, Martin CO, Younis N, Tamer P, Douard E, Thébault-Dagher F, Côté V, Charlebois AR, Deguire F, Maillard AM, Rodriguez-Herreros B, Pain A, Richetin S, Melie-Garcia L, Kushan L, Silva AI, van den Bree MBM, Linden DEJ, Owen MJ, Hall J, Lippé S, Chakravarty M, Bzdok D, Bearden CE, Draganski B, Jacquemont S. Effects of eight neuropsychiatric copy number variants on human brain structure. Transl Psychiatry 2021; 11:399. [PMID: 34285187 PMCID: PMC8292542 DOI: 10.1038/s41398-021-01490-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/03/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Many copy number variants (CNVs) confer risk for the same range of neurodevelopmental symptoms and psychiatric conditions including autism and schizophrenia. Yet, to date neuroimaging studies have typically been carried out one mutation at a time, showing that CNVs have large effects on brain anatomy. Here, we aimed to characterize and quantify the distinct brain morphometry effects and latent dimensions across 8 neuropsychiatric CNVs. We analyzed T1-weighted MRI data from clinically and non-clinically ascertained CNV carriers (deletion/duplication) at the 1q21.1 (n = 39/28), 16p11.2 (n = 87/78), 22q11.2 (n = 75/30), and 15q11.2 (n = 72/76) loci as well as 1296 non-carriers (controls). Case-control contrasts of all examined genomic loci demonstrated effects on brain anatomy, with deletions and duplications showing mirror effects at the global and regional levels. Although CNVs mainly showed distinct brain patterns, principal component analysis (PCA) loaded subsets of CNVs on two latent brain dimensions, which explained 32 and 29% of the variance of the 8 Cohen's d maps. The cingulate gyrus, insula, supplementary motor cortex, and cerebellum were identified by PCA and multi-view pattern learning as top regions contributing to latent dimension shared across subsets of CNVs. The large proportion of distinct CNV effects on brain morphology may explain the small neuroimaging effect sizes reported in polygenic psychiatric conditions. Nevertheless, latent gene brain morphology dimensions will help subgroup the rapidly expanding landscape of neuropsychiatric variants and dissect the heterogeneity of idiopathic conditions.
Collapse
Affiliation(s)
- Claudia Modenato
- LREN - Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Kuldeep Kumar
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | - Clara Moreau
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | - Sandra Martin-Brevet
- LREN - Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Guillaume Huguet
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | - Catherine Schramm
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | - Martineau Jean-Louis
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | | | - Nadine Younis
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | - Petra Tamer
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | - Elise Douard
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | | | - Valérie Côté
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | | | - Florence Deguire
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | - Anne M Maillard
- Service des Troubles du Spectre de l'Autisme et apparentés, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Borja Rodriguez-Herreros
- Service des Troubles du Spectre de l'Autisme et apparentés, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Aurèlie Pain
- Service des Troubles du Spectre de l'Autisme et apparentés, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Sonia Richetin
- Service des Troubles du Spectre de l'Autisme et apparentés, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Lester Melie-Garcia
- Applied Signal Processing Group (ASPG), Swiss Federal Institute Lausanne (EPFL), Lausanne, Switzerland
| | - Leila Kushan
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Psychology, UCLA, Los Angeles, USA
| | - Ana I Silva
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Marianne B M van den Bree
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - David E J Linden
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Sarah Lippé
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada
| | | | - Danilo Bzdok
- Department of Biomedical Engineering, McConnell Brain Imaging Centre; Montreal Neurological Institute, McGill University, Montréal, QC, Canada
- Mila - Quebec Artificial Intelligence Institute, Montréal, QC, Canada
| | - Carrie E Bearden
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Psychology, UCLA, Los Angeles, USA
| | - Bogdan Draganski
- LREN - Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- Neurology Department, Max-Planck-Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Sébastien Jacquemont
- Centre de recherche CHU Sainte-Justine and University of Montréal, Montréal, Canada.
