48 research outputs found

    Effect of 16P11.2 copy number variants on cognitive traits and brain structures

    Get PDF
    The 600kb 16p11.2 CNVs (breakpoints 4–5, 29.6-30.2 Mb-Hg19) are among the most frequent genetic risk factors for neurodevelopmental and psychiatric conditions: A 10-fold enrichment of deletions and duplications is observed in autism cohorts and a 10-fold enrichment of duplications in schizophrenia cohorts. Previous studies demonstrated “mirror” effects of both CNVs on body mass index and head circumference (deletion>control>duplication). However, the large global effect of brain size and the sample size of the two previous neuroimaging studies limited the interpretation of the analyses on regional brain structures, any estimate of the effect size, and the generalizability of the results across different ascertainments of the patients. In the first part of my Ph.D., I analyze structural magnetic resonance imaging (MRI) on 78 deletion carriers, 71 duplication carriers, and 212 controls. I show that both CNVs affect in a “mirror” way the volume and the cortical surface of the insula (Cohen’s d>1), whilst other brain regions are preferentially altered in either the deletion carriers (calcarine cortex and superior, middle, transverse temporal gyri, Cohen’s d>1) or the duplication carriers (caudate and hippocampus, Cohen’s d of 0.5 to 1). Results are generalizable across scanning sites, computational methods, age, sex, ascertainment for psychiatric disorders. They partially overlap with results of meta-analyses performed across psychiatric disorders. In the second part, I characterize the developmental trajectory of global brain metrics and regional brain structures in the 16p11.2 CNV carriers. I adapt a previously published longitudinal pipeline and normalizing method, derived from 339 typically developing individuals aged from 4.5 to 20 years old. From this population of reference, I Z-score our cross-sectional 16p11.2 dataset and show that all the brain alterations in the 16p11.2 carriers are already present at 4.5 years old and follow parallel trajectories to the controls. In summary, my results suggest that brain alterations, present in childhood and stable across adolescence and adulthood, are related to the risk conferred by the 16p11.2 CNVs, regardless of the carriers’ symptoms. Additional factors are therefore likely required for the development of psychiatric disorders. I highlight the relevance of studying genetic risk factors and mechanisms as a complement to groups defined by behavioral criteria. Further studies comparing multiple CNVs and monogenic conditions, from the earliest age, are required to understand the onset of neuroanatomical alterations and their overlap between different genetic risk factors for neurodevelopmental disorders. -- Les variations en nombre de copies (CNV), au locus 16p11.2 et d’une taille d’600kb (points de cassure 4–5, 29.6-30.2 Mb-Hg19) reprĂ©sentent un des facteurs de risque gĂ©nĂ©tique les plus frĂ©quents parmi les troubles psychiatriques : 10% d’enrichissement en dĂ©lĂ©tion et duplication pour les troubles du spectre autistique, 10% d’enrichissement en duplication pour la schizophrĂ©nie. Les effets « miroirs » des deux CNVs sur l’indice de masse corporelle et le pĂ©rimĂštre cranien ont dĂ©jĂ  Ă©tĂ© dĂ©montrĂ©s (dĂ©lĂ©tion>contrĂŽle>duplication). Cependant, les diffĂ©rences en taille de cerveau et les Ă©chantillons des deux prĂ©cĂ©dentes Ă©tudes de neuro- imagerie ont limitĂ© les analyses des rĂ©gions cĂ©rĂ©brales, l’estimation de la taille des effets, et la gĂ©nĂ©ralisation des rĂ©sultats selon les modes de recrutement des patients. Dans cette thĂšse, j’analyse les images par rĂ©sonance magnĂ©tique (IRM) de 78 porteurs de la dĂ©lĂ©tion, 71 porteurs de la duplication et 212 participants contrĂŽles. Je montre que les deux CNVs sont associĂ©es Ă  des diffĂ©rences « en miroir » du volume et de la surface corticale de l’insula (Cohen’s d>1), tandis que le cortex calcarin, les gyri temporaux supĂ©rieur, moyen et transverse sont prĂ©fĂ©rentiellement altĂ©rĂ©s par la dĂ©lĂ©tion (Cohen’s d>1), les noyaux caudĂ©s et l’hippocampe sont prĂ©fĂ©rentiellement altĂ©rĂ©s par la duplication (0.5<Cohen’s d<1). Les rĂ©sultats sont gĂ©nĂ©ralisables Ă  travers les differents sites d’IRM, les mĂ©thodes d’analyse computationnelle, les Ăąges, les sexes et les divers diagnostiques psychiatriques des patients. Les rĂ©sultats chevauchent partiellement ceux d’une mĂ©ta-analyse sur plusieurs diagnostiques psychiatriques. Dans un second temps, je caractĂ©rise la trajectoire dĂ©veloppementale de ces diffĂ©rences cĂ©rĂ©brales. J’adapte un pipeline longitunal et une mĂ©thode de normalisation dĂ©jĂ  publiĂ©s, construits Ă  partir de 339 participants contrĂŽles de 4.5 Ă  20 ans. Je calcule des Z-scores pour nos donnĂ©es transversales et montre que les diffĂ©rences cĂ©rĂ©brales liĂ©es aux CNVs sont dĂ©jĂ  prĂ©sentes Ă  4.5 ans, avec les mĂȘmes tailles d’effet et une trajectoire parallĂšle aux contrĂŽles. En rĂ©sumĂ©, mes rĂ©sultats suggĂšrent que les diffĂ©rences cĂ©rĂ©brales, prĂ©sentes dans la jeune enfance et stables Ă  l’adolescence et l’ñge adulte, sont liĂ©es au risque confĂ©rĂ© par les CNVs en 16p11.2, quelque soient les symptĂŽmes. Des facteurs additionnels sont probablement nĂ©cessaires pour le dĂ©veloppement de maladies psychiatriques. Je montre la pertinence d’étudier les facteurs de risque gĂ©nĂ©tiques en complĂ©ment des groupes de patients dĂ©finis sur des critĂšres comportementaux. Des Ă©tudes comparant diverses conditions gĂ©nĂ©tiques, dĂšs la naissance, sont nĂ©cessaires pour comprendre le dĂ©but et le chevauchement des diffĂ©rences neuro-anatomiques observĂ©es pour diffĂ©rents facteurs de risque gĂ©nĂ©tiques

    Effects of copy number variations on brain structure and risk for psychiatric illness: Large-scale studies from the ENIGMA working groups on CNVs

    Get PDF
    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

    Dose response of the 16p11.2 distal copy number variant on intracranial volume and basal ganglia

    Get PDF
    Carriers of large recurrent copy number variants (CNVs) have a higher risk of developing neurodevelopmental disorders. The 16p11.2 distal CNV predisposes carriers to e.g., autism spectrum disorder and schizophrenia. We compared subcortical brain volumes of 12 16p11.2 distal deletion and 12 duplication carriers to 6882 non-carriers from the large-scale brain Magnetic Resonance Imaging collaboration, ENIGMA-CNV. After stringent CNV calling procedures, and standardized FreeSurfer image analysis, we found negative dose-response associations with copy number on intracranial volume and on regional caudate, pallidum and putamen volumes (??=??0.71 to ?1.37; P?<?0.0005). In an independent sample, consistent results were obtained, with significant effects in the pallidum (??=??0.95, P?=?0.0042). The two data sets combined showed significant negative dose-response for the accumbens, caudate, pallidum, putamen and ICV (P?=?0.0032, 8.9?×?10?6, 1.7?×?10?9, 3.5?×?10?12 and 1.0?×?10?4, respectively). Full scale IQ was lower in both deletion and duplication carriers compared to non-carriers. This is the first brain MRI study of the impact of the 16p11.2 distal CNV, and we demonstrate a specific effect on subcortical brain structures, suggesting a neuropathological pattern underlying the neurodevelopmental syndromes.1000BRAINS is a populationbased cohort based on the Heinz-Nixdorf Recall Study and is supported in part by the German National Cohort. We thank the Heinz Nixdorf Foundation (Germany) for their generous support in terms of the Heinz Nixdorf Study. The HNR study is also supported by the German Ministry of Education and Science (FKZ 01EG940), and the German Research Council (DFG, ER 155/6-1). The authors are supported by the Initiative and Networking Fund of the Helmholtz Association (Svenja Caspers) and the European Union’s Horizon 2020 Research and Innovation Programme under Grant Agreement 7202070 (Human Brain Project SGA1; Katrin Amunts, Sven Cichon). This work was further supported by the German Federal Ministry of Education and Research (BMBF) through the Integrated Network 592 I. E. SĂžnderby et al. IntegraMent (Integrated Understanding of Causes and Mechanisms in Mental Disorders) under the auspices of the e:Med Program (grant 01ZX1314A to M.M.N. and S.C.), and by the Swiss National Science Foundation (SNSF, grant 156791 to S.C.). 16p.11.2 European Consortium: B.D. is supported by the Swiss National Science Foundation (NCCR Synapsy, project grant Nr 32003B_159780) and Foundation Synapsis. LREN is very grateful to the Roger De Spoelberch and Partridge Foundations for their generous financial support. This work was supported by grants from the Simons Foundation (SFARI274424) and the Swiss National Science Foundation (31003A_160203) to A.R. and S.J. Betula: The relevant Betula data collection and analyses were supported by a grant from the Knut & Alice Wallenberg (KAW) to L. Nyberg. Brainscale: the Brainscale study was supported by the Netherlands Organization for Scientific Research MagW 480-04-004 (Dorret Boomsma), 51.02.060 (Hilleke Hulshoff Pol), 668.772 (Dorret Boomsma & Hilleke Hulshoff Pol); NWO/SPI 56-464-14192 (Dorret Boomsma), the European Research Council (ERC-230374) (Dorret Boomsma), High Potential Grant Utrecht University (Hilleke Hulshoff Pol), NWO Brain and Cognition 433-09-220 (Hilleke Hulshoff Pol). Brain Imaging Genetics (BIG): This work makes use of the BIG database, first established in Nijmegen, The Netherlands, in 2007. This resource is now part of Cognomics (www.cognomics.nl), a joint initiative by researchers of the Donders Centre for Cognitive Neuroimaging, the Human Genetics and Cognitive Neuroscience departments of the Radboud university medical centre and the Max Planck Institute for Psycholinguistics in Nijmegen. The Cognomics Initiative has received supported from the participating departments and centres and from external grants, i.e., the Biobanking and Biomolecular Resources Research Infrastructure (the Netherlands) (BBMRI-NL), the Hersenstichting Nederland, and the Netherlands Organisation for Scientific Research (NWO). The research leading to these results also receives funding from the NWO Gravitation grant ‘Language in Interaction’, the European Community’s Seventh Framework Programme (FP7/2007–2013) under grant agreements n° 602450 (IMAGEMEND), n°278948 (TACTICS), and n°602805 (Aggressotype) as well as from the European Community’s Horizon 2020 programme under grant agreement n° 643051 (MiND) and from ERC-2010-AdG 268800-NEUROSCHEMA. In addition, the work was supported by a grant for the ENIGMA Consortium (grant number U54 EB020403) from the BD2K Initiative of a cross-NIH partnership. COBRE: This work was supported by a NIH COBRE Phase I grant (1P20RR021938, Lauriello, PI and 2P20GM103472, Calhoun, PI) awarded to the Mind Research Network. We wish to express our gratitude to numerous investigators who were either external consultants to the Cores and projects, mentors on the projects, members of the external advisory committee and members of the internal advisory committee. Decode: The research leading to these results has received financial contribution from the European Union’s Seventh Framework Programme (EU-FP7/2007–2013), EU-FP7 funded grant no. 602450 (IMAGEMEND) as well as support from the Innovative Medicines Initiative Joint Undertaking under grant agreement no.115300 (EUAIMS). DemGene: Norwegian Health Association and Research Council of Norway. Dublin: Work was supported by Science Foundation Ireland (SFI grant 12/IP/1359 to Gary Donohoe and SFI08/IN.1/B1916-Corvin to Aidan C Corvin) and the European Research Council (ERC-StG-2015-677467). EPIGEN-UK (SMS, CL): The work was partly undertaken at UCLH/UCL, which received a proportion of funding from the UK Department of Health’s NIHR Biomedical Research Centres funding scheme. We are grateful to the Wolfson Trust and the Epilepsy Society for supporting the Epilepsy Society MRI scanner, and the Epilepsy Society for supporting CL. Haavik: The work at the K.G.Jebsen center for neuropsychiatric disorders at the University of Bergen, Norway, was supported by Stiftelsen K.G. Jebsen, European Community’s Seventh Framework Program under grant agreement no 602805 and the H2020 Research and Innovation Program under grant agreement numbers 643051 and 667302. HUNT: The HUNT Study is a collaboration between HUNT Research Centre (Faculty of Medicine, Norwegian University of Science and Technology), Nord-TrĂžndelag County Council, Central Norway Health Authority, and the Norwegian Institute of Public Health. HUNT-MRI was funded by the Liaison Committee between the Central Norway Regional Health Authority and the Norwegian University of Science and Technology, and the Norwegian National Advisory Unit for functional MRI. IMAGEN: The work received support from the European Union-funded FP6Integrated Project IMAGEN (Reinforcement-related behaviour in normal brain function and psychopathology) (LSHM-CT- 2007-037286), the Horizon 2020 funded ERC Advanced Grant ‘STRATIFY’ (Brain network based stratification of reinforcement-related disorders) (695313), ERANID (Understanding the Interplay between Cultural, Biological and Subjective Factors in Drug Use Pathways) (PR-ST-0416-10004), BRIDGET (JPND: BRain Imaging, cognition Dementia and next generation GEnomics) (MR/N027558/1), the FP7 projects IMAGEMEND (602450; IMAging GEnetics for MENtal Disorders) and MATRICS (603016), the Innovative Medicine Initiative Project EUAIMS (115300), the Medical Research Council Grant ‘c-VEDA’ (Consortium on Vulnerability to Externalizing Disorders and Addictions) (MR/N000390/1), the Swedish Research Council FORMAS, the Medical Research Council, the National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, the BundesministeriumfĂŒr Bildung und Forschung (BMBF grants 01GS08152; 01EV0711; eMED SysAlc01ZX1311A; Forschungsnetz AERIAL), the Deutsche Forschungsgemeinschaft (DFG grants SM 80/7-1, SM 80/7-2, SFB 940/1). Further support was provided by grants from: ANR (project AF12-NEUR0008-01—WM2NA, and ANR-12-SAMA-0004), the Fondation de France, the Fondation pour la Recherche MĂ©dicale, the Mission InterministĂ©rielle de Lutte-contreles-Drogues-et-les-Conduites-Addictives (MILDECA), the AssistancePublique-HĂŽpitaux-de-Paris and INSERM (interface grant), Paris Sud University IDEX 2012; the National Institutes of Health, Science Foundation Ireland (16/ERCD/3797), USA (Axon, Testosterone and Mental Health during Adolescence; RO1 MH085772-01A1), and by NIH Consortium grant U54 EB020403, supported by a cross-NIH alliance that funds Big Data to Knowledge Centres of Excellence. MCIC: This work was supported primarily by the Department of Energy DE-FG02-99ER62764 through its support of the Mind Research Network and the consortium as well as by the National Association for Research in Schizophrenia and Affective Disorders (NARSAD) Young Investigator Award (to SE) as well as through the Blowitz-Ridgeway and Essel Foundations, and through NWO ZonMw TOP 91211021, the DFG research fellowship (to SE), the Mind Research Network, National Institutes of Health through NCRR 5 month-RR001066 (MGH General Clinical Research Center), NIMH K08 MH068540, the Biomedical Informatics Research Network with NCRR Supplements to P41 RR14075 (MGH), M01 RR 01066 (MGH), NIBIB R01EB006841 (MRN), R01EB005846 (MRN), 2R01 EB000840 (MRN), 1RC1MH089257 (MRN), as well as grant U24 RR021992. NCNG: this sample collection was supported by grants from the Bergen Research Foundation and the University of Bergen, the Dr Einar Martens Fund, the K.G. Jebsen Foundation, the Research Council of Norway, to SLH, VMS and TE. The Bergen group was supported by grants from the Western Norway Regional Health Authority (Grant 911593 to AL, Grant 911397 and 911687 to AJL). NESDA: Funding for NESDA was obtained from the Netherlands Organization for Scientific Research (Geestkracht program grant 10-000-1002); the Center for Medical Systems Biology (CSMB, NWO Genomics), Biobanking and Biomolecular Resources Research Infrastructure (BBMRI-NL), VU University’s Institutes for Health and Care Research (EMGO+) and Neuroscience Campus Amsterdam, University Medical Center Groningen, Leiden University Medical Center, National Institutes of Health (NIH, R01D0042157-01A, MH081802, 16p11.2 distal copy number variant brain structure 593 Grand Opportunity grants 1RC2 MH089951 and 1RC2 MH089995). Part of the genotyping and analyses were funded by the Genetic Association Information Network (GAIN) of the Foundation for the National Institutes of Health.Computing was supported by BiG Grid, the Dutch e-Science Grid, which is financially supported by NWO. NTR: The NTR study was supported by the Netherlands Organization for Scientific Research (NWO), MW904-61-193 (Eco de Geus & Dorret Boomsma), MaGW-nr: 400-07- 080 (Dennis van ‘t Ent), MagW 480-04-004 (Dorret Boomsma), NWO/SPI 56-464-14192 (Dorret Boomsma), the European Research Council, ERC-230374 (Dorret Boomsma), and Amsterdam Neuroscience. OATS: OATS (Older Australian Twins Study) was facilitated by access to Twins Research Australia, which is funded by a National Health and Medical Research Council (NHMRC) Enabling Grant 310667. OATS is also supported via a NHMRC/Australian Research Council Strategic Award (401162) and a NHMRC Project Grant (1045325). DNA extraction was performed by Genetic Repositories Australia, which was funded by a NHMRC Enabling Grant (401184). OATS genotyping was partly funded by a Commonwealth Scientific and Industrial Research Organisation Flagship Collaboration Fund Grant. PAFIP: PAFIP data were collected at the Hospital Universitario MarquĂ©s de Valdecilla, University of Cantabria, Santander, Spain, under the following grant support: Carlos III Health Institute PIE14/00031 and SAF2013-46292-R and SAF2015-71526-REDT. We wish to acknowledge IDIVAL Neuroimaging Unit for imaging acquirement and analysis.We want to particularly acknowledge the patients and the BioBankValdecilla (PT13/0010/0024) integrated in the Spanish National Biobanks Network for its collaboration. QTIM: The QTIM study was supported by grants from the US National Institute of Child Health and Human Development (R01 HD050735) and the Australian National Health and Medical Research Council (NHMRC) (486682, 1009064). Genotyping was supported by NHMRC (389875). Lachlan Strike is supported by an Australian Postgraduate Award (APA). AFM is supported by NHMRC CDF 1083656. We thank the twins and siblings for their participation, the many research assistants, as well as the radiographers, for their contribution to data collection and processing of the samples. SHIP: SHIP is part of the Community Medicine Research net of the University of Greifswald, Germany, which is funded by the Federal Ministry of Education and Research (grants no. 01ZZ9603, 01ZZ0103, 01ZZ0403 and 01ZZ0701), the Ministry of Cultural Affairs as well as the Social Ministry of the Federal State of Mecklenburg-West Pomerania, and the network ‘Greifswald Approach to Individualized Medicine (GANI_MED)’ funded by the Federal Ministry of Education and Research (grant 03IS2061A). Genome-wide data have been supported by the Federal Ministry of Education and Research (grant no. 03ZIK012) and a joint grant from Siemens Healthineers, Erlangen, Germany and the Federal State of Mecklenburg- West Pomerania. Whole-body MR imaging was supported by a joint grant from Siemens Healthineers, Erlangen, Germany and the Federal State of Mecklenburg West Pomerania. The University of Greifswald is a member of the CachĂ© Campus program of the InterSystems GmbH. StrokeMRI: StrokeMRI has been supported by the Research Council of Norway (249795), the South-Eastern Norway Regional Health Authority (2014097, 2015044, 2015073) and the Norwegian ExtraFoundation for Health and Rehabilitation. TOP: TOP is supported by the Research Council of Norway (223273, 213837, 249711), the South East Norway Health Authority (2017-112), the Kristian Gerhard Jebsen Stiftelsen (SKGJ‐MED‐008) and the European Community’s Seventh Framework Programme (FP7/2007–2013), grant agreement no. 602450 (IMAGEMEND). We acknowledge the technical support and service from the Genomics Core Facility at the Department of Clinical Science, the University of Bergen for the 16p11.2 European Consortium; for the ENIGMA-CNV working group Ida Elken SĂžnderby (NORMENT, K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway), Ómar GĂșstafsson (deCODE Genetics/Amgen, Reykjavik, Iceland), Nhat Trung Doan (NORMENT, K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway), Derrek Paul Hibar (Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, USA), (Janssen Research and Development, La Jolla, CA USA, Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, U. S.A), Sandra Martin-Brevet (Service of Medical Genetics, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 46, 1011 Lausanne, Switzerland), Abdel Abdellaoui (Biological Psychology, Vrije Universiteit Amsterdam, van Boechorststraat 1, 1081 BT Amsterdam, The Netherlands), (Department of Psychiatry, Academic Medical Center, Amsterdam, the Netherlands), David Ames (National Ageing Research Institute, Melbourne, Australia), (Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, Australia), Katrin Amunts (Institute of Neuroscience and Medicine (INM-1), Research Centre Juelich, Wilhelm-Johnen-Str., 52425 Juelich, Germany), (C. and O. Vogt Institute for Brain Research, Medical Faculty, University of Dusseldorf, Merowingerplatz 1A, 40225 Dusseldorf, Germany), (JARA-BRAIN, Juelich-Aachen Research Alliance, Wilhelm-Johnen-Str., 52425 Juelich, Germany), Michael Andersson (UmeĂ„ Center for Functional Brain Imaging (UFBI), UmeĂ„ University, 90187 UmeĂ„, Sweden), Nicola J. Armstrong (Mathematics and Statistics, Murdoch University, Perth, Australia), Manon Bernard (The Hospital for Sick Children, University of Toronto, Toronto, M5G 1X8, Canada), Nicholas Blackburn (South Texas Diabetes and Obesity Institute, Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, One West University Blvd., 78520 Brownsville, TX, USA), John Blangero (South Texas Diabetes and