151
|
Yuan S, Li H, Xie J, Sun X. Quantitative Trait Module-Based Genetic Analysis of Alzheimer's Disease. Int J Mol Sci 2019; 20:E5912. [PMID: 31775305 PMCID: PMC6928939 DOI: 10.3390/ijms20235912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 01/02/2023] Open
Abstract
The pathological features of Alzheimer's Disease (AD) first appear in the medial temporal lobe and then in other brain structures with the development of the disease. In this work, we investigated the association between genetic loci and subcortical structure volumes of AD on 393 samples in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Brain subcortical structures were clustered into modules using Pearson's correlation coefficient of volumes across all samples. Module volumes were used as quantitative traits to identify not only the main effect loci but also the interactive effect loci for each module. Thirty-five subcortical structures were clustered into five modules, each corresponding to a particular brain structure/area, including the limbic system (module I), the corpus callosum (module II), thalamus-cerebellum-brainstem-pallidum (module III), the basal ganglia neostriatum (module IV), and the ventricular system (module V). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment results indicate that the gene annotations of the five modules were distinct, with few overlaps between different modules. We identified several main effect loci and interactive effect loci for each module. All these loci are related to the function of module structures and basic biological processes such as material transport and signal transduction.
Collapse
Affiliation(s)
| | | | | | - Xiao Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; (S.Y.)
| |
Collapse
|
152
|
Yu L, Boyle PA, Dawe RJ, Bennett DA, Arfanakis K, Schneider JA. Contribution of TDP and hippocampal sclerosis to hippocampal volume loss in older-old persons. Neurology 2019; 94:e142-e152. [PMID: 31757868 DOI: 10.1212/wnl.0000000000008679] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To investigate the contribution of Alzheimer disease (AD) vs non-AD neuropathologies to hippocampal atrophy. METHODS The Religious Orders Study and Rush Memory and Aging Project are clinicopathologic cohort studies of aging. The current study included 547 participants who had undergone brain autopsy and postmortem hippocampal volume measurement by November 1, 2018. Hippocampal volume was measured with postmortem MRI via a 3D region of interest applied to the hippocampal formation. Neuropathologies were measured via uniform structured evaluations. Linear regression analyses estimated the proportion of variance of hippocampal volume attributable to AD and non-AD neuropathologies. RESULTS The average age at death was 90 years, and the average hippocampal volume was 2.1 mL. AD, transactive response DNA-binding protein 43 (TDP), hippocampal sclerosis (HS), and atherosclerosis were associated with hippocampal volume. After demographics and total hemisphere volume were controlled for, 7.0% of the variance (95% bootstrapped confidence interval [CI] 4.3%-10.5%) of hippocampal volume was attributable to AD pathology. TDP/HS explained an additional 4.5% (95% CI 2.2%-7.6%). Among individuals with Alzheimer dementia (n = 232), 3.1% (95% CI 0.6%-7.7%) of the variance was attributable to AD pathology, and TDP/HS explained an additional 6.1% (95% CI 2.2%-11.6%). Among those without Alzheimer dementia (n = 307), 3.2% (95% CI 0.9%-7.3%) of the variance was attributable to AD pathology, and TDP/HS explained an additional 1.1%, which did not reach statistical significance. Lewy bodies and vascular diseases had modest contribution to the variance of hippocampal volume. CONCLUSIONS Both AD and TDP/HS contribute to hippocampal volume loss in older-old persons, with TDP/HS more strongly associated with hippocampal volume than AD in Alzheimer dementia.
Collapse
Affiliation(s)
- Lei Yu
- From the Rush Alzheimer's Disease Center (L.Y., P.A.B., R.J.D., D.A.B., K.A., J.A.S.), Department of Neurological Sciences (L.Y., D.A.B., J.A.S.), Department of Behavioral Sciences (P.A.B.), Department of Diagnostic Radiology and Nuclear Medicine (R.J.D., K.A.), and Department of Pathology (J.A.S.), Rush University Medical Center; and Department of Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago.
| | - Patricia A Boyle
- From the Rush Alzheimer's Disease Center (L.Y., P.A.B., R.J.D., D.A.B., K.A., J.A.S.), Department of Neurological Sciences (L.Y., D.A.B., J.A.S.), Department of Behavioral Sciences (P.A.B.), Department of Diagnostic Radiology and Nuclear Medicine (R.J.D., K.A.), and Department of Pathology (J.A.S.), Rush University Medical Center; and Department of Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago
| | - Robert J Dawe
- From the Rush Alzheimer's Disease Center (L.Y., P.A.B., R.J.D., D.A.B., K.A., J.A.S.), Department of Neurological Sciences (L.Y., D.A.B., J.A.S.), Department of Behavioral Sciences (P.A.B.), Department of Diagnostic Radiology and Nuclear Medicine (R.J.D., K.A.), and Department of Pathology (J.A.S.), Rush University Medical Center; and Department of Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago
| | - David A Bennett
- From the Rush Alzheimer's Disease Center (L.Y., P.A.B., R.J.D., D.A.B., K.A., J.A.S.), Department of Neurological Sciences (L.Y., D.A.B., J.A.S.), Department of Behavioral Sciences (P.A.B.), Department of Diagnostic Radiology and Nuclear Medicine (R.J.D., K.A.), and Department of Pathology (J.A.S.), Rush University Medical Center; and Department of Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago
| | - Konstantinos Arfanakis
- From the Rush Alzheimer's Disease Center (L.Y., P.A.B., R.J.D., D.A.B., K.A., J.A.S.), Department of Neurological Sciences (L.Y., D.A.B., J.A.S.), Department of Behavioral Sciences (P.A.B.), Department of Diagnostic Radiology and Nuclear Medicine (R.J.D., K.A.), and Department of Pathology (J.A.S.), Rush University Medical Center; and Department of Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago
| | - Julie A Schneider
- From the Rush Alzheimer's Disease Center (L.Y., P.A.B., R.J.D., D.A.B., K.A., J.A.S.), Department of Neurological Sciences (L.Y., D.A.B., J.A.S.), Department of Behavioral Sciences (P.A.B.), Department of Diagnostic Radiology and Nuclear Medicine (R.J.D., K.A.), and Department of Pathology (J.A.S.), Rush University Medical Center; and Department of Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago
| |
Collapse
|
153
|
Zhao B, Luo T, Li T, Li Y, Zhang J, Shan Y, Wang X, Yang L, Zhou F, Zhu Z, Zhu H. Genome-wide association analysis of 19,629 individuals identifies variants influencing regional brain volumes and refines their genetic co-architecture with cognitive and mental health traits. Nat Genet 2019; 51:1637-1644. [PMID: 31676860 PMCID: PMC6858580 DOI: 10.1038/s41588-019-0516-6] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 09/23/2019] [Indexed: 12/19/2022]
Abstract
Volumetric variations of the human brain are heritable and are associated with many brain-related complex traits. Here we performed genome-wide association studies (GWAS) of 101 brain volumetric phenotypes using the UK Biobank sample including 19,629 participants. GWAS identified 365 independent genetic variants exceeding a significance threshold of 4.9 × 10-10, adjusted for testing multiple phenotypes. A gene-based association study found 157 associated genes (124 new), and functional gene mapping analysis linked 146 additional genes. Many of the discovered genetic variants and genes have previously been implicated in cognitive and mental health traits. Through genome-wide polygenic-risk-score prediction, more than 6% of the phenotypic variance (P = 3.13 × 10-24) in four other independent studies could be explained by the UK Biobank GWAS results. In conclusion, our study identifies many new genetic associations at the variant, locus and gene levels and advances our understanding of the pleiotropy and genetic co-architecture between brain volumes and other traits.
Collapse
Affiliation(s)
- Bingxin Zhao
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tianyou Luo
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jingwen Zhang
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Yue Shan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Liuqing Yang
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Fan Zhou
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ziliang Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
154
|
Short AK, Baram TZ. Early-life adversity and neurological disease: age-old questions and novel answers. Nat Rev Neurol 2019; 15:657-669. [PMID: 31530940 PMCID: PMC7261498 DOI: 10.1038/s41582-019-0246-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
Neurological illnesses, including cognitive impairment, memory decline and dementia, affect over 50 million people worldwide, imposing a substantial burden on individuals and society. These disorders arise from a combination of genetic, environmental and experiential factors, with the latter two factors having the greatest impact during sensitive periods in development. In this Review, we focus on the contribution of adverse early-life experiences to aberrant brain maturation, which might underlie vulnerability to cognitive brain disorders. Specifically, we draw on recent robust discoveries from diverse disciplines, encompassing human studies and experimental models. These discoveries suggest that early-life adversity, especially in the perinatal period, influences the maturation of brain circuits involved in cognition. Importantly, new findings suggest that fragmented and unpredictable environmental and parental signals comprise a novel potent type of adversity, which contributes to subsequent vulnerabilities to cognitive illnesses via mechanisms involving disordered maturation of brain 'wiring'.
Collapse
Affiliation(s)
- Annabel K Short
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA.
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA.
- Departments of Neurology, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
155
|
Satizabal CL, Adams HHH, Hibar DP, White CC, Knol MJ, Stein JL, Scholz M, Sargurupremraj M, Jahanshad N, Roshchupkin GV, Smith AV, Bis JC, Jian X, Luciano M, Hofer E, Teumer A, van der Lee SJ, Yang J, Yanek LR, Lee TV, Li S, Hu Y, Koh JY, Eicher JD, Desrivières S, Arias-Vasquez A, Chauhan G, Athanasiu L, Rentería ME, Kim S, Hoehn D, Armstrong NJ, Chen Q, Holmes AJ, den Braber A, Kloszewska I, Andersson M, Espeseth T, Grimm O, Abramovic L, Alhusaini S, Milaneschi Y, Papmeyer M, Axelsson T, Ehrlich S, Roiz-Santiañez R, Kraemer B, Håberg AK, Jones HJ, Pike GB, Stein DJ, Stevens A, Bralten J, Vernooij MW, Harris TB, Filippi I, Witte AV, Guadalupe T, Wittfeld K, Mosley TH, Becker JT, Doan NT, Hagenaars SP, Saba Y, Cuellar-Partida G, Amin N, Hilal S, Nho K, Mirza-Schreiber N, Arfanakis K, Becker DM, Ames D, Goldman AL, Lee PH, Boomsma DI, Lovestone S, Giddaluru S, Le Hellard S, Mattheisen M, Bohlken MM, Kasperaviciute D, Schmaal L, Lawrie SM, Agartz I, Walton E, Tordesillas-Gutierrez D, Davies GE, Shin J, Ipser JC, Vinke LN, Hoogman M, Jia T, Burkhardt R, Klein M, Crivello F, Janowitz D, Carmichael O, Haukvik UK, Aribisala BS, Schmidt H, et alSatizabal CL, Adams HHH, Hibar DP, White CC, Knol MJ, Stein JL, Scholz M, Sargurupremraj M, Jahanshad N, Roshchupkin GV, Smith AV, Bis JC, Jian X, Luciano M, Hofer E, Teumer A, van der Lee SJ, Yang J, Yanek LR, Lee TV, Li S, Hu Y, Koh JY, Eicher JD, Desrivières S, Arias-Vasquez A, Chauhan G, Athanasiu L, Rentería ME, Kim S, Hoehn D, Armstrong NJ, Chen Q, Holmes AJ, den Braber A, Kloszewska I, Andersson M, Espeseth T, Grimm O, Abramovic L, Alhusaini S, Milaneschi Y, Papmeyer M, Axelsson T, Ehrlich S, Roiz-Santiañez R, Kraemer B, Håberg AK, Jones HJ, Pike GB, Stein DJ, Stevens A, Bralten J, Vernooij MW, Harris TB, Filippi I, Witte AV, Guadalupe T, Wittfeld K, Mosley TH, Becker JT, Doan NT, Hagenaars SP, Saba Y, Cuellar-Partida G, Amin N, Hilal S, Nho K, Mirza-Schreiber N, Arfanakis K, Becker DM, Ames D, Goldman AL, Lee PH, Boomsma DI, Lovestone S, Giddaluru S, Le Hellard S, Mattheisen M, Bohlken MM, Kasperaviciute D, Schmaal L, Lawrie SM, Agartz I, Walton E, Tordesillas-Gutierrez D, Davies GE, Shin J, Ipser JC, Vinke LN, Hoogman M, Jia T, Burkhardt R, Klein M, Crivello F, Janowitz D, Carmichael O, Haukvik UK, Aribisala BS, Schmidt H, Strike LT, Cheng CY, Risacher SL, Pütz B, Fleischman DA, Assareh AA, Mattay VS, Buckner RL, Mecocci P, Dale AM, Cichon S, Boks MP, Matarin M, Penninx BWJH, Calhoun VD, Chakravarty MM, Marquand AF, Macare C, Kharabian Masouleh S, Oosterlaan J, Amouyel P, Hegenscheid K, Rotter JI, Schork AJ, Liewald DCM, de Zubicaray GI, Wong TY, Shen L, Sämann PG, Brodaty H, Roffman JL, de Geus EJC, Tsolaki M, Erk S, van Eijk KR, Cavalleri GL, van der Wee NJA, McIntosh AM, Gollub RL, Bulayeva KB, Bernard M, Richards JS, Himali JJ, Loeffler M, Rommelse N, Hoffmann W, Westlye LT, Valdés Hernández MC, Hansell NK, van Erp TGM, Wolf C, Kwok JBJ, Vellas B, Heinz A, Olde Loohuis LM, Delanty N, Ho BC, Ching CRK, Shumskaya E, Singh B, Hofman A, van der Meer D, Homuth G, Psaty BM, Bastin ME, Montgomery GW, Foroud TM, Reppermund S, Hottenga JJ, Simmons A, Meyer-Lindenberg A, Cahn W, Whelan CD, van Donkelaar MMJ, Yang Q, Hosten N, Green RC, Thalamuthu A, Mohnke S, Hulshoff Pol HE, Lin H, Jack CR, Schofield PR, Mühleisen TW, Maillard P, Potkin SG, Wen W, Fletcher E, Toga AW, Gruber O, Huentelman M, Davey Smith G, Launer LJ, Nyberg L, Jönsson EG, Crespo-Facorro B, Koen N, Greve DN, Uitterlinden AG, Weinberger DR, Steen VM, Fedko IO, Groenewold NA, Niessen WJ, Toro R, Tzourio C, Longstreth WT, Ikram MK, Smoller JW, van Tol MJ, Sussmann JE, Paus T, Lemaître H, Schroeter ML, Mazoyer B, Andreassen OA, Holsboer F, Depondt C, Veltman DJ, Turner JA, Pausova Z, Schumann G, van Rooij D, Djurovic S, Deary IJ, McMahon KL, Müller-Myhsok B, Brouwer RM, Soininen H, Pandolfo M, Wassink TH, Cheung JW, Wolfers T, Martinot JL, Zwiers MP, Nauck M, Melle I, Martin NG, Kanai R, Westman E, Kahn RS, Sisodiya SM, White T, Saremi A, van Bokhoven H, Brunner HG, Völzke H, Wright MJ, van 't Ent D, Nöthen MM, Ophoff RA, Buitelaar JK, Fernández G, Sachdev PS, Rietschel M, van Haren NEM, Fisher SE, Beiser AS, Francks C, Saykin AJ, Mather KA, Romanczuk-Seiferth N, Hartman CA, DeStefano AL, Heslenfeld DJ, Weiner MW, Walter H, Hoekstra PJ, Nyquist PA, Franke B, Bennett DA, Grabe HJ, Johnson AD, Chen C, van Duijn CM, Lopez OL, Fornage M, Wardlaw JM, Schmidt R, DeCarli C, De Jager PL, Villringer A, Debette S, Gudnason V, Medland SE, Shulman JM, Thompson PM, Seshadri S, Ikram MA. Genetic architecture of subcortical brain structures in 38,851 individuals. Nat Genet 2019; 51:1624-1636. [PMID: 31636452 PMCID: PMC7055269 DOI: 10.1038/s41588-019-0511-y] [Show More Authors] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 09/05/2019] [Indexed: 12/15/2022]
Abstract
Subcortical brain structures are integral to motion, consciousness, emotions and learning. We identified common genetic variation related to the volumes of the nucleus accumbens, amygdala, brainstem, caudate nucleus, globus pallidus, putamen and thalamus, using genome-wide association analyses in almost 40,000 individuals from CHARGE, ENIGMA and UK Biobank. We show that variability in subcortical volumes is heritable, and identify 48 significantly associated loci (40 novel at the time of analysis). Annotation of these loci by utilizing gene expression, methylation and neuropathological data identified 199 genes putatively implicated in neurodevelopment, synaptic signaling, axonal transport, apoptosis, inflammation/infection and susceptibility to neurological disorders. This set of genes is significantly enriched for Drosophila orthologs associated with neurodevelopmental phenotypes, suggesting evolutionarily conserved mechanisms. Our findings uncover novel biology and potential drug targets underlying brain development and disease.
Collapse
Affiliation(s)
- Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA.
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA.
- The Framingham Heart Study, Framingham, MA, USA.
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
- Department of Clinical Genetics, Erasmus MC, Rotterdam, the Netherlands
| | - Derrek P Hibar
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Charles C White
- Cell Circuits Program, Broad Institute, Cambridge, MA, USA
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Jason L Stein
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE: The Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Muralidharan Sargurupremraj
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, Bordeaux, France
| | - Neda Jahanshad
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gennady V Roshchupkin
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
- Department of Medical Informatics, Erasmus MC, Rotterdam, the Netherlands
| | - Albert V Smith
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Xueqiu Jian
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michelle Luciano
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | | | - Jingyun Yang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Lisa R Yanek
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tom V Lee
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Shuo Li
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Jia Yu Koh
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - John D Eicher
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung and Blood Institute, Framingham, MA, USA
| | - Sylvane Desrivières
- MRC-SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alejandro Arias-Vasquez
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Ganesh Chauhan
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, Bordeaux, France
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Lavinia Athanasiu
- CoE NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- CoE NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Miguel E Rentería
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sungeun Kim
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David Hoehn
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Nicola J Armstrong
- Mathematics and Statistics, Murdoch University, Perth, Western Australia, Australia
| | - Qiang Chen
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Avram J Holmes
- Department of Psychology, Yale University, New Haven, CT, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Anouk den Braber
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Netherlands Twin Register, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, VU Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Micael Andersson
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Umeå Centre for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Thomas Espeseth
- CoE NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Oliver Grimm
- Central Institute of Mental Health, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Lucija Abramovic
- Department of Psychiatry, UMC Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Saud Alhusaini
- The Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Martina Papmeyer
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
- Division of Systems Neuroscience of Psychopathology, Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| | - Tomas Axelsson
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Stefan Ehrlich
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Roberto Roiz-Santiañez
- Department of Psychiatry, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria-IDIVAL, Santander, Spain
- Department of Medicine, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria-IDIVAL, Santander, Spain
- Centro Investigación Biomédica en Red Salud Mental, Santander, Spain
| | - Bernd Kraemer
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, Germany
| | - Asta K Håberg
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Radiology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Hannah J Jones
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and University of Bristol, Bristol, UK
| | - G Bruce Pike
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Dan J Stein
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Cape Town, South Africa
| | - Allison Stevens
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Janita Bralten
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Irina Filippi
- INSERM, Research Unit 1000 'Neuroimaging and Psychiatry', Paris Saclay University and Paris Descartes University-DIGITEO Labs, Gif sur Yvette, France
| | - A Veronica Witte
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Faculty of Medicine, CRC 1052 'Obesity Mechanisms', University of Leipzig, Leipzig, Germany
| | - Tulio Guadalupe
- International Max Planck Research School for Language Sciences, Nijmegen, the Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Katharina Wittfeld
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases, Greifswald, Germany
| | - Thomas H Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - James T Becker
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nhat Trung Doan
- CoE NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Saskia P Hagenaars
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Yasaman Saba
- Research Unit-Genetic Epidemiology, Gottfried Schatz Research Centre for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | | | - Najaf Amin
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Saima Hilal
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Center, National University Health System, Singapore, Singapore
| | - Kwangsik Nho
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nazanin Mirza-Schreiber
- Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Diane M Becker
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David Ames
- Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, Victoria, Australia
- National Ageing Research Institute, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | | | - Phil H Lee
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Lexington, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Netherlands Twin Register, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, the Netherlands
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, UK
- NIHR Dementia Biomedical Research Unit, King's College London, London, UK
| | - Sudheer Giddaluru
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Stephanie Le Hellard
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Manuel Mattheisen
- Centre for integrated Sequencing, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
- Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Marc M Bohlken
- Department of Psychiatry, UMC Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Dalia Kasperaviciute
- UCL Queen Square Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Bucks, UK
| | - Lianne Schmaal
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, Melbourne, Victoria, Australia
| | - Stephen M Lawrie
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Ingrid Agartz
- CoE NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
- Department of Research and Development, Diakonhjemmet Hospital, Oslo, Norway
| | - Esther Walton
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
- Department of Psychology, University of Bath, Bath, UK
| | - Diana Tordesillas-Gutierrez
- Centro Investigación Biomédica en Red Salud Mental, Santander, Spain
- Neuroimaging Unit, Technological Facilities, Valdecilla Biomedical Research Institute IDIVAL, Santander, Spain
| | | | - Jean Shin
- Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan C Ipser
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Louis N Vinke
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Martine Hoogman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Tianye Jia
- MRC-SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ralph Burkhardt
- LIFE: The Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Marieke Klein
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Fabrice Crivello
- Neurodegeneratives Diseases Institute, CNRS UMR 5293, Université de Bordeaux, Bordeaux, France
| | - Deborah Janowitz
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany
| | | | - Unn K Haukvik
- CoE NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Adult Psychiatry, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Benjamin S Aribisala
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh, UK
- Department of Computer Science, Lagos State University, Ojo, Nigeria
| | - Helena Schmidt
- Research Unit-Genetic Epidemiology, Gottfried Schatz Research Centre for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Lachlan T Strike
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
| | - Shannon L Risacher
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Benno Pütz
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Debra A Fleischman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Amelia A Assareh
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Venkata S Mattay
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Randy L Buckner
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Patrizia Mecocci
- Section of Gerontology and Geriatrics, Department of Medicine, University of Perugia, Perugia, Italy
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, University of California, San Diego, San Diego, CA, USA
- Department of Cognitive Sciences, University of California, San Diego, San Diego, CA, USA
- Department of Neurosciences, University of California. San Diego, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, San Diego, CA, USA
- Department of Radiology, University of California, San Diego, San Diego, CA, USA
| | - Sven Cichon
- Division of Medical Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Institute for Neuroscience and Medicine: Structural and Functional Organisation of the Brain (INM-1), Research Centre Jülich, Jülich, Germany
| | - Marco P Boks
- Department of Psychiatry, UMC Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Mar Matarin
- UCL Queen Square Institute of Neurology, London, UK
- Reta Lila Weston Institute, UCL Institute of Neurology, London, UK
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Vince D Calhoun
- Department of ECE, University of New Mexico, Albuquerque, NM, USA
- The Mind Research Network and LBERI, Albuquerque, NM, USA
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science, Georgia State University, Atlanta, GA, USA
| | - M Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Québec, Canada
- Departments of Psychiatry and Biological and Biomedical Engineering, McGill University, Montreal, Québec, Canada
| | - Andre F Marquand
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands
| | - Christine Macare
- MRC-SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Shahrzad Kharabian Masouleh
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute for Neuroscience and Medicine: Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
| | - Jaap Oosterlaan
- Clinical Neuropsychology Section, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Emma Neuroscience Group, Department of Pediatrics, Emma Children's Hospital, Amsterdam Reproduction & Development, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Philippe Amouyel
- LabEx DISTALZ-U1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille, France
- Inserm U1167, Lille, France
- Centre Hospitalier Universitaire Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Katrin Hegenscheid
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Andrew J Schork
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Roskilde, Denmark
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - David C M Liewald
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Greig I de Zubicaray
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Academic Medicine Research Institute, Duke-NUS Medical School, Singapore, Singapore
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Dementia Centre for Research Collaboration, UNSW, Sydney, New South Wales, Australia
| | - Joshua L Roffman
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Netherlands Twin Register, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, the Netherlands
| | - Magda Tsolaki
- 1st Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Susanne Erk
- Division of Mind and Brain Research, D, Corporate member of Freie Universität Berliepartment of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Kristel R van Eijk
- Brain Center Rudolf Magnus, Human Neurogenetics Unit, UMC Utrecht, Utrecht, the Netherlands
| | - Gianpiero L Cavalleri
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Nic J A van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden University Medical Center, Leiden, the Netherlands
| | - Andrew M McIntosh
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Randy L Gollub
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kazima B Bulayeva
- Department of Evolution and Genetics, Dagestan State University, Makhachkala, Russia
| | - Manon Bernard
- Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer S Richards
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- University Medical Center Groningen, Department of Psychiatry, University of Groningen, Groningen, the Netherlands
| | - Jayandra J Himali
- The Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE: The Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Nanda Rommelse
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands
| | - Wolfgang Hoffmann
- German Center for Neurodegenerative Diseases, Greifswald, Germany
- Section Epidemiology of Health Care and Community Health, Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Lars T Westlye
- CoE NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- CoE NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Maria C Valdés Hernández
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Narelle K Hansell
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Theo G M van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA
| | - Christiane Wolf
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - John B J Kwok
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- University of New South Wales, Sydney, New South Wales, Australia
| | - Bruno Vellas
- Department of Internal Medicine, INSERM U 1027, University of Toulouse, Toulouse, France
- Department of Geriatric Medicine, INSERM U 1027, University of Toulouse, Toulouse, France
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Loes M Olde Loohuis
- Center for Neurobehavioral Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Norman Delanty
- The Royal College of Surgeons in Ireland, Dublin, Ireland
- Neurology Division, Beaumont Hospital, Dublin, Ireland
| | - Beng-Choon Ho
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Christopher R K Ching
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Interdepartmental Neuroscience Graduate Program, UCLA School of Medicine, Los Angeles, CA, USA
| | - Elena Shumskaya
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands
| | - Baljeet Singh
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Dennis van der Meer
- CoE NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- CoE NORMENT, 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
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Kaiser Permanent Washington Health Research Institute, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
| | - Mark E Bastin
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Grant W Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Tatiana M Foroud
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Simone Reppermund
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Department of Developmental Disability Neuropsychiatry, School of Psychiatry, UNSW Medicine, Sydney, New South Wales, Australia
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Netherlands Twin Register, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, the Netherlands
| | - Andrew Simmons
- Biomedical Research Unit for Dementia, King's College London, London, UK
- Department of Neuroimaging, Institute of Psychiatry, King's College London, London, UK
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Andreas Meyer-Lindenberg
- Central Institute of Mental Health, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Wiepke Cahn
- Department of Psychiatry, UMC Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Christopher D Whelan
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Marjolein M J van Donkelaar
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Norbert Hosten
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Robert C Green
- Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Sebastian Mohnke
- Division of Mind and Brain Research, D, Corporate member of Freie Universität Berliepartment of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Hilleke E Hulshoff Pol
- Department of Psychiatry, UMC Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Honghuang Lin
- The Framingham Heart Study, Framingham, MA, USA
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | | | - Peter R Schofield
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Medical Sciences, UNSW, Sydney, New South Wales, Australia
| | - Thomas W Mühleisen
- Institute for Neuroscience and Medicine: Structural and Functional Organisation of the Brain (INM-1), Research Centre Jülich, Jülich, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Cécile and Oskar Vogt Institute for Brain Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Pauline Maillard
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Steven G Potkin
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Wei Wen
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Evan Fletcher
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, Germany
| | - Matthew Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Lars Nyberg
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Umeå Centre for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Radiation Sciences, Umeå University, Umeå, Sweden
| | - Erik G Jönsson
- CoE NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Benedicto Crespo-Facorro
- Department of Medicine, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria-IDIVAL, Santander, Spain
- Centro Investigación Biomédica en Red Salud Mental, Santander, Spain
| | - Nastassja Koen
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Cape Town, South Africa
| | - Douglas N Greve
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vidar M Steen
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Iryna O Fedko
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Netherlands Twin Register, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, the Netherlands
| | - Nynke A Groenewold
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Wiro J Niessen
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
- Department of Medical Informatics, Erasmus MC, Rotterdam, the Netherlands
- Imaging Physics, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | | | | | - William T Longstreth
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Neurology, Erasmus MC, Rotterdam, the Netherlands
| | - Jordan W Smoller
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Marie-Jose van Tol
- Cognitive Neuroscience Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jessika E Sussmann
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Tomas Paus
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Hervé Lemaître
- INSERM, Research Unit 1000 'Neuroimaging and Psychiatry', Paris Saclay University and Paris Descartes University-DIGITEO Labs, Gif sur Yvette, France
| | - Matthias L Schroeter
- LIFE: The Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Clinic for Cognitive Neurology, University Clinic Leipzig, Leipzig, Germany
| | - Bernard Mazoyer
- Neurodegeneratives Diseases Institute, CNRS UMR 5293, Université de Bordeaux, Bordeaux, France
| | - Ole A Andreassen
- CoE NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- CoE NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Florian Holsboer
- Max Planck Institute of Psychiatry, Munich, Germany
- HMNC Brain Health, Munich, Germany
| | - Chantal Depondt
- Department of Neurology, Hopital Erasme, Universite Libre de Bruxelles, Brussels, Belgium
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Jessica A Turner
- The Mind Research Network and LBERI, Albuquerque, NM, USA
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science, Georgia State University, Atlanta, GA, USA
- Department of Psychology, Georgia State University, Atlanta, GA, USA
| | - Zdenka Pausova
- Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Gunter Schumann
- MRC-SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Daan van Rooij
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- University Medical Center Groningen, Department of Psychiatry, University of Groningen, Groningen, the Netherlands
| | - Srdjan Djurovic
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ian J Deary
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Katie L McMahon
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Rachel M Brouwer
- Department of Psychiatry, UMC Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Hilkka Soininen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
- Neurocentre Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Massimo Pandolfo
- Department of Neurology, Hopital Erasme, Universite Libre de Bruxelles, Brussels, Belgium
| | - Thomas H Wassink
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Joshua W Cheung
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Thomas Wolfers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Jean-Luc Martinot
- INSERM, Research Unit 1000 'Neuroimaging and Psychiatry', Paris Saclay University and Paris Descartes University-DIGITEO Labs, Gif sur Yvette, France
| | - Marcel P Zwiers
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (partner site Greifswald), Greifswald, Germany
| | - Ingrid Melle
- CoE NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- CoE NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Ryota Kanai
- Department of Neuroinformatics, Araya, Tokyo, Japan
- Institute of Cognitive Neuroscience, University College London, London, UK
- School of Psychology, University of Sussex, Brighton, UK
| | - Eric Westman
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - René S Kahn
- Department of Psychiatry, UMC Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanjay M Sisodiya
- UCL Queen Square Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Bucks, UK
| | - Tonya White
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Arvin Saremi
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hans van Bokhoven
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Han G Brunner
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht, the Netherlands
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (partner site Greifswald), Greifswald, Germany
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Dennis van 't Ent
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Netherlands Twin Register, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, the Netherlands
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands
| | - Guillén Fernández
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Marcella Rietschel
- Central Institute of Mental Health, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Neeltje E M van Haren
- Department of Psychiatry, UMC Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Simon E Fisher
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Alexa S Beiser
- The Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Clyde Francks
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Andrew J Saykin
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Sydney, New South Wales, Australia
| | - Nina Romanczuk-Seiferth
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Catharina A Hartman
- University Medical Center Groningen, Department of Psychiatry, University of Groningen, Groningen, the Netherlands
| | - Anita L DeStefano
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Dirk J Heslenfeld
- Department of Psychology, VU University Amsterdam, Amsterdam, the Netherlands
| | - Michael W Weiner
- Center for Imaging of Neurodegenerative Disease, San Francisco VA Medical Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Henrik Walter
- Division of Mind and Brain Research, D, Corporate member of Freie Universität Berliepartment of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Pieter J Hoekstra
- University Medical Center Groningen, Department of Psychiatry, University of Groningen, Groningen, the Netherlands
| | - Paul A Nyquist
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara Franke
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Hans J Grabe
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases, Greifswald, Germany
| | - Andrew D Johnson
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung and Blood Institute, Framingham, MA, USA
| | - Christopher Chen
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Center, National University Health System, Singapore, Singapore
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Myriam Fornage
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joanna M Wardlaw
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Charles DeCarli
- Department of Neurology, Center for Neuroscience, University of California, Davis, Sacramento, CA, USA
| | - Philip L De Jager
- Cell Circuits Program, Broad Institute, Cambridge, MA, USA
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Faculty of Medicine, CRC 1052 'Obesity Mechanisms', University of Leipzig, Leipzig, Germany
| | - Stéphanie Debette
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, Bordeaux, France
- Department of Neurology, CHU de Bordeaux, Bordeaux, France
| | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Joshua M Shulman
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Paul M Thompson
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sudha Seshadri
- The Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands.
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
156
|
Vangberg TR, Eikenes L, Håberg AK. The effect of white matter hyperintensities on regional brain volumes and white matter microstructure, a population-based study in HUNT. Neuroimage 2019; 203:116158. [PMID: 31493533 DOI: 10.1016/j.neuroimage.2019.116158] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/03/2019] [Accepted: 09/02/2019] [Indexed: 12/19/2022] Open
Abstract
Even though age-related white matter hyperintensities (WMH) begin to emerge in middle age, their effect on brain micro- and macrostructure in this age group is not fully elucidated. We have examined how presence of WMH and load of WMH affect regional brain volumes and microstructure in a validated, representative general population sample of 873 individuals between 50 and 66 years. Presence of WMH was determined as Fazakas grade ≥1. WMH load was WMH volume from manual tracing of WMHs divided on intracranial volume. The impact of age appropriate WMH (Fazakas grade 1) on the brain was also investigated. Major novel findings were that even the age appropriate WMH group had widespread macro- and microstructural changes in gray and white matter, showing that the mere presence of WMH, not just WMH load is an important clinical indicator of brain health. With increasing WMH load, structural changes spread centrifugally. Further, we found three major patterns of FA and MD changes related to increasing WMH load, demonstrating a heterogeneous effect on white matter microstructure, where distinct patterns were found in the proximity of the lesions, in deep white matter and in white matter near the cortex. This study also raises several questions about the onset of WMH related pathology, in particular, whether some of the aberrant brain structural and microstructural findings are present before the emergence of WMH. We also found, similar to other studies, that WMH risk factors had low explanatory power for WMH, making it unclear which factors lead to WMH.
Collapse
Affiliation(s)
- Torgil Riise Vangberg
- Medical Imaging Research Group, Department of Clinical Medicine, UiT the Arctic University of Norway, Tromsø, Norway; PET Center, University Hospital North Norway, Tromsø, Norway
| | - Live Eikenes
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Asta K Håberg
- Department of Radiology and Nuclear Medicine, St. Olav University Hospital, Trondheim, Norway; Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
157
|
Caciagli L, Wandschneider B, Xiao F, Vollmar C, Centeno M, Vos SB, Trimmel K, Sidhu MK, Thompson PJ, Winston GP, Duncan JS, Koepp MJ. Abnormal hippocampal structure and function in juvenile myoclonic epilepsy and unaffected siblings. Brain 2019; 142:2670-2687. [PMID: 31365054 PMCID: PMC6776114 DOI: 10.1093/brain/awz215] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 04/09/2019] [Accepted: 05/27/2019] [Indexed: 02/05/2023] Open
Abstract
Juvenile myoclonic epilepsy is the most common genetic generalized epilepsy syndrome, characterized by a complex polygenetic aetiology. Structural and functional MRI studies demonstrated mesial or lateral frontal cortical derangements and impaired fronto-cortico-subcortical connectivity in patients and their unaffected siblings. The presence of hippocampal abnormalities and associated memory deficits is controversial, and functional MRI studies in juvenile myoclonic epilepsy have not tested hippocampal activation. In this observational study, we implemented multi-modal MRI and neuropsychological data to investigate hippocampal structure and function in 37 patients with juvenile myoclonic epilepsy, 16 unaffected siblings and 20 healthy controls, comparable for age, gender, handedness and hemispheric dominance as assessed with language laterality indices. Automated hippocampal volumetry was complemented by validated qualitative and quantitative morphological criteria to detect hippocampal malrotation, assumed to represent a neurodevelopmental marker. Neuropsychological measures of verbal and visuo-spatial learning and an event-related verbal and visual memory functional MRI paradigm addressed mesiotemporal function. We detected a reduction of mean left hippocampal volume in patients and their siblings compared with controls (P < 0.01). Unilateral or bilateral hippocampal malrotation was identified in 51% of patients and 50% of siblings, against 15% of controls (P < 0.05). For bilateral hippocampi, quantitative markers of verticalization had significantly larger values in patients and siblings compared with controls (P < 0.05). In the patient subgroup, there was no relationship between structural measures and age at disease onset or degree of seizure control. No overt impairment of verbal and visual memory was identified with neuropsychological tests. Functional mapping highlighted atypical patterns of hippocampal activation, pointing to abnormal recruitment during verbal encoding in patients and their siblings [P < 0.05, familywise error (FWE)-corrected]. Subgroup analyses indicated distinct profiles of hypoactivation along the hippocampal long axis in juvenile myoclonic epilepsy patients with and without malrotation; patients with malrotation also exhibited reduced frontal recruitment for verbal memory, and more pronounced left posterior hippocampal involvement for visual memory. Linear models across the entire study cohort indicated significant associations between morphological markers of hippocampal positioning and hippocampal activation for verbal items (all P < 0.05, FWE-corrected). We demonstrate abnormalities of hippocampal volume, shape and positioning in patients with juvenile myoclonic epilepsy and their siblings, which are associated with reorganization of function and imply an underlying neurodevelopmental mechanism with expression during the prenatal stage. Co-segregation of abnormal hippocampal morphology in patients and their siblings is suggestive of a genetic imaging phenotype, independent of disease activity, and can be construed as a novel endophenotype of juvenile myoclonic epilepsy.
Collapse
Affiliation(s)
- Lorenzo Caciagli
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
| | - Britta Wandschneider
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
| | - Fenglai Xiao
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Christian Vollmar
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
- Department of Neurology, Ludwig-Maximilians-Universität, Marchioninistrasse 15, Munich, Germany
| | - Maria Centeno
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
| | - Sjoerd B Vos
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences, University College London, London, UK
- Centre for Medical Image Computing, University College London, London, UK
| | - Karin Trimmel
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Meneka K Sidhu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
| | - Pamela J Thompson
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
| | - Gavin P Winston
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
- Department of Medicine, Division of Neurology, Queen’s University, Kingston, Ontario, Canada
| | - John S Duncan
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
| | - Matthias J Koepp
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- MRI Unit, Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
| |
Collapse
|
158
|
Khramtsova EA, Heldman R, Derks EM, Yu D, Davis LK, Stranger BE. Sex differences in the genetic architecture of obsessive-compulsive disorder. Am J Med Genet B Neuropsychiatr Genet 2019; 180:351-364. [PMID: 30456828 PMCID: PMC6527502 DOI: 10.1002/ajmg.b.32687] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 12/31/2022]
Abstract
Obsessive-compulsive disorder (OCD) is a highly heritable complex phenotype that demonstrates sex differences in age of onset and clinical presentation, suggesting a possible sex difference in underlying genetic architecture. We present the first genome-wide characterization of the sex-specific genetic architecture of OCD, utilizing the largest set of OCD cases and controls available from the Psychiatric Genomics Consortium. We assessed evidence for several mechanisms that may contribute to sex differences including a sex-dependent liability threshold, the presence of individual sex-specific risk variants on the autosomes and the X chromosome, and sex-specific pleiotropic effects. Furthermore, we tested the hypothesis that genetic heterogeneity between the sexes may obscure associations in a sex-combined genome-wide association study. We observed a strong genetic correlation between male and female OCD and no evidence for a sex-dependent liability threshold model, suggesting that sex-combined analysis does not suffer from widespread loss of power because of genetic heterogeneity between the sexes. While we did not detect any significant sex-specific genome-wide single nucleotide polymorphisms (SNP) associations, we did identify two significant gene-based associations in females: GRID2 and GRP135, which showed no association in males. We observed that the SNPs with sexually differentiated effects showed an enrichment of regulatory variants influencing expression of genes in brain and immune tissues. These findings suggest that future studies with larger sample sizes hold great promise for the identification of sex-specific genetic risk factors for OCD.
Collapse
Affiliation(s)
- Ekaterina A Khramtsova
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, Illinois
| | | | - Eske M Derks
- Queensland Institute of Medical Research, Brisbane, Australia
| | - Dongmei Yu
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Lea K Davis
- Vanderbilt Genetics Institute; Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Medical Genetics, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Barbara E Stranger
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, Illinois
- Center for Data Intensive Science, University of Chicago, Chicago, Illinois
| |
Collapse
|
159
|
Absolute and relative estimates of genetic and environmental variance in brain structure volumes. Brain Struct Funct 2019; 224:2805-2821. [PMID: 31428865 DOI: 10.1007/s00429-019-01931-8] [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: 10/30/2018] [Accepted: 07/30/2019] [Indexed: 10/26/2022]
Abstract
Comparing estimates of the amount of genetic and environmental variance for different brain structures may elucidate differences in the genetic architecture or developmental constraints of individual brain structures. However, most studies compare estimates of relative genetic (heritability) and environmental variance in brain structure, which do not reflect differences in absolute variance between brain regions. Here we used a population sample of young adult twins and singleton siblings of twins (n = 791; M = 23 years, Queensland Twin IMaging study) to estimate the absolute genetic and environmental variance, standardised by the phenotypic mean, in the size of cortical, subcortical, and ventricular brain structures. Mean-standardised genetic variance differed widely across structures [23.5-fold range 0.52% (hippocampus) to 12.28% (lateral ventricles)], but the range of estimates within cortical, subcortical, or ventricular structures was more moderate (two to fivefold range). There was no association between mean-standardised and relative measures of genetic variance (i.e., heritability) in brain structure volumes. We found similar results in an independent sample (n = 1075, M = 29 years, Human Connectome Project). These findings open important new lines of enquiry: namely, understanding the bases of these variance patterns, and their implications regarding the genetic architecture, evolution, and development of the human brain.
Collapse
|
160
|
van der Lee SJ, Knol MJ, Chauhan G, Satizabal CL, Smith AV, Hofer E, Bis JC, Hibar DP, Hilal S, van den Akker EB, Arfanakis K, Bernard M, Yanek LR, Amin N, Crivello F, Cheung JW, Harris TB, Saba Y, Lopez OL, Li S, van der Grond J, Yu L, Paus T, Roshchupkin GV, Amouyel P, Jahanshad N, Taylor KD, Yang Q, Mathias RA, Boehringer S, Mazoyer B, Rice K, Cheng CY, Maillard P, van Heemst D, Wong TY, Niessen WJ, Beiser AS, Beekman M, Zhao W, Nyquist PA, Chen C, Launer LJ, Psaty BM, Ikram MK, Vernooij MW, Schmidt H, Pausova Z, Becker DM, De Jager PL, Thompson PM, van Duijn CM, Bennett DA, Slagboom PE, Schmidt R, Longstreth WT, Ikram MA, Seshadri S, Debette S, Gudnason V, Adams HHH, DeCarli C. A genome-wide association study identifies genetic loci associated with specific lobar brain volumes. Commun Biol 2019; 2:285. [PMID: 31396565 PMCID: PMC6677735 DOI: 10.1038/s42003-019-0537-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/14/2019] [Indexed: 12/26/2022] Open
Abstract
Brain lobar volumes are heritable but genetic studies are limited. We performed genome-wide association studies of frontal, occipital, parietal and temporal lobe volumes in 16,016 individuals, and replicated our findings in 8,789 individuals. We identified six genetic loci associated with specific lobar volumes independent of intracranial volume. Two loci, associated with occipital (6q22.32) and temporal lobe volume (12q14.3), were previously reported to associate with intracranial and hippocampal volume, respectively. We identified four loci previously unknown to affect brain volumes: 3q24 for parietal lobe volume, and 1q22, 4p16.3 and 14q23.1 for occipital lobe volume. The associated variants were located in regions enriched for histone modifications (DAAM1 and THBS3), or close to genes causing Mendelian brain-related diseases (ZIC4 and FGFRL1). No genetic overlap between lobar volumes and neurological or psychiatric diseases was observed. Our findings reveal part of the complex genetics underlying brain development and suggest a role for regulatory regions in determining brain volumes.
Collapse
Affiliation(s)
- Sven J. van der Lee
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
| | - Maria J. Knol
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
| | - Ganesh Chauhan
- University of Bordeaux, Bordeaux Population Health Research Center, INSERM UMR 1219, 33000 Bordeaux, France
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012 India
| | - Claudia L. Satizabal
- The Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229 USA
- Boston University School of Medicine and the Framingham Heart Study, Boston, MA 02118 USA
| | - Albert Vernon Smith
- Icelandic Heart Association, 201 Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, 8036 Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, 8036 Austria
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101 USA
| | - Derrek P. Hibar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90292 USA
| | - Saima Hilal
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
- Department of Pharmacology, National University of Singapore, Singapore, 117600 Singapore
- Memory, Aging and Cognition Center, National University Health System, Singapore, 119228 Singapore
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
| | - Erik B. van den Akker
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, 2333ZA the Netherlands
- Pattern Recognition & Bioinformatics, Delft University of Technology, Delft, 2628XE the Netherlands
- Department of Biomedical Data Sciences, Statistical Genetics, Leiden University Medical Center, Leiden, 2333ZA the Netherlands
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616 USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612 USA
| | - Manon Bernard
- The Hospital for Sick Children, University of Toronto, Toronto, M5G 1X8 ON Canada
| | - Lisa R. Yanek
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
| | - Fabrice Crivello
- Neurofunctional Imaging Group - Neurodegenerative Diseases Institute, UMR 5293, Team 5 - CEA - CNRS - Bordeaux University, Bordeaux, 33076 France
| | - Josh W. Cheung
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90292 USA
| | - Tamara B. Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD 20892 USA
| | - Yasaman Saba
- Research Unit-Genetic Epidemiology, Gottfried Schatz Research Centre for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Shuo Li
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA 02118 USA
| | - Jeroen van der Grond
- Department of Radiology, Leiden University Medical Center, Leiden, 2333ZA the Netherlands
| | - Lei Yu
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612 USA
| | - Tomas Paus
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, M4G 1R8 Canada
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, M5S 1A1 Canada
| | - Gennady V. Roshchupkin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
- Department of Medical Informatics, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
| | - Philippe Amouyel
- Univ. Lille, Inserm, Centre Hosp. Univ Lille, Institut Pasteur de Lille, LabEx DISTALZ-UMR1167 - RID-AGE - Risk factors and molecular determinants of aging-related, 59000 Lille, France
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90292 USA
| | - Kent D. Taylor
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics at LABioMed-Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Qiong Yang
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA 02118 USA
| | - Rasika A. Mathias
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Stefan Boehringer
- Department of Biomedical Data Sciences, Statistical Genetics, Leiden University Medical Center, Leiden, 2333ZA the Netherlands
| | - Bernard Mazoyer
- Neurofunctional Imaging Group - Neurodegenerative Diseases Institute, UMR 5293, Team 5 - CEA - CNRS - Bordeaux University, Bordeaux, 33076 France
| | - Ken Rice
- Department of Biostatistics, University of Washington, Seattle, WA 98195 USA
| | - Ching Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, 169857 Singapore
| | - Pauline Maillard
- Imaging of Dementia and Aging (IDeA) Laboratory, University of California-Davis, Davis, CA 95817 USA
| | - Diana van Heemst
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, 2333ZA the Netherlands
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, 169857 Singapore
| | - Wiro J. Niessen
- Department of Medical Informatics, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
- Faculty of Applied Sciences, Delft University of Technology, Delft, 2629HZ the Netherlands
| | - Alexa S. Beiser
- Boston University School of Medicine and the Framingham Heart Study, Boston, MA 02118 USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA 02118 USA
| | - Marian Beekman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, 2333ZA the Netherlands
| | - Wanting Zhao
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, 169857 Singapore
| | - Paul A. Nyquist
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Christopher Chen
- Department of Pharmacology, National University of Singapore, Singapore, 117600 Singapore
- Memory, Aging and Cognition Center, National University Health System, Singapore, 119228 Singapore
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD 20892 USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101 USA
- Department of Epidemiology, University of Washington, Seattle, WA 98195 USA
- Department of Health Services, University of Washington, Seattle, WA 98195 USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101 USA
| | - M. Kamran Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
| | - Meike W. Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
| | - Helena Schmidt
- Research Unit-Genetic Epidemiology, Gottfried Schatz Research Centre for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, M5G 1X8 ON Canada
- Departments of Physiology and Nutritional Sciences, The Hospital for Sick Children, University of Toronto, Toronto, M5G 1X8 Canada
| | - Diane M. Becker
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY 10032 USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90292 USA
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612 USA
| | - P. Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, 2333ZA the Netherlands
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, 8036 Austria
| | - W. T. Longstreth
- Department of Epidemiology, University of Washington, Seattle, WA 98195 USA
- Department of Neurology, University of Washington, Seattle, WA 98195 USA
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
| | - Sudha Seshadri
- The Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229 USA
- Boston University School of Medicine and the Framingham Heart Study, Boston, MA 02118 USA
| | - Stéphanie Debette
- University of Bordeaux, Bordeaux Population Health Research Center, INSERM UMR 1219, 33000 Bordeaux, France
- Department of Neurology, University Hospital of Bordeaux, Bordeaux, 33000 France
| | - Vilmundur Gudnason
- Icelandic Heart Association, 201 Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Hieab H. H. Adams
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, 3015CN the Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Davis, CA 95817 USA
| |
Collapse
|
161
|
Lancaster T, Hill M, Sims R, Williams J. Microglia - mediated immunity partly contributes to the genetic association between Alzheimer's disease and hippocampal volume. Brain Behav Immun 2019; 79:267-273. [PMID: 30776473 PMCID: PMC6605284 DOI: 10.1016/j.bbi.2019.02.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/14/2019] [Accepted: 02/14/2019] [Indexed: 12/17/2022] Open
Abstract
Genome-wide association studies (GWAS) suggest that Alzheimer's disease (AD) is partly explained by a burden of risk alleles (single nucleotide polymorphisms; SNPs) with relatively small effects. However, the mechanisms by which these loci cumulatively confer susceptibility remain largely unknown. Accumulating evidence suggests an association between increased AD risk allele burden (measured via a polygenic risk profile score; AD-RPS) with reduced hippocampal volume (HV) across a number of independent cohorts. These lines of research suggest that the reduced HV may be a causal mechanism of risk in the development of late-onset Alzheimer's disease (AD). However, as RPS assesses broad, cumulative genetic risk, little is known about the biological processes which may explain this observation. Here, we leverage GWAS data from i) 17,008 late onset AD cases & 37,154 controls and ii) hippocampal volume (N = 12,147; N = 9707) to explore putative pathways that may explain this association. We first demonstrate an association between whole genome AD-RPS and HV (PT < 0.5, Z = -2.07, P = 0.038), confirming previous associations. Second, we restrict our analysis to SNPs within AD genes within a microglia mediated immunity network (NGENES = 56). A microglia AD-RPS was further associated with HV (PT < 0.01; Z = -2.152, P = 0.031). Last, using a competitive, permutation based approach, we show that the common variation within this candidate gene-set is associated with HV, controlling for SNP set-size (P = 0.024). Together, the observations suggest that the relationship between AD and HV is partially explained by genes within an AD-linked microglia mediated immunity network.
Collapse
Affiliation(s)
- T.M. Lancaster
- UK Dementia Research Institute, School of Medicine, Cardiff University, UK,Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, UK,MRC Centre of Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, UK,Corresponding author at: Cardiff University Brain Research Imaging Centre, School of Medicine, Cardiff University, Maindy Road, Cardiff CF24 4HQ, Wales, UK.
| | - M.J. Hill
- UK Dementia Research Institute, School of Medicine, Cardiff University, UK,MRC Centre of Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, UK
| | - R. Sims
- UK Dementia Research Institute, School of Medicine, Cardiff University, UK,MRC Centre of Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, UK
| | - J. Williams
- UK Dementia Research Institute, School of Medicine, Cardiff University, UK,MRC Centre of Neuropsychiatric Genetics & Genomics, School of Medicine, Cardiff University, UK
| |
Collapse
|
162
|
Le BD, Stein JL. Mapping causal pathways from genetics to neuropsychiatric disorders using genome-wide imaging genetics: Current status and future directions. Psychiatry Clin Neurosci 2019; 73:357-369. [PMID: 30864184 PMCID: PMC6625892 DOI: 10.1111/pcn.12839] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/17/2022]
Abstract
Imaging genetics aims to identify genetic variants associated with the structure and function of the human brain. Recently, collaborative consortia have been successful in this goal, identifying and replicating common genetic variants influencing gross human brain structure as measured through magnetic resonance imaging. In this review, we contextualize imaging genetic associations as one important link in understanding the causal chain from genetic variant to increased risk for neuropsychiatric disorders. We provide examples in other fields of how identifying genetic variant associations to disease and multiple phenotypes along the causal chain has revealed a mechanistic understanding of disease risk, with implications for how imaging genetics can be similarly applied. We discuss current findings in the imaging genetics research domain, including that common genetic variants can have a slightly larger effect on brain structure than on risk for disorders like schizophrenia, indicating a somewhat simpler genetic architecture. Also, gross brain structure measurements share a genetic basis with some, but not all, neuropsychiatric disorders, invalidating the previously held belief that they are broad endophenotypes, yet pinpointing brain regions likely involved in the pathology of specific disorders. Finally, we suggest that in order to build a more detailed mechanistic understanding of the effects of genetic variants on the brain, future directions in imaging genetics research will require observations of cellular and synaptic structure in specific brain regions beyond the resolution of magnetic resonance imaging. We expect that integrating genetic associations at biological levels from synapse to sulcus will reveal specific causal pathways impacting risk for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Brandon D. Le
- Department of Genetics & UNC Neuroscience Center, University of North Carolina at Chapel Hill, USA
| | - Jason L. Stein
- Department of Genetics & UNC Neuroscience Center, University of North Carolina at Chapel Hill, USA
| |
Collapse
|
163
|
Dezhina Z, Ranlund S, Kyriakopoulos M, Williams SCR, Dima D. A systematic review of associations between functional MRI activity and polygenic risk for schizophrenia and bipolar disorder. Brain Imaging Behav 2019; 13:862-877. [PMID: 29748770 PMCID: PMC6538577 DOI: 10.1007/s11682-018-9879-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Genetic factors account for up to 80% of the liability for schizophrenia (SCZ) and bipolar disorder (BD). Genome-wide association studies have successfully identified several genes associated with increased risk for both disorders. This has allowed researchers to model the aggregate effect of genes associated with disease status and create a polygenic risk score (PGRS) for each individual. The interest in imaging genetics using PGRS has grown in recent years, with several studies now published. We have conducted a systematic review to examine the effects of PGRS of SCZ, BD and cross psychiatric disorders on brain function and connectivity using fMRI data. Results indicate that the effect of genetic load for SCZ and BD on brain function affects task-related recruitment, with frontal areas having a more prominent role, independent of task. Additionally, the results suggest that the polygenic architecture of psychotic disorders is not regionally confined but impacts on the task-dependent recruitment of multiple brain regions. Future imaging genetics studies with large samples, especially population studies, would be uniquely informative in mapping the spatial distribution of the genetic risk to psychiatric disorders on brain processes during various cognitive tasks and may lead to the discovery of biological pathways that could be crucial in mediating the link between genetic factors and alterations in brain networks.
Collapse
Affiliation(s)
- Zalina Dezhina
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Siri Ranlund
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marinos Kyriakopoulos
- National and Specialist Acorn Lodge Inpatient Children Unit, South London and Maudsley NHS Foundation Trust, London, UK
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Steve C R Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Danai Dima
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- Department of Psychology, School of Arts and Social Sciences, City, University of London, 10 Northampton Square, London, EC1V 0HB, UK.
| |
Collapse
|
164
|
Whelan CD, Altmann A, Botía JA, Jahanshad N, Hibar DP, Absil J, Alhusaini S, Alvim MKM, Auvinen P, Bartolini E, Bergo FPG, Bernardes T, Blackmon K, Braga B, Caligiuri ME, Calvo A, Carr SJ, Chen J, Chen S, Cherubini A, David P, Domin M, Foley S, França W, Haaker G, Isaev D, Keller SS, Kotikalapudi R, Kowalczyk MA, Kuzniecky R, Langner S, Lenge M, Leyden KM, Liu M, Loi RQ, Martin P, Mascalchi M, Morita ME, Pariente JC, Rodríguez-Cruces R, Rummel C, Saavalainen T, Semmelroch MK, Severino M, Thomas RH, Tondelli M, Tortora D, Vaudano AE, Vivash L, von Podewils F, Wagner J, Weber B, Yao Y, Yasuda CL, Zhang G, Bargalló N, Bender B, Bernasconi N, Bernasconi A, Bernhardt BC, Blümcke I, Carlson C, Cavalleri GL, Cendes F, Concha L, Delanty N, Depondt C, Devinsky O, Doherty CP, Focke NK, Gambardella A, Guerrini R, Hamandi K, Jackson GD, Kälviäinen R, Kochunov P, Kwan P, Labate A, McDonald CR, Meletti S, O'Brien TJ, Ourselin S, Richardson MP, Striano P, Thesen T, Wiest R, Zhang J, Vezzani A, Ryten M, Thompson PM, Sisodiya SM. Structural brain abnormalities in the common epilepsies assessed in a worldwide ENIGMA study. Brain 2019; 141:391-408. [PMID: 29365066 PMCID: PMC5837616 DOI: 10.1093/brain/awx341] [Citation(s) in RCA: 328] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/24/2017] [Indexed: 12/02/2022] Open
Abstract
Progressive functional decline in the epilepsies is largely unexplained. We formed the ENIGMA-Epilepsy consortium to understand factors that influence brain measures in epilepsy, pooling data from 24 research centres in 14 countries across Europe, North and South America, Asia, and Australia. Structural brain measures were extracted from MRI brain scans across 2149 individuals with epilepsy, divided into four epilepsy subgroups including idiopathic generalized epilepsies (n =367), mesial temporal lobe epilepsies with hippocampal sclerosis (MTLE; left, n = 415; right, n = 339), and all other epilepsies in aggregate (n = 1026), and compared to 1727 matched healthy controls. We ranked brain structures in order of greatest differences between patients and controls, by meta-analysing effect sizes across 16 subcortical and 68 cortical brain regions. We also tested effects of duration of disease, age at onset, and age-by-diagnosis interactions on structural measures. We observed widespread patterns of altered subcortical volume and reduced cortical grey matter thickness. Compared to controls, all epilepsy groups showed lower volume in the right thalamus (Cohen’s d = −0.24 to −0.73; P < 1.49 × 10−4), and lower thickness in the precentral gyri bilaterally (d = −0.34 to −0.52; P < 4.31 × 10−6). Both MTLE subgroups showed profound volume reduction in the ipsilateral hippocampus (d = −1.73 to −1.91, P < 1.4 × 10−19), and lower thickness in extrahippocampal cortical regions, including the precentral and paracentral gyri, compared to controls (d = −0.36 to −0.52; P < 1.49 × 10−4). Thickness differences of the ipsilateral temporopolar, parahippocampal, entorhinal, and fusiform gyri, contralateral pars triangularis, and bilateral precuneus, superior frontal and caudal middle frontal gyri were observed in left, but not right, MTLE (d = −0.29 to −0.54; P < 1.49 × 10−4). Contrastingly, thickness differences of the ipsilateral pars opercularis, and contralateral transverse temporal gyrus, were observed in right, but not left, MTLE (d = −0.27 to −0.51; P < 1.49 × 10−4). Lower subcortical volume and cortical thickness associated with a longer duration of epilepsy in the all-epilepsies, all-other-epilepsies, and right MTLE groups (beta, b < −0.0018; P < 1.49 × 10−4). In the largest neuroimaging study of epilepsy to date, we provide information on the common epilepsies that could not be realistically acquired in any other way. Our study provides a robust ranking of brain measures that can be further targeted for study in genetic and neuropathological studies. This worldwide initiative identifies patterns of shared grey matter reduction across epilepsy syndromes, and distinctive abnormalities between epilepsy syndromes, which inform our understanding of epilepsy as a network disorder, and indicate that certain epilepsy syndromes involve more widespread structural compromise than previously assumed.
Collapse
Affiliation(s)
- Christopher D Whelan
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, USA.,Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Andre Altmann
- Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Juan A Botía
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, USA
| | - Derrek P Hibar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, USA
| | - Julie Absil
- Department of Radiology, Hôpital Erasme, Universite Libre de Bruxelles, Brussels 1070, Belgium
| | - Saud Alhusaini
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Marina K M Alvim
- Department of Neurology, University of Campinas, Campinas, Brazil
| | - Pia Auvinen
- Epilepsy Center, Department of Neurology, Kuopio University, Kuopio, Finland.,Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Emanuele Bartolini
- Pediatric Neurology Unit, Children's Hospital A. Meyer-University of Florence, Italy.,IRCCS Stella Maris Foundation, Pisa, Italy
| | - Felipe P G Bergo
- Department of Neurology, University of Campinas, Campinas, Brazil
| | - Tauana Bernardes
- Department of Neurology, University of Campinas, Campinas, Brazil
| | - Karen Blackmon
- Comprehensive Epilepsy Center, Department of Neurology, New York University School of Medicine, New York, USA.,Department of Physiology, Neuroscience and Behavioral Science, St. George's University, Grenada, West Indies
| | - Barbara Braga
- Department of Neurology, University of Campinas, Campinas, Brazil
| | - Maria Eugenia Caligiuri
- Institute of Molecular Bioimaging and Physiology of the National Research Council (IBFM-CNR), Catanzaro, Italy
| | - Anna Calvo
- Magnetic Resonance Image Core Facility, IDIBAPS, Barcelona, Spain
| | - Sarah J Carr
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Jian Chen
- Department of Computer Science and Engineering, The Ohio State University, USA
| | - Shuai Chen
- Cognitive Science Department, Xiamen University, Xiamen, China.,Fujian Key Laboratory of the Brain-like Intelligent Systems, China
| | - Andrea Cherubini
- Institute of Molecular Bioimaging and Physiology of the National Research Council (IBFM-CNR), Catanzaro, Italy
| | - Philippe David
- Department of Radiology, Hôpital Erasme, Universite Libre de Bruxelles, Brussels 1070, Belgium
| | - Martin Domin
- Functional Imaging Unit, Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Sonya Foley
- Cardiff University Brain Research Imaging Centre, School of Psychology, Wales, UK
| | - Wendy França
- Department of Neurology, University of Campinas, Campinas, Brazil
| | - Gerrit Haaker
- Department of Neurosurgery, University Hospital, Freiburg, Germany.,Department of Neuropathology, University Hospital Erlangen, Germany
| | - Dmitry Isaev
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, USA
| | - Simon S Keller
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, UK
| | - Raviteja Kotikalapudi
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Diagnostic and Interventional Neuroradiology, University of Tübingen, Tübingen, Germany
| | - Magdalena A Kowalczyk
- The Florey Institute of Neuroscience and Mental Health, Austin Campus, Melbourne, VIC, Australia
| | - Ruben Kuzniecky
- Comprehensive Epilepsy Center, Department of Neurology, New York University School of Medicine, New York, USA
| | - Soenke Langner
- Functional Imaging Unit, Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Matteo Lenge
- Pediatric Neurology Unit, Children's Hospital A. Meyer-University of Florence, Italy
| | - Kelly M Leyden
- Multimodal Imaging Laboratory, University of California San Diego, San Diego, California, USA.,Department of Psychiatry, University of California San Diego, San Diego, California, USA
| | - Min Liu
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute and Hospital, Mcgill University, Montreal, Quebec, Canada
| | - Richard Q Loi
- Multimodal Imaging Laboratory, University of California San Diego, San Diego, California, USA.,Department of Psychiatry, University of California San Diego, San Diego, California, USA
| | - Pascal Martin
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Mario Mascalchi
- Neuroradiology Unit, Children's Hospital A. Meyer, Florence, Italy.,"Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Marcia E Morita
- Department of Neurology, University of Campinas, Campinas, Brazil
| | - Jose C Pariente
- Magnetic Resonance Image Core Facility, IDIBAPS, Barcelona, Spain
| | - Raul Rodríguez-Cruces
- Instituto de Neurobiología, Universidad Nacional Autónoma de México. Querétaro, Querétaro, México
| | - Christian Rummel
- Support Center for Advanced Neuroimaging (SCAN), University Institute for Diagnostic and Interventional Neuroradiology, Inselspital, University of Bern, Bern, Switzerland
| | - Taavi Saavalainen
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland.,Central Finland Central Hospital, Medical Imaging Unit, Jyväskylä, Finland
| | - Mira K Semmelroch
- The Florey Institute of Neuroscience and Mental Health, Austin Campus, Melbourne, VIC, Australia
| | - Mariasavina Severino
- Neuroradiology Unit, Department of Head and Neck and Neurosciences, Istituto Giannina Gaslini, Genova, Italy
| | - Rhys H Thomas
- Institute of Psychological Medicine and Clinical Neurosciences, Hadyn Ellis Building, Maindy Road, Cardiff, UK.,Department of Neurology, University Hospital of Wales, Cardiff, UK
| | - Manuela Tondelli
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, NOCSE Hospital, Modena, Italy
| | - Domenico Tortora
- Neuroradiology Unit, Department of Head and Neck and Neurosciences, Istituto Giannina Gaslini, Genova, Italy
| | - Anna Elisabetta Vaudano
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, NOCSE Hospital, Modena, Italy
| | - Lucy Vivash
- Melbourne Brain Centre, Department of Medicine, University of Melbourne, Parkville, VIC, 3052, Australia.,Department of Neurology, Royal Melbourne Hospital, Parkville, 3050, Australia
| | - Felix von Podewils
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Jan Wagner
- Department of Epileptology, University Hospital Bonn, Bonn, Germany.,Department of Neurology, Philips University of Marburg, Marburg Germany
| | - Bernd Weber
- Department of Epileptology, University Hospital Bonn, Bonn, Germany.,Department of Neurocognition / Imaging, Life&Brain Research Centre, Bonn, Germany
| | - Yi Yao
- The Affiliated Chenggong Hospital of Xiamen University, Xiamen, China
| | | | - Guohao Zhang
- Department of Computer Science and Electrical Engineering, University of Maryland, Baltimore County, USA
| | - Nuria Bargalló
- Magnetic Resonance Image Core Facility, IDIBAPS, Barcelona, Spain.,Centre de Diagnostic Per la Imatge (CDIC), Hospital Clinic, Barcelona, Spain
| | - Benjamin Bender
- Department of Diagnostic and Interventional Neuroradiology, University of Tübingen, Tübingen, Germany
| | - Neda Bernasconi
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute and Hospital, Mcgill University, Montreal, Quebec, Canada
| | - Andrea Bernasconi
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute and Hospital, Mcgill University, Montreal, Quebec, Canada
| | - Boris C Bernhardt
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute and Hospital, Mcgill University, Montreal, Quebec, Canada.,Multimodal Imaging and Connectome Analysis Lab, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Germany
| | - Chad Carlson
- Comprehensive Epilepsy Center, Department of Neurology, New York University School of Medicine, New York, USA.,Medical College of Wisconsin, Department of Neurology, Milwaukee, WI, USA
| | - Gianpiero L Cavalleri
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, RCSI, Dublin, Ireland
| | - Fernando Cendes
- Department of Neurology, University of Campinas, Campinas, Brazil
| | - Luis Concha
- Instituto de Neurobiología, Universidad Nacional Autónoma de México. Querétaro, Querétaro, México
| | - Norman Delanty
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, RCSI, Dublin, Ireland.,Division of Neurology, Beaumont Hospital, Dublin 9, Ireland
| | - Chantal Depondt
- Department of Neurology, Hôpital Erasme, Universite Libre de Bruxelles, Brussels 1070, Belgium
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, Department of Neurology, New York University School of Medicine, New York, USA
| | - Colin P Doherty
- FutureNeuro Research Centre, RCSI, Dublin, Ireland.,Neurology Department, St. James's Hospital, Dublin 8, Ireland
| | - Niels K Focke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Clinical Neurophysiology, University Medicine Göttingen, Göttingen, Germany
| | - Antonio Gambardella
- Institute of Molecular Bioimaging and Physiology of the National Research Council (IBFM-CNR), Catanzaro, Italy.,Institute of Neurology, University "Magna Græcia", Catanzaro, Italy
| | - Renzo Guerrini
- Pediatric Neurology Unit, Children's Hospital A. Meyer-University of Florence, Italy.,IRCCS Stella Maris Foundation, Pisa, Italy
| | - Khalid Hamandi
- Institute of Psychological Medicine and Clinical Neurosciences, Hadyn Ellis Building, Maindy Road, Cardiff, UK.,Department of Neurology, University Hospital of Wales, Cardiff, UK
| | - Graeme D Jackson
- The Florey Institute of Neuroscience and Mental Health, Austin Campus, Melbourne, VIC, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Reetta Kälviäinen
- Epilepsy Center, Department of Neurology, Kuopio University, Kuopio, Finland.,Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Maryland, USA
| | - Patrick Kwan
- Department of Neurology, Royal Melbourne Hospital, Parkville, 3050, Australia
| | - Angelo Labate
- Institute of Molecular Bioimaging and Physiology of the National Research Council (IBFM-CNR), Catanzaro, Italy.,Institute of Neurology, University "Magna Græcia", Catanzaro, Italy
| | - Carrie R McDonald
- Multimodal Imaging Laboratory, University of California San Diego, San Diego, California, USA.,Department of Psychiatry, University of California San Diego, San Diego, California, USA
| | - Stefano Meletti
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, NOCSE Hospital, Modena, Italy
| | - Terence J O'Brien
- Department of Neurology, Royal Melbourne Hospital, Parkville, 3050, Australia.,Department of Medicine, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Sebastien Ourselin
- Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Mark P Richardson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK.,Department of Neurology, King's College Hospital, London, UK
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Thomas Thesen
- Comprehensive Epilepsy Center, Department of Neurology, New York University School of Medicine, New York, USA.,Department of Physiology, Neuroscience and Behavioral Science, St. George's University, Grenada, West Indies
| | - Roland Wiest
- Support Center for Advanced Neuroimaging (SCAN), University Institute for Diagnostic and Interventional Neuroradiology, Inselspital, University of Bern, Bern, Switzerland
| | - Junsong Zhang
- Cognitive Science Department, Xiamen University, Xiamen, China.,Fujian Key Laboratory of the Brain-like Intelligent Systems, China
| | - Annamaria Vezzani
- Dept of Neuroscience, Mario Negri Institute for Pharmacological Research, Via G. La Masa 19, 20156 Milano, Italy
| | - Mina Ryten
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK.,Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, UK
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, USA
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK.,Chalfont Centre for Epilepsy, Bucks, UK
| |
Collapse
|
165
|
Fan CC, Smeland OB, Schork AJ, Chen CH, Holland D, Lo MT, Sundar VS, Frei O, Jernigan TL, Andreassen OA, Dale AM. Beyond heritability: improving discoverability in imaging genetics. Hum Mol Genet 2019. [PMID: 29522091 DOI: 10.1093/hmg/ddy082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Structural neuroimaging measures based on magnetic resonance imaging have been at the forefront of imaging genetics. Global efforts to ensure homogeneity of measurements across study sites have enabled large-scale imaging genetic projects, accumulating nearly 50K samples for genome-wide association studies (GWAS). However, not many novel genetic variants have been identified by these GWAS, despite the high heritability of structural neuroimaging measures. Here, we discuss the limitations of using heritability as a guidance for assessing statistical power of GWAS, and highlight the importance of discoverability-which is the power to detect genetic variants for a given phenotype depending on its unique genomic architecture and GWAS sample size. Further, we present newly developed methods that boost genetic discovery in imaging genetics. By redefining imaging measures independent of traditional anatomical conventions, it is possible to improve discoverability, enabling identification of more genetic effects. Moreover, by leveraging enrichment priors from genomic annotations and independent GWAS of pleiotropic traits, we can better characterize effect size distributions, and identify reliable and replicable loci associated with structural neuroimaging measures. Statistical tools leveraging novel insights into the genetic discoverability of human traits, promises to accelerate the identification of genetic underpinnings underlying brain structural variation.
Collapse
Affiliation(s)
- Chun Chieh Fan
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Olav B Smeland
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andrew J Schork
- Institute for Biological Psychiatry, Mental Health Center Sct. Hans, Capital Region of Denmark, Denmark
| | - Chi-Hua Chen
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Dominic Holland
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Min-Tzu Lo
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - V S Sundar
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Oleksandr Frei
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Terry L Jernigan
- Center for Human Development, University of California San Diego, La Jolla, CA 92093, USA
| | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA.,Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
166
|
Chen J, Liu J, Calhoun VD. The Translational Potential of Neuroimaging Genomic Analyses To Diagnosis And Treatment In The Mental Disorders. PROCEEDINGS OF THE IEEE. INSTITUTE OF ELECTRICAL AND ELECTRONICS ENGINEERS 2019; 107:912-927. [PMID: 32051642 PMCID: PMC7015534 DOI: 10.1109/jproc.2019.2913145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Imaging genomics focuses on characterizing genomic influence on the variation of neurobiological traits, holding promise for illuminating the pathogenesis, reforming the diagnostic system, and precision medicine of mental disorders. This paper aims to provide an overall picture of the current status of neuroimaging-genomic analyses in mental disorders, and how we can increase their translational potential into clinical practice. The review is organized around three perspectives. (a) Towards reliability, generalizability and interpretability, where we summarize the multivariate models and discuss the considerations and trade-offs of using these methods and how reliable findings may be reached, to serve as ground for further delineation. (b) Towards improved diagnosis, where we outline the advantages and challenges of constructing a dimensional transdiagnostic model and how imaging genomic analyses map into this framework to aid in deconstructing heterogeneity and achieving an optimal stratification of patients that better inform treatment planning. (c) Towards improved treatment. Here we highlight recent efforts and progress in elucidating the functional annotations that bridge between genomic risk and neurobiological abnormalities, in detecting genomic predisposition and prodromal neurodevelopmental changes, as well as in identifying imaging genomic biomarkers for predicting treatment response. Providing an overview of the challenges and promises, this review hopefully motivates imaging genomic studies with multivariate, dimensional and transdiagnostic designs for generalizable and interpretable findings that facilitate development of personalized treatment.
Collapse
Affiliation(s)
- Jiayu Chen
- The Mind Research Network, Albuquerque, NM 87106 USA
| | - Jingyu Liu
- The Mind Research Network, Albuquerque, NM 87106 USA, and also with the Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM 87131 USA
| | - Vince D Calhoun
- The Mind Research Network, Albuquerque, NM 87106 USA, and also with the Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM 87131 USA
| |
Collapse
|
167
|
The spatial correspondence and genetic influence of interhemispheric connectivity with white matter microstructure. Nat Neurosci 2019; 22:809-819. [PMID: 30988526 DOI: 10.1038/s41593-019-0379-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/08/2019] [Indexed: 12/14/2022]
Abstract
Microscopic features (that is, microstructure) of axons affect neural circuit activity through characteristics such as conduction speed. To what extent axonal microstructure in white matter relates to functional connectivity (synchrony) between brain regions is largely unknown. Using MRI data in 11,354 subjects, we constructed multivariate models that predict functional connectivity of pairs of brain regions from the microstructural signature of white matter pathways that connect them. Microstructure-derived models provided predictions of functional connectivity that explained 3.5% of cross-subject variance on average (ranging from 1-13%, or r = 0.1-0.36) and reached statistical significance in 90% of the brain regions considered. The microstructure-function relationships were associated with genetic variants, co-located with genes DAAM1 and LPAR1, that have previously been linked to neural development. Our results demonstrate that variation in white matter microstructure predicts a fraction of functional connectivity across individuals, and that this relationship is underpinned by genetic variability in certain brain areas.
Collapse
|
168
|
Elliott ML, Knodt AR, Cooke M, Kim MJ, Melzer TR, Keenan R, Ireland D, Ramrakha S, Poulton R, Caspi A, Moffitt TE, Hariri AR. General functional connectivity: Shared features of resting-state and task fMRI drive reliable and heritable individual differences in functional brain networks. Neuroimage 2019; 189:516-532. [PMID: 30708106 PMCID: PMC6462481 DOI: 10.1016/j.neuroimage.2019.01.068] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/22/2019] [Accepted: 01/27/2019] [Indexed: 01/15/2023] Open
Abstract
Intrinsic connectivity, measured using resting-state fMRI, has emerged as a fundamental tool in the study of the human brain. However, due to practical limitations, many studies do not collect enough resting-state data to generate reliable measures of intrinsic connectivity necessary for studying individual differences. Here we present general functional connectivity (GFC) as a method for leveraging shared features across resting-state and task fMRI and demonstrate in the Human Connectome Project and the Dunedin Study that GFC offers better test-retest reliability than intrinsic connectivity estimated from the same amount of resting-state data alone. Furthermore, at equivalent scan lengths, GFC displayed higher estimates of heritability than resting-state functional connectivity. We also found that predictions of cognitive ability from GFC generalized across datasets, performing as well or better than resting-state or task data alone. Collectively, our work suggests that GFC can improve the reliability of intrinsic connectivity estimates in existing datasets and, subsequently, the opportunity to identify meaningful correlates of individual differences in behavior. Given that task and resting-state data are often collected together, many researchers can immediately derive more reliable measures of intrinsic connectivity through the adoption of GFC rather than solely using resting-state data. Moreover, by better capturing heritable variation in intrinsic connectivity, GFC represents a novel endophenotype with broad applications in clinical neuroscience and biomarker discovery.
Collapse
Affiliation(s)
- Maxwell L Elliott
- Department of Psychology & Neuroscience, Duke University, Box 104410, Durham, NC, 27708, USA.
| | - Annchen R Knodt
- Department of Psychology & Neuroscience, Duke University, Box 104410, Durham, NC, 27708, USA
| | - Megan Cooke
- Department of Psychology & Neuroscience, Duke University, Box 104410, Durham, NC, 27708, USA
| | - M Justin Kim
- Department of Psychology, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Tracy R Melzer
- New Zealand Brain Research Institute, Christchurch, New Zealand; Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Ross Keenan
- New Zealand Brain Research Institute, Christchurch, New Zealand; Christchurch Radiology Group, Christchurch, New Zealand
| | - David Ireland
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, 163 Union St E, Dunedin, 9016, New Zealand
| | - Sandhya Ramrakha
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, 163 Union St E, Dunedin, 9016, New Zealand
| | - Richie Poulton
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, 163 Union St E, Dunedin, 9016, New Zealand
| | - Avshalom Caspi
- Department of Psychology & Neuroscience, Duke University, Box 104410, Durham, NC, 27708, USA; Social, Genetic, & Developmental Psychiatry Research Centre, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK; Department of Psychiatry & Behavioral Sciences, Duke University School of Medicine, Durham, NC, 27708, USA; Center for Genomic and Computational Biology, Duke University, Box 90338, Durham, NC, 27708, USA
| | - Terrie E Moffitt
- Department of Psychology & Neuroscience, Duke University, Box 104410, Durham, NC, 27708, USA; Social, Genetic, & Developmental Psychiatry Research Centre, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK; Department of Psychiatry & Behavioral Sciences, Duke University School of Medicine, Durham, NC, 27708, USA; Center for Genomic and Computational Biology, Duke University, Box 90338, Durham, NC, 27708, USA
| | - Ahmad R Hariri
- Department of Psychology & Neuroscience, Duke University, Box 104410, Durham, NC, 27708, USA
| |
Collapse
|
169
|
Niego A, Benítez-Burraco A. Williams Syndrome, Human Self-Domestication, and Language Evolution. Front Psychol 2019; 10:521. [PMID: 30936846 PMCID: PMC6431629 DOI: 10.3389/fpsyg.2019.00521] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/22/2019] [Indexed: 01/06/2023] Open
Abstract
Language evolution resulted from changes in our biology, behavior, and culture. One source of these changes might be human self-domestication. Williams syndrome (WS) is a clinical condition with a clearly defined genetic basis which results in a distinctive behavioral and cognitive profile, including enhanced sociability. In this paper we show evidence that the WS phenotype can be satisfactorily construed as a hyper-domesticated human phenotype, plausibly resulting from the effect of the WS hemideletion on selected candidates for domestication and neural crest (NC) function. Specifically, we show that genes involved in animal domestication and NC development and function are significantly dysregulated in the blood of subjects with WS. We also discuss the consequences of this link between domestication and WS for our current understanding of language evolution.
Collapse
Affiliation(s)
- Amy Niego
- Ph.D. Program, Faculty of Humanities, University of Huelva, Huelva, Spain
| | - Antonio Benítez-Burraco
- Department of Spanish, Linguistics, and Theory of Literature, Faculty of Philology, University of Seville, Seville, Spain
| |
Collapse
|
170
|
Klein M, Walters RK, Demontis D, Stein JL, Hibar DP, Adams HH, Bralten J, Roth Mota N, Schachar R, Sonuga-Barke E, Mattheisen M, Neale BM, Thompson PM, Medland SE, Børglum AD, Faraone SV, Arias-Vasquez A, Franke B. Genetic Markers of ADHD-Related Variations in Intracranial Volume. Am J Psychiatry 2019; 176:228-238. [PMID: 30818988 PMCID: PMC7780894 DOI: 10.1176/appi.ajp.2018.18020149] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Attention deficit hyperactivity disorder (ADHD) is a common and highly heritable neurodevelopmental disorder with a complex pathophysiology. Intracranial volume (ICV) and volumes of the nucleus accumbens, amygdala, caudate nucleus, hippocampus, and putamen are smaller in people with ADHD compared with healthy individuals. The authors investigated the overlap between common genetic variation associated with ADHD risk and these brain volume measures to identify underlying biological processes contributing to the disorder. METHODS The authors combined genome-wide association results from the largest available studies of ADHD (N=55,374) and brain volumes (N=11,221-24,704), using a set of complementary methods to investigate overlap at the level of global common variant genetic architecture and at the single variant level. RESULTS Analyses revealed a significant negative genetic correlation between ADHD and ICV (rg=-0.22). Meta-analysis of single variants revealed two significant loci of interest associated with both ADHD risk and ICV; four additional loci were identified for ADHD and volumes of the amygdala, caudate nucleus, and putamen. Exploratory gene-based and gene-set analyses in the ADHD-ICV meta-analytic data showed association with variation in neurite outgrowth-related genes. CONCLUSIONS This is the first genome-wide study to show significant genetic overlap between brain volume measures and ADHD, both on the global and the single variant level. Variants linked to smaller ICV were associated with increased ADHD risk. These findings can help us develop new hypotheses about biological mechanisms by which brain structure alterations may be involved in ADHD disease etiology.
Collapse
Affiliation(s)
- Marieke Klein
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Raymond K. Walters
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Ditte Demontis
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Jason L. Stein
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Derrek P. Hibar
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Hieab H. Adams
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Janita Bralten
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Nina Roth Mota
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Russell Schachar
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Edmund Sonuga-Barke
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Manuel Mattheisen
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Benjamin M. Neale
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Paul M. Thompson
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Sarah E. Medland
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Anders D. Børglum
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Stephen V. Faraone
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Alejandro Arias-Vasquez
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| | - Barbara Franke
- The Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands (Klein, Bralten, Roth Mota, Arias-Vasquez, Franke); University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Psychiatry, Utrecht, the Netherlands (Klein); the Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (Walters, Neale); Program in Medical and Population
| |
Collapse
|
171
|
Comparison of methods for multivariate gene-based association tests for complex diseases using common variants. Eur J Hum Genet 2019; 27:811-823. [PMID: 30683923 PMCID: PMC6461986 DOI: 10.1038/s41431-018-0327-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 10/30/2018] [Accepted: 12/04/2018] [Indexed: 12/29/2022] Open
Abstract
Complex diseases are usually associated with multiple correlated phenotypes, and the analysis of composite scores or disease status may not fully capture the complexity (or multidimensionality). Joint analysis of multiple disease-related phenotypes in genetic tests could potentially increase power to detect association of a disease with common SNPs (or genes). Gene-based tests are designed to identify genes containing multiple risk variants that individually are weakly associated with a univariate trait. We combined three multivariate association tests (O'Brien method, TATES, and MultiPhen) with two gene-based association tests (GATES and VEGAS) and compared performance (type I error and power) of six multivariate gene-based methods using simulated data. Data (n = 2000) for genetic sequence and correlated phenotypes were simulated by varying causal variant proportions and phenotype correlations for various scenarios. These simulations showed that two multivariate association tests (TATES and MultiPhen, but not O'Brien) paired with VEGAS have inflated type I error in all scenarios, while the three multivariate association tests paired with GATES have correct type I error. MultiPhen paired with GATES has higher power than competing methods if the correlations among phenotypes are low (r < 0.57). We applied these gene-based association methods to a GWAS dataset from the Alzheimer's Disease Genetics Consortium containing three neuropathological traits related to Alzheimer disease (neuritic plaque, neurofibrillary tangles, and cerebral amyloid angiopathy) measured in 3500 autopsied brains. Gene-level significant evidence (P < 2.7 × 10-6) was identified in a region containing three contiguous genes (TRAPPC12, TRAPPC12-AS1, ADI1) using O'Brien and VEGAS. Gene-wide significant associations were not observed in univariate gene-based tests.
Collapse
|
172
|
Gao W, Grewen K, Knickmeyer RC, Qiu A, Salzwedel A, Lin W, Gilmore JH. A review on neuroimaging studies of genetic and environmental influences on early brain development. Neuroimage 2019; 185:802-812. [PMID: 29673965 PMCID: PMC6191379 DOI: 10.1016/j.neuroimage.2018.04.032] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 12/11/2022] Open
Abstract
The past decades witnessed a surge of interest in neuroimaging study of normal and abnormal early brain development. Structural and functional studies of normal early brain development revealed massive structural maturation as well as sequential, coordinated, and hierarchical emergence of functional networks during the infancy period, providing a great foundation for the investigation of abnormal early brain development mechanisms. Indeed, studies of altered brain development associated with either genetic or environmental risks emerged and thrived. In this paper, we will review selected studies of genetic and environmental risks that have been relatively more extensively investigated-familial risks, candidate risk genes, and genome-wide association studies (GWAS) on the genetic side; maternal mood disorders and prenatal drug exposures on the environmental side. Emerging studies on environment-gene interactions will also be reviewed. Our goal was not to perform an exhaustive review of all studies in the field but to leverage some representative ones to summarize the current state, point out potential limitations, and elicit discussions on important future directions.
Collapse
Affiliation(s)
- Wei Gao
- Biomedical Imaging Research Institute (BIRI), Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, CA, United States; Department of Medicine, University of California, Los Angeles, CA, United States.
| | - Karen Grewen
- Department of Psychiatry, Neurobiology, and Psychology, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Rebecca C Knickmeyer
- Department of Psychiatry, University of North Carolina at Chapel Hill, N.C, United States
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Andrew Salzwedel
- Biomedical Imaging Research Institute (BIRI), Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, CA, United States
| | - Weili Lin
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, NC, United States
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina at Chapel Hill, N.C, United States
| |
Collapse
|
173
|
Alexander-Bloch AF, Mathias SR, Fox PT, Olvera RL, Göring HHH, Duggirala R, Curran JE, Blangero J, Glahn DC. Human Cortical Thickness Organized into Genetically-determined Communities across Spatial Resolutions. Cereb Cortex 2019; 29:106-118. [PMID: 29190330 PMCID: PMC6676978 DOI: 10.1093/cercor/bhx309] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/19/2017] [Indexed: 12/13/2022] Open
Abstract
The cerebral cortex may be organized into anatomical genetic modules, communities of brain regions with shared genetic influences via pleiotropy. Such modules could represent novel phenotypes amenable to large-scale gene discovery. This modular structure was investigated with network analysis of in vivo MRI of extended pedigrees, revealing a "multiscale" structure where smaller and larger modules exist simultaneously and in partially overlapping fashion across spatial scales, in contrast to prior work suggesting a specific number of cortical thickness modules. Inter-regional genetic correlations, gene co-expression patterns and computational models indicate that two simple organizational principles account for a large proportion of the apparent complexity in the network of genetic correlations. First, regions are strongly genetically correlated with their homologs in the opposite cerebral hemisphere. Second, regions are strongly genetically correlated with nearby regions in the same hemisphere, with an initial steep decrease in genetic correlation with anatomical distance, followed by a more gradual decline. Understanding underlying organizational principles of genetic influence is a critical step towards a mechanistic model of how specific genes influence brain anatomy and mediate neuropsychiatric risk.
Collapse
Affiliation(s)
| | - Samuel R Mathias
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, USA
| | - Peter T Fox
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - Rene L Olvera
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - Harold H H Göring
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - Ravi Duggirala
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - Joanne E Curran
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, TX, USA
- University of Texas of the Rio Grande Valley, Brownsville, TX, USA
| | - David C Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, USA
| |
Collapse
|
174
|
Otsuka Y, Kakeda S, Sugimoto K, Katsuki A, Nguyen LH, Igata R, Watanabe K, Ueda I, Kishi T, Iwata N, Korogi Y, Yoshimura R. COMT polymorphism regulates the hippocampal subfield volumes in first-episode, drug-naive patients with major depressive disorder. Neuropsychiatr Dis Treat 2019; 15:1537-1545. [PMID: 31239688 PMCID: PMC6560253 DOI: 10.2147/ndt.s199598] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
Purpose: Compared with healthy subjects (HS), patients with major depressive disorder (MDD) exhibit volume differences that affect the volume changes in several areas such as the limbic, cortical, subcortical, and white matter. Catechol-O-methyltransferase (COMT) is a methylation enzyme that catalyzes endogenous catecholamines. The Val158Met polymorphism of COMT has been reported to affect the dopamine (DA) levels, which plays an important role in psychiatric diseases. However, the relationships among both DA levels, COMT genotype, and brain morphology are complicated and controversial. In previous studies that investigated the hippocampal subfields, the greatest brain abnormalities in MDD patients were observed in Cornu Ammonis (CA)1 and the subiculum, followed by that in CA2-3. We have prospectively demonstrated the relationship between the single-nucleotide polymorphism of the Val158Met COMT gene (rs4680) and the hippocampal subfields in drug-naive MDD patients. Patients and methods: In this study, we compared 27 MDD patients and 42 HS who were divided into groups based on their COMT genotype. The effects of the diagnosis, genotype, and genotype-diagnosis interaction related to CA1 and the subiculum volumes, as well as the whole-brain cortical thickness, were evaluated by performing a FreeSurfer statistical analysis of high-resolution magnetic resonance imaging (MRI) findings. Results: The results revealed that there was a statistically significant interaction between the effects of diagnosis and genotype on the right subiculum (a component of the hippocampus). Conclusion: This Val158Met COMT polymorphism may influence the subiculum volume in drug-naive, first-episode MDD patients.
Collapse
Affiliation(s)
- Yuka Otsuka
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shingo Kakeda
- Department of Radiology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Koichiro Sugimoto
- Department of Radiology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Asuka Katsuki
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Le Hoa Nguyen
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Ryohei Igata
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Keita Watanabe
- Department of Radiology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Issei Ueda
- Department of Radiology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Taro Kishi
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yukunori Korogi
- Department of Radiology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Reiji Yoshimura
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
175
|
Panayides AS, Pattichis MS, Leandrou S, Pitris C, Constantinidou A, Pattichis CS. Radiogenomics for Precision Medicine With a Big Data Analytics Perspective. IEEE J Biomed Health Inform 2018; 23:2063-2079. [PMID: 30596591 DOI: 10.1109/jbhi.2018.2879381] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Precision medicine promises better healthcare delivery by improving clinical practice. Using evidence-based substratification of patients, the objective is to achieve better prognosis, diagnosis, and treatment that will transform existing clinical pathways toward optimizing care for the specific needs of each patient. The wealth of today's healthcare data, often characterized as big data, provides invaluable resources toward new knowledge discovery that has the potential to advance precision medicine. The latter requires interdisciplinary efforts that will capitalize the information, know-how, and medical data of newly formed groups fusing different backgrounds and expertise. The objective of this paper is to provide insights with respect to the state-of-the-art research in precision medicine. More specifically, our goal is to highlight the fundamental challenges in emerging fields of radiomics and radiogenomics by reviewing the case studies of Cancer and Alzheimer's disease, describe the computational challenges from a big data analytics perspective, and discuss standardization and open data initiatives that will facilitate the adoption of precision medicine methods and practices.
Collapse
|
176
|
Gunz P, Tilot AK, Wittfeld K, Teumer A, Shapland CY, van Erp TGM, Dannemann M, Vernot B, Neubauer S, Guadalupe T, Fernández G, Brunner HG, Enard W, Fallon J, Hosten N, Völker U, Profico A, Di Vincenzo F, Manzi G, Kelso J, St Pourcain B, Hublin JJ, Franke B, Pääbo S, Macciardi F, Grabe HJ, Fisher SE. Neandertal Introgression Sheds Light on Modern Human Endocranial Globularity. Curr Biol 2018; 29:120-127.e5. [PMID: 30554901 PMCID: PMC6380688 DOI: 10.1016/j.cub.2018.10.065] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/21/2018] [Accepted: 10/31/2018] [Indexed: 12/15/2022]
Abstract
One of the features that distinguishes modern humans from our extinct relatives and ancestors is a globular shape of the braincase [1-4]. As the endocranium closely mirrors the outer shape of the brain, these differences might reflect altered neural architecture [4, 5]. However, in the absence of fossil brain tissue, the underlying neuroanatomical changes as well as their genetic bases remain elusive. To better understand the biological foundations of modern human endocranial shape, we turn to our closest extinct relatives: the Neandertals. Interbreeding between modern humans and Neandertals has resulted in introgressed fragments of Neandertal DNA in the genomes of present-day non-Africans [6, 7]. Based on shape analyses of fossil skull endocasts, we derive a measure of endocranial globularity from structural MRI scans of thousands of modern humans and study the effects of introgressed fragments of Neandertal DNA on this phenotype. We find that Neandertal alleles on chromosomes 1 and 18 are associated with reduced endocranial globularity. These alleles influence expression of two nearby genes, UBR4 and PHLPP1, which are involved in neurogenesis and myelination, respectively. Our findings show how integration of fossil skull data with archaic genomics and neuroimaging can suggest developmental mechanisms that may contribute to the unique modern human endocranial shape.
Collapse
Affiliation(s)
- Philipp Gunz
- Department of Human Evolution, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany.
| | - Amanda K Tilot
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, P.O. Box 310, 6500 AH, Nijmegen, the Netherlands
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University of Greifswald, Ellernholzstr. 1-2, 17489 Greifswald, Germany; German Center for Neurodegenerative Diseases (DZNE), Partner Site Rostock/Greifswald, Ellernholzstr. 1-2, 17489 Greifswald, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Walter-Rathenau Str. 48, 17475 Greifswald, Germany
| | - Chin Yang Shapland
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, P.O. Box 310, 6500 AH, Nijmegen, the Netherlands
| | - Theo G M van Erp
- Clinical and Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, 5251 California Ave, Irvine, CA 92617, USA
| | - Michael Dannemann
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Benjamin Vernot
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Simon Neubauer
- Department of Human Evolution, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Tulio Guadalupe
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, P.O. Box 310, 6500 AH, Nijmegen, the Netherlands
| | - Guillén Fernández
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 GA, Nijmegen, the Netherlands
| | - Han G Brunner
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 GA, Nijmegen, the Netherlands; Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht University Medical Center, 6202 AZ, Maastricht, the Netherlands
| | - Wolfgang Enard
- Anthropology and Human Genomics, Department Biology II, Ludwig Maximilians University Munich, Grosshaderner Str. 2, D-82152 Martinsried, Germany
| | - James Fallon
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Norbert Hosten
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine, Ernst-Moritz-Arndt-University Greifswald, Ferdinand-Sauerbruch-Str. 1, 17475 Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany
| | - Antonio Profico
- Università degli Studi di Roma La Sapienza, Department of Environmental Biology, Piazzale Aldo Moro, 5, 00185, Roma, Italy
| | - Fabio Di Vincenzo
- Università degli Studi di Roma La Sapienza, Department of Environmental Biology, Piazzale Aldo Moro, 5, 00185, Roma, Italy; Istituto Italiano di Paleontologia Umana, Via Ulisse Aldrovandi, 18, 00197, Roma, Italy
| | - Giorgio Manzi
- Università degli Studi di Roma La Sapienza, Department of Environmental Biology, Piazzale Aldo Moro, 5, 00185, Roma, Italy
| | - Janet Kelso
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Beate St Pourcain
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, P.O. Box 310, 6500 AH, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Jean-Jacques Hublin
- Department of Human Evolution, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Barbara Franke
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands; Departments of Human Genetics and Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Svante Pääbo
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Fabio Macciardi
- Department of Psychiatry and Human Behavior, University of California, Irvine, Sprague Hall - Room 312, Gillespie Neuroscience - Laboratory, Mail Code: 3960, Irvine, CA 92697, USA
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University of Greifswald, Ellernholzstr. 1-2, 17489 Greifswald, Germany
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, P.O. Box 310, 6500 AH, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands.
| |
Collapse
|
177
|
Elman JA, Panizzon MS, Gillespie NA, Hagler DJ, Fennema‐Notestine C, Eyler LT, McEvoy LK, Neale MC, Lyons MJ, Franz CE, Dale AM, Kremen WS. Genetic architecture of hippocampal subfields on standard resolution MRI: How the parts relate to the whole. Hum Brain Mapp 2018; 40:1528-1540. [PMID: 30430703 PMCID: PMC6397064 DOI: 10.1002/hbm.24464] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/19/2018] [Accepted: 10/29/2018] [Indexed: 01/30/2023] Open
Abstract
The human hippocampus can be subdivided into subfields with unique functional properties and differential vulnerability to disease or neuropsychiatric conditions. Identifying genes that confer susceptibility to such processes is an important goal in developing treatments. Recent advances in automatic subfield segmentation from magnetic resonance images make it possible to use these measures as phenotypes in large-scale genome-wide association studies. Such analyses are likely to rely largely on standard resolution (~1 mm isotropic) T1 -weighted images acquired on 3.0T scanners. Determining whether the genetic architecture of subfields can be detected from such images is therefore an important step. We used Freesurfer v6.0 to segment hippocampal subfields in two large twin studies, the Vietnam Era Twin Study of Aging and the Human Connectome Project. We estimated heritability of subfields and the genetic overlap with total hippocampal volume. Heritability was similar across samples, but little genetic variance remained after accounting for genetic influences on total hippocampal volume. Importantly, we examined genetic relationships between subfields to determine whether subfields can be grouped based on a smaller number of underlying, genetically independent factors. We identified three genetic factors in both samples, but the high degree of cross loadings precluded formation of genetically distinct groupings of subfields. These results confirm the reliability of Freesurfer v6.0 generated subfields across samples for phenotypic analyses. However, the current results suggest that it will be difficult for large-scale genetic analyses to identify subfield-specific genes that are distinct from both total hippocampal volume and other subfields using segmentations generated from standard resolution T1 -weighted images.
Collapse
Affiliation(s)
- Jeremy A. Elman
- Department of PsychiatryUniversity of California San DiegoSan DiegoCalifornia,Center for Behavior Genetics of AgingUniversity of California San DiegoSan DiegoCalifornia
| | - Matthew S. Panizzon
- Department of PsychiatryUniversity of California San DiegoSan DiegoCalifornia,Center for Behavior Genetics of AgingUniversity of California San DiegoSan DiegoCalifornia
| | - Nathan A. Gillespie
- Virginia Institute for Psychiatric and Behavior GeneticsVirginia Commonwealth UniversityRichmondVirginia
| | - Donald J. Hagler
- Department of RadiologyUniversity of California San DiegoSan DiegoCalifornia
| | - Christine Fennema‐Notestine
- Department of PsychiatryUniversity of California San DiegoSan DiegoCalifornia,Department of RadiologyUniversity of California San DiegoSan DiegoCalifornia
| | - Lisa T. Eyler
- Department of PsychiatryUniversity of California San DiegoSan DiegoCalifornia,VA San Diego Health Care SystemSan DiegoCalifornia
| | - Linda K. McEvoy
- Department of RadiologyUniversity of California San DiegoSan DiegoCalifornia
| | - Michael C. Neale
- Virginia Institute for Psychiatric and Behavior GeneticsVirginia Commonwealth UniversityRichmondVirginia
| | - Michael J. Lyons
- Department of Psychological and Brain SciencesBoston UniversityBostonMassachusetts
| | - Carol E. Franz
- Department of PsychiatryUniversity of California San DiegoSan DiegoCalifornia,Center for Behavior Genetics of AgingUniversity of California San DiegoSan DiegoCalifornia
| | - Anders M. Dale
- Department of RadiologyUniversity of California San DiegoSan DiegoCalifornia,Department of NeurosciencesUniversity of California San DiegoSan DiegoCalifornia
| | - William S. Kremen
- Department of PsychiatryUniversity of California San DiegoSan DiegoCalifornia,Center for Behavior Genetics of AgingUniversity of California San DiegoSan DiegoCalifornia,Center of Excellence for Stress and Mental HealthVA San Diego Health Care SystemSan DiegoCalifornia
| |
Collapse
|
178
|
Wolfers T, Doan NT, Kaufmann T, Alnæs D, Moberget T, Agartz I, Buitelaar JK, Ueland T, Melle I, Franke B, Andreassen OA, Beckmann CF, Westlye LT, Marquand AF. Mapping the Heterogeneous Phenotype of Schizophrenia and Bipolar Disorder Using Normative Models. JAMA Psychiatry 2018; 75:1146-1155. [PMID: 30304337 PMCID: PMC6248110 DOI: 10.1001/jamapsychiatry.2018.2467] [Citation(s) in RCA: 264] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
IMPORTANCE Schizophrenia and bipolar disorder are severe and complex brain disorders characterized by substantial clinical and biological heterogeneity. However, case-control studies often ignore such heterogeneity through their focus on the average patient, which may be the core reason for a lack of robust biomarkers indicative of an individual's treatment response and outcome. OBJECTIVES To investigate the degree to which case-control analyses disguise interindividual differences in brain structure among patients with schizophrenia and bipolar disorder and to map the brain alterations linked to these disorders at the level of individual patients. DESIGN, SETTING, AND PARTICIPANTS This study used cross-sectional, T1-weighted magnetic resonance imaging data from participants recruited for the Thematically Organized Psychosis study from October 27, 2004, to October 17, 2012. Data were reanalyzed in 2017 and 2018. Patients were recruited from inpatient and outpatient clinics in the Oslo area of Norway, and healthy individuals from the same catchment area were drawn from the national population registry. MAIN OUTCOMES AND MEASURES Interindividual differences in brain structure among patients with schizophrenia and bipolar disorder. Voxel-based morphometry maps were computed, which were used for normative modeling to map the range of interindividual differences in brain structure. RESULTS This study included 218 patients with schizophrenia spectrum disorders (mean [SD] age, 30 [9.3] years; 126 [57.8%] male), of whom 163 had schizophrenia (mean [SD] age, 31 [8.7] years; 105 [64.4%] male) and 190 had bipolar disorder (mean [SD] age, 34 [11.3] years; 79 [41.6%] male), and 256 healthy individuals (mean [SD] age, 34 [9.5] years; 140 [54.7%] male). At the level of the individual, deviations from the normative model were frequent in both disorders but highly heterogeneous. Overlap of more than 2% among patients was observed in only a few loci, primarily in frontal, temporal, and cerebellar regions. The proportion of alterations was associated with diagnosis and cognitive and clinical characteristics within clinical groups. Patients with schizophrenia, on average, had significantly reduced gray matter in frontal regions, cerebellum, and temporal cortex. In patients with bipolar disorder, mean deviations were primarily present in cerebellar regions. CONCLUSIONS AND RELEVANCE This study found that group-level differences disguised biological heterogeneity and interindividual differences among patients with the same diagnosis. This finding suggests that the idea of the average patient is a noninformative construct in psychiatry that falls apart when mapping abnormalities at the level of the individual patient. This study presents a workable route toward precision medicine in psychiatry.
Collapse
Affiliation(s)
- Thomas Wolfers
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands,Donders Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Nhat Trung Doan
- Norwegian Centre for Mental Disorders Research, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- Norwegian Centre for Mental Disorders Research, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dag Alnæs
- Norwegian Centre for Mental Disorders Research, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torgeir Moberget
- Norwegian Centre for Mental Disorders Research, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid Agartz
- Norwegian Centre for Mental Disorders Research, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Jan K. Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands,Karakter Child and Adolescent Psychiatry University Centre, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Torill Ueland
- Norwegian Centre for Mental Disorders Research, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Psychology, University of Oslo, Oslo, Norway
| | - Ingrid Melle
- Norwegian Centre for Mental Disorders Research, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands,Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Christian F. Beckmann
- Donders Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands,Centre for Functional MRI of the Brain, University of Oxford, Oxford, United Kingdom
| | - Lars T. Westlye
- Norwegian Centre for Mental Disorders Research, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Psychology, University of Oslo, Oslo, Norway
| | - Andre F. Marquand
- Donders Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands,Department of Neuroimaging, Institute of Psychiatry, King’s College London, London, United Kingdom
| |
Collapse
|
179
|
Veitch DP, Weiner MW, Aisen PS, Beckett LA, Cairns NJ, Green RC, Harvey D, Jack CR, Jagust W, Morris JC, Petersen RC, Saykin AJ, Shaw LM, Toga AW, Trojanowski JQ. Understanding disease progression and improving Alzheimer's disease clinical trials: Recent highlights from the Alzheimer's Disease Neuroimaging Initiative. Alzheimers Dement 2018; 15:106-152. [PMID: 30321505 DOI: 10.1016/j.jalz.2018.08.005] [Citation(s) in RCA: 286] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The overall goal of the Alzheimer's Disease Neuroimaging Initiative (ADNI) is to validate biomarkers for Alzheimer's disease (AD) clinical trials. ADNI is a multisite, longitudinal, observational study that has collected many biomarkers since 2004. Recent publications highlight the multifactorial nature of late-onset AD. We discuss selected topics that provide insights into AD progression and outline how this knowledge may improve clinical trials. METHODS We used standard methods to identify nearly 600 publications using ADNI data from 2016 and 2017 (listed in Supplementary Material and searchable at http://adni.loni.usc.edu/news-publications/publications/). RESULTS (1) Data-driven AD progression models supported multifactorial interactions rather than a linear cascade of events. (2) β-Amyloid (Aβ) deposition occurred concurrently with functional connectivity changes within the default mode network in preclinical subjects and was followed by specific and progressive disconnection of functional and anatomical networks. (3) Changes in functional connectivity, volumetric measures, regional hypometabolism, and cognition were detectable at subthreshold levels of Aβ deposition. 4. Tau positron emission tomography imaging studies detailed a specific temporal and spatial pattern of tau pathology dependent on prior Aβ deposition, and related to subsequent cognitive decline. 5. Clustering studies using a wide range of modalities consistently identified a "typical AD" subgroup and a second subgroup characterized by executive impairment and widespread cortical atrophy in preclinical and prodromal subjects. 6. Vascular pathology burden may act through both Aβ dependent and independent mechanisms to exacerbate AD progression. 7. The APOE ε4 allele interacted with cerebrovascular disease to impede Aβ clearance mechanisms. 8. Genetic approaches identified novel genetic risk factors involving a wide range of processes, and demonstrated shared genetic risk for AD and vascular disorders, as well as the temporal and regional pathological associations of established AD risk alleles. 9. Knowledge of early pathological changes guided the development of novel prognostic biomarkers for preclinical subjects. 10. Placebo populations of randomized controlled clinical trials had highly variable trajectories of cognitive change, underscoring the importance of subject selection and monitoring. 11. Selection criteria based on Aβ positivity, hippocampal volume, baseline cognitive/functional measures, and APOE ε4 status in combination with improved cognitive outcome measures were projected to decrease clinical trial duration and cost. 12. Multiple concurrent therapies targeting vascular health and other AD pathology in addition to Aβ may be more effective than single therapies. DISCUSSION ADNI publications from 2016 and 2017 supported the idea of AD as a multifactorial disease and provided insights into the complexities of AD disease progression. These findings guided the development of novel biomarkers and suggested that subject selection on the basis of multiple factors may lower AD clinical trial costs and duration. The use of multiple concurrent therapies in these trials may prove more effective in reversing AD disease progression.
Collapse
Affiliation(s)
- Dallas P Veitch
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA; Northern California Institute for Research and Education (NCIRE), Department of Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Michael W Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA; Department of Radiology, University of California, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, CA, USA.
| | - Paul S Aisen
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, CA, USA
| | - Laurel A Beckett
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Nigel J Cairns
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Robert C Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | | | - William Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - John C Morris
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute of Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Alzheimer's Disease Core Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Udall Parkinson's Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
180
|
Elliott LT, Sharp K, Alfaro-Almagro F, Shi S, Miller KL, Douaud G, Marchini J, Smith SM. Genome-wide association studies of brain imaging phenotypes in UK Biobank. Nature 2018; 562:210-216. [PMID: 30305740 PMCID: PMC6786974 DOI: 10.1038/s41586-018-0571-7] [Citation(s) in RCA: 489] [Impact Index Per Article: 69.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 09/04/2018] [Indexed: 12/16/2022]
Abstract
The genetic architecture of brain structure and function is largely unknown. To investigate this, we carried out genome-wide association studies of 3,144 functional and structural brain imaging phenotypes from UK Biobank (discovery dataset 8,428 subjects). Here we show that many of these phenotypes are heritable. We identify 148 clusters of associations between single nucleotide polymorphisms and imaging phenotypes that replicate at P < 0.05, when we would expect 21 to replicate by chance. Notable significant, interpretable associations include: iron transport and storage genes, related to magnetic susceptibility of subcortical brain tissue; extracellular matrix and epidermal growth factor genes, associated with white matter micro-structure and lesions; genes that regulate mid-line axon development, associated with organization of the pontine crossing tract; and overall 17 genes involved in development, pathway signalling and plasticity. Our results provide insights into the genetic architecture of the brain that are relevant to neurological and psychiatric disorders, brain development and ageing.
Collapse
Affiliation(s)
| | - Kevin Sharp
- Department of Statistics, University of Oxford, Oxford, UK
| | - Fidel Alfaro-Almagro
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Sinan Shi
- Department of Statistics, University of Oxford, Oxford, UK
| | - Karla L Miller
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Gwenaëlle Douaud
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Jonathan Marchini
- Department of Statistics, University of Oxford, Oxford, UK.
- The Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Stephen M Smith
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK.
| |
Collapse
|
181
|
Inkster B, Simmons A, Cole J, Schoof E, Linding R, Nichols T, Muglia P, Holsboer F, Saemann P, McGuffin P, Fu C, Miskowiak K, Matthews PM, Zai G, Nicodemus K. Unravelling the GSK3β-related genotypic interaction network influencing hippocampal volume in recurrent major depressive disorder. Psychiatr Genet 2018; 28:77-84. [PMID: 30080747 PMCID: PMC6531290 DOI: 10.1097/ypg.0000000000000203] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Glycogen synthase kinase 3β (GSK3β) has been implicated in mood disorders. We previously reported associations between a GSK3β polymorphism and hippocampal volume in major depressive disorder (MDD). We then reported similar associations for a subset of GSK3β-regulated genes. We now investigate an algorithm-derived comprehensive list of genes encoding proteins that directly interact with GSK3β to identify a genotypic network influencing hippocampal volume in MDD. PARTICIPANTS AND METHODS We used discovery (N=141) and replication (N=77) recurrent MDD samples. Our gene list was generated from the NetworKIN database. Hippocampal measures were derived using an optimized Freesurfer protocol. We identified interacting single nucleotide polymorphisms using the machine learning algorithm Random Forest and verified interactions using likelihood ratio tests between nested linear regression models. RESULTS The discovery sample showed multiple two-single nucleotide polymorphism interactions with hippocampal volume. The replication sample showed a replicable interaction (likelihood ratio test: P=0.0088, replication sample; P=0.017, discovery sample; Stouffer's combined P=0.0007) between genes associated previously with endoplasmic reticulum stress, calcium regulation and histone modifications. CONCLUSION Our results provide genetic evidence supporting associations between hippocampal volume and MDD, which may reflect underlying cellular stress responses. Our study provides evidence of biological mechanisms that should be further explored in the search for disease-modifying therapeutic targets for depression.