| |
Collapse
|
44
|
Reversal of synaptic and behavioral deficits in a 16p11.2 duplication mouse model via restoration of the GABA synapse regulator Npas4. Mol Psychiatry 2021; 26:1967-1979. [PMID: 32099100 PMCID: PMC7483162 DOI: 10.1038/s41380-020-0693-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/09/2020] [Accepted: 02/14/2020] [Indexed: 12/18/2022]
Abstract
The human 16p11.2 gene locus is a hot spot for copy number variations, which predispose carriers to a range of neuropsychiatric phenotypes. Microduplications of 16p11.2 are associated with autism spectrum disorder (ASD), intellectual disability (ID), and schizophrenia (SZ). Despite the debilitating nature of 16p11.2 duplications, the underlying molecular mechanisms remain poorly understood. Here we performed a comprehensive behavioral characterization of 16p11.2 duplication mice (16p11.2dp/+) and identified social and cognitive deficits reminiscent of ASD and ID phenotypes. 16p11.2dp/+ mice did not exhibit the SZ-related sensorimotor gating deficits, psychostimulant-induced hypersensitivity, or motor impairment. Electrophysiological recordings of 16p11.2dp/+ mice found deficient GABAergic synaptic transmission and elevated neuronal excitability in the prefrontal cortex (PFC), a brain region critical for social and cognitive functions. RNA-sequencing identified genome-wide transcriptional aberrance in the PFC of 16p11.2dp/+ mice, including downregulation of the GABA synapse regulator Npas4. Restoring Npas4 expression in PFC of 16p11.2dp/+ mice ameliorated the social and cognitive deficits and reversed GABAergic synaptic impairment and neuronal hyperexcitability. These findings suggest that prefrontal cortical GABAergic synaptic circuitry and Npas4 are strongly implicated in 16p11.2 duplication pathology, and may represent potential targets for therapeutic intervention in ASD.
Collapse
|
45
|
Huguet G, Schramm C, Douard E, Tamer P, Main A, Monin P, England J, Jizi K, Renne T, Poirier M, Nowak S, Martin CO, Younis N, Knoth IS, Jean-Louis M, Saci Z, Auger M, Tihy F, Mathonnet G, Maftei C, Léveillé F, Porteous D, Davies G, Redmond P, Harris SE, Hill WD, Lemyre E, Schumann G, Bourgeron T, Pausova Z, Paus T, Karama S, Lippe S, Deary IJ, Almasy L, Labbe A, Glahn D, Greenwood CMT, Jacquemont S. Genome-wide analysis of gene dosage in 24,092 individuals estimates that 10,000 genes modulate cognitive ability. Mol Psychiatry 2021; 26:2663-2676. [PMID: 33414497 PMCID: PMC8953148 DOI: 10.1038/s41380-020-00985-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/30/2020] [Accepted: 11/30/2020] [Indexed: 11/09/2022]
Abstract
Genomic copy number variants (CNVs) are routinely identified and reported back to patients with neuropsychiatric disorders, but their quantitative effects on essential traits such as cognitive ability are poorly documented. We have recently shown that the effect size of deletions on cognitive ability can be statistically predicted using measures of intolerance to haploinsufficiency. However, the effect sizes of duplications remain unknown. It is also unknown if the effect of multigenic CNVs are driven by a few genes intolerant to haploinsufficiency or distributed across tolerant genes as well. Here, we identified all CNVs > 50 kilobases in 24,092 individuals from unselected and autism cohorts with assessments of general intelligence. Statistical models used measures of intolerance to haploinsufficiency of genes included in CNVs to predict their effect size on intelligence. Intolerant genes decrease general intelligence by 0.8 and 2.6 points of intelligence quotient when duplicated or deleted, respectively. Effect sizes showed no heterogeneity across cohorts. Validation analyses demonstrated that models could predict CNV effect sizes with 78% accuracy. Data on the inheritance of 27,766 CNVs showed that deletions and duplications with the same effect size on intelligence occur de novo at the same frequency. We estimated that around 10,000 intolerant and tolerant genes negatively affect intelligence when deleted, and less than 2% have large effect sizes. Genes encompassed in CNVs were not enriched in any GOterms but gene regulation and brain expression were GOterms overrepresented in the intolerant subgroup. Such pervasive effects on cognition may be related to emergent properties of the genome not restricted to a limited number of biological pathways.
Collapse
Affiliation(s)
- Guillaume Huguet
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada.
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada.
| | - Catherine Schramm
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Elise Douard
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Petra Tamer
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Antoine Main
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Département de Sciences de la Décision, HEC Montreal, Montreal, QC, Canada
| | - Pauline Monin
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Human Genetics and Cognitive Functions, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Jade England
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Khadije Jizi
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Thomas Renne
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Universite de Rouen Normandie, UFR des Sciences et Techniques, Rouen, France
| | - Myriam Poirier
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Sabrina Nowak
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Charles-Olivier Martin
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Nadine Younis
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Inga Sophia Knoth
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Martineau Jean-Louis
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Zohra Saci
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Maude Auger
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Frédérique Tihy
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Géraldine Mathonnet
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Catalina Maftei
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - France Léveillé
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - David Porteous
- Lothian Birth Cohorts Group, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Medical Genetics Section, Centre for Genomic & Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Gail Davies
- Lothian Birth Cohorts Group, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Paul Redmond
- Lothian Birth Cohorts Group, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Sarah E Harris
- Lothian Birth Cohorts Group, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - W David Hill
- Lothian Birth Cohorts Group, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Emmanuelle Lemyre
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Gunter Schumann
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, England
| | - Thomas Bourgeron
- Department of Neurosciences, Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique Genes, Synapses and Cognition Laboratory, Institut Pasteur, Paris, France
- Human Genetics and Cognitive Functions, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Tomas Paus
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Sherif Karama
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Center, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Sarah Lippe
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Psychology, Université de Montréal, Montreal, QC, Canada
| | - Ian J Deary
- Medical Genetics Section, Centre for Genomic & Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Laura Almasy
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aurélie Labbe
- Human Genetics and Cognitive Functions, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - David Glahn
- Department of Psychiatry, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Olin Neuropsychiatric Research Center, Institute of Living, Hartford Hospital, Hartford, CT, USA
| | - Celia M T Greenwood
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, Departments of Epidemiology, Biostatistics & Occupational Health and Human Genetics, McGill University, Montreal, QC, Canada
| | - Sébastien Jacquemont
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada.
- Centre de recherche et Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada.
| |
Collapse
|
46
|
16p11.2 deletion is associated with hyperactivation of human iPSC-derived dopaminergic neuron networks and is rescued by RHOA inhibition in vitro. Nat Commun 2021; 12:2897. [PMID: 34006844 PMCID: PMC8131375 DOI: 10.1038/s41467-021-23113-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 04/16/2021] [Indexed: 02/03/2023] Open
Abstract
Reciprocal copy number variations (CNVs) of 16p11.2 are associated with a wide spectrum of neuropsychiatric and neurodevelopmental disorders. Here, we use human induced pluripotent stem cells (iPSCs)-derived dopaminergic (DA) neurons carrying CNVs of 16p11.2 duplication (16pdup) and 16p11.2 deletion (16pdel), engineered using CRISPR-Cas9. We show that 16pdel iPSC-derived DA neurons have increased soma size and synaptic marker expression compared to isogenic control lines, while 16pdup iPSC-derived DA neurons show deficits in neuronal differentiation and reduced synaptic marker expression. The 16pdel iPSC-derived DA neurons have impaired neurophysiological properties. The 16pdel iPSC-derived DA neuronal networks are hyperactive and have increased bursting in culture compared to controls. We also show that the expression of RHOA is increased in the 16pdel iPSC-derived DA neurons and that treatment with a specific RHOA-inhibitor, Rhosin, rescues the network activity of the 16pdel iPSC-derived DA neurons. Our data suggest that 16p11.2 deletion-associated iPSC-derived DA neuron hyperactivation can be rescued by RHOA inhibition.
Collapse
|
47
|
To eat, or not to eat, that is the question: Neural stem cells escape phagocytosis in autism with macrocephaly. Proc Natl Acad Sci U S A 2021; 118:2104888118. [PMID: 33883269 DOI: 10.1073/pnas.2104888118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
48
|
Overexpression of CD47 is associated with brain overgrowth and 16p11.2 deletion syndrome. Proc Natl Acad Sci U S A 2021; 118:2005483118. [PMID: 33833053 DOI: 10.1073/pnas.2005483118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Copy number variation (CNV) at the 16p11.2 locus is associated with neuropsychiatric disorders, such as autism spectrum disorder and schizophrenia. CNVs of the 16p gene can manifest in opposing head sizes. Carriers of 16p11.2 deletion tend to have macrocephaly (or brain enlargement), while those with 16p11.2 duplication frequently have microcephaly. Increases in both gray and white matter volume have been observed in brain imaging studies in 16p11.2 deletion carriers with macrocephaly. Here, we use human induced pluripotent stem cells (hiPSCs) derived from controls and subjects with 16p11.2 deletion and 16p11.2 duplication to understand the underlying mechanisms regulating brain overgrowth. To model both gray and white matter, we differentiated patient-derived iPSCs into neural progenitor cells (NPCs) and oligodendrocyte progenitor cells (OPCs). In both NPCs and OPCs, we show that CD47 (a "don't eat me" signal) is overexpressed in the 16p11.2 deletion carriers contributing to reduced phagocytosis both in vitro and in vivo. Furthermore, 16p11.2 deletion NPCs and OPCs up-regulate cell surface expression of calreticulin (a prophagocytic "eat me" signal) and its binding sites, indicating that these cells should be phagocytosed but fail to be eliminated due to elevations in CD47. Treatment of 16p11.2 deletion NPCs and OPCs with an anti-CD47 antibody to block CD47 restores phagocytosis to control levels. While the CD47 pathway is commonly implicated in cancer progression, we document a role for CD47 in psychiatric disorders associated with brain overgrowth.
Collapse
|
49
|
Sønderby IE, van der Meer D, Moreau C, Kaufmann T, Walters GB, Ellegaard M, Abdellaoui A, Ames D, Amunts K, Andersson M, Armstrong NJ, Bernard M, Blackburn NB, Blangero J, Boomsma DI, Brodaty H, Brouwer RM, Bülow R, Bøen R, Cahn W, Calhoun VD, Caspers S, Ching CRK, Cichon S, Ciufolini S, Crespo-Facorro B, Curran JE, Dale AM, Dalvie S, Dazzan P, de Geus EJC, de Zubicaray GI, de Zwarte SMC, Desrivieres S, Doherty JL, Donohoe G, Draganski B, Ehrlich S, Eising E, Espeseth T, Fejgin K, Fisher SE, Fladby T, Frei O, Frouin V, Fukunaga M, Gareau T, Ge T, Glahn DC, Grabe HJ, Groenewold NA, Gústafsson Ó, Haavik J, Haberg AK, Hall J, Hashimoto R, Hehir-Kwa JY, Hibar DP, Hillegers MHJ, Hoffmann P, Holleran L, Holmes AJ, Homuth G, Hottenga JJ, Hulshoff Pol HE, Ikeda M, Jahanshad N, Jockwitz C, Johansson S, Jönsson EG, Jørgensen NR, Kikuchi M, Knowles EEM, Kumar K, Le Hellard S, Leu C, Linden DEJ, Liu J, Lundervold A, Lundervold AJ, Maillard AM, Martin NG, Martin-Brevet S, Mather KA, Mathias SR, McMahon KL, McRae AF, Medland SE, Meyer-Lindenberg A, Moberget T, Modenato C, Sánchez JM, Morris DW, Mühleisen TW, Murray RM, Nielsen J, Nordvik JE, Nyberg L, Loohuis LMO, Ophoff RA, et alSønderby IE, van der Meer D, Moreau C, Kaufmann T, Walters GB, Ellegaard M, Abdellaoui A, Ames D, Amunts K, Andersson M, Armstrong NJ, Bernard M, Blackburn NB, Blangero J, Boomsma DI, Brodaty H, Brouwer RM, Bülow R, Bøen R, Cahn W, Calhoun VD, Caspers S, Ching CRK, Cichon S, Ciufolini S, Crespo-Facorro B, Curran JE, Dale AM, Dalvie S, Dazzan P, de Geus EJC, de Zubicaray GI, de Zwarte SMC, Desrivieres S, Doherty JL, Donohoe G, Draganski B, Ehrlich S, Eising E, Espeseth T, Fejgin K, Fisher SE, Fladby T, Frei O, Frouin V, Fukunaga M, Gareau T, Ge T, Glahn DC, Grabe HJ, Groenewold NA, Gústafsson Ó, Haavik J, Haberg AK, Hall J, Hashimoto R, Hehir-Kwa JY, Hibar DP, Hillegers MHJ, Hoffmann P, Holleran L, Holmes AJ, Homuth G, Hottenga JJ, Hulshoff Pol HE, Ikeda M, Jahanshad N, Jockwitz C, Johansson S, Jönsson EG, Jørgensen NR, Kikuchi M, Knowles EEM, Kumar K, Le Hellard S, Leu C, Linden DEJ, Liu J, Lundervold A, Lundervold AJ, Maillard AM, Martin NG, Martin-Brevet S, Mather KA, Mathias SR, McMahon KL, McRae AF, Medland SE, Meyer-Lindenberg A, Moberget T, Modenato C, Sánchez JM, Morris DW, Mühleisen TW, Murray RM, Nielsen J, Nordvik JE, Nyberg L, Loohuis LMO, Ophoff RA, Owen MJ, Paus T, Pausova Z, Peralta JM, Pike GB, Prieto C, Quinlan EB, Reinbold CS, Marques TR, Rucker JJH, Sachdev PS, Sando SB, Schofield PR, Schork AJ, Schumann G, Shin J, Shumskaya E, Silva AI, Sisodiya SM, Steen VM, Stein DJ, Strike LT, Suzuki IK, Tamnes CK, Teumer A, Thalamuthu A, Tordesillas-Gutiérrez D, Uhlmann A, Ulfarsson MO, van 't Ent D, van den Bree MBM, Vanderhaeghen P, Vassos E, Wen W, Wittfeld K, Wright MJ, Agartz I, Djurovic S, Westlye LT, Stefansson H, Stefansson K, Jacquemont S, Thompson PM, Andreassen OA. 1q21.1 distal copy number variants are associated with cerebral and cognitive alterations in humans. Transl Psychiatry 2021; 11:182. [PMID: 33753722 PMCID: PMC7985307 DOI: 10.1038/s41398-021-01213-0] [Show More Authors] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/23/2020] [Accepted: 01/08/2021] [Indexed: 01/07/2023] Open
Abstract
Low-frequency 1q21.1 distal deletion and duplication copy number variant (CNV) carriers are predisposed to multiple neurodevelopmental disorders, including schizophrenia, autism and intellectual disability. Human carriers display a high prevalence of micro- and macrocephaly in deletion and duplication carriers, respectively. The underlying brain structural diversity remains largely unknown. We systematically called CNVs in 38 cohorts from the large-scale ENIGMA-CNV collaboration and the UK Biobank and identified 28 1q21.1 distal deletion and 22 duplication carriers and 37,088 non-carriers (48% male) derived from 15 distinct magnetic resonance imaging scanner sites. With standardized methods, we compared subcortical and cortical brain measures (all) and cognitive performance (UK Biobank only) between carrier groups also testing for mediation of brain structure on cognition. We identified positive dosage effects of copy number on intracranial volume (ICV) and total cortical surface area, with the largest effects in frontal and cingulate cortices, and negative dosage effects on caudate and hippocampal volumes. The carriers displayed distinct cognitive deficit profiles in cognitive tasks from the UK Biobank with intermediate decreases in duplication carriers and somewhat larger in deletion carriers-the latter potentially mediated by ICV or cortical surface area. These results shed light on pathobiological mechanisms of neurodevelopmental disorders, by demonstrating gene dose effect on specific brain structures and effect on cognitive function.
Collapse
Affiliation(s)
- Ida E Sønderby
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway.
| | - Dennis van der Meer
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Clara Moreau
- Sainte Justine Hospital Research Center, Montreal, Quebec, Canada
- Centre de recherche de l'Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada
| | - Tobias Kaufmann
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - G Bragi Walters
- deCODE Genetics (Amgen), Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Maria Ellegaard
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| | - Abdel Abdellaoui
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Biological Psychology and Netherlands Twin Register, VU University Amsterdam, Amsterdam, the Netherlands
| | - David Ames
- University of Melbourne Academic Unit for Psychiatry of Old Age, Kew, Australia
- National Ageing Research Institute, Parkville, Australia
| | - Katrin Amunts
- Institute of Neuroscience and Medicine, INM-1, Research Centre Jülich, Jülich, Germany
- C. and O. Vogt Institute for Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Duesseldorf, Düsseldorf, Germany
| | - Micael Andersson
- Umeå Centre for Functional Brain Imaging, Umeå University, Umeå, Sweden
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | - Manon Bernard
- Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicholas B Blackburn
- South Texas Diabetes and Obesity Institute, Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Brownsville, USA
| | - John Blangero
- South Texas Diabetes and Obesity Institute, Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Brownsville, USA
| | - Dorret I Boomsma
- Department of Biological Psychology and Netherlands Twin Register, VU University Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, the Netherlands
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
- Dementia Centre for Research Collaboration, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Rachel M Brouwer
- Department of Psychiatry, University Medical Center Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Rune Bøen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Wiepke Cahn
- Department of Psychiatry, University Medical Center Brain Center, Utrecht University, Utrecht, the Netherlands
- Altrecht Science, Utrecht, the Netherlands
| | - Vince D Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, USA
- The Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, USA
| | - Svenja Caspers
- Institute of Neuroscience and Medicine, INM-1, Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christopher R K Ching
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, USA
| | - Sven Cichon
- Institute of Neuroscience and Medicine, INM-1, Research Centre Jülich, Jülich, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Simone Ciufolini
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Benedicto Crespo-Facorro
- University Hospital Marqués de Valdecilla, IDIVAL, Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), Santander, Spain
- University Hospital Virgen del Rocío, IBiS, Centre de Investigació Biomédica en Red Salud Mental (CIBERSAM), Sevilla, Spain
| | - Joanne E Curran
- South Texas Diabetes and Obesity Institute, Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Brownsville, USA
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, University of California, San Diego, USA
| | - Shareefa Dalvie
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, Western Cape, South Africa
| | - Paola Dazzan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Eco J C de Geus
- Department of Biological Psychology and Netherlands Twin Register, VU University Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, the Netherlands
| | | | - Sonja M C de Zwarte
- Department of Psychiatry, University Medical Center Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Sylvane Desrivieres
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Joanne L Doherty
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Cardiff University Brain Research Imaging Centre School of Psychology, Cardiff University, Cardiff, United Kingdom
| | - Gary Donohoe
- Centre for Neuroimaging and Cognitive Genomics, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
| | - Bogdan Draganski
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Neurology Department, Max-Planck-Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Stefan Ehrlich
- Division of Psychological and Social Medicine, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Else Eising
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Thomas Espeseth
- Department of Psychology, University of Oslo, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Kim Fejgin
- Signal Transduction, H. Lundbeck A/S, Ottiliavej 9, DK-2500, Valby, Denmark
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, 1474, Nordbyhagen, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Oleksandr Frei
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vincent Frouin
- Université Paris-Saclay, CEA, Neurospin, 91191, Gif-sur-Yvette, France
| | - Masaki Fukunaga
- Division of Cerebral Integration, National Institute for Physiological Sciences, Okazaki, Japan
- Department of Life Science, Sokendai, Hayama, Japan
| | - Thomas Gareau
- Université Paris-Saclay, CEA, Neurospin, 91191, Gif-sur-Yvette, France
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David C Glahn
- Boston Children's Hospital, Boston, Massachusetts, USA
- Institute of Living, Hartford, Connecticut, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center of Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Greifswald, Germany
| | - Nynke A Groenewold
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, Western Cape, South Africa
| | | | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Asta K Haberg
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
- St Olav's Hospital, Department of Radiology and Nuclear Medicine, Trondheim, Norway
| | - Jeremy Hall
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Ryota Hashimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Japan
- Osaka University, Osaka, Japan
| | - Jayne Y Hehir-Kwa
- Princess Màxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Manon H J Hillegers
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-Sophia, Rotterdam, the Netherlands
| | - Per Hoffmann
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Institute of Human Genetics, University of Bonn Medical School, Bonn, Germany
| | - Laurena Holleran
- Centre for Neuroimaging and Cognitive Genomics, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
| | - Avram J Holmes
- Psychology Department, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Jouke-Jan Hottenga
- Department of Biological Psychology and Netherlands Twin Register, VU University Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, the Netherlands
| | - Hilleke E Hulshoff Pol
- Department of Psychiatry, University Medical Center Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Masashi Ikeda
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, USA
| | - Christiane Jockwitz
- Institute of Neuroscience and Medicine, INM-1, Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefan Johansson
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Erik G Jönsson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Niklas R Jørgensen
- Department of Clinical Biochemistry, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Masataka Kikuchi
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Emma E M Knowles
- Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Kuldeep Kumar
- Sainte Justine Hospital Research Center, Montreal, Quebec, Canada
| | - Stephanie Le Hellard
- Norwegian Centre for Mental Disorders Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Costin Leu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States
- Chalfont Centre for Epilepsy, Chalfont-St-Peter, United Kingdom
| | - David E J Linden
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
| | - Jingyu Liu
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, USA
| | - Arvid Lundervold
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | | | - Anne M Maillard
- Service des Troubles du Spectre de l'Autisme et apparentés, Lausanne University Hospital, Lausanne, Switzerland
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Sandra Martin-Brevet
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
- Neuroscience Research Australia, Randwick, Australia
| | - Samuel R Mathias
- Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Katie L McMahon
- Herston Imaging Research Facility and School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Sarah E Medland
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Torgeir Moberget
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Claudia Modenato
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Jennifer Monereo Sánchez
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Derek W Morris
- Centre for Neuroimaging and Cognitive Genomics, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
| | - Thomas W Mühleisen
- Institute of Neuroscience and Medicine, INM-1, Research Centre Jülich, Jülich, Germany
- C. and O. Vogt Institute for Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Duesseldorf, Düsseldorf, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Robin M Murray
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Jacob Nielsen
- Signal Transduction, H. Lundbeck A/S, Ottiliavej 9, DK-2500, Valby, Denmark
| | | | - Lars Nyberg
- Umeå Centre for Functional Brain Imaging, Umeå University, Umeå, Sweden
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Loes M Olde Loohuis
- Center for Neurobehavioral Genetics, University of California, Los Angeles, USA
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, University of California, Los Angeles, USA
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
| | - Tomas Paus
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
- Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Zdenka Pausova
- Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Juan M Peralta
- South Texas Diabetes and Obesity Institute, Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Brownsville, USA
| | - G Bruce Pike
- Departments of Radiology and Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Carlos Prieto
- Bioinformatics Service, Nucleus, University of Salamanca, Salamanca, Spain
| | - Erin B Quinlan
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Céline S Reinbold
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Tiago Reis Marques
- Department of Psychosis, Institute of Psychiatry, Psychology & Neuroscience, Kings College, London, United Kingdom
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Hammersmith Hospital, Imperial College, London, United Kingdom
| | - James J H Rucker
- Institute of Psychiatry, Psychology and Neuroscience, London, London, United Kingdom
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
- Neuropsychiatric Institute, The Prince of Wales Hospital, Sydney, Australia
| | - Sigrid B Sando
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
- University Hospital of Trondheim,Department of Neurology and Clinical Neurophysiology, Trondheim, Norway
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Andrew J Schork
- Institute of Biological Psychiatry, Roskilde, Denmark
- The Translational Genetics Institute (TGEN), Phoenix, AZ, United States
| | - Gunter Schumann
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Jean Shin
- Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Elena Shumskaya