Obesity Institute, Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, One West University Blvd., 78520 Brownsville, TX, USA), Dorret I Boomsma (Netherlands Twin Register, Vrije Universiteit, van der Boechorststraat 1, 1081BT Amsterdam, Netherlands), Janita Bralten (Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands), Hans-Richard Brattbak (Department of Clinical Science, University of Bergen, Bergen, Norway), (Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway), Henry Brodaty (Centre for Healthy Brain Ageing and Dementia Collaborative Research Centre, UNSW, Sydney, Australia), Rachel M. Brouwer (Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands), Robin BĂŒlow (Department of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany), Vince Calhoun (The Mind Research Network, The University of New Mexico, Albuquerque, NM), Svenja Caspers (Institute of Neuroscience and Medicine (INM-1), Research Centre Juelich, Wilhelm-Johnen-Str., 52425 Juelich, Germany), (C. and O. Vogt Institute for Brain Research, Medical Faculty, University of Dusseldorf, Merowingerplatz 1A, 40225 Dusseldorf, Germany), (JARA-BRAIN, Juelich-Aachen Research Alliance, Wilhelm-Johnen-Str., 52425 Juelich, Germany), Gianpiero Cavalleri (The Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland), Chi-Hua Chen (Department of Radiology, University of California San Diego, La Jolla, USA), (Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, USA), Sven Cichon (Institute of Neuroscience and Medicine (INM-1), Structural and Functional Organisation of the Brain, Genomic Imaging, Research Centre Juelich, Leo-Brandt-Strasse 5, 52425 JĂŒlich, Germany), (Human Genomics Research Group, 594 I. E. SĂžnderby et al. Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland), (Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland), Simone Ciufolini (Psychosis Studies, Insitute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespingy Park, SE5 8AF London, United Kingdom), Aiden Corvin (Neuropsychiatric Genetics Research Group, Discipline of Psychiatry, School of Medicine, Trinity College Dublin, Dublin 2, Ireland.), Benedicto Crespo-Facorro (Department of Medicine and Psychiatry, University Hospital Marque?s de Valdecilla, School of Medicine, University of Cantabria-IDIVAL, 39008 Santander, Spain), (CIBERSAM (Centro InvestigaciĂłn BiomĂ©dica en Red Salud Mental), Santander, 39011, Spain), Joanne E. Curran (South Texas Diabetes and Obesity Institute, Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, One West University Blvd., 78520 Brownsville, TX, USA), Anders M Dale (Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, USA), Shareefa Dalvie (Department of Psychiatry and Mental Health, Anzio Road, 7925 Cape Town, South Africa), Paola Dazzan (Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, SE5 8AF London, United Kingdom), (National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, United Kingdom), Eco JC de Geus (Department of Biological Psychology, Behavioral and Movement Sciences, Vrije Universiteit, van der Boechorststraat 1, 1081 BT Amsterdam, Netherlands), (Amsterdam Neuroscience, VU University medical center, van der Boechorststraat 1, 1081 BT Amsterdam, NH, Netherlands), Greig I. de Zubicaray (Faculty of Health and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia), Sonja M.C. de Zwarte (Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands), Norman Delanty (The Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland), (Imaging of Dementia and Aging (IDeA) Laboratory, Department of Neurology and Center for Neuroscience, University of California at Davis, 4860 Y Street, Suite 3700, Sacramento, California 95817, USA.), Anouk den Braber (Department of Biological Psychology, Behavioral and Movement Sciences, Vrije Universiteit, van der Boechorststraat 1, 1081 BT Amsterdam, Netherlands), (Alzheimer Center and Department of Neurology, VU University Medical Center, De Boelelaan 1105, 1081HV Amsterdam Amsterdam, Amsterdam), Sylvane DesriviĂšres (Medical Research Council - Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom), Gary Donohoe (Cognitive Genetics