Collapse
Affiliation(s)
- Becky Inkster
- Department of Psychiatry, University of Cambridge, UK
- Wolfson College, University of Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, UK
| | - Andy Simmons
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - James Cole
- The Computational, Cognitive & Clinical Neuroimaging Lab, Department of Medicine, Imperial College London, UK
| | - Erwin Schoof
- Biotech Research & Innovation Centre, University of Copenhagen
| | - Rune Linding
- Biotech Research & Innovation Centre, University of Copenhagen
| | - Tom Nichols
- Department of Statistics, Warwick University, UK
| | - Pierandrea Muglia
- Genetics Division, Drug Discovery, Medicine Development Centre, GlaxoSmithKline, R&D, Verona, Italy
| | | | | | - Peter McGuffin
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Cynthia Fu
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Kamilla Miskowiak
- Department of Psychiatry, Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Paul M Matthews
- Department of Medicine, Imperial College London and UK Dementia Research Institute
| | - Gwyneth Zai
- Neurogenetics Section, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, and Mood & Anxiety Division, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Kristin Nicodemus
- Centre for Genomics and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, UK
| |
Collapse
|
182
|
Ikram MA, Zonneveld HI, Roshchupkin G, Smith AV, Franco OH, Sigurdsson S, van Duijn C, Uitterlinden AG, Launer LJ, Vernooij MW, Gudnason V, Adams HH. Heritability and genome-wide associations studies of cerebral blood flow in the general population. J Cereb Blood Flow Metab 2018; 38. [PMID: 28627999 PMCID: PMC6120124 DOI: 10.1177/0271678x17715861] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cerebral blood flow is an important process for brain functioning and its dysregulation is implicated in multiple neurological disorders. While environmental risk factors have been identified, it remains unclear to what extent the flow is regulated by genetics. Here we performed heritability and genome-wide association analyses of cerebral blood flow in a population-based cohort study. We included 4472 persons free of cortical infarcts who underwent genotyping and phase-contrast magnetic resonance flow imaging (mean age 64.8 ± 10.8 years). The flow rate, cross-sectional area of the vessel, and flow velocity through the vessel were measured in the basilar artery and bilateral carotids. We found that the flow rate of the basilar artery is most heritable (h2 (SE) = 24.1 (9.8), p-value = 0.0056), and this increased over age. The association studies revealed two significant loci for the right carotid artery area (rs12546630, p-value = 2.0 × 10-8) and velocity (rs2971609, p-value = 1.4 × 10-8), with the latter showing a concordant effect in an independent sample (N = 1350, p-value = 0.057, meta-analyzed p-value = 2.5 × 10-9). These loci were also associated with other cerebral blood flow parameters below genome-wide significance, and rs2971609 lies in a known migraine locus. These findings establish that cerebral blood flow is under genetic control with potential relevance for neurological diseases.
Collapse
Affiliation(s)
- M Arfan Ikram
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands.,2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands.,3 Department of Neurology, Erasmus MC, Rotterdam, the Netherlands
| | - Hazel I Zonneveld
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands.,2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Gennady Roshchupkin
- 2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands.,4 Department of Medical Informatics, Erasmus MC, Rotterdam, the Netherlands
| | - Albert V Smith
- 5 Icelandic Heart Association, Kopavogur, Iceland.,6 Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Oscar H Franco
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | | | | | | | | | - Meike W Vernooij
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands.,2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Vilmundur Gudnason
- 5 Icelandic Heart Association, Kopavogur, Iceland.,6 Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Hieab Hh Adams
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands.,2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
183
|
Vélez JI, Lopera F, Creagh PK, Piñeros LB, Das D, Cervantes-Henríquez ML, Acosta-López JE, Isaza-Ruget MA, Espinosa LG, Easteal S, Quintero GA, Silva CT, Mastronardi CA, Arcos-Burgos M. Targeting Neuroplasticity, Cardiovascular, and Cognitive-Associated Genomic Variants in Familial Alzheimer's Disease. Mol Neurobiol 2018; 56:3235-3243. [PMID: 30112632 PMCID: PMC6476862 DOI: 10.1007/s12035-018-1298-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/02/2018] [Indexed: 11/24/2022]
Abstract
The identification of novel genetic variants contributing to the widespread in the age of onset (AOO) of Alzheimer’s disease (AD) could aid in the prognosis and/or development of new therapeutic strategies focused on early interventions. We recruited 78 individuals with AD from the Paisa genetic isolate in Antioquia, Colombia. These individuals belong to the world largest multigenerational and extended pedigree segregating AD as a consequence of a dominant fully penetrant mutation in the PSEN1 gene and exhibit an AOO ranging from the early 1930s to the late 1970s. To shed light on the genetic underpinning that could explain the large spread of the age of onset (AOO) of AD, 64 single nucleotide polymorphisms (SNP) associated with neuroanatomical, cardiovascular, and cognitive measures in AD were genotyped. Standard quality control and filtering procedures were applied, and single- and multi-locus linear mixed-effects models were used to identify AOO-associated SNPs. A full two-locus interaction model was fitted to define how identified SNPs interact to modulate AOO. We identified two key epistatic interactions between the APOE*E2 allele and SNPs ASTN2-rs7852878 and SNTG1-rs16914781 that delay AOO by up to ~ 8 years (95% CI 3.2–12.7, P = 1.83 × 10−3) and ~ 7.6 years (95% CI 3.3–11.8, P = 8.69 × 10−4), respectively, and validated our previous finding indicating that APOE*E2 delays AOO of AD in PSEN1 E280 mutation carriers. This new evidence involving APOE*E2 as an AOO delayer could be used for developing precision medicine approaches and predictive genomics models to potentially determine AOO in individuals genetically predisposed to AD.
Collapse
Affiliation(s)
- Jorge I. Vélez
- Genomics and Predictive Medicine Group, Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600 Australia
- Universidad del Norte, Barranquilla, Colombia
| | - Francisco Lopera
- Neuroscience Research Group, University of Antioquia, Medellín, Colombia
| | - Penelope K. Creagh
- Genomics and Predictive Medicine Group, Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600 Australia
| | - Laura B. Piñeros
- GENIUROS, Center for Research in Genetics and Genomics, Institute of Translational Medicine, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Debjani Das
- Genome Diversity and Health Group, Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, ACT, Canberra, 2600 Australia
| | - Martha L. Cervantes-Henríquez
- Universidad del Norte, Barranquilla, Colombia
- Grupo de Neurociencias del Caribe, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Johan E. Acosta-López
- Grupo de Neurociencias del Caribe, Universidad Simón Bolívar, Barranquilla, Colombia
| | | | - Lady G. Espinosa
- INPAC Research Group, Fundación Universitaria Sanitas, Bogotá, Colombia
| | - Simon Easteal
- Genome Diversity and Health Group, Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, ACT, Canberra, 2600 Australia
| | - Gustavo A. Quintero
- Studies in Translational Microbiology and Emerging Diseases (MICROS) Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Claudia Tamar Silva
- GENIUROS, Center for Research in Genetics and Genomics, Institute of Translational Medicine, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Claudio A. Mastronardi
- Genomics and Predictive Medicine Group, Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600 Australia
- Neuroscience Group (NeUROS), Institute of Translational Medicine, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mauricio Arcos-Burgos
- Genomics and Predictive Medicine Group, Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600 Australia
- GENIUROS, Center for Research in Genetics and Genomics, Institute of Translational Medicine, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
184
|
De Jager PL, Ma Y, McCabe C, Xu J, Vardarajan BN, Felsky D, Klein HU, White CC, Peters MA, Lodgson B, Nejad P, Tang A, Mangravite LM, Yu L, Gaiteri C, Mostafavi S, Schneider JA, Bennett DA. A multi-omic atlas of the human frontal cortex for aging and Alzheimer's disease research. Sci Data 2018; 5:180142. [PMID: 30084846 PMCID: PMC6080491 DOI: 10.1038/sdata.2018.142] [Citation(s) in RCA: 360] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/26/2018] [Indexed: 12/14/2022] Open
Abstract
We initiated the systematic profiling of the dorsolateral prefrontal cortex obtained from a subset of autopsied individuals enrolled in the Religious Orders Study (ROS) or the Rush Memory and Aging Project (MAP), which are jointly designed prospective studies of aging and dementia with detailed, longitudinal cognitive phenotyping during life and a quantitative, structured neuropathologic examination after death. They include over 3,322 subjects. Here, we outline the first generation of data including genome-wide genotypes (n=2,090), whole genome sequencing (n=1,179), DNA methylation (n=740), chromatin immunoprecipitation with sequencing using an anti-Histone 3 Lysine 9 acetylation (H3K9Ac) antibody (n=712), RNA sequencing (n=638), and miRNA profile (n=702). Generation of other omic data including ATACseq, proteomic and metabolomics profiles is ongoing. Thanks to its prospective design and recruitment of older, non-demented individuals, these data can be repurposed to investigate a large number of syndromic and quantitative neuroscience phenotypes. The many subjects that are cognitively non-impaired at death also offer insights into the biology of the human brain in older non-impaired individuals.
Collapse
Affiliation(s)
- Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, 630 West 168 street, New York, NY 10032, USA
- Cell Circuits Program, Broad Institute, 415 Main street, Cambridge MA 02142, USA
| | - Yiyi Ma
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, 630 West 168 street, New York, NY 10032, USA
| | - Cristin McCabe
- Cell Circuits Program, Broad Institute, 415 Main street, Cambridge MA 02142, USA
| | - Jishu Xu
- Cell Circuits Program, Broad Institute, 415 Main street, Cambridge MA 02142, USA
| | - Badri N. Vardarajan
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, 630 West 168 street, New York, NY 10032, USA
| | - Daniel Felsky
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, 630 West 168 street, New York, NY 10032, USA
- Cell Circuits Program, Broad Institute, 415 Main street, Cambridge MA 02142, USA
| | - Hans-Ulrich Klein
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, 630 West 168 street, New York, NY 10032, USA
- Cell Circuits Program, Broad Institute, 415 Main street, Cambridge MA 02142, USA
| | - Charles C. White
- Cell Circuits Program, Broad Institute, 415 Main street, Cambridge MA 02142, USA
| | - Mette A. Peters
- Sage Bionetworks, 1100 Fairview Avenue N. Seattle WA 98109, USA
| | - Ben Lodgson
- Sage Bionetworks, 1100 Fairview Avenue N. Seattle WA 98109, USA
| | - Parham Nejad
- Cell Circuits Program, Broad Institute, 415 Main street, Cambridge MA 02142, USA
| | - Anna Tang
- Cell Circuits Program, Broad Institute, 415 Main street, Cambridge MA 02142, USA
| | | | - Lei Yu
- Rush Alzehimer Disease Center, RUSH University, 600 South Paulina Street, Chicago IL 60612, USA
| | - Chris Gaiteri
- Rush Alzehimer Disease Center, RUSH University, 600 South Paulina Street, Chicago IL 60612, USA
| | - Sara Mostafavi
- Departments of Statistics and Medical Genetics and Centre for Molecular Medicine and Therapeutics, University of British Columbia, 950 West 28 Avenue, Vancouver, British Columbia BC V5Z 4H4, Canada
| | - Julie A. Schneider
- Rush Alzehimer Disease Center, RUSH University, 600 South Paulina Street, Chicago IL 60612, USA
| | - David A. Bennett
- Rush Alzehimer Disease Center, RUSH University, 600 South Paulina Street, Chicago IL 60612, USA
| |
Collapse
|
185
|
Whitton L, Apostolova G, Rieder D, Dechant G, Rea S, Donohoe G, Morris DW. Genes regulated by SATB2 during neurodevelopment contribute to schizophrenia and educational attainment. PLoS Genet 2018; 14:e1007515. [PMID: 30040823 PMCID: PMC6097700 DOI: 10.1371/journal.pgen.1007515] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/17/2018] [Accepted: 06/26/2018] [Indexed: 12/20/2022] Open
Abstract
SATB2 is associated with schizophrenia and is an important transcription factor regulating neocortical organization and circuitry. Rare mutations in SATB2 cause a syndrome that includes developmental delay, and mouse studies identify an important role for SATB2 in learning and memory. Interacting partners BCL11B and GATAD2A are also schizophrenia risk genes indicating that other genes interacting with or are regulated by SATB2 are making a contribution to schizophrenia and cognition. We used data from Satb2 mouse models to generate three gene-sets that contain genes either functionally related to SATB2 or targeted by SATB2 at different stages of development. Each was tested for enrichment using the largest available genome-wide association studies (GWAS) datasets for schizophrenia and educational attainment (EA) and enrichment analysis was also performed for schizophrenia and other neurodevelopmental disorders using data from rare variant sequencing studies. These SATB2 gene-sets were enriched for genes containing common variants associated with schizophrenia and EA, and were enriched for genes containing rare variants reported in studies of schizophrenia, autism and intellectual disability. In the developing cortex, genes targeted by SATB2 based on ChIP-seq data, and functionally affected when SATB2 is not expressed based on differential expression analysis using RNA-seq data, show strong enrichment for genes associated with EA. For genes expressed in the hippocampus or at the synapse, those targeted by SATB2 are more strongly enriched for genes associated EA than gene-sets not targeted by SATB2. This study demonstrates that single gene findings from GWAS can provide important insights to pathobiological processes. In this case we find evidence that genes influenced by SATB2 and involved in synaptic transmission, axon guidance and formation of the corpus callosum are contributing to schizophrenia and cognition. Schizophrenia is a complex disorder caused by many genes. Using new gene discoveries to understand pathobiology is a foundation for development of new treatments. Current drugs for schizophrenia are only partially effective and do not treat cognitive deficits, which are key factors for explaining disability, leading to unemployment, homelessness and social isolation. Genome-wide association studies (GWAS) of schizophrenia have been effective at identifying individual SNPs and genes that contribute to risk but have struggled to immediately uncover the bigger picture of the underlying biology of the disorder. Here we take an individual gene identified in a schizophrenia GWAS called SATB2, which on its own is a very important regulator of brain development. We use functional genomics data from mouse studies to identify sets of others genes that are influenced by SATB2 during development. We show that these gene sets are enriched for common variants associated with schizophrenia and educational attainment (used as a proxy for cognition), and for rare variants that increase risk of various neurodevelopmental disorders. This study provides evidence that the molecular mechanisms that underpin schizophrenia and cognitive function include disruption of biological processes influenced by SATB2 as the brain is being organized and wired during development.
Collapse
Affiliation(s)
- Laura Whitton
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging, Cognition and Genomics (NICOG) Centre and NCBES Galway Neuroscience Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
| | - Galina Apostolova
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Dietmar Rieder
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Dechant
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Stephen Rea
- Centre for Chromosome Biology, Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
| | - Gary Donohoe
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging, Cognition and Genomics (NICOG) Centre and NCBES Galway Neuroscience Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
| | - Derek W. Morris
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging, Cognition and Genomics (NICOG) Centre and NCBES Galway Neuroscience Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
- * E-mail:
| |
Collapse
|
186
|
Vilor-Tejedor N, Alemany S, Cáceres A, Bustamante M, Pujol J, Sunyer J, González JR. Strategies for integrated analysis in imaging genetics studies. Neurosci Biobehav Rev 2018; 93:57-70. [PMID: 29944960 DOI: 10.1016/j.neubiorev.2018.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/30/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023]
Abstract
Imaging Genetics (IG) integrates neuroimaging and genomic data from the same individual, deepening our knowledge of the biological mechanisms behind neurodevelopmental domains and neurological disorders. Although the literature on IG has exponentially grown over the past years, the majority of studies have mainly analyzed associations between candidate brain regions and individual genetic variants. However, this strategy is not designed to deal with the complexity of neurobiological mechanisms underlying behavioral and neurodevelopmental domains. Moreover, larger sample sizes and increased multidimensionality of this type of data represents a challenge for standardizing modeling procedures in IG research. This review provides a systematic update of the methods and strategies currently used in IG studies, and serves as an analytical framework for researchers working in this field. To complement the functionalities of the Neuroconductor framework, we also describe existing R packages that implement these methodologies. In addition, we present an overview of how these methodological approaches are applied in integrating neuroimaging and genetic data.
Collapse
Affiliation(s)
- Natàlia Vilor-Tejedor
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Barcelona Beta Brain Research Center (BBRC) - Pasqual Maragall Foundation, Barcelona, Spain.
| | - Silvia Alemany
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Alejandro Cáceres
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Mariona Bustamante
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Jesús Pujol
- MRI Research Unit, Hospital del Mar, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM G21, Barcelona, Spain
| | - Jordi Sunyer
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Juan R González
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain.
| |
Collapse
|
187
|
m6A RNA Methylation Controls Neural Development and Is Involved in Human Diseases. Mol Neurobiol 2018; 56:1596-1606. [DOI: 10.1007/s12035-018-1138-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/18/2018] [Indexed: 12/31/2022]
|
188
|
Wolters FJ, Adams HH, Bos D, Licher S, Ikram MA. Three Decades of Dementia Research: Insights from One Small Community of Indomitable Rotterdammers. J Alzheimers Dis 2018; 64:S145-S159. [DOI: 10.3233/jad-179938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Frank J. Wolters
- Department of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Neurology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hieab H.H. Adams
- Department of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Silvan Licher
- Department of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
189
|
Smeland OB, Wang Y, Frei O, Li W, Hibar DP, Franke B, Bettella F, Witoelar A, Djurovic S, Chen CH, Thompson PM, Dale AM, Andreassen OA. Genetic Overlap Between Schizophrenia and Volumes of Hippocampus, Putamen, and Intracranial Volume Indicates Shared Molecular Genetic Mechanisms. Schizophr Bull 2018; 44:854-864. [PMID: 29136250 PMCID: PMC6007549 DOI: 10.1093/schbul/sbx148] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Schizophrenia (SCZ) is associated with differences in subcortical brain volumes and intracranial volume (ICV). However, little is known about the underlying etiology of these brain alterations. Here, we explored whether brain structure volumes and SCZ share genetic risk factors. Using conditional false discovery rate (FDR) analysis, we integrated genome-wide association study (GWAS) data on SCZ (n = 82315) and GWAS data on 7 subcortical brain volumes and ICV (n = 11840). By conditioning the FDR on overlapping associations, this statistical approach increases power to discover genetic loci. To assess the credibility of our approach, we studied the identified loci in larger GWAS samples on ICV (n = 26577) and hippocampal volume (n = 26814). We observed polygenic overlap between SCZ and volumes of hippocampus, putamen, and ICV. Based on conjunctional FDR < 0.05, we identified 2 loci shared between SCZ and ICV implicating genes FOXO3 (rs10457180) and ITIH4 (rs4687658), 2 loci shared between SCZ and hippocampal volume implicating SLC4A10 (rs4664442) and SPATS2L (rs1653290), and 2 loci shared between SCZ and volume of putamen implicating DCC (rs4632195) and DLG2 (rs11233632). The loci shared between SCZ and hippocampal volume or ICV had not reached significance in the primary GWAS on brain phenotypes. Proving our point of increased power, 2 loci did reach genome-wide significance with ICV (rs10457180) and hippocampal volume (rs4664442) in the larger GWAS. Three of the 6 identified loci are novel for SCZ. Altogether, the findings provide new insights into the relationship between SCZ and brain structure volumes, suggesting that their genetic architectures are not independent.
