51
|
Liu S, Abdellaoui A, Verweij KJH, van Wingen GA. Replicable brain-phenotype associations require large-scale neuroimaging data. Nat Hum Behav 2023; 7:1344-1356. [PMID: 37365408 DOI: 10.1038/s41562-023-01642-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 05/25/2023] [Indexed: 06/28/2023]
Abstract
Numerous neuroimaging studies have investigated the neural basis of interindividual differences but the replicability of brain-phenotype associations remains largely unknown. We used the UK Biobank neuroimaging dataset (N = 37,447) to examine associations with six variables related to physical and mental health: age, body mass index, intelligence, memory, neuroticism and alcohol consumption, and assessed the improvement of replicability for brain-phenotype associations with increasing sampling sizes. Age may require only 300 individuals to provide highly replicable associations but other phenotypes required 1,500 to 3,900 individuals. The required sample size showed a negative power law relation with the estimated effect size. When only comparing the upper and lower quarters, the minimally required sample sizes for imaging decreased by 15-75%. Our findings demonstrate that large-scale neuroimaging data are required for replicable brain-phenotype associations, that this can be mitigated by preselection of individuals and that small-scale studies may have reported false positive findings.
Collapse
Affiliation(s)
- Shu Liu
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
- Amsterdam Neuroscience, Amsterdam, the Netherlands.
| | - Abdel Abdellaoui
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Karin J H Verweij
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Guido A van Wingen
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
- Amsterdam Neuroscience, Amsterdam, the Netherlands.
| |
Collapse
|
52
|
Ding P, Xu R. Causal association of COVID-19 with brain structure changes: Findings from a non-overlapping 2-sample Mendelian randomization study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.16.23292735. [PMID: 37502838 PMCID: PMC10371182 DOI: 10.1101/2023.07.16.23292735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Recent cohort studies suggested that SARS-CoV-2 infection is associated with changes in brain structