551
|
Franke B, Stein JL, Ripke S, Anttila V, Hibar DP, van Hulzen KJE, Arias-Vasquez A, Smoller JW, Nichols TE, Neale MC, McIntosh AM, Lee P, McMahon FJ, Meyer-Lindenberg A, Mattheisen M, Andreassen OA, Gruber O, Sachdev PS, Roiz-Santiañez R, Saykin AJ, Ehrlich S, Mather KA, Turner JA, Schwarz E, Thalamuthu A, Shugart YY, Ho YYW, Martin NG, Wright MJ, Schizophrenia Working Group of the Psychiatric Genomics Consortium, ENIGMA Consortium, O'Donovan MC, Thompson PM, Neale BM, Medland SE, Sullivan PF. Genetic influences on schizophrenia and subcortical brain volumes: large-scale proof of concept. Nat Neurosci 2016; 19:420-431. [PMID: 26854805 PMCID: PMC4852730 DOI: 10.1038/nn.4228] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/22/2015] [Indexed: 12/12/2022]
Abstract
Schizophrenia is a devastating psychiatric illness with high heritability. Brain structure and function differ, on average, between people with schizophrenia and healthy individuals. As common genetic associations are emerging for both schizophrenia and brain imaging phenotypes, we can now use genome-wide data to investigate genetic overlap. Here we integrated results from common variant studies of schizophrenia (33,636 cases, 43,008 controls) and volumes of several (mainly subcortical) brain structures (11,840 subjects). We did not find evidence of genetic overlap between schizophrenia risk and subcortical volume measures either at the level of common variant genetic architecture or for single genetic markers. These results provide a proof of concept (albeit based on a limited set of structural brain measures) and define a roadmap for future studies investigating the genetic covariance between structural or functional brain phenotypes and risk for psychiatric disorders.
Collapse
Affiliation(s)
- 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
| | - Jason L Stein
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA, USA
- Neurogenetics Program, Department of Neurology, UCLA School of Medicine, Los Angeles, USA
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, CCM, Berlin, Germany
| | - Verneri Anttila
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - 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, USA
| | - Kimm J E van Hulzen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Alejandro Arias-Vasquez
- 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
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jordan W Smoller
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Thomas E Nichols
- FMRIB Centre, University of Oxford, United Kingdom
- Department of Statistics & WMG, University of Warwick, Coventry, United Kingdom
| | - Michael C Neale
- Departments of Psychiatry & Human Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew M McIntosh
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Phil Lee
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Francis J McMahon
- Intramural Research Program, National Institutes of Health, US Dept of Health & Human Services, Bethesda, USA
| | - Andreas Meyer-Lindenberg
- Central Institute of Mental Health, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Manuel Mattheisen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus and Copenhagen, Denmark
- Center for integrated Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
| | - Ole A Andreassen
- NORMENT - KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Oliver Gruber
- Center for Translational Research in Systems Neuroscience and Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center, Goettingen, Germany
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales (UNSW), Sydney, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, Australia
| | - Roberto Roiz-Santiañez
- Department of Psychiatry, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria-IDIVAL, Santander, Spain
- Cibersam (Centro Investigación Biomédica en Red Salud Mental), Madrid, Spain
| | - Andrew J Saykin
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, USA
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Stefan Ehrlich
- Department of Child and Adolescent Psychiatry, Faculty of Medicine and University Hospital, TU Dresden, Dresden, Germany
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales (UNSW), Sydney, Australia
| | - Jessica A Turner
- Georgia State University, Atlanta, USA
- Mind Research Network, Albuquerque, NM, USA
| | - Emanuel Schwarz
- Central Institute of Mental Health, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales (UNSW), Sydney, Australia
| | - Yin Yao Shugart
- Intramural Research Program, National Institutes of Health, US Dept of Health & Human Services, Bethesda, USA
| | - Yvonne YW Ho
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Margaret J Wright
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- School of Psychology, University of Queensland, Brisbane, Australia
| | | | | | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
- National Centre for Mental Health, Cardiff University, Cardiff, UK
| | - 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, USA
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
552
|
Whelan CD, Hibar DP, van Velzen LS, Zannas AS, Carrillo-Roa T, McMahon K, Prasad G, Kelly S, Faskowitz J, deZubiracay G, Iglesias JE, van Erp TGM, Frodl T, Martin NG, Wright MJ, Jahanshad N, Schmaal L, Sämann PG, Thompson PM. Heritability and reliability of automatically segmented human hippocampal formation subregions. Neuroimage 2016; 128:125-137. [PMID: 26747746 PMCID: PMC4883013 DOI: 10.1016/j.neuroimage.2015.12.039] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/28/2015] [Accepted: 12/23/2015] [Indexed: 12/01/2022] Open
Abstract
The human hippocampal formation can be divided into a set of cytoarchitecturally and functionally distinct subregions, involved in different aspects of memory formation. Neuroanatomical disruptions within these subregions are associated with several debilitating brain disorders including Alzheimer's disease, major depression, schizophrenia, and bipolar disorder. Multi-center brain imaging consortia, such as the Enhancing Neuro Imaging Genetics through Meta-Analysis (ENIGMA) consortium, are interested in studying disease effects on these subregions, and in the genetic factors that affect them. For large-scale studies, automated extraction and subsequent genomic association studies of these hippocampal subregion measures may provide additional insight. Here, we evaluated the test-retest reliability and transplatform reliability (1.5T versus 3T) of the subregion segmentation module in the FreeSurfer software package using three independent cohorts of healthy adults, one young (Queensland Twins Imaging Study, N=39), another elderly (Alzheimer's Disease Neuroimaging Initiative, ADNI-2, N=163) and another mixed cohort of healthy and depressed participants (Max Planck Institute, MPIP, N=598). We also investigated agreement between the most recent version of this algorithm (v6.0) and an older version (v5.3), again using the ADNI-2 and MPIP cohorts in addition to a sample from the Netherlands Study for Depression and Anxiety (NESDA) (N=221). Finally, we estimated the heritability (h(2)) of the segmented subregion volumes using the full sample of young, healthy QTIM twins (N=728). Test-retest reliability was high for all twelve subregions in the 3T ADNI-2 sample (intraclass correlation coefficient (ICC)=0.70-0.97) and moderate-to-high in the 4T QTIM sample (ICC=0.5-0.89). Transplatform reliability was strong for eleven of the twelve subregions (ICC=0.66-0.96); however, the hippocampal fissure was not consistently reconstructed across 1.5T and 3T field strengths (ICC=0.47-0.57). Between-version agreement was moderate for the hippocampal tail, subiculum and presubiculum (ICC=0.78-0.84; Dice Similarity Coefficient (DSC)=0.55-0.70), and poor for all other subregions (ICC=0.34-0.81; DSC=0.28-0.51). All hippocampal subregion volumes were highly heritable (h(2)=0.67-0.91). Our findings indicate that eleven of the twelve human hippocampal subregions segmented using FreeSurfer version 6.0 may serve as reliable and informative quantitative phenotypes for future multi-site imaging genetics initiatives such as those of the ENIGMA consortium.
Collapse
Affiliation(s)
- Christopher D Whelan
- Imaging Genetics Center, University of Southern California, Marina del Rey, CA, USA
| | - Derrek P Hibar
- Imaging Genetics Center, University of Southern California, Marina del Rey, CA, USA
| | - Laura S van Velzen
- Department of Psychiatry and Neuroscience Campus Amsterdam, VU University Medical Center and GGZ inGeest, Amsterdam, The Netherlands
| | - Anthony S Zannas
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Tania Carrillo-Roa
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Katie McMahon
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - Gautam Prasad
- Imaging Genetics Center, University of Southern California, Marina del Rey, CA, USA
| | - Sinéad Kelly
- Imaging Genetics Center, University of Southern California, Marina del Rey, CA, USA
| | - Joshua Faskowitz
- Imaging Genetics Center, University of Southern California, Marina del Rey, CA, USA
| | - Greig deZubiracay
- Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Juan E Iglesias
- Basque Center on Cognition, Brain and Language, Donostia, Gipuzkoa, Spain
| | - Theo G M van Erp
- Department of Psychiatry and Human Behavior, University of California, Irvine, USA
| | - Thomas Frodl
- Department of Psychiatry, Otto-von Guericke-University of Magdeburg, Germany
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Neda Jahanshad
- Imaging Genetics Center, University of Southern California, Marina del Rey, CA, USA
| | - Lianne Schmaal
- Department of Psychiatry and Neuroscience Campus Amsterdam, VU University Medical Center and GGZ inGeest, Amsterdam, The Netherlands
| | - Philipp G Sämann
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Paul M Thompson
- Imaging Genetics Center, University of Southern California, Marina del Rey, CA, USA.
| |
Collapse
|
553
|
Abstract
Although genetic studies of Bipolar Disorder have been pursued for decades, it has only been in the last several years that clearly replicated findings have emerged. These findings, typically of modest effects, point to a polygenic genetic architecture consisting of multiple common and rare susceptibility variants. While larger genome-wide association studies are ongoing, the advent of whole exome and genome sequencing should lead to the identification of rare, and potentially more penetrant, variants. Progress along both fronts will provide novel insights into the biology of Bipolar Disorder and help usher in a new era of personalized medicine and improved treatments.
Collapse
|
554
|
Zhao Y, Castellanos FX. Annual Research Review: Discovery science strategies in studies of the pathophysiology of child and adolescent psychiatric disorders--promises and limitations. J Child Psychol Psychiatry 2016; 57:421-39. [PMID: 26732133 PMCID: PMC4760897 DOI: 10.1111/jcpp.12503] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/17/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Psychiatric science remains descriptive, with a categorical nosology intended to enhance interobserver reliability. Increased awareness of the mismatch between categorical classifications and the complexity of biological systems drives the search for novel frameworks including discovery science in Big Data. In this review, we provide an overview of incipient approaches, primarily focused on classically categorical diagnoses such as schizophrenia (SZ), autism spectrum disorder (ASD), and attention-deficit/hyperactivity disorder (ADHD), but also reference convincing, if focal, advances in cancer biology, to describe the challenges of Big Data and discovery science, and outline approaches being formulated to overcome existing obstacles. FINDINGS A paradigm shift from categorical diagnoses to a domain/structure-based nosology and from linear causal chains to complex causal network models of brain-behavior relationship is ongoing. This (r)evolution involves appreciating the complexity, dimensionality, and heterogeneity of neuropsychiatric data collected from multiple sources ('broad' data) along with data obtained at multiple levels of analysis, ranging from genes to molecules, cells, circuits, and behaviors ('deep' data). Both of these types of Big Data landscapes require the use and development of robust and powerful informatics and statistical approaches. Thus, we describe Big Data analysis pipelines and the promise and potential limitations in using Big Data approaches to study psychiatric disorders. CONCLUSIONS We highlight key resources available for psychopathological studies and call for the application and development of Big Data approaches to dissect the causes and mechanisms of neuropsychiatric disorders and identify corresponding biomarkers for early diagnosis.
Collapse
Affiliation(s)
- Yihong Zhao
- Department of Child and Adolescent Psychiatry, NYU Child Study Center at NYU Langone Medical Center, New York, NY 10016, USA
| | - F. Xavier Castellanos
- Department of Child and Adolescent Psychiatry, NYU Child Study Center at NYU Langone Medical Center, New York, NY 10016, USA,Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| |
Collapse
|
555
|
Lee A, Qiu A. Modulative effects of COMT haplotype on age-related associations with brain morphology. Hum Brain Mapp 2016; 37:2068-82. [PMID: 26920810 DOI: 10.1002/hbm.23161] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 02/09/2016] [Accepted: 02/16/2016] [Indexed: 12/25/2022] Open
Abstract
Catechol-O-methyltransferase (COMT), located on chromosome 22q11.2, encodes an enzyme critical for dopamine flux in the prefrontal cortex. Genetic variants of COMT have been suggested to functionally manipulate prefrontal morphology and function in healthy adults. This study aims to investigate modulative roles of individuals COMT SNPs (rs737865, val158met, rs165599) and its haplotypes in age-related brain morphology using an Asian sample with 174 adults aged from 21 to 80 years. We showed an age-related decline in cortical thickness of the dorsal visual pathway, including the left dorsolateral prefrontal cortex, bilateral angular gyrus, right superior frontal cortex, and age-related shape compression in the basal ganglia as a function of the genotypes of the individual COMT SNPs, especially COMT val158met. Using haplotype trend regression analysis, COMT haplotype probabilities were estimated and further revealed an age-related decline in cortical thickness in the default mode network (DMN), including the posterior cingulate, precuneus, supramarginal and paracentral cortex, and the ventral visual system, including the occipital cortex and left inferior temporal cortex, as a function of the COMT haplotype. Our results provided new evidence on an antagonistic pleiotropic effect in COMT, suggesting that genetically programmed neural benefits in early life may have a potential bearing towards neural susceptibility in later life. Hum Brain Mapp 37:2068-2082, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Annie Lee
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore.,Clinical Imaging Research Center, National University of Singapore, Singapore, 117456, Singapore.,Singapore Institute for Clinical Sciences, the Agency for Science, Technology and Research, Singapore, 117609, Singapore
| |
Collapse
|
556
|
Becker M, Guadalupe T, Franke B, Hibar DP, Renteria ME, Stein JL, Thompson PM, Francks C, Vernes SC, Fisher SE. Early developmental gene enhancers affect subcortical volumes in the adult human brain. Hum Brain Mapp 2016; 37:1788-800. [PMID: 26890892 DOI: 10.1002/hbm.23136] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 12/30/2015] [Accepted: 01/26/2016] [Indexed: 11/08/2022] Open
Abstract
Genome-wide association screens aim to identify common genetic variants contributing to the phenotypic variability of complex traits, such as human height or brain morphology. The identified genetic variants are mostly within noncoding genomic regions and the biology of the genotype-phenotype association typically remains unclear. In this article, we propose a complementary targeted strategy to reveal the genetic underpinnings of variability in subcortical brain volumes, by specifically selecting genomic loci that are experimentally validated forebrain enhancers, active in early embryonic development. We hypothesized that genetic variation within these enhancers may affect the development and ultimately the structure of subcortical brain regions in adults. We tested whether variants in forebrain enhancer regions showed an overall enrichment of association with volumetric variation in subcortical structures of >13,000 healthy adults. We observed significant enrichment of genomic loci that affect the volume of the hippocampus within forebrain enhancers (empirical P = 0.0015), a finding which robustly passed the adjusted threshold for testing of multiple brain phenotypes (cutoff of P < 0.0083 at an alpha of 0.05). In analyses of individual single nucleotide polymorphisms (SNPs), we identified an association upstream of the ID2 gene with rs7588305 and variation in hippocampal volume. This SNP-based association survived multiple-testing correction for the number of SNPs analyzed but not for the number of subcortical structures. Targeting known regulatory regions offers a way to understand the underlying biology that connects genotypes to phenotypes, particularly in the context of neuroimaging genetics. This biology-driven approach generates testable hypotheses regarding the functional biology of identified associations. Hum Brain Mapp 37:1788-1800, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Martin Becker
- Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Tulio Guadalupe
- Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Barbara Franke
- Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands.,Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Derrek P Hibar
- Imaging Genetics Center, Keck School of Medicine, University of Southern California, Marina Del Rey, California
| | - Miguel E Renteria
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jason L Stein
- Imaging Genetics Center, Keck School of Medicine, University of Southern California, Marina Del Rey, California.,Department of Neurology, Neurogenetics Program, UCLA School of Medicine, Los Angeles, California
| | - Paul M Thompson
- Imaging Genetics Center, Keck School of Medicine, University of Southern California, Marina Del Rey, California
| | - Clyde Francks
- Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| | - Sonja C Vernes
- Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| | - Simon E Fisher
- Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| |
Collapse
|
557
|
Holland D, Wang Y, Thompson WK, Schork A, Chen CH, Lo MT, Witoelar A, Werge T, O'Donovan M, Andreassen OA, Dale AM. Estimating Effect Sizes and Expected Replication Probabilities from GWAS Summary Statistics. Front Genet 2016; 7:15. [PMID: 26909100 PMCID: PMC4754432 DOI: 10.3389/fgene.2016.00015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/28/2016] [Indexed: 12/19/2022] Open
Abstract
Genome-wide Association Studies (GWAS) result in millions of summary statistics (“z-scores”) for single nucleotide polymorphism (SNP) associations with phenotypes. These rich datasets afford deep insights into the nature and extent of genetic contributions to complex phenotypes such as psychiatric disorders, which are understood to have substantial genetic components that arise from very large numbers of SNPs. The complexity of the datasets, however, poses a significant challenge to maximizing their utility. This is reflected in a need for better understanding the landscape of z-scores, as such knowledge would enhance causal SNP and gene discovery, help elucidate mechanistic pathways, and inform future study design. Here we present a parsimonious methodology for modeling effect sizes and replication probabilities, relying only on summary statistics from GWAS substudies, and a scheme allowing for direct empirical validation. We show that modeling z-scores as a mixture of Gaussians is conceptually appropriate, in particular taking into account ubiquitous non-null effects that are likely in the datasets due to weak linkage disequilibrium with causal SNPs. The four-parameter model allows for estimating the degree of polygenicity of the phenotype and predicting the proportion of chip heritability explainable by genome-wide significant SNPs in future studies with larger sample sizes. We apply the model to recent GWAS of schizophrenia (N = 82,315) and putamen volume (N = 12,596), with approximately 9.3 million SNP z-scores in both cases. We show that, over a broad range of z-scores and sample sizes, the model accurately predicts expectation estimates of true effect sizes and replication probabilities in multistage GWAS designs. We assess the degree to which effect sizes are over-estimated when based on linear-regression association coefficients. We estimate the polygenicity of schizophrenia to be 0.037 and the putamen to be 0.001, while the respective sample sizes required to approach fully explaining the chip heritability are 106 and 105. The model can be extended to incorporate prior knowledge such as pleiotropy and SNP annotation. The current findings suggest that the model is applicable to a broad array of complex phenotypes and will enhance understanding of their genetic architectures.
Collapse
Affiliation(s)
- Dominic Holland
- Multimodal Imaging Laboratory, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Neurosciences, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Yunpeng Wang
- Multimodal Imaging Laboratory, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Neurosciences, University of CaliforniaSan Diego, La Jolla, CA, USA; NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of OsloOslo, Norway; Division of Mental Health and Addiction, Oslo University HospitalOslo, Norway
| | - Wesley K Thompson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Andrew Schork
- Multimodal Imaging Laboratory, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Cognitive Sciences, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Chi-Hua Chen
- Multimodal Imaging Laboratory, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Radiology, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Min-Tzu Lo
- Multimodal Imaging Laboratory, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Radiology, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Aree Witoelar
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of OsloOslo, Norway; Division of Mental Health and Addiction, Oslo University HospitalOslo, Norway
| | | | | | - Thomas Werge
- Institute of Biological Psychiatry, MHC, Sct. Hans Hospital and University of Copenhagen Copenhagen, Denmark
| | - Michael O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University Cardiff, UK
| | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of OsloOslo, Norway; Division of Mental Health and Addiction, Oslo University HospitalOslo, Norway
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Neurosciences, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Psychiatry, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Radiology, University of CaliforniaSan Diego, La Jolla, CA, USA
| |
Collapse
|
558
|
Lorio S, Kherif F, Ruef A, Melie-Garcia L, Frackowiak R, Ashburner J, Helms G, Lutti A, Draganski B. Neurobiological origin of spurious brain morphological changes: A quantitative MRI study. Hum Brain Mapp 2016; 37:1801-15. [PMID: 26876452 PMCID: PMC4855623 DOI: 10.1002/hbm.23137] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 01/18/2016] [Accepted: 01/26/2016] [Indexed: 01/04/2023] Open
Abstract
The high gray‐white matter contrast and spatial resolution provided by T1‐weighted magnetic resonance imaging (MRI) has made it a widely used imaging protocol for computational anatomy studies of the brain. While the image intensity in T1‐weighted images is predominantly driven by T1, other MRI parameters affect the image contrast, and hence brain morphological measures derived from the data. Because MRI parameters are correlates of different histological properties of brain tissue, this mixed contribution hampers the neurobiological interpretation of morphometry findings, an issue which remains largely ignored in the community. We acquired quantitative maps of the MRI parameters that determine signal intensities in T1‐weighted images (R1 (=1/T1), R2*, and PD) in a large cohort of healthy subjects (n = 120, aged 18–87 years). Synthetic T1‐weighted images were calculated from these quantitative maps and used to extract morphometry features—gray matter volume and cortical thickness. We observed significant variations in morphometry measures obtained from synthetic images derived from different subsets of MRI parameters. We also detected a modulation of these variations by age. Our findings highlight the impact of microstructural properties of brain tissue—myelination, iron, and water content—on automated measures of brain morphology and show that microstructural tissue changes might lead to the detection of spurious morphological changes in computational anatomy studies. They motivate a review of previous morphological results obtained from standard anatomical MRI images and highlight the value of quantitative MRI data for the inference of microscopic tissue changes in the healthy and diseased brain. Hum Brain Mapp 37:1801–1815, 2016. © 2016 The Authors. Human Brain Mapping Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sara Lorio
- LREN - Department of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - Ferath Kherif
- LREN - Department of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - Anne Ruef
- LREN - Department of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - Lester Melie-Garcia
- LREN - Department of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - Richard Frackowiak
- LREN - Department of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - John Ashburner
- Wellcome Trust Centre for Neuroimaging, UCL Institute of Neurology, UCL, London, United Kingdom
| | - Gunther Helms
- Department of Clinical Sciences, Lund University, Medical Radiation Physics, Lund, Sweden
| | - Antoine Lutti
- LREN - Department of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - Bodgan Draganski
- LREN - Department of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland.,Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
559
|
Doshi J, Erus G, Ou Y, Resnick SM, Gur RC, Gur RE, Satterthwaite TD, Furth S, Davatzikos C, The Alzheimer's Neuroimaging Initiative. MUSE: MUlti-atlas region Segmentation utilizing Ensembles of registration algorithms and parameters, and locally optimal atlas selection. Neuroimage 2016; 127:186-195. [PMID: 26679328 PMCID: PMC4806537 DOI: 10.1016/j.neuroimage.2015.11.073] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 11/30/2015] [Accepted: 11/30/2015] [Indexed: 11/21/2022] Open
Abstract
Atlas-based automated anatomical labeling is a fundamental tool in medical image segmentation, as it defines regions of interest for subsequent analysis of structural and functional image data. The extensive investigation of multi-atlas warping and fusion techniques over the past 5 or more years has clearly demonstrated the advantages of consensus-based segmentation. However, the common approach is to use multiple atlases with a single registration method and parameter set, which is not necessarily optimal for every individual scan, anatomical region, and problem/data-type. Different registration criteria and parameter sets yield different solutions, each providing complementary information. Herein, we present a consensus labeling framework that generates a broad ensemble of labeled atlases in target image space via the use of several warping algorithms, regularization parameters, and atlases. The label fusion integrates two complementary sources of information: a local similarity ranking to select locally optimal atlases and a boundary modulation term to refine the segmentation consistently with the target image's intensity profile. The ensemble approach consistently outperforms segmentations using individual warping methods alone, achieving high accuracy on several benchmark datasets. The MUSE methodology has been used for processing thousands of scans from various datasets, producing robust and consistent results. MUSE is publicly available both as a downloadable software package, and as an application that can be run on the CBICA Image Processing Portal (https://ipp.cbica.upenn.edu), a web based platform for remote processing of medical images.
Collapse
Affiliation(s)
- Jimit Doshi
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Yangming Ou
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Martinos Biomedical Imaging Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, USA
| | - Ruben C. Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Raquel E. Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Theodore D. Satterthwaite
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Susan Furth
- Division of Nephrology, Childrens Hospital of Philadelphia, 34th and Civic Center Boulevard, Philadelphia PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
560
|
Giddaluru S, Espeseth T, Salami A, Westlye LT, Lundquist A, Christoforou A, Cichon S, Adolfsson R, Steen VM, Reinvang I, Nilsson LG, Le Hellard S, Nyberg L. Genetics of structural connectivity and information processing in the brain. Brain Struct Funct 2016; 221:4643-4661. [PMID: 26852023 PMCID: PMC5102980 DOI: 10.1007/s00429-016-1194-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 01/22/2016] [Indexed: 12/20/2022]
Abstract
Understanding the genetic factors underlying brain structural connectivity is a major challenge in imaging genetics. Here, we present results from genome-wide association studies (GWASs) of whole-brain white matter (WM) fractional anisotropy (FA), an index of microstructural coherence measured using diffusion tensor imaging. Data from independent GWASs of 355 Swedish and 250 Norwegian healthy adults were integrated by meta-analysis to enhance power. Complementary GWASs on behavioral data reflecting processing speed, which is related to microstructural properties of WM pathways, were performed and integrated with WM FA results via multimodal analysis to identify shared genetic associations. One locus on chromosome 17 (rs145994492) showed genome-wide significant association with WM FA (meta P value = 1.87 × 10-08). Suggestive associations (Meta P value <1 × 10-06) were observed for 12 loci, including one containing ZFPM2 (lowest meta P value = 7.44 × 10-08). This locus was also implicated in multimodal analysis of WM FA and processing speed (lowest Fisher P value = 8.56 × 10-07). ZFPM2 is relevant in specification of corticothalamic neurons during brain development. Analysis of SNPs associated with processing speed revealed association with a locus that included SSPO (lowest meta P value = 4.37 × 10-08), which has been linked to commissural axon growth. An intergenic SNP (rs183854424) 14 kb downstream of CSMD1, which is implicated in schizophrenia, showed suggestive evidence of association in the WM FA meta-analysis (meta P value = 1.43 × 10-07) and the multimodal analysis (Fisher P value = 1 × 10-07). These findings provide novel data on the genetics of WM pathways and processing speed, and highlight a role of ZFPM2 and CSMD1 in information processing in the brain.