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ana I Silva
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Cardiff University Brain Research Imaging Centre School of Psychology, Cardiff University, Cardiff, United Kingdom
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Chalfont Centre for Epilepsy, Chalfont-St-Peter, United Kingdom
| | - Vidar M Steen
- Norwegian Centre for Mental Disorders Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Dan J Stein
- South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Lachlan T Strike
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Ikuo K Suzuki
- VIB Center for Brain & Disease Research, Stem Cell and Developmental Neurobiology Lab, Leuven, Belgium
- University of Brussels (ULB), Institute of Interdisciplinary Research (IRIBHM) ULB Neuroscience Institute, Brussels, Belgium
- The University of Tokyo, Department of Biological Sciences, Graduate School of Science, Tokyo, Japan
| | - Christian K Tamnes
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- PROMENTA Research Center, Department of Psychology, University of Oslo, Oslo, Norway
- Department of Psychiatry, Diakonhjemmet Hospital, Oslo, Norway
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Diana Tordesillas-Gutiérrez
- University Hospital Marqués de Valdecilla, IDIVAL, Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), Santander, Spain
- Department of Radiology, Marqués de Valdecilla University Hospital, Valdecilla Biomedical Research Institute IDIVAL, Santander, Spain
| | - Anne Uhlmann
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, Western Cape, South Africa
| | - Magnus O Ulfarsson
- deCODE Genetics (Amgen), Reykjavík, Iceland
- Faculty of Electrical and Computer Engineering, University of Iceland, Reykjavík, Iceland
| | - Dennis van 't Ent
- Department of Biological Psychology and Netherlands Twin Register, VU University Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Marianne B M van den Bree
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences & Leuven Brain Institute, 3000, Leuven, Belgium
- Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070, Brussels, Belgium
| | - Evangelos Vassos
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research, Mental Health Biomedical Research Centre, South London and Maudsley National Health Service Foundation Trust and King's College London, London, United Kingdom
| | - Wei Wen
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center of Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Greifswald, Germany
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - Ingrid Agartz
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry, Diakonhjemmet Hospital, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars T Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | | | - Kari Stefansson
- deCODE Genetics (Amgen), Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Sébastien Jacquemont
- Sainte Justine Hospital Research Center, Montreal, Quebec, Canada
- Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, USA
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
50
|
Szelest M, Stefaniak M, Ręka G, Jaszczuk I, Lejman M. Three case reports of patients indicating the diversity of molecular and clinical features of 16p11.2 microdeletion anomaly. BMC Med Genomics 2021; 14:76. [PMID: 33691695 PMCID: PMC7945342 DOI: 10.1186/s12920-021-00929-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND 16p11.2 microdeletion is a known chromosomal anomaly associated mainly with neurocognitive developmental delay, predisposition to obesity, and variable dysmorphism. Although this deletion is relatively rare among the general population, it is one of the serious known genetic aetiologies of obesity and autism spectrum disorder. CASE PRESENTATION This study presents three cases of deletions within the 16p11.2 region. Every child had mild variable craniofacial abnormalities, hand or foot anomalies and developmental and language delays. The first proband had obesity, epilepsy, moderate intellectual disability, aphasia, motor delay, hyperinsulinism, and café au lait spots. The second proband suffered from cardiac, pulmonary, and haematological problems. The third proband had motor and language delays, bronchial asthma, and umbilical hernia. Although each patient presented some features of the syndrome, the children differed in terms of their clinical pictures. Genetic diagnosis of 16p11.2 microdeletion syndrome was made in children at different ages based on multiplex ligation probe-dependent amplification analysis and/or microarray methods. CONCLUSIONS Our reports allow us to analyse and better understand the biology of 16p11.2 microdeletion throughout development. However, the variability of presented cases supports the alternate conclusion to this presented in available literature regarding 16p11.2 deletion, as we observed no direct cause-and-effect genotype/phenotype relationships. The reported cases indicate the key role of the interdisciplinary approach in 16p11.2 deletion diagnostics. The care of patients with this anomaly is based on regular health assessment and adjustment of nervous system development therapy.
Collapse
Affiliation(s)
- Monika Szelest
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, Gębali 6, 20-093, Lublin, Poland
| | - Martyna Stefaniak
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, Gębali 6, 20-093, Lublin, Poland
| | - Gabriela Ręka
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, Gębali 6, 20-093, Lublin, Poland
| | - Ilona Jaszczuk
- Department of Cancer Genetics With Cytogenetics Laboratory, Medical University of Lublin, Radziwiłłowska 11, 20-080, Lublin, Poland
| | - Monika Lejman
- Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093, Lublin, Poland.
| |
Collapse
|