    Multi-level analysis of the gut-brain axis shows autism spectrum disorder-associated molecular and microbial profiles

    Get PDF
    Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by heterogeneous cognitive, behavioral and communication impairments. Disruption of the gut-brain axis (GBA) has been implicated in ASD although with limited reproducibility across studies. In this study, we developed a Bayesian differential ranking algorithm to identify ASD-associated molecular and taxa profiles across 10 cross-sectional microbiome datasets and 15 other datasets, including dietary patterns, metabolomics, cytokine profiles and human brain gene expression profiles. We found a functional architecture along the GBA that correlates with heterogeneity of ASD phenotypes, and it is characterized by ASD-associated amino acid, carbohydrate and lipid profiles predominantly encoded by microbial species in the genera Prevotella, Bifidobacterium, Desulfovibrio and Bacteroides and correlates with brain gene expression changes, restrictive dietary patterns and pro-inflammatory cytokine profiles. The functional architecture revealed in age-matched and sex-matched cohorts is not present in sibling-matched cohorts. We also show a strong association between temporal changes in microbiome composition and ASD phenotypes. In summary, we propose a framework to leverage multi-omic datasets from well-defined cohorts and investigate how the GBA influences ASD

    Developmental trajectories of neuroanatomical alterations associated with the 16p11.2 Copy Number Variations