Collapse
Affiliation(s)
- Sudheer Giddaluru
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, 5021, Bergen, Norway.,K.G.Jebsen Center for Psychosis Research and the Norwegian Center for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, 5021, Bergen, Norway
| | - Thomas Espeseth
- K.G. Jebsen Center for Psychosis Research, Norwegian Center for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0424, Oslo, Norway.,Department of Psychology, University of Oslo, 0317, Oslo, Norway
| | - Alireza Salami
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, 90187, Umeå, Sweden.,Aging Research Center, Karolinska Institutet and Stockholm University, 11330, Stockholm, Sweden
| | - Lars T Westlye
- K.G. Jebsen Center for Psychosis Research, Norwegian Center for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0424, Oslo, Norway.,Department of Psychology, University of Oslo, 0317, Oslo, Norway
| | - Anders Lundquist
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, 90187, Umeå, Sweden.,Department of Statistics, USBF, Umeå University, 90187, Umeå, Sweden
| | - Andrea Christoforou
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, 5021, Bergen, Norway.,K.G.Jebsen Center for Psychosis Research and the Norwegian Center for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, 5021, Bergen, Norway
| | - Sven Cichon
- Division of Medical Genetics, Department of Biomedicine, University of Basel, 4058, Basel, Switzerland.,Institute of Neuroscience and Medicine (INM-1), Research Center Juelich, 52425, Juelich, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, 53127, Bonn, Germany
| | - Rolf Adolfsson
- Department of Clinical Sciences, Psychiatry, Umeå University, 90187, Umeå, Sweden
| | - Vidar M Steen
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, 5021, Bergen, Norway.,K.G.Jebsen Center for Psychosis Research and the Norwegian Center for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, 5021, Bergen, Norway
| | - Ivar Reinvang
- Department of Psychology, University of Oslo, 0317, Oslo, Norway
| | - Lars Göran Nilsson
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, 90187, Umeå, Sweden.,ARC, Karolinska Institutet, Stockholm, Sweden
| | - Stéphanie Le Hellard
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, 5021, Bergen, Norway.,K.G.Jebsen Center for Psychosis Research and the Norwegian Center for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, 5021, Bergen, Norway
| | - Lars Nyberg
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, 90187, Umeå, Sweden. .,Department of Radiation Sciences, Umeå University, 90187, Umeå, Sweden. .,Department of Integrative Medical Biology, Umeå University, 90187, Umeå, Sweden.
| |
Collapse
|
561
|
Wang Y, Thompson WK, Schork AJ, Holland D, Chen CH, Bettella F, Desikan RS, Li W, Witoelar A, Zuber V, Devor A, Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium, Enhancing Neuro Imaging Genetics through Meta Analysis Consortium, Nöthen MM, Rietschel M, Chen Q, Werge T, Cichon S, Weinberger DR, Djurovic S, O’Donovan M, Visscher PM, Andreassen OA, Dale AM. Leveraging Genomic Annotations and Pleiotropic Enrichment for Improved Replication Rates in Schizophrenia GWAS. PLoS Genet 2016; 12:e1005803. [PMID: 26808560 PMCID: PMC4726519 DOI: 10.1371/journal.pgen.1005803] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 12/21/2015] [Indexed: 02/05/2023] Open
Abstract
Most of the genetic architecture of schizophrenia (SCZ) has not yet been identified. Here, we apply a novel statistical algorithm called Covariate-Modulated Mixture Modeling (CM3), which incorporates auxiliary information (heterozygosity, total linkage disequilibrium, genomic annotations, pleiotropy) for each single nucleotide polymorphism (SNP) to enable more accurate estimation of replication probabilities, conditional on the observed test statistic (“z-score”) of the SNP. We use a multiple logistic regression on z-scores to combine information from auxiliary information to derive a “relative enrichment score” for each SNP. For each stratum of these relative enrichment scores, we obtain nonparametric estimates of posterior expected test statistics and replication probabilities as a function of discovery z-scores, using a resampling-based approach that repeatedly and randomly partitions meta-analysis sub-studies into training and replication samples. We fit a scale mixture of two Gaussians model to each stratum, obtaining parameter estimates that minimize the sum of squared differences of the scale-mixture model with the stratified nonparametric estimates. We apply this approach to the recent genome-wide association study (GWAS) of SCZ (n = 82,315), obtaining a good fit between the model-based and observed effect sizes and replication probabilities. We observed that SNPs with low enrichment scores replicate with a lower probability than SNPs with high enrichment scores even when both they are genome-wide significant (p < 5x10-8). There were 693 and 219 independent loci with model-based replication rates ≥80% and ≥90%, respectively. Compared to analyses not incorporating relative enrichment scores, CM3 increased out-of-sample yield for SNPs that replicate at a given rate. This demonstrates that replication probabilities can be more accurately estimated using prior enrichment information with CM3. Genome-wide association studies (GWAS) have thus far identified only a small fraction of the heritability of common complex disorders, such as schizophrenia. Here, we demonstrate that by using auxiliary information we can improve estimates of replication probabilities from GWAS summary statistics. The proposed Covariate-Modulated Mixture Model (CM3) incorporates auxiliary information to construct an “enrichment score” for each single nucleotide polymorphism (SNP). We show that a scale mixture of two Gaussians provides a good fit to the observed effect size distribution stratified by the predicted enrichment score when applied the method to a recent genome-wide association study (GWAS) of SCZ (n = 82,315). Compared to estimates performed not using auxiliary information, the CM3 more accurately models the observed replication rates by stratifying on covariate-modulated enrichment scores. We observed that SNPs with low enrichment scores replicate with a lower probability compared to SNPs with high enrichment scores, even when both are genome-wide significant (p < 5x10-8). At model-based replication rates ≥80% and ≥90% there were 693 and 219 independent loci, respectively. Increased out-of-sample yield for SNPs ranked according to CM3 demonstrate the utility of incorporating auxiliary information via CM3.
Collapse
Affiliation(s)
- Yunpeng Wang
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Neurosciences, University of California, San Diego, La Jolla, California, United States of America
- Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, California, United States of America
| | - Wesley K. Thompson
- Department of Psychiatry, University of California, San Diego, La Jolla, California, United States of America
| | - Andrew J. Schork
- Department of Cognitive Sciences, University of California at San Diego, La Jolla, California, United States of America
| | - Dominic Holland
- Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, California, United States of America
| | - Chi-Hua Chen
- Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, California, United States of America
- Department of Radiology, University of California, San Diego, La Jolla, California, United States of America
| | - Francesco Bettella
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Rahul S. Desikan
- Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, California, United States of America
- Department of Radiology, University of California, San Diego, La Jolla, California, United States of America
| | - Wen Li
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- 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, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Verena Zuber
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Anna Devor
- Department of Neurosciences, University of California, San Diego, La Jolla, California, United States of America
- Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, California, United States of America
| | | | | | | | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Qiang Chen
- Lieber Institute for Brain Development, Baltimore, Maryland, United States of America
| | - Thomas Werge
- Institute of Biological Psychiatry, MHC, Sct. Hans Hospital and University of Copenhagen, Copenhagen, Denmark
| | - Sven Cichon
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Daniel R. Weinberger
- Lieber Institute for Brain Development, Baltimore, Maryland, United States of America
| | - Srdjan Djurovic
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Michael O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| | - Peter M. Visscher
- The Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute (TRI), Brisbane, Australia
| | - Ole A. Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- * E-mail: (AMD); (OAA)
| | - Anders M. Dale
- Department of Neurosciences, University of California, San Diego, La Jolla, California, United States of America
- Multimodal Imaging Laboratory, University of California at San Diego, La Jolla, California, United States of America
- Department of Psychiatry, University of California, San Diego, La Jolla, California, United States of America
- Department of Radiology, University of California, San Diego, La Jolla, California, United States of America
- * E-mail: (AMD); (OAA)
| |
Collapse
|
562
|
Jiang Q, Liu G. REST rs3796529 variant does not influence human subcortical brain structures. Ann Neurol 2016; 79:334-5. [DOI: 10.1002/ana.24590] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Qinghua Jiang
- School of Life Science and Technology, Harbin Institute of Technology; Harbin China
| | - Guiyou Liu
- School of Life Science and Technology, Harbin Institute of Technology; Harbin China
- Genome Analysis Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; Tianjin China
| |
Collapse
|
563
|
Bogdan R, Pagliaccio D, Baranger DAA, Hariri AR. Genetic Moderation of Stress Effects on Corticolimbic Circuitry. Neuropsychopharmacology 2016; 41:275-96. [PMID: 26189450 PMCID: PMC4677127 DOI: 10.1038/npp.2015.216] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/09/2015] [Accepted: 07/11/2015] [Indexed: 02/06/2023]
Abstract
Stress exposure is associated with individual differences in corticolimbic structure and function that often mirror patterns observed in psychopathology. Gene x environment interaction research suggests that genetic variation moderates the impact of stress on risk for psychopathology. On the basis of these findings, imaging genetics, which attempts to link variability in DNA sequence and structure to neural phenotypes, has begun to incorporate measures of the environment. This research paradigm, known as imaging gene x environment interaction (iGxE), is beginning to contribute to our understanding of the neural mechanisms through which genetic variation and stress increase psychopathology risk. Although awaiting replication, evidence suggests that genetic variation within the canonical neuroendocrine stress hormone system, the hypothalamic-pituitary-adrenal axis, contributes to variability in stress-related corticolimbic structure and function, which, in turn, confers risk for psychopathology. For iGxE research to reach its full potential it will have to address many challenges, of which we discuss: (i) small effects, (ii) measuring the environment and neural phenotypes, (iii) the absence of detailed mechanisms, and (iv) incorporating development. By actively addressing these challenges, iGxE research is poised to help identify the neural mechanisms underlying genetic and environmental associations with psychopathology.
Collapse
Affiliation(s)
- Ryan Bogdan
- Department of Psychology, BRAIN Lab, Washington University in St Louis, St Louis, MO, USA
- Neurosciences Program, Division of Biology and Biomedical Sciences, Washington University in St Louis, St Louis, MO, USA
| | - David Pagliaccio
- Neurosciences Program, Division of Biology and Biomedical Sciences, Washington University in St Louis, St Louis, MO, USA
| | - David AA Baranger
- Department of Psychology, BRAIN Lab, Washington University in St Louis, St Louis, MO, USA
- Neurosciences Program, Division of Biology and Biomedical Sciences, Washington University in St Louis, St Louis, MO, USA
| | - Ahmad R Hariri
- Department of Psychology and Neuroscience, Laboratory of NeuroGenetics, Duke University, Durham, NC, USA
| |
Collapse
|
564
|
Sumner JA, Powers A, Jovanovic T, Koenen KC. Genetic influences on the neural and physiological bases of acute threat: A research domain criteria (RDoC) perspective. Am J Med Genet B Neuropsychiatr Genet 2016; 171B:44-64. [PMID: 26377804 PMCID: PMC4715467 DOI: 10.1002/ajmg.b.32384] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 09/01/2015] [Indexed: 01/13/2023]
Abstract
The NIMH Research Domain Criteria (RDoC) initiative aims to describe key dimensional constructs underlying mental function across multiple units of analysis-from genes to observable behaviors-in order to better understand psychopathology. The acute threat ("fear") construct of the RDoC Negative Valence System has been studied extensively from a translational perspective, and is highly pertinent to numerous psychiatric conditions, including anxiety and trauma-related disorders. We examined genetic contributions to the construct of acute threat at two units of analysis within the RDoC framework: (1) neural circuits and (2) physiology. Specifically, we focused on genetic influences on activation patterns of frontolimbic neural circuitry and on startle, skin conductance, and heart rate responses. Research on the heritability of activation in threat-related frontolimbic neural circuitry is lacking, but physiological indicators of acute threat have been found to be moderately heritable (35-50%). Genetic studies of the neural circuitry and physiology of acute threat have almost exclusively relied on the candidate gene method and, as in the broader psychiatric genetics literature, most findings have failed to replicate. The most robust support has been demonstrated for associations between variation in the serotonin transporter (SLC6A4) and catechol-O-methyltransferase (COMT) genes with threat-related neural activation and physiological responses. However, unbiased genome-wide approaches using very large samples are needed for gene discovery, and these can be accomplished with collaborative consortium-based research efforts, such as those of the Psychiatric Genomics Consortium (PGC) and Enhancing Neuro Imaging Genetics through Meta-Analysis (ENIGMA) Consortium.
Collapse
Affiliation(s)
- Jennifer A Sumner
- Center for Behavioral Cardiovascular Health, Columbia University Medical Center, New York, New York
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Abigail Powers
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Tanja Jovanovic
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Psychiatric and Neurodevelopmental Genetics Unit and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts
- The Stanley Center for Psychiatric Research at the Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
565
|
Opel N, Zwanzger P, Redlich R, Grotegerd D, Dohm K, Arolt V, Heindel W, Kugel H, Dannlowski U. Differing brain structural correlates of familial and environmental risk for major depressive disorder revealed by a combined VBM/pattern recognition approach. Psychol Med 2016; 46:277-290. [PMID: 26355299 DOI: 10.1017/s0033291715001683] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Neuroimaging traits of either familial or environmental risk for major depressive disorder (MDD) have been interpreted as possibly useful vulnerability markers. However, the simultaneous occurrence of familial and environmental risk might prove to be a major obstacle in the attempt of recent studies to confine the precise impact of each of these conditions on brain structure. Moreover, the exclusive use of group-level analyses does not permit prediction of individual illness risk which would be the basic requirement for the clinical application of imaging vulnerability markers. Hence, we aimed to distinguish between brain structural characteristics of familial predisposition and environmental stress by using both group- and individual-level analyses. METHOD We investigated grey matter alterations between 20 healthy control subjects (HC) and 20 MDD patients; 16 healthy first-degree relatives of MDD patients (FH+) and 20 healthy subjects exposed to former childhood maltreatment (CM+) by using a combined VBM/pattern recognition approach. RESULTS We found similar grey matter reductions in the insula and the orbitofrontal cortex in patients and FH+ subjects and in the hippocampus in patients and CM+ subjects. No direct overlap in grey matter alterations was found between FH+ and CM+ subjects. Pattern classification successfully detected subjects at risk for the disease even by strictly focusing on morphological traits of MDD. CONCLUSIONS Familial and environmental risk factors for MDD are associated with differing morphometric anomalies. Pattern recognition might be a promising instrument in the search for and future application of vulnerability markers for MDD.
Collapse
Affiliation(s)
- N Opel
- Department of Psychiatry,University of Münster,Münster,Germany
| | - P Zwanzger
- Department of Psychiatry,University of Münster,Münster,Germany
| | - R Redlich
- Department of Psychiatry,University of Münster,Münster,Germany
| | - D Grotegerd
- Department of Psychiatry,University of Münster,Münster,Germany
| | - K Dohm
- Department of Psychiatry,University of Münster,Münster,Germany
| | - V Arolt
- Department of Psychiatry,University of Münster,Münster,Germany
| | - W Heindel
- Department of Clinical Radiology,University of Münster,Münster,Germany
| | - H Kugel
- Department of Clinical Radiology,University of Münster,Münster,Germany
| | - U Dannlowski
- Department of Psychiatry,University of Münster,Münster,Germany
| |
Collapse
|
566
|
Smoller JW. The Genetics of Stress-Related Disorders: PTSD, Depression, and Anxiety Disorders. Neuropsychopharmacology 2016; 41:297-319. [PMID: 26321314 PMCID: PMC4677147 DOI: 10.1038/npp.2015.266] [Citation(s) in RCA: 295] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/05/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023]
Abstract
Research into the causes of psychopathology has largely focused on two broad etiologic factors: genetic vulnerability and environmental stressors. An important role for familial/heritable factors in the etiology of a broad range of psychiatric disorders was established well before the modern era of genomic research. This review focuses on the genetic basis of three disorder categories-posttraumatic stress disorder (PTSD), major depressive disorder (MDD), and the anxiety disorders-for which environmental stressors and stress responses are understood to be central to pathogenesis. Each of these disorders aggregates in families and is moderately heritable. More recently, molecular genetic approaches, including genome-wide studies of genetic variation, have been applied to identify specific risk variants. In this review, I summarize evidence for genetic contributions to PTSD, MDD, and the anxiety disorders including genetic epidemiology, the role of common genetic variation, the role of rare and structural variation, and the role of gene-environment interaction. Available data suggest that stress-related disorders are highly complex and polygenic and, despite substantial progress in other areas of psychiatric genetics, few risk loci have been identified for these disorders. Progress in this area will likely require analysis of much larger sample sizes than have been reported to date. The phenotypic complexity and genetic overlap among these disorders present further challenges. The review concludes with a discussion of prospects for clinical translation of genetic findings and future directions for research.
Collapse
Affiliation(s)
- Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
567
|
Jean-Pierre P, McDonald B. Neuroepidemiology of cancer and treatment-related neurocognitive dysfunction in adult-onset cancer patients and survivors. Neuroepidemiology 2016; 138:297-309. [DOI: 10.1016/b978-0-12-802973-2.00017-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
568
|
Nho K, Saykin AJ. Reply. Ann Neurol 2015; 79:335. [PMID: 26710319 DOI: 10.1002/ana.24588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
569
|
Orellana C, Ferreira D, Muehlboeck JS, Mecocci P, Vellas B, Tsolaki M, Kłoszewska I, Soininen H, Lovestone S, Simmons A, Wahlund LO, Westman E. Measuring Global Brain Atrophy with the Brain Volume/Cerebrospinal Fluid Index: Normative Values, Cut-Offs and Clinical Associations. NEURODEGENER DIS 2015; 16:77-86. [PMID: 26726737 DOI: 10.1159/000442443] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 11/11/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Global brain atrophy is present in normal aging and different neurodegenerative disorders such as Alzheimer's disease (AD) and is becoming widely used to monitor disease progression. SUMMARY The brain volume/cerebrospinal fluid index (BV/CSF index) is validated in this study as a measurement of global brain atrophy. We tested the ability of the BV/CSF index to detect global brain atrophy, investigated the influence of confounders, provided normative values and cut-offs for mild, moderate and severe brain atrophy, and studied associations with different outcome variables. A total of 1,009 individuals were included [324 healthy controls, 408 patients with mild cognitive impairment (MCI) and 277 patients with AD]. Magnetic resonance images were segmented using FreeSurfer, and the BV/CSF index was calculated and studied both cross-sectionally and longitudinally (1-year follow-up). Both AD patients and MCI patients who progressed to AD showed greater global brain atrophy compared to stable MCI patients and controls. Atrophy was associated with older age, larger intracranial volume, less education and presence of the ApoE ε4 allele. Significant correlations were found with clinical variables, CSF biomarkers and several cognitive tests. KEY MESSAGES The BV/CSF index may be useful for staging individuals according to the degree of global brain atrophy, and for monitoring disease progression. It also shows potential for predicting clinical changes and for being used in the clinical routine.
Collapse
|
570
|
Wong JE, Cao J, Dorris DM, Meitzen J. Genetic sex and the volumes of the caudate-putamen, nucleus accumbens core and shell: original data and a review. Brain Struct Funct 2015; 221:4257-4267. [PMID: 26666530 DOI: 10.1007/s00429-015-1158-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 11/24/2015] [Indexed: 11/24/2022]
Abstract
Sex differences are widespread across vertebrate nervous systems. Such differences are sometimes reflected in the neural substrate via neuroanatomical differences in brain region volume. One brain region that displays sex differences in its associated functions and pathologies is the striatum, including the caudate-putamen (dorsal striatum), nucleus accumbens core and shell (ventral striatum). The extent to which these differences can be attributed to alterations in volume is unclear. We thus tested whether the volumes of the caudate-putamen, nucleus accumbens core, and nucleus accumbens shell differed by region, sex, and hemisphere in adult Sprague-Dawley rats. As a positive control for detecting sex differences in brain region volume, we measured the sexually dimorphic nucleus of the medial preoptic area (SDN-POA). As expected, SDN-POA volume was larger in males than in females. No sex differences were detected in the volumes of the caudate-putamen, nucleus accumbens core or shell. Nucleus accumbens core volume was larger in the right than left hemisphere across males and females. These findings complement previous reports of lateralized nucleus accumbens volume in humans, and suggest that this may possibly be driven via hemispheric differences in nucleus accumbens core volume. In contrast, striatal sex differences seem to be mediated by factors other than striatal region volume. This conclusion is presented within the context of a detailed review of studies addressing sex differences and similarities in striatal neuroanatomy.
Collapse
Affiliation(s)
- Jordan E Wong
- Department of Biological Sciences, North Carolina State University, Campus Box 7617, Raleigh, NC, 27695-7617, USA
| | - Jinyan Cao
- Department of Biological Sciences, North Carolina State University, Campus Box 7617, Raleigh, NC, 27695-7617, USA.,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA
| | - David M Dorris
- Department of Biological Sciences, North Carolina State University, Campus Box 7617, Raleigh, NC, 27695-7617, USA
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Campus Box 7617, Raleigh, NC, 27695-7617, USA. .,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA. .,Center for Human Health and the Environment, Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
571
|
Ikram MA, van der Lugt A, Niessen WJ, Koudstaal PJ, Krestin GP, Hofman A, Bos D, Vernooij MW. The Rotterdam Scan Study: design update 2016 and main findings. Eur J Epidemiol 2015; 30:1299-315. [PMID: 26650042 PMCID: PMC4690838 DOI: 10.1007/s10654-015-0105-7] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/25/2015] [Indexed: 12/20/2022]
Abstract
Imaging plays an essential role in research on neurological diseases in the elderly. The Rotterdam Scan Study was initiated as part of the ongoing Rotterdam Study with the aim to elucidate the causes of neurological disease by performing imaging of the brain in a prospective population-based setting. Initially, in 1995 and 1999, random subsamples of participants from the Rotterdam Study underwent neuroimaging, whereas from 2005 onwards MRI has been implemented into the core protocol of the Rotterdam Study. In this paper, we discuss the background and rationale of the Rotterdam Scan Study. Moreover, we describe the imaging protocol, image post-processing techniques, and the main findings to date. Finally, we provide recommendations for future research, which will also be topics of investigation in the Rotterdam Scan Study.
Collapse
Affiliation(s)
- M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
- Department of Radiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| | - Aad van der Lugt
- Department of Radiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Wiro J Niessen
- Biomedical Imaging Group Rotterdam, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Peter J Koudstaal
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Gabriel P Krestin
- Department of Radiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Radiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Radiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
572
|
Bootsman F, Brouwer RM, Kemner SM, Schnack HG, van der Schot AC, Vonk R, Hillegers MHJ, Boomsma DI, Hulshoff Pol HE, Nolen WA, Kahn RS, van Haren NEM. Contribution of genes and unique environment to cross-sectional and longitudinal measures of subcortical volumes in bipolar disorder. Eur Neuropsychopharmacol 2015; 25:2197-209. [PMID: 26481908 DOI: 10.1016/j.euroneuro.2015.09.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/19/2015] [Accepted: 09/28/2015] [Indexed: 02/06/2023]
Abstract
The influence of genes and environment on the association between bipolar disorder (BD) and volumes of subcortical brain regions involved in emotion processing has rarely been studied. Furthermore, as far as we know, longitudinal twin studies of subcortical brain volume change in BD have not been carried out at all. In this study, we focused on the genetic and environmental contributions to cross-sectional and longitudinal measures of subcortical brain volumes in BD. A total of 99 twins from monozygotic and dizygotic pairs concordant or discordant for BD and 129 twins from monozygotic and dizygotic healthy control pairs underwent magnetic resonance imaging at baseline. Longitudinal assessment was carried out in 48 twins from monozygotic and dizygotic patient pairs and 52 twins from monozygotic and dizygotic control pairs. Subcortical volume measures were obtained with Freesurfer software and analyzed with structural equation modeling software OpenMx. At baseline, BD was phenotypically and genetically associated with smaller volumes of the thalamus, putamen and nucleus accumbens. BD was not associated with subcortical brain volume change over time in any of the examined regions. Heritability of subcortical volumes at baseline was high, whereas subcortical volume change had low heritability. Genes contributing to BD showed overlap with those associated with smaller volumes of the thalamus, putamen and nucleus accumbens at baseline. Further evaluation of genetic contributions to abnormalities in subcortical brain regions assumed to be involved in emotion processing is recommended.
Collapse
Affiliation(s)
- Florian Bootsman
- University Medical Center Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands.
| | - Rachel M Brouwer
- University Medical Center Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Sanne M Kemner
- University Medical Center Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Hugo G Schnack
- University Medical Center Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | | | - Ronald Vonk
- Reinier van Arkel Group, ׳s-Hertogenbosch, The Netherlands
| | - Manon H J Hillegers
- University Medical Center Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Dorret I Boomsma
- Free University Amsterdam, Department of Biological Psychology, Amsterdam, The Netherlands
| | | | - Willem A Nolen
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - René S Kahn
- University Medical Center Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Neeltje E M van Haren
- University Medical Center Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| |
Collapse
|
573
|
|
574
|
Mackey S, Kan KJ, Chaarani B, Alia-Klein N, Batalla A, Brooks S, Cousijn J, Dagher A, de Ruiter M, Desrivieres S, Feldstein Ewing SW, Goldstein RZ, Goudriaan AE, Heitzeg MM, Hutchison K, Li CSR, London ED, Lorenzetti V, Luijten M, Martin-Santos R, Morales AM, Paulus MP, Paus T, Pearlson G, Schluter R, Momenan R, Schmaal L, Schumann G, Sinha R, Sjoerds Z, Stein DJ, Stein EA, Solowij N, Tapert S, Uhlmann A, Veltman D, van Holst R, Walter H, Wright MJ, Yucel M, Yurgelun-Todd D, Hibar DP, Jahanshad N, Thompson PM, Glahn DC, Garavan H, Conrod P. Genetic imaging consortium for addiction medicine: From neuroimaging to genes. PROGRESS IN BRAIN RESEARCH 2015; 224:203-23. [PMID: 26822360 PMCID: PMC4820288 DOI: 10.1016/bs.pbr.2015.07.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Since the sample size of a typical neuroimaging study lacks sufficient statistical power to explore unknown genomic associations with brain phenotypes, several international genetic imaging consortia have been organized in recent years to pool data across sites. The challenges and achievements of these consortia are considered here with the goal of leveraging these resources to study addiction. The authors of this review have joined together to form an Addiction working group within the framework of the ENIGMA project, a meta-analytic approach to multisite genetic imaging data. Collectively, the Addiction working group possesses neuroimaging and genomic data obtained from over 10,000 subjects. The deadline for contributing data to the first round of analyses occurred at the beginning of May 2015. The studies performed on this data should significantly impact our understanding of the genetic and neurobiological basis of addiction.
Collapse
Affiliation(s)
- Scott Mackey
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, USA.
| | - Kees-Jan Kan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, USA
| | - Bader Chaarani
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, USA
| | - Nelly Alia-Klein
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Albert Batalla
- Department of Psychiatry and Psychology, Hospital Clínic, IDIBAPS, CIBERSAM, University of Barcelona, Barcelona, Spain; Department of Psychiatry, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Samantha Brooks
- Department of Psychiatry and MRC Unit on Anxiety & Stress Disorders, University of Cape Town, Cape Town, South Africa
| | - Janna Cousijn
- Department of Psychiatry and MRC Unit on Anxiety & Stress Disorders, University of Cape Town, Cape Town, South Africa
| | - Alain Dagher
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Michiel de Ruiter
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Rita Z Goldstein
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna E Goudriaan
- Department of Psychiatry and MRC Unit on Anxiety & Stress Disorders, University of Cape Town, Cape Town, South Africa; Department of Psychiatry, University of Amsterdam, Amsterdam, The Netherlands
| | - Mary M Heitzeg
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Kent Hutchison
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Edythe D London
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Valentina Lorenzetti
- School of Psychological Sciences, Monash Institute of Cognitive and Clinical Neurosciences and Monash Biomedical Imaging, Monash University, Melbourne, Australia
| | - Maartje Luijten
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| | - Rocio Martin-Santos
- Department of Psychiatry and Psychology, Hospital Clínic, IDIBAPS, CIBERSAM, University of Barcelona, Barcelona, Spain
| | - Angelica M Morales
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Martin P Paulus
- VA San Diego Healthcare System and Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Laureate Institute for Brain Research, Tulsa, OK, USA
| | - Tomas Paus
- Rotman Research Institute, University of Toronto, Toronto, ON, Canada
| | - Godfrey Pearlson
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Renée Schluter
- Department of Psychiatry, University of Amsterdam, Amsterdam, The Netherlands
| | - Reza Momenan
- Section on Brain Electrophysiology and Imaging, Institute on Alcohol Abuse and Alcoholism, Bethesda, USA
| | - Lianne Schmaal
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Rajita Sinha
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Zsuzsika Sjoerds
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Dan J Stein
- Department of Psychiatry and MRC Unit on Anxiety & Stress Disorders, University of Cape Town, Cape Town, South Africa
| | - Elliot A Stein
- Intramural Research Program-Neuroimaging Research Branch, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Nadia Solowij
- School of Psychology, University of Wollongong, Wollongong, NSW, Australia
| | - Susan Tapert
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | - Anne Uhlmann
- Department of Psychiatry and MRC Unit on Anxiety & Stress Disorders, University of Cape Town, Cape Town, South Africa
| | - Dick Veltman
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Ruth van Holst
- Department of Psychiatry, University of Amsterdam, Amsterdam, The Netherlands
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Charité Universitatsmedizin, Berlin, Germany
| | | | - Murat Yucel
- School of Psychological Sciences, Monash Institute of Cognitive and Clinical Neurosciences and Monash Biomedical Imaging, Monash University, Melbourne, Australia
| | - Deborah Yurgelun-Todd
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Derrek P Hibar
- Department of Neurology, Imaging Genetics Center, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Neda Jahanshad
- Department of Neurology, Imaging Genetics Center, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Paul M Thompson
- Department of Neurology, Imaging Genetics Center, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - David C Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, USA
| | - Patricia Conrod
- Department of Psychiatry, Université de Montreal, CHU Ste Justine Hospital, Montreal, QC, Canada
| |
Collapse
|
575
|
Han S, Ma Y. A Culture–Behavior–Brain Loop Model of Human Development. Trends Cogn Sci 2015; 19:666-676. [DOI: 10.1016/j.tics.2015.08.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 08/06/2015] [Accepted: 08/14/2015] [Indexed: 01/01/2023]
|
576
|
Abstract
Among the common mental illnesses in childhood and adolescence, the unipolar depressions are the most concerning. These mental illnesses are aetiologically and clinically heterogeneous and little is known about their pathophysiology. This selected review considers the contribution of genetic and environmental factors in the emergence of these illnesses in the second decade of life.
Collapse
|
577
|
Kim J, Pan W. A cautionary note on using secondary phenotypes in neuroimaging genetic studies. Neuroimage 2015; 121:136-45. [PMID: 26220747 PMCID: PMC4604049 DOI: 10.1016/j.neuroimage.2015.07.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/12/2015] [Accepted: 07/20/2015] [Indexed: 11/18/2022] Open
Abstract
Almost all genome-wide association studies (GWASs), including Alzheimer's Disease Neuroimaging Initiative (ADNI), are based on the case-control study design, implying that the resulting case-control data are likely a biased, not random, sample of the target population. Although association analysis of the disease (e.g. Alzheimer's disease in the ADNI) can be conducted using a standard logistic regression by ignoring the biased case-control sampling, a standard linear regression analysis on a secondary phenotype (e.g. any neuroimaging phenotype in the ADNI) may in general lead to biased inference, including biased parameter estimates, inflated Type I errors and reduced power for association testing. Despite of this well known result in genetic epidemiology, to our surprise, all the published studies on secondary phenotypes with the ADNI data have ignored this potential problem. Here we aim to answer whether such a standard analysis of a secondary phenotype is valid or problematic with the ADNI data. Through both real data analyses and simulation studies, we found that, strikingly, such an analysis was generally valid (with only small biases or slightly inflated Type I errors) for the ADNI data, though cautions must be taken when analyzing other data. We also illustrate applications and possible problems of two methods specifically developed for valid analysis of secondary phenotypes.
Collapse
Affiliation(s)
- Junghi Kim
- Division of Biostatistics, University of Minnesota, USA
| | - Wei Pan
- Division of Biostatistics, University of Minnesota, USA.
| |
Collapse
|
578
|
Carey CE, Agrawal A, Zhang B, Conley ED, Degenhardt L, Heath AC, Li D, Lynskey MT, Martin NG, Montgomery GW, Wang T, Bierut LJ, Hariri AR, Nelson EC, Bogdan R. Monoacylglycerol lipase (MGLL) polymorphism rs604300 interacts with childhood adversity to predict cannabis dependence symptoms and amygdala habituation: Evidence from an endocannabinoid system-level analysis. JOURNAL OF ABNORMAL PSYCHOLOGY 2015; 124:860-77. [PMID: 26595473 PMCID: PMC4700831 DOI: 10.1037/abn0000079] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite evidence for heritable variation in cannabis involvement and the discovery of cannabinoid receptors and their endogenous ligands, no consistent patterns have emerged from candidate endocannabinoid (eCB) genetic association studies of cannabis involvement. Given interactions between eCB and stress systems and associations between childhood stress and cannabis involvement, it may be important to consider childhood adversity in the context of eCB-related genetic variation. We employed a system-level gene-based analysis of data from the Comorbidity and Trauma Study (N = 1,558) to examine whether genetic variation in six eCB genes (anabolism: DAGLA, DAGLB, NAPEPLD; catabolism: MGLL, FAAH; binding: CNR1; SNPs N = 65) and childhood sexual abuse (CSA) predict cannabis dependence symptoms. Significant interactions with CSA emerged for MGLL at the gene level (p = .009), and for rs604300 within MGLL (ΔR2 = .007, p < .001), the latter of which survived SNP-level Bonferroni correction and was significant in an additional sample with similar directional effects (N = 859; ΔR2 = .005, p = .026). Furthermore, in a third sample (N = 312), there was evidence that rs604300 genotype interacts with early life adversity to predict threat-related basolateral amygdala habituation, a neural phenotype linked to the eCB system and addiction (ΔR2 = .013, p = .047). Rs604300 may be related to epigenetic modulation of MGLL expression. These results are consistent with rodent models implicating 2-arachidonoylglycerol (2-AG), an endogenous cannabinoid metabolized by the enzyme encoded by MGLL, in the etiology of stress adaptation related to cannabis dependence, but require further replication.
Collapse
Affiliation(s)
- Caitlin E Carey
- Department of Psychology, Washington University in St. Louis
| | - Arpana Agrawal
- Department of Psychiatry, Washington University in St. Louis
| | - Bo Zhang
- Department of Genetics, Washington University in St. Louis
| | | | - Louisa Degenhardt
- National Drug and Alcohol Research Centre, University of New South Wales
| | - Andrew C Heath
- Department of Psychiatry, Washington University in St. Louis
| | - Daofeng Li
- Department of Genetics, Washington University in St. Louis
| | | | | | | | - Ting Wang
- Department of Genetics, Washington University in St. Louis
| | - Laura J Bierut
- Department of Psychiatry, Washington University in St. Louis
| | - Ahmad R Hariri
- Department of Psychology and Neuroscience, Duke University
| | - Elliot C Nelson
- Department of Psychiatry, Washington University in St. Louis
| | - Ryan Bogdan
- Department of Psychology, Washington University in St. Louis
| |
Collapse
|
579
|
Kowal P. Chronic medical disease and cognitive aging: Toward a healthy body and brain. Edited by KristineYaffe. 314 pp. New York: Oxford University Press. 2013. $79.00 (cloth), $77.99 (e-book). Am J Hum Biol 2015. [DOI: 10.1002/ajhb.22788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Paul Kowal
- Study on global AGEing and adult health (SAGE); World Health Organization; Geneva Switzerland
| |
Collapse
|
580
|
Li J, Chen C, Wu K, Zhang M, Zhu B, Chen C, Moyzis RK, Dong Q. Genetic variations in the serotonergic system contribute to amygdala volume in humans. Front Neuroanat 2015; 9:129. [PMID: 26500508 PMCID: PMC4598478 DOI: 10.3389/fnana.2015.00129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/17/2015] [Indexed: 11/13/2022] Open
Abstract
The amygdala plays a critical role in emotion processing and psychiatric disorders associated with emotion dysfunction. Accumulating evidence suggests that amygdala structure is modulated by serotonin-related genes. However, there is a gap between the small contributions of single loci (less than 1%) and the reported 63–65% heritability of amygdala structure. To understand the “missing heritability,” we systematically explored the contribution of serotonin genes on amygdala structure at the gene set level. The present study of 417 healthy Chinese volunteers examined 129 representative polymorphisms in genes from multiple biological mechanisms in the regulation of serotonin neurotransmission. A system-level approach using multiple regression analyses identified that nine SNPs collectively accounted for approximately 8% of the variance in amygdala volume. Permutation analyses showed that the probability of obtaining these findings by chance was low (p = 0.043, permuted for 1000 times). Findings showed that serotonin genes contribute moderately to individual differences in amygdala volume in a healthy Chinese sample. These results indicate that the system-level approach can help us to understand the genetic basis of a complex trait such as amygdala structure.
Collapse
Affiliation(s)
- Jin Li
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University Beijing, China ; Brainnetome Center, Institute of Automation, Chinese Academy of Sciences Beijing, China ; National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences Beijing, China
| | - Chunhui Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University Beijing, China ; Center for Collaboration and Innovation in Brain and Learning Sciences, Beijing Normal University Beijing, China
| | - Karen Wu
- Department of Psychology and Social Behavior, University of California, Irvine Irvine, CA, USA
| | - Mingxia Zhang
- Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences Beijing, China
| | - Bi Zhu
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University Beijing, China ; Center for Collaboration and Innovation in Brain and Learning Sciences, Beijing Normal University Beijing, China
| | - Chuansheng Chen
- Department of Psychology and Social Behavior, University of California, Irvine Irvine, CA, USA
| | - Robert K Moyzis
- Department of Biological Chemistry, University of California, Irvine Irvine, CA, USA ; Institute of Genomics and Bioinformatics, University of California, Irvine Irvine, CA, USA
| | - Qi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University Beijing, China ; Center for Collaboration and Innovation in Brain and Learning Sciences, Beijing Normal University Beijing, China
| |
Collapse
|
581
|
Mathias SR, Knowles EEM, Kent JW, McKay DR, Curran JE, de Almeida MAA, Dyer TD, Göring HHH, Olvera RL, Duggirala R, Fox PT, Almasy L, Blangero J, Glahn DC. Recurrent major depression and right hippocampal volume: A bivariate linkage and association study. Hum Brain Mapp 2015; 37:191-202. [PMID: 26485182 DOI: 10.1002/hbm.23025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 10/02/2015] [Indexed: 01/04/2023] Open
Abstract
Previous work has shown that the hippocampus is smaller in the brains of individuals suffering from major depressive disorder (MDD) than those of healthy controls. Moreover, right hippocampal volume specifically has been found to predict the probability of subsequent depressive episodes. This study explored the utility of right hippocampal volume as an endophenotype of recurrent MDD (rMDD). We observed a significant genetic correlation between the two traits in a large sample of Mexican American individuals from extended pedigrees (ρg = -0.34, p = 0.013). A bivariate linkage scan revealed a significant pleiotropic quantitative trait locus on chromosome 18p11.31-32 (LOD = 3.61). Bivariate association analysis conducted under the linkage peak revealed a variant (rs574972) within an intron of the gene SMCHD1 meeting the corrected significance level (χ(2) = 19.0, p = 7.4 × 10(-5)). Univariate association analyses of each phenotype separately revealed that the same variant was significant for right hippocampal volume alone, and also revealed a suggestively significant variant (rs12455524) within the gene DLGAP1 for rMDD alone. The results implicate right-hemisphere hippocampal volume as a possible endophenotype of rMDD, and in so doing highlight a potential gene of interest for rMDD risk.
Collapse
Affiliation(s)
- Samuel R Mathias
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,Olin Neuropsychiatry Research Center, Institute of Living, Hartford, Connecticut
| | - Emma E M Knowles
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,Olin Neuropsychiatry Research Center, Institute of Living, Hartford, Connecticut
| | - Jack W Kent
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas
| | - D Reese McKay
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Joanne E Curran
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, Texas.,University of Texas of the Rio Grande Valley, Brownsville, Texas
| | - Marcio A A de Almeida
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, Texas.,University of Texas of the Rio Grande Valley, Brownsville, Texas
| | - Thomas D Dyer
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, Texas.,University of Texas of the Rio Grande Valley, Brownsville, Texas
| | - Harald H H Göring
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas.,South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, Texas.,University of Texas of the Rio Grande Valley, Brownsville, Texas
| | - Rene L Olvera
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Ravi Duggirala
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, Texas.,University of Texas of the Rio Grande Valley, Brownsville, Texas
| | - Peter T Fox
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas.,South Texas Veterans Health System, San Antonio, Texas
| | - Laura Almasy
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, Texas.,University of Texas of the Rio Grande Valley, Brownsville, Texas
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio, Texas.,University of Texas of the Rio Grande Valley, Brownsville, Texas
| | - David C Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,Olin Neuropsychiatry Research Center, Institute of Living, Hartford, Connecticut
| |
Collapse
|
582
|
Wang P, Lin M, Pedrosa E, Hrabovsky A, Zhang Z, Guo W, Lachman HM, Zheng D. CRISPR/Cas9-mediated heterozygous knockout of the autism gene CHD8 and characterization of its transcriptional networks in neurodevelopment. Mol Autism 2015; 6:55. [PMID: 26491539 PMCID: PMC4612430 DOI: 10.1186/s13229-015-0048-6] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/10/2015] [Indexed: 01/24/2023] Open
Abstract
Background Disruptive mutation in the CHD8 gene is one of the top genetic risk factors in autism spectrum disorders (ASDs). Previous analyses of genome-wide CHD8 occupancy and reduced expression of CHD8 by shRNA knockdown in committed neural cells showed that CHD8 regulates multiple cell processes critical for neural functions, and its targets are enriched with ASD-associated genes. Methods To further understand the molecular links between CHD8 functions and ASD, we have applied the CRISPR/Cas9 technology to knockout one copy of CHD8 in induced pluripotent stem cells (iPSCs) to better mimic the loss-of-function status that would exist in the developing human embryo prior to neuronal differentiation. We then carried out transcriptomic and bioinformatic analyses of neural progenitors and neurons derived from the CHD8 mutant iPSCs. Results Transcriptome profiling revealed that CHD8 hemizygosity (CHD8+/−) affected the expression of several thousands of genes in neural progenitors and early differentiating neurons. The differentially expressed genes were enriched for functions of neural development, β-catenin/Wnt signaling, extracellular matrix, and skeletal system development. They also exhibited significant overlap with genes previously associated with autism and schizophrenia, as well as the downstream transcriptional targets of multiple genes implicated in autism. Providing important insight into how CHD8 mutations might give rise to macrocephaly, we found that seven of the twelve genes associated with human brain volume or head size by genome-wide association studies (e.g., HGMA2) were dysregulated in CHD8+/− neural progenitors or neurons. Conclusions We have established a renewable source of CHD8+/− iPSC lines that would be valuable for investigating the molecular and cellular functions of CHD8. Transcriptomic profiling showed that CHD8 regulates multiple genes implicated in ASD pathogenesis and genes associated with brain volume. Electronic supplementary material The online version of this article (doi:10.1186/s13229-015-0048-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ping Wang
- Department of Neurology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| | - Mingyan Lin
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| | - Anastasia Hrabovsky
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| | - Zheng Zhang
- Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA ; Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| | - Wenjun Guo
- Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA ; Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| | - Herbert M Lachman
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA ; Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA ; Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA ; Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, New York USA
| | - Deyou Zheng
- Department of Neurology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA ; Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA ; Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| |
Collapse
|
583
|
Foster PP. Mild traumatic brain injury and delayed alteration of memory processing. Front Neurosci 2015; 9:369. [PMID: 26528118 PMCID: PMC4604240 DOI: 10.3389/fnins.2015.00369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 09/23/2015] [Indexed: 11/17/2022] Open
Affiliation(s)
- Philip P Foster
- Department of Nanomedicine and Biomedical Engineering, The Brown Foundation, Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston - Medical School Houston, TX, USA ; Pulmonary, Sleep and Critical Care Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston - Medical School Houston, TX, USA
| |
Collapse
|
584
|
|
585
|
Abstract
Language is a defining characteristic of the human species, but its foundations remain mysterious. Heritable disorders offer a gateway into biological underpinnings, as illustrated by the discovery that FOXP2 disruptions cause a rare form of speech and language impairment. The genetic architecture underlying language-related disorders is complex, and although some progress has been made, it has proved challenging to pinpoint additional relevant genes with confidence. Next-generation sequencing and genome-wide association studies are revolutionizing understanding of the genetic bases of other neurodevelopmental disorders, like autism and schizophrenia, and providing fundamental insights into the molecular networks crucial for typical brain development. We discuss how a similar genomic perspective, brought to the investigation of language-related phenotypes, promises to yield equally informative discoveries. Moreover, we outline how follow-up studies of genetic findings using cellular systems and animal models can help to elucidate the biological mechanisms involved in the development of brain circuits supporting language.
Collapse
Affiliation(s)
- Sarah A Graham
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands;
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands; .,Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 EN Nijmegen, The Netherlands;
| |
Collapse
|
586
|
Zhu D, Zhan L, Faskowitz J, Daianu M, Jahanshad N, de Zubicaray GI, McMahon KL, Martin NG, Wright MJ, Thompson PM. GENETIC ANALYSIS OF STRUCTURAL BRAIN CONNECTIVITY USING DICCCOL MODELS OF DIFFUSION MRI IN 522 TWINS. PROCEEDINGS. IEEE INTERNATIONAL SYMPOSIUM ON BIOMEDICAL IMAGING 2015; 2015:1167-1171. [PMID: 26413210 DOI: 10.1109/isbi.2015.7164080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Genetic and environmental factors affect white matter connectivity in the normal brain, and they also influence diseases in which brain connectivity is altered. Little is known about genetic influences on brain connectivity, despite wide variations in the brain's neural pathways. Here we applied the "DICCCOL" framework to analyze structural connectivity, in 261 twin pairs (522 participants, mean age: 21.8 y ± 2.7SD). We encoded connectivity patterns by projecting the white matter (WM) bundles of all "DICCCOLs" as a tracemap (TM). Next we fitted an A/C/E structural equation model to estimate additive genetic (A), common environmental (C), and unique environmental/error (E) components of the observed variations in brain connectivity. We found 44 "heritable DICCCOLs" whose connectivity was genetically influenced (a2>1%); half of them showed significant heritability (a2>20%). Our analysis of genetic influences on WM structural connectivity suggests high heritability for some WM projection patterns, yielding new targets for genome-wide association studies.
Collapse
Affiliation(s)
- Dajiang Zhu
- Imaging Genetics Center, University of Southern California, CA, USA
| | - Liang Zhan
- Imaging Genetics Center, University of Southern California, CA, USA
| | - Joshua Faskowitz
- Imaging Genetics Center, University of Southern California, CA, USA
| | - Madelaine Daianu
- Imaging Genetics Center, University of Southern California, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, University of Southern California, CA, USA
| | | | - Katie L McMahon
- Center for Advanced Imaging, University of Queensland, Brisbane, Australia
| | | | | | - Paul M Thompson
- Imaging Genetics Center, University of Southern California, CA, USA
| |
Collapse
|
587
|
Boraxbekk CJ, Ames D, Kochan NA, Lee T, Thalamuthu A, Wen W, Armstrong NJ, Kwok JBJ, Schofield PR, Reppermund S, Wright MJ, Trollor JN, Brodaty H, Sachdev P, Mather KA. Investigating the influence of KIBRA and CLSTN2 genetic polymorphisms on cross-sectional and longitudinal measures of memory performance and hippocampal volume in older individuals. Neuropsychologia 2015; 78:10-7. [PMID: 26415670 DOI: 10.1016/j.neuropsychologia.2015.09.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 11/15/2022]
Abstract
The variability of episodic memory decline and hippocampal atrophy observed with increasing age may partly be explained by genetic factors. KIBRA (kidney and brain expressed protein) and CLSTN2 (calsyntenin 2) are two candidate genes previously linked to episodic memory performance and volume of the hippocampus, a key memory structure. However, whether polymorphisms in these two genes also influence age-related longitudinal memory decline and hippocampal atrophy is still unknown. Using data from two independent cohorts, the Sydney Memory and Ageing Study and the Older Australian Twins Study, we investigated whether the KIBRA and CLSTN2 genetic polymorphisms (rs17070145 and rs6439886) are associated with episodic memory performance and hippocampal volume in older adults (65-90 years at baseline). We were able to examine these polymorphisms in relation to memory and hippocampal volume using cross-sectional data and, more importantly, also using longitudinal data (2 years between testing occasions). Overall we did not find support for an association of KIBRA either alone or in combination with CLSTN2 with memory performance or hippocampal volume, nor did variation in these genes influence longitudinal memory decline or hippocampal atrophy in two cohorts of older adults.
Collapse
Affiliation(s)
- C J Boraxbekk
- CEDAR, Center for Demographic and Aging Research, Umeå University, S-901 87 Umeå, Sweden; UFBI, Umeå centre for Functional Brain Imaging, Umeå University, Sweden.
| | - David Ames
- National Ageing Research Institute, Parkville, Victoria, Australia; Department of Psychiatry, University of Melbourne, Victoria, Australia
| | - Nicole A Kochan
- Centre for Healthy Brain Ageing, UNSW Australia, Sydney, NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Teresa Lee
- Centre for Healthy Brain Ageing, UNSW Australia, Sydney, NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, NSW, Australia
| | | | - Wei Wen
- Centre for Healthy Brain Ageing, UNSW Australia, Sydney, NSW, Australia
| | - Nicola J Armstrong
- Centre for Healthy Brain Ageing, UNSW Australia, Sydney, NSW, Australia; Mathematics and Statistics, Murdoch University, WA, Australia
| | - John B J Kwok
- Neuroscience Research Australia, Sydney, NSW, Australia; School of Medical Sciences, UNSW, Sydney, NSW, Australia
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia; School of Medical Sciences, UNSW, Sydney, NSW, Australia
| | - Simone Reppermund
- Centre for Healthy Brain Ageing, UNSW Australia, Sydney, NSW, Australia; Department of Developmental Disability Neuropsychiatry, UNSW Australia, Sydney, NSW, Australia
| | | | - Julian N Trollor
- Centre for Healthy Brain Ageing, UNSW Australia, Sydney, NSW, Australia; Department of Developmental Disability Neuropsychiatry, UNSW Australia, Sydney, NSW, Australia
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, UNSW Australia, Sydney, NSW, Australia; Dementia Collaborative Research Centre, UNSW Australia, Sydney, NSW, Australia
| | - Perminder Sachdev
- Centre for Healthy Brain Ageing, UNSW Australia, Sydney, NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Karen A Mather
- National Ageing Research Institute, Parkville, Victoria, Australia
| |
Collapse
|
588
|
Abstract
Large-scale genomic investigations have just begun to illuminate the molecular genetic contributions to major psychiatric illnesses, ranging from small-effect-size common variants to larger-effect-size rare mutations. The findings provide causal anchors from which to understand their neurobiological basis. Although these studies represent enormous success, they highlight major challenges reflected in the heterogeneity and polygenicity of all of these conditions and the difficulty of connecting multiple levels of molecular, cellular, and circuit functions to complex human behavior. Nevertheless, these advances place us on the threshold of a new frontier in the pathophysiological understanding, diagnosis, and treatment of psychiatric disease.
Collapse
Affiliation(s)
- Daniel H Geschwind
- Departments of Neurology, Psychiatry, and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Jonathan Flint
- Wellcome Trust Center for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
589
|
Logue MW, Amstadter AB, Baker DG, Duncan L, Koenen KC, Liberzon I, Miller MW, Morey RA, Nievergelt CM, Ressler KJ, Smith AK, Smoller JW, Stein MB, Sumner JA, Uddin M. The Psychiatric Genomics Consortium Posttraumatic Stress Disorder Workgroup: Posttraumatic Stress Disorder Enters the Age of Large-Scale Genomic Collaboration. Neuropsychopharmacology 2015; 40:2287-97. [PMID: 25904361 PMCID: PMC4538342 DOI: 10.1038/npp.2015.118] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/10/2015] [Accepted: 03/25/2015] [Indexed: 11/09/2022]
Abstract
The development of posttraumatic stress disorder (PTSD) is influenced by genetic factors. Although there have been some replicated candidates, the identification of risk variants for PTSD has lagged behind genetic research of other psychiatric disorders such as schizophrenia, autism, and bipolar disorder. Psychiatric genetics has moved beyond examination of specific candidate genes in favor of the genome-wide association study (GWAS) strategy of very large numbers of samples, which allows for the discovery of previously unsuspected genes and molecular pathways. The successes of genetic studies of schizophrenia and bipolar disorder have been aided by the formation of a large-scale GWAS consortium: the Psychiatric Genomics Consortium (PGC). In contrast, only a handful of GWAS of PTSD have appeared in the literature to date. Here we describe the formation of a group dedicated to large-scale study of PTSD genetics: the PGC-PTSD. The PGC-PTSD faces challenges related to the contingency on trauma exposure and the large degree of ancestral genetic diversity within and across participating studies. Using the PGC analysis pipeline supplemented by analyses tailored to address these challenges, we anticipate that our first large-scale GWAS of PTSD will comprise over 10 000 cases and 30 000 trauma-exposed controls. Following in the footsteps of our PGC forerunners, this collaboration-of a scope that is unprecedented in the field of traumatic stress-will lead the search for replicable genetic associations and new insights into the biological underpinnings of PTSD.
Collapse
Affiliation(s)
- Mark W Logue
- Research, VA Boston Healthcare System, Boston, MA, USA
- Biomedical Genetics, Boston University School of Medicine, Boston, MA, USA
- Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Ananda B Amstadter
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Dewleen G Baker
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, VA Center of Excellence for Stress and Mental Health (CESAMH), La Jolla, CA, USA
| | - Laramie Duncan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Israel Liberzon
- Department of Psychiatry, University of Michigan, Ann Arbor, MI
- Veterans Affairs Ann Arbor Health System, Ann Arbor, MI, USA
| | - Mark W Miller
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Rajendra A Morey
- Duke-UNC Brain Imaging and Analysis Center, Duke University Medical Center, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
- Mental Illness Research Education and Clinical Center for Post Deployment Mental Health, Durham VA Medical Center, Durham, NC, USA
| | - Caroline M Nievergelt
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, VA Center of Excellence for Stress and Mental Health (CESAMH), La Jolla, CA, USA
| | - Kerry J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Center for Behavioral Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA
- Howard Hughes Medical Institute, Bethesda, MD, USA
| | - Alicia K Smith
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Jordan W Smoller
- Center of Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Murray B Stein
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Jennifer A Sumner
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Monica Uddin
- Carl R Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Psychology, University of Illinois Urbana-Champaign, Champaign, IL, USA
| |
Collapse
|
590
|
Ota K, Oishi N, Ito K, Fukuyama H. Effects of imaging modalities, brain atlases and feature selection on prediction of Alzheimer's disease. J Neurosci Methods 2015; 256:168-83. [PMID: 26318777 DOI: 10.1016/j.jneumeth.2015.08.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 07/27/2015] [Accepted: 08/18/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND The choice of biomarkers for early detection of Alzheimer's disease (AD) is important for improving the accuracy of imaging-based prediction of conversion from mild cognitive impairment (MCI) to AD. The primary goal of this study was to assess the effects of imaging modalities and brain atlases on prediction. We also investigated the influence of support vector machine recursive feature elimination (SVM-RFE) on predictive performance. METHODS Eighty individuals with amnestic MCI [40 developed AD within 3 years] underwent structural magnetic resonance imaging (MRI) and (18)F-fluorodeoxyglucose positron emission tomography (FDG-PET) scans at baseline. Using Automated Anatomical Labeling (AAL) and LONI Probabilistic Brain Atlas (LPBA40), we extracted features representing gray matter density and relative cerebral metabolic rate for glucose in each region of interest from the baseline MRI and FDG-PET data, respectively. We used linear SVM ensemble with bagging and computed the area under the receiver operating characteristic curve (AUC) as a measure of classification performance. We performed multiple SVM-RFE to compute feature ranking. We performed analysis of variance on the mean AUCs for eight feature sets. RESULTS The interactions between atlas and modality choices were significant. The main effect of SVM-RFE was significant, but the interactions with the other factors were not significant. COMPARISON WITH EXISTING METHOD Multimodal features were found to be better than unimodal features to predict AD. FDG-PET was found to be better than MRI. CONCLUSIONS Imaging modalities and brain atlases interact with each other and affect prediction. SVM-RFE can improve the predictive accuracy when using atlas-based features.
Collapse
Affiliation(s)
- Kenichi Ota
- Human Brain Research Center, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Naoya Oishi
- Human Brain Research Center, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Kengo Ito
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu-shi, Aichi 474-8511, Japan
| | - Hidenao Fukuyama
- Human Brain Research Center, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | |
Collapse
|
591
|
Wolthusen RPF, Hass J, Walton E, Turner JA, Rössner V, Sponheim SR, Ho BC, Holt DJ, Gollub RL, Calhoun V, Ehrlich S. Genetic underpinnings of left superior temporal gyrus thickness in patients with schizophrenia. World J Biol Psychiatry 2015:1-11. [PMID: 26249676 PMCID: PMC4795983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
OBJECTIVES Schizophrenia is a highly disabling psychiatric disorder with a heterogeneous phenotypic appearance. We aimed to further the understanding of some of the underlying genetics of schizophrenia, using left superior temporal gyrus (STG) grey matter thickness reduction as an endophenoptype in a genome-wide association (GWA) study. METHODS Structural magnetic resonance imaging (MRI) and genetic data of the Mind Clinical Imaging Consortium (MCIC) study of schizophrenia were used to analyse the interaction effects between 1,067,955 single nucleotide polymorphisms (SNPs) and disease status on left STG thickness in 126 healthy controls and 113 patients with schizophrenia. We next used a pathway approach to detect underlying pathophysiological pathways that may be related to schizophrenia. RESULTS No SNP by diagnosis interaction effect reached genome-wide significance (5 × 10-8) in our GWA study, but 10 SNPs reached P-values less than 10-6. The most prominent pathways included those involved in insulin, calcium, PI3K-Akt and MAPK signalling. CONCLUSIONS Our strongest findings in the GWA study and pathway analysis point towards an involvement of glucose metabolism in left STG thickness reduction in patients with schizophrenia only. These results are in line with recently published studies, which showed an increased prevalence of psychosis among patients with metabolic syndrome-related illnesses including diabetes.
Collapse
Affiliation(s)
- Rick P F Wolthusen
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine Carl Gustav Carus of the Technische Universität Dresden , Dresden , Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
592
|
Wolfers T, Buitelaar JK, Beckmann CF, Franke B, Marquand AF. From estimating activation locality to predicting disorder: A review of pattern recognition for neuroimaging-based psychiatric diagnostics. Neurosci Biobehav Rev 2015; 57:328-49. [PMID: 26254595 DOI: 10.1016/j.neubiorev.2015.08.001] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/29/2015] [Accepted: 08/02/2015] [Indexed: 12/19/2022]
Abstract
Psychiatric disorders are increasingly being recognised as having a biological basis, but their diagnosis is made exclusively behaviourally. A promising approach for 'biomarker' discovery has been based on pattern recognition methods applied to neuroimaging data, which could yield clinical utility in future. In this review we survey the literature on pattern recognition for making diagnostic predictions in psychiatric disorders, and evaluate progress made in translating such findings towards clinical application. We evaluate studies on many criteria, including data modalities used, the types of features extracted and algorithm applied. We identify problems common to many studies, such as a relatively small sample size and a primary focus on estimating generalisability within a single study. Furthermore, we highlight challenges that are not widely acknowledged in the field including the importance of accommodating disease prevalence, the necessity of more extensive validation using large carefully acquired samples, the need for methodological innovations to improve accuracy and to discriminate between multiple disorders simultaneously. Finally, we identify specific clinical contexts in which pattern recognition can add value in the short to medium term.
Collapse
Affiliation(s)
- Thomas Wolfers
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands; Donders Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - 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, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Christian F Beckmann
- Donders Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University, PO Box 9101, 6500 HB 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 (FMRIB), University of Oxford, Oxford, United Kingdom
| | - Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands; Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Andre F Marquand
- Donders Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University, PO Box 9101, 6500 HB Nijmegen, The Netherlands; Department of Neuroimaging, Institute of Psychiatry, King's College London, LondonUnited Kingdom
| |
Collapse
|
593
|
Stewart AM, Nguyen M, Song C, Kalueff AV. Understanding the genetic architectonics of complex CNS traits: Lost by the association, but found in the interaction? J Psychopharmacol 2015; 29:872-7. [PMID: 26156859 DOI: 10.1177/0269881115593904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recent evidence supports the value of endophenotypes and genome-wide association studies in psychiatric genetics, and their importance for dissecting the neural pathways and molecular mechanisms of complex neuropsychiatric disorders. Continuing this important discussion, here we outline three new mechanisms by which novel classes of genes may facilitate CNS pathogenesis without directly worsening its individual 'established' endophenotypes. These putative genetic mechanisms can apply to other human disorders in general, and may also be used for designing novel effective CNS drug treatments.
Collapse
Affiliation(s)
| | - Michael Nguyen
- ZENEREI Institute, Slidell, LA, USA Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Cai Song
- Research Institute for Marine Drugs and Nutrition, College for Food Science and Technology, Guangdong Ocean University, Zhanjiang, China Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Allan V Kalueff
- ZENEREI Institute, Slidell, LA, USA Research Institute for Marine Drugs and Nutrition, College for Food Science and Technology, Guangdong Ocean University, Zhanjiang, China Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
594
|
Saykin AJ, Shen L, Yao X, Kim S, Nho K, Risacher SL, Ramanan VK, Foroud TM, Faber KM, Sarwar N, Munsie LM, Hu X, Soares HD, Potkin SG, Thompson PM, Kauwe JSK, Kaddurah-Daouk R, Green RC, Toga AW, Weiner MW. Genetic studies of quantitative MCI and AD phenotypes in ADNI: Progress, opportunities, and plans. Alzheimers Dement 2015; 11:792-814. [PMID: 26194313 PMCID: PMC4510473 DOI: 10.1016/j.jalz.2015.05.009] [Citation(s) in RCA: 232] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 05/08/2015] [Accepted: 05/08/2015] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Genetic data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) have been crucial in advancing the understanding of Alzheimer's disease (AD) pathophysiology. Here, we provide an update on sample collection, scientific progress and opportunities, conceptual issues, and future plans. METHODS Lymphoblastoid cell lines and DNA and RNA samples from blood have been collected and banked, and data and biosamples have been widely disseminated. To date, APOE genotyping, genome-wide association study (GWAS), and whole exome and whole genome sequencing data have been obtained and disseminated. RESULTS ADNI genetic data have been downloaded thousands of times, and >300 publications have resulted, including reports of large-scale GWAS by consortia to which ADNI contributed. Many of the first applications of quantitative endophenotype association studies used ADNI data, including some of the earliest GWAS and pathway-based studies of biospecimen and imaging biomarkers, as well as memory and other clinical/cognitive variables. Other contributions include some of the first whole exome and whole genome sequencing data sets and reports in healthy controls, mild cognitive impairment, and AD. DISCUSSION Numerous genetic susceptibility and protective markers for AD and disease biomarkers have been identified and replicated using ADNI data and have heavily implicated immune, mitochondrial, cell cycle/fate, and other biological processes. Early sequencing studies suggest that rare and structural variants are likely to account for significant additional phenotypic variation. Longitudinal analyses of transcriptomic, proteomic, metabolomic, and epigenomic changes will also further elucidate dynamic processes underlying preclinical and prodromal stages of disease. Integration of this unique collection of multiomics data within a systems biology framework will help to separate truly informative markers of early disease mechanisms and potential novel therapeutic targets from the vast background of less relevant biological processes. Fortunately, a broad swath of the scientific community has accepted this grand challenge.
Collapse
Affiliation(s)
- Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Li Shen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiaohui Yao
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; School of Informatics and Computing, Indiana University, Purdue University - Indianapolis, Indianapolis, IN, USA
| | - Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Vijay K Ramanan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tatiana M Foroud
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kelley M Faber
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Xiaolan Hu
- Bristol-Myers Squibb, Wallingford, CT, USA
| | | | - Steven G Potkin
- Department of Psychiatry and Human Behavior, University of California - Irvine, Irvine, CA, USA
| | - Paul M Thompson
- Department of Neurology, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA; Imaging Genetics Center, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT, USA; Department of Neuroscience, Brigham Young University, Provo, UT, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA; Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Robert C Green
- Partners Center for Personalized Genetic Medicine, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute for Neuroimaging and Neuroinformatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Michael W Weiner
- Department of Radiology, University of California-San Francisco, San Francisco, CA, USA; Department of Medicine, University of California-San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California-San Francisco, San Francisco, CA, USA; Center for Imaging of Neurodegenerative Diseases, San Francisco VA Medical Center, San Francisco, CA, USA
| |
Collapse
|
595
|
Yang C, Li C, Wang Q, Chung D, Zhao H. Implications of pleiotropy: challenges and opportunities for mining Big Data in biomedicine. Front Genet 2015; 6:229. [PMID: 26175753 PMCID: PMC4485215 DOI: 10.3389/fgene.2015.00229] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/15/2015] [Indexed: 01/23/2023] Open
Abstract
Pleiotropy arises when a locus influences multiple traits. Rich GWAS findings of various traits in the past decade reveal many examples of this phenomenon, suggesting the wide existence of pleiotropic effects. What underlies this phenomenon is the biological connection among seemingly unrelated traits/diseases. Characterizing the molecular mechanisms of pleiotropy not only helps to explain the relationship between diseases, but may also contribute to novel insights concerning the pathological mechanism of each specific disease, leading to better disease prevention, diagnosis and treatment. However, most pleiotropic effects remain elusive because their functional roles have not been systematically examined. A systematic investigation requires availability of qualified measurements at multilayered biological processes (e.g., transcription and translation). The rise of Big Data in biomedicine, such as high-quality multi-omics data, biomedical imaging data and electronic medical records of patients, offers us an unprecedented opportunity to investigate pleiotropy. There will be a great need of computationally efficient and statistically rigorous methods for integrative analysis of these Big Data in biomedicine. In this review, we outline many opportunities and challenges in methodology developments for systematic analysis of pleiotropy, and highlight its implications on disease prevention, diagnosis and treatment.
Collapse
Affiliation(s)
- Can Yang
- Department of Mathematics, Hong Kong Baptist UniversityHong Kong, Hong Kong
- Hong Kong Baptist University Institute of Research and Continuing EducationShenzhen, China
| | - Cong Li
- Program in Computational Biology and Bioinformatics, Yale UniversityNew Haven, CT, USA
| | - Qian Wang
- Program in Computational Biology and Bioinformatics, Yale UniversityNew Haven, CT, USA
| | - Dongjun Chung
- Department of Public Health Sciences, Medical University of South CarolinaCharleston, SC, USA
| | - Hongyu Zhao
- Program in Computational Biology and Bioinformatics, Yale UniversityNew Haven, CT, USA
- Department of Biostatistics, Yale School of Public HealthNew Haven, CT, USA
- Department of Genetics, Yale School of MedicineNew Haven, CT, USA
- VA Cooperative Studies Program Coordinating CenterWest Haven, CT, USA
| |
Collapse
|
596
|
Foster PP. Role of physical and mental training in brain network configuration. Front Aging Neurosci 2015; 7:117. [PMID: 26157387 PMCID: PMC4477154 DOI: 10.3389/fnagi.2015.00117] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/01/2015] [Indexed: 01/21/2023] Open
Abstract
It is hypothesized that the topology of brain networks is constructed by connecting nodes which may be continuously remodeled by appropriate training. Efficiency of physical and/or mental training on the brain relies on the flexibility of networks' architecture molded by local remodeling of proteins and synapses of excitatory neurons producing transformations in network topology. Continuous remodeling of proteins of excitatory neurons is fine-tuning the scaling and strength of excitatory synapses up or down via regulation of intra-cellular metabolic and regulatory networks of the genome-transcriptome-proteome interface. Alzheimer's disease is a model of “energy cost-driven small-world network disorder” with dysfunction of high-energy cost wiring as the network global efficiency is impaired by the deposition of an informed agent, the amyloid-β, selectively targeting high-degree nodes. In schizophrenia, the interconnectivity and density of rich-club networks are significantly reduced. Training-induced homeostatic synaptogenesis-enhancement, presumably via reconfiguration of brain networks into greater small-worldness, appears essential in learning, memory, and executive functions. A macroscopic cartography of creation-removal of synaptic connections in a macro-network, and at the intra-cellular scale, micro-networks regulate the physiological mechanisms for the preferential attachment of synapses. The strongest molecular relationship of exercise and functional connectivity was identified for brain-derived neurotrophic factor (BDNF). The allele variant, rs7294919, also shows a powerful relationship with the hippocampal volume. How the brain achieves this unique quest of reconfiguration remains a puzzle. What are the underlying mechanisms of synaptogenesis promoting communications brain ↔ muscle and brain ↔ brain in such trainings? What is the respective role of independent mental, physical, or combined-mental-physical trainings? Physical practice seems to be playing an instrumental role in the cognitive enhancement (brain ↔ muscle com.). However, mental training, meditation or virtual reality (films, games) require only minimal motor activity and cardio-respiratory stimulation. Therefore, other potential paths (brain ↔ brain com.) molding brain networks are nonetheless essential. Patients with motor neuron disease/injury (e.g., amyotrophic lateral sclerosis, traumatism) also achieve successful cognitive enhancement albeit they may only elicit mental practice.
Collapse
Affiliation(s)
- Philip P Foster
- Department of Nano Medicine and Biomedical Engineering, The Brown Foundation, Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston - Medical School Houston, TX, USA ; Pulmonary, Sleep and Critical Care Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston - Medical School Houston, TX, USA
| |
Collapse
|
597
|
Lateralization of gene expression in human language cortex. Cortex 2015; 67:30-6. [DOI: 10.1016/j.cortex.2015.03.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/09/2015] [Accepted: 03/04/2015] [Indexed: 01/03/2023]
|
598
|
Abstract
In much of the developed world, the HIV epidemic has largely been controlled by antiretroviral treatment. Even so, there is growing concern that HIV-infected individuals may be at risk for accelerated brain aging and a range of cognitive impairments. What promotes or resists these changes is largely unknown. There is also interest in discovering factors that promote resilience to HIV and combat its adverse effects in children. Here, we review recent developments in brain imaging that reveal how the virus affects the brain. We relate these brain changes to changes in blood markers, cognitive function, and other patient outcomes or symptoms, such as apathy or neuropathic pain. We focus on new and emerging techniques, including new variants of brain MRI. Diffusion tensor imaging, for example, can map the brain's structural connections, while fMRI can uncover functional connections. Finally, we suggest how large-scale global research alliances, such as ENIGMA, may resolve controversies over effects where evidence is now lacking. These efforts pool scans from tens of thousands of individuals and offer a source of power not previously imaginable for brain imaging studies.
Collapse
Affiliation(s)
- Paul Thompson
- Dept. of Neurology, Keck USC School of Medicine, Imaging Genetics Center, University of Southern California, 4676 Admiralty Way, Marina del Rey, CA 90292, Phone: (323) 44-BRAIN Fax: (323) 442-0137
| | - Neda Jahanshad
- Dept. of Neurology, Keck USC School of Medicine, Imaging Genetics Center, University of Southern California, 4676 Admiralty Way, Marina del Rey, CA 90292, Phone: (323) 44-BRAIN Fax: (323) 442-0137
| |
Collapse
|
599
|
Neuroimaging data sharing on the neuroinformatics database platform. Neuroimage 2015; 124:1089-1092. [PMID: 25888923 DOI: 10.1016/j.neuroimage.2015.04.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/06/2015] [Accepted: 04/07/2015] [Indexed: 01/20/2023] Open
Abstract
We describe the Neuroinformatics Database (NiDB), an open-source database platform for archiving, analysis, and sharing of neuroimaging data. Data from the multi-site projects Autism Brain Imaging Data Exchange (ABIDE), Bipolar-Schizophrenia Network on Intermediate Phenotypes parts one and two (B-SNIP1, B-SNIP2), and Monetary Incentive Delay task (MID) are available for download from the public instance of NiDB, with more projects sharing data as it becomes available. As demonstrated by making several large datasets available, NiDB is an extensible platform appropriately suited to archive and distribute shared neuroimaging data.
Collapse
|
600
|
Yang T, Wang J, Sun Q, Hibar DP, Jahanshad N, Liu L, Wang Y, Zhan L, Thompson PM, Ye J. Detecting Genetic Risk Factors for Alzheimer's Disease in Whole Genome Sequence Data via Lasso Screening. PROCEEDINGS. IEEE INTERNATIONAL SYMPOSIUM ON BIOMEDICAL IMAGING 2015; 2015:985-989. [PMID: 26413209 DOI: 10.1109/isbi.2015.7164036] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Genetic factors play a key role in Alzheimer's disease (AD). The Alzheimer's Disease Neuroimaging Initiative (ADNI) whole genome sequence (WGS) data offers new power to investigate mechanisms of AD by combining entire genome sequences with neuroimaging and clinical data. Here we explore the ADNI WGS SNP (single nucleotide polymorphism) data in depth and extract approximately six million valid SNP features. We investigate imaging genetics associations using Lasso regression-a widely used sparse learning technique. To solve the large-scale Lasso problem more efficiently, we employ a highly efficient screening rule for Lasso-called dual polytope projections (DPP)-to remove irrelevant features from the optimization problem. Experiments demonstrate that the DPP can effectively identify irrelevant features and leads to a 400× speedup. This allows us for the first time to run the compute-intensive model selection procedure called stability selection to rank SNPs that may affect the brain and AD risk.
Collapse
Affiliation(s)
- Tao Yang
- Dept. of Computer Science and Engineering, Arizona State Univ., Tempe, AZ, USA ; Center for Evolutionary Medicine and Informatics, The Biodesign Institute, Arizona State Univ., Tempe, AZ, USA
| | - Jie Wang
- Dept. of Computer Science and Engineering, Arizona State Univ., Tempe, AZ, USA ; Center for Evolutionary Medicine and Informatics, The Biodesign Institute, Arizona State Univ., Tempe, AZ, USA
| | - Qian Sun
- Dept. of Computer Science and Engineering, Arizona State Univ., Tempe, AZ, USA ; Center for Evolutionary Medicine and Informatics, The Biodesign Institute, Arizona State Univ., Tempe, AZ, USA
| | - Derrek P Hibar
- Imaging Genetics Center, Keck School of Medicine, Univ. of Southern California, Los Angeles, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Keck School of Medicine, Univ. of Southern California, Los Angeles, CA, USA
| | - Li Liu
- Center for Evolutionary Medicine and Informatics, The Biodesign Institute, Arizona State Univ., Tempe, AZ, USA
| | - Yalin Wang
- Dept. of Computer Science and Engineering, Arizona State Univ., Tempe, AZ, USA
| | - Liang Zhan
- Imaging Genetics Center, Keck School of Medicine, Univ. of Southern California, Los Angeles, CA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Keck School of Medicine, Univ. of Southern California, Los Angeles, CA, USA
| | - Jieping Ye
- Dept. of Computational Medicine and Bioinformatics, Univ. of Michigan, Ann Arbor, MI, USA ; Dept. of Electrical Engineering and Computer Science, Univ. of Michigan, Ann Arbor, MI, USA
| |
Collapse
|