Collapse
Affiliation(s)
- Olav B Smeland
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Department of Neurosciences, University of California San Diego, La Jolla, CA,To whom correspondence should be addressed; Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Kirkeveien 166, 0424 Oslo, Norway; tel: +1-858-568-4915, fax: +47-230-273-33, e-mail:
| | - Yunpeng Wang
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Department of Radiology, University of California San Diego, La Jolla, CA,Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA
| | - Oleksandr Frei
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Wen Li
- NORMENT, KG 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 P Hibar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA
| | - Barbara Franke
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands,Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Francesco Bettella
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Aree Witoelar
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway,NORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Chi-Hua Chen
- Department of Radiology, University of California San Diego, La Jolla, CA,Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA
| | - Anders M Dale
- Department of Neurosciences, University of California San Diego, La Jolla, CA,Department of Radiology, University of California San Diego, La Jolla, CA,Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA,Department of Psychiatry, University of California San Diego, La Jolla, CA
| | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
190
|
Rao S, Ghani M, Guo Z, Deming Y, Wang K, Sims R, Mao C, Yao Y, Cruchaga C, Stephan DA, Rogaeva E. An APOE-independent cis-eSNP on chromosome 19q13.32 influences tau levels and late-onset Alzheimer's disease risk. Neurobiol Aging 2018; 66:178.e1-178.e8. [PMID: 29395286 PMCID: PMC7050280 DOI: 10.1016/j.neurobiolaging.2017.12.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/18/2017] [Accepted: 12/27/2017] [Indexed: 10/18/2022]
Abstract
Although multiple susceptibility loci for late-onset Alzheimer's disease (LOAD) have been identified, a large portion of the genetic risk for this disease remains unexplained. LOAD risk may be associated with single-nucleotide polymorphisms responsible for changes in gene expression (eSNPs). To detect eSNPs associated with LOAD, we integrated data from LOAD genome-wide association studies and expression quantitative trait loci using Sherlock (a Bayesian statistical method). We identified a cis-regulatory eSNP (rs2927438) located on chromosome 19q13.32, for which subsequent analyses confirmed the association with both LOAD risk and the expression level of several nearby genes. Importantly, rs2927438 may represent an APOE-independent LOAD eSNP according to the weak linkage disequilibrium of rs2927438 with the 2 polymorphisms (rs7412 and rs429358) defining the APOE-ε2, -ε3, and -ε4 alleles. Furthermore, rs2927438 does not influence chromatin interaction events at the APOE locus or cis-regulation of APOE expression. Further exploratory analysis revealed that rs2927438 is significantly associated with tau levels in the cerebrospinal fluid. Our findings suggest that rs2927438 may confer APOE-independent risk for LOAD.
Collapse
Affiliation(s)
- Shuquan Rao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.
| | - Mahdi Ghani
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhiyun Guo
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Yuetiva Deming
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Kesheng Wang
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| | - Rebecca Sims
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Canquan Mao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Yao Yao
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dietrich A Stephan
- Department of Human Genetics, Graduate School of Public Health, Pittsburgh, PA, USA
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
191
|
Precision medicine screening using whole-genome sequencing and advanced imaging to identify disease risk in adults. Proc Natl Acad Sci U S A 2018; 115:3686-3691. [PMID: 29555771 PMCID: PMC5889622 DOI: 10.1073/pnas.1706096114] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Advances in technology are enabling evaluation for prevention and early detection of age-related chronic diseases associated with premature mortality, such as cancer and cardiovascular diseases. These diseases kill about one-third of men and one-quarter of women between the ages of 50 and 74 years old in the United States. We used whole-genome sequencing, advanced imaging, and other clinical testing to screen 209 active, symptom-free adults. We identified a broad set of complementary age-related chronic disease risks associated with premature mortality. Reducing premature mortality associated with age-related chronic diseases, such as cancer and cardiovascular disease, is an urgent priority. We report early results using genomics in combination with advanced imaging and other clinical testing to proactively screen for age-related chronic disease risk among adults. We enrolled active, symptom-free adults in a study of screening for age-related chronic diseases associated with premature mortality. In addition to personal and family medical history and other clinical testing, we obtained whole-genome sequencing (WGS), noncontrast whole-body MRI, dual-energy X-ray absorptiometry (DXA), global metabolomics, a new blood test for prediabetes (Quantose IR), echocardiography (ECHO), ECG, and cardiac rhythm monitoring to identify age-related chronic disease risks. Precision medicine screening using WGS and advanced imaging along with other testing among active, symptom-free adults identified a broad set of complementary age-related chronic disease risks associated with premature mortality and strengthened WGS variant interpretation. This and other similarly designed screening approaches anchored by WGS and advanced imaging may have the potential to extend healthy life among active adults through improved prevention and early detection of age-related chronic diseases (and their risk factors) associated with premature mortality.
Collapse
|
192
|
Chen J, Rashid B, Yu Q, Liu J, Lin D, Du Y, Sui J, Calhoun VD. Variability in Resting State Network and Functional Network Connectivity Associated With Schizophrenia Genetic Risk: A Pilot Study. Front Neurosci 2018; 12:114. [PMID: 29545739 PMCID: PMC5838400 DOI: 10.3389/fnins.2018.00114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/13/2018] [Indexed: 12/19/2022] Open
Abstract
Imaging genetics posits a valuable strategy for elucidating genetic influences on brain abnormalities in psychiatric disorders. However, association analysis between 2D genetic data (subject × genetic variable) and 3D first-level functional magnetic resonance imaging (fMRI) data (subject × voxel × time) has been challenging given the asymmetry in data dimension. A summary feature needs to be derived for the imaging modality to compute inter-modality association at subject level. In this work, we propose to use variability in resting state networks (RSNs) and functional network connectivity (FNC) as potential features for purpose of association analysis. We conducted a pilot study to investigate the proposed features in a dataset of 171 healthy controls and 134 patients with schizophrenia (SZ). We computed variability in RSN and FNC in a group independent component analysis framework and tested three types of variability metrics, namely Euclidean distance, Pearson correlation and Kullback-Leibler (KL) divergence. Euclidean distance and Pearson correlation metrics more effectively discriminated controls from patients than KL divergence. The group differences observed with variability in RSN and FNC were highly consistent, indicating patients presenting increased deviation from the cohort-common pattern of RSN and FNC than controls. The variability in RSN and FNC showed significant associations with network global efficiency, the more the deviation, the lower the efficiency. Furthermore, the RSN and FNC variability were found to associate with individual SZ risk SNPs as well as cumulative polygenic risk score for SZ. Collectively the current findings provide preliminary evidence for variability in RSN and FNC being promising imaging features that may find applications as biomarkers and in imaging genetic association analysis.
Collapse
Affiliation(s)
- Jiayu Chen
- Mind Research Network, Albuquerque, NM, United States
| | - Barnaly Rashid
- Mind Research Network, Albuquerque, NM, United States
- Harvard Medical School, Harvard University, Boston, MA, United States
| | - Qingbao Yu
- Mind Research Network, Albuquerque, NM, United States
| | - Jingyu Liu
- Mind Research Network, Albuquerque, NM, United States
- Department of Electrical Engineering, University of New Mexico, Albuquerque, NM, United States
| | - Dongdong Lin
- Mind Research Network, Albuquerque, NM, United States
| | - Yuhui Du
- Mind Research Network, Albuquerque, NM, United States
- School of Computer & Information Technology, Shanxi University, Taiyuan, China
| | - Jing Sui
- Mind Research Network, Albuquerque, NM, United States
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Vince D. Calhoun
- Mind Research Network, Albuquerque, NM, United States
- Department of Electrical Engineering, University of New Mexico, Albuquerque, NM, United States
- Departments of Neurosciences and Psychiatry, University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
193
|
Roostaei T, Sadaghiani S, Mashhadi R, Falahatian M, Mohamadi E, Javadian N, Nazeri A, Doosti R, Naser Moghadasi A, Owji M, Hashemi Taheri AP, Shakouri Rad A, Azimi A, Voineskos AN, Nazeri A, Sahraian MA. Convergent effects of a functional C3 variant on brain atrophy, demyelination, and cognitive impairment in multiple sclerosis. Mult Scler 2018; 25:532-540. [PMID: 29485352 DOI: 10.1177/1352458518760715] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Complement system activation products are present in areas of neuroinflammation, demyelination, and neurodegeneration in brains of patients with multiple sclerosis (MS). C3 is a central element in the activation of complement cascades. A common coding variant in the C3 gene (rs2230199, C3R102G) affects C3 activity. OBJECTIVES To assess the effects of rs2230199 on MS severity using clinical, cognitive, and imaging measures. METHODS In total, 161 relapse-onset MS patients (Expanded Disability Status Scale (EDSS) ≤ 6) underwent physical assessments, cognitive tests (Paced Auditory Serial Addition Test (PASAT), Symbol Digit Modalities Test (SDMT), and California Verbal Learning Test (CVLT)), and magnetic resonance imaging (MRI). Lesion volumes were quantified semi-automatically. Voxel-wise analyses were performed to assess the effects of rs2230199 genotype on gray matter (GM) atrophy ( n = 155), white matter (WM) fractional anisotropy (FA; n = 105), and WM magnetization transfer ratio (MTR; n = 90). RESULTS While rs2230199 minor-allele dosage (C3-102G) showed no significant effect on EDSS and Multiple Sclerosis Functional Composite (MSFC), it was associated with worse cognitive performance ( p = 0.02), lower brain parenchymal fraction ( p = 0.003), and higher lesion burden ( p = 0.02). Moreover, voxel-wise analyses showed lower GM volume in subcortical structures and insula, and lower FA and MTR in several WM areas with higher copies of rs2230199 minor allele. CONCLUSION C3-rs2230199 affects white and GM damage as well as cognitive impairment in MS patients. Our findings support a causal role for complement system activity in the pathophysiology of MS.
Collapse
Affiliation(s)
- Tina Roostaei
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran/Interdisciplinary Neuroscience Research Program, Tehran University of Medical Sciences, Tehran, Iran/Kimel Family Translational Imaging-Genetics Laboratory, Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada/Department of Psychiatry, University of Toronto, Toronto, ON, Canada/Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Shokufeh Sadaghiani
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran/Interdisciplinary Neuroscience Research Program, Tehran University of Medical Sciences, Tehran, Iran
| | - Rahil Mashhadi
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masih Falahatian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran/School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Esmaeil Mohamadi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran
| | - Nina Javadian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran
| | - Aria Nazeri
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran/Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rozita Doosti
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran
| | - Abdorreza Naser Moghadasi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran/Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Owji
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran
| | | | - Ali Shakouri Rad
- Department of Radiology, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirreza Azimi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran/Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Aristotle N Voineskos
- Kimel Family Translational Imaging-Genetics Laboratory, Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada/Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Arash Nazeri
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran/Interdisciplinary Neuroscience Research Program, Tehran University of Medical Sciences, Tehran, Iran/Kimel Family Translational Imaging-Genetics Laboratory, Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mohammad Ali Sahraian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences and Sina Hospital, Tehran, Iran/Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
194
|
Unschuld PG. Novel Translational Research Methodology and the Prospect to a Better Understanding of Neurodegenerative Disease. NEURODEGENER DIS 2018; 18:1-4. [PMID: 29339665 DOI: 10.1159/000486565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Paul G Unschuld
- Hospital for Psychogeriatric Medicine, Psychiatric University Hospital Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| |
Collapse
|
195
|
Arslan A. Mapping the Schizophrenia Genes by Neuroimaging: The Opportunities and the Challenges. Int J Mol Sci 2018; 19:ijms19010219. [PMID: 29324666 PMCID: PMC5796168 DOI: 10.3390/ijms19010219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/05/2018] [Accepted: 01/07/2018] [Indexed: 12/18/2022] Open
Abstract
Schizophrenia (SZ) is a heritable brain disease originating from a complex interaction of genetic and environmental factors. The genes underpinning the neurobiology of SZ are largely unknown but recent data suggest strong evidence for genetic variations, such as single nucleotide polymorphisms, making the brain vulnerable to the risk of SZ. Structural and functional brain mapping of these genetic variations are essential for the development of agents and tools for better diagnosis, treatment and prevention of SZ. Addressing this, neuroimaging methods in combination with genetic analysis have been increasingly used for almost 20 years. So-called imaging genetics, the opportunities of this approach along with its limitations for SZ research will be outlined in this invited paper. While the problems such as reproducibility, genetic effect size, specificity and sensitivity exist, opportunities such as multivariate analysis, development of multisite consortia for large-scale data collection, emergence of non-candidate gene (hypothesis-free) approach of neuroimaging genetics are likely to contribute to a rapid progress for gene discovery besides to gene validation studies that are related to SZ.
Collapse
Affiliation(s)
- Ayla Arslan
- Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnica cesta, 15 Ilidza, Sarajevo 71210, Bosnia and Herzegovina.
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul 34662, Turkey.
| |
Collapse
|
196
|
Gan EH, Razvi S. The role of local thyroid hormone perturbation in hippocampal sclerosis dementia-commentary on a multi-modality study. Gland Surg 2018; 6:604-607. [PMID: 29302474 DOI: 10.21037/gs.2017.05.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Earn H Gan
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, UK.,Department of Endocrinology, Queen Elizabeth Hospital, Gateshead NE9 6SX, UK
| | - Salman Razvi
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, UK.,Department of Endocrinology, Queen Elizabeth Hospital, Gateshead NE9 6SX, UK
| |
Collapse
|
197
|
Affiliation(s)
- Kevin J. Mitchell
- Institutes of Genetics and Neuroscience; Trinity College Dublin; Dublin 2 Ireland
| |
Collapse
|
198
|
Adams SL, Benayoun L, Tilton K, Mellott TJ, Seshadri S, Blusztajn JK, Delalle I. Immunohistochemical Analysis of Activin Receptor-Like Kinase 1 (ACVRL1/ALK1) Expression in the Rat and Human Hippocampus: Decline in CA3 During Progression of Alzheimer's Disease. J Alzheimers Dis 2018; 63:1433-1443. [PMID: 29843236 PMCID: PMC5988976 DOI: 10.3233/jad-171065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The pathophysiology of Alzheimer's disease (AD) includes signaling defects mediated by the transforming growth factor β-bone morphogenetic protein-growth and differentiation factor (TGFβ-BMP-GDF) family of proteins. In animal models of AD, administration of BMP9/GDF2 improves memory and reduces amyloidosis. The best characterized type I receptor of BMP9 is ALK1. We characterized ALK1 expression in the hippocampus using immunohistochemistry. In the rat, ALK1 immunoreactivity was found in CA pyramidal neurons, most frequently and robustly in the CA2 and CA3 fields. In addition, there were sporadic ALK1-immunoreactive cells in the stratum oriens, mainly in CA1. The ALK1 expression pattern in human hippocampus was similar to that of rat. Pyramidal neurons within the CA2, CA3, and CA4 were strongly ALK1-immunoreactive in hippocampi of cognitively intact subjects with no neurofibrillary tangles. ALK1 signal was found in the axons of alveus and fimbria, and in the neuropil across CA fields. Relatively strongest ALK1 neuropil signal was observed in CA1 where pyramidal neurons were occasionally ALK1-immunoractive. As in the rat, horizontally oriented neurons in the stratum oriens of CA1 were both ALK1- and GAD67-immunoreactive. Analysis of ALK1 immunoreactivity across stages of AD pathology revealed that disease progression was characterized by overall reduction of the ALK1 signal in CA3 in advanced, but not early, stages of AD. These data suggest that the CA3 pyramidal neurons may remain responsive to the ALK1 ligands, e.g., BMP9, during initial stages of AD and that ALK1 may constitute a therapeutic target in early and moderate AD.
Collapse
Affiliation(s)
- Stephanie L. Adams
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Laurent Benayoun
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Kathy Tilton
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Tiffany J. Mellott
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Sudha Seshadri
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Jan Krzysztof Blusztajn
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
199
|
Fiandaca MS, Mapstone M, Connors E, Jacobson M, Monuki ES, Malik S, Macciardi F, Federoff HJ. Systems healthcare: a holistic paradigm for tomorrow. BMC SYSTEMS BIOLOGY 2017; 11:142. [PMID: 29258513 PMCID: PMC5738174 DOI: 10.1186/s12918-017-0521-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 12/01/2017] [Indexed: 12/13/2022]
Abstract
Systems healthcare is a holistic approach to health premised on systems biology and medicine. The approach integrates data from molecules, cells, organs, the individual, families, communities, and the natural and man-made environment. Both extrinsic and intrinsic influences constantly challenge the biological networks associated with wellness. Such influences may dysregulate networks and allow pathobiology to evolve, resulting in early clinical presentation that requires astute assessment and timely intervention for successful mitigation. Herein, we describe the components of relevant biological systems and the nature of progression from at-risk to manifest disease. We illustrate the systems approach by examining two relevant clinical examples: Alzheimer's and cardiovascular diseases. The implications of systems healthcare management are examined through the lens of economics, ethics, policy and the law. Finally, we propose the need to develop new educational paradigms to enhance the training of the health professional in an era of systems medicine.
Collapse
Affiliation(s)
- Massimo S Fiandaca
- Department of Neurology, School of Medicine, Irvine, USA
- Department of Neurological Surgery, School of Medicine, Irvine, USA
- Department of Anatomy & Neurobiology, School of Medicine, Irvine, USA
| | - Mark Mapstone
- Department of Neurology, School of Medicine, Irvine, USA
| | | | - Mireille Jacobson
- Department of Economics, Paul Merage School of Business, Irvine, USA
| | - Edwin S Monuki
- Department of Pathology & Laboratory Medicine, School of Medicine, Irvine, USA
| | - Shaista Malik
- Department of Medicine, School of Medicine, Irvine, USA
| | - Fabio Macciardi
- Department of Psychiatry & Human Behavior, School of Medicine, Irvine, USA
| | - Howard J Federoff
- Department of Neurology, School of Medicine, Irvine, USA.
- University of California Irvine (UCI), Irvine, CA, USA.
| |
Collapse
|
200
|
Mufford MS, Stein DJ, Dalvie S, Groenewold NA, Thompson PM, Jahanshad N. Neuroimaging genomics in psychiatry-a translational approach. Genome Med 2017; 9:102. [PMID: 29179742 PMCID: PMC5704437 DOI: 10.1186/s13073-017-0496-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Neuroimaging genomics is a relatively new field focused on integrating genomic and imaging data in order to investigate the mechanisms underlying brain phenotypes and neuropsychiatric disorders. While early work in neuroimaging genomics focused on mapping the associations of candidate gene variants with neuroimaging measures in small cohorts, the lack of reproducible results inspired better-powered and unbiased large-scale approaches. Notably, genome-wide association studies (GWAS) of brain imaging in thousands of individuals around the world have led to a range of promising findings. Extensions of such approaches are now addressing epigenetics, gene–gene epistasis, and gene–environment interactions, not only in brain structure, but also in brain function. Complementary developments in systems biology might facilitate the translation of findings from basic neuroscience and neuroimaging genomics to clinical practice. Here, we review recent approaches in neuroimaging genomics—we highlight the latest discoveries, discuss advantages and limitations of current approaches, and consider directions by which the field can move forward to shed light on brain disorders.
Collapse
Affiliation(s)
- Mary S Mufford
- UCT/MRC Human Genetics Research Unit, Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa, 7925
| | - Dan J Stein
- MRC Unit on Risk and Resilience, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa, 7925.,Department of Psychiatry and Mental Health, Groote Schuur Hospital, Cape Town, South Africa, 7925
| | - Shareefa Dalvie
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa, 7925
| | - Nynke A Groenewold
- Department of Psychiatry and Mental Health, Groote Schuur Hospital, Cape Town, South Africa, 7925
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90292, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90292, USA.
| |
Collapse
|