    Get PDF

    Defining the Effect of the 16p11.2 Duplication on Cognition, Behavior, and Medical Comorbidities

    Full text link
    IMPORTANCE The 16p11.2 BP4-BP5 duplication is the copy number variant most frequently associated with autism spectrum disorder (ASD), schizophrenia, and comorbidities such as decreased body mass index (BMI). OBJECTIVES To characterize the effects of the 16p11.2 duplication on cognitive, behavioral, medical, and anthropometric traits and to understand the specificity of these effects by systematically comparing results in duplication carriers and reciprocal deletion carriers, who are also at risk for ASD. DESIGN, SETTING, AND PARTICIPANTS This international cohort study of 1006 study participants compared 270 duplication carriers with their 102 intrafamilial control individuals, 390 reciprocal deletion carriers, and 244 deletion controls from European and North American cohorts. Data were collected from August 1, 2010, to May 31, 2015 and analyzed from January 1 to August 14, 2015. Linear mixed models were used to estimate the effect of the duplication and deletion on clinical traits by comparison with noncarrier relatives. MAIN OUTCOMES AND MEASURES Findings on the Full-Scale IQ (FSIQ), Nonverbal IQ, and Verbal IQ; the presence of ASD or other DSM-IV diagnoses; BMI; head circumference; and medical data. RESULTS Among the 1006 study participants, the duplication was associated with a mean FSIQ score that was lower by 26.3 points between proband carriers and noncarrier relatives and a lower mean FSIQ score (16.2-11.4 points) in nonproband carriers. The mean overall effect of the deletion was similar (-22.1 points; P 100) compared with the deletion group (P < .001). Parental FSIQ predicted part of this variation (approximately 36.0% in hereditary probands). Although the frequency of ASD was similar in deletion and duplication proband carriers (16.0% and 20.0%, respectively), the FSIQ was significantly lower (by 26.3 points) in the duplication probands with ASD. There also were lower head circumference and BMI measurements among duplication carriers, which is consistent with the findings of previous studies. CONCLUSIONS AND RELEVANCE The mean effect of the duplication on cognition is similar to that of the reciprocal deletion, but the variance in the duplication is significantly higher, with severe and mild subgroups not observed with the deletion. These results suggest that additional genetic and familial factors contribute to this variability. Additional studies will be necessary to characterize the predictors of cognitive deficits

    Effects of eight neuropsychiatric copy number variants on human brain structure

    Get PDF
    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

    The Effect of Uncertainty on Pain Decisions for Self and Others

    No full text
    Estimating others’ pain is a challenging inferential process, associated with a high degree of uncertainty. The present study exploited models of probabilistic decision-making to investigate how uncertainty influences the assessment of another’s pain. We engaged sixty-three dyads (43 strangers and 20 romantic couples) in a task where individual choices affected the pain delivered to either oneself (the agent) or the other member of the dyad. At each trial, agents were presented with cues predicting a given pain intensity with an associated probability of occurrences. Agents chose either a sure (mild decrease of pain) or risky (50% chance of avoiding pain altogether) management option, following which they were asked to bid on their choice. A heat stimulation was then issued to the target (self or other), whose intensity was rated. We found that, the higher the expected pain, the more risk-averse agents became, in line with findings in value-based decision-making. Furthermore, agents gambled less on another individual’s pain (especially strangers) and placed higher bids on pain relief than they did for themselves. Most critically, the uncertainty associated with expected pain dampened ratings made for strangers’ pain. This contrasted with the effect on an agent’s own pain, for which risk had a marginal hyperalgesic effect. Overall, our results suggested that risk selectively affects decision-making on a stranger’s suffering, both at the level of assessment and treatment selection, by 1) leading to underestimation, 2) privileging sure options, and 3) altruistically allocating more money to insure the treatment’s success
    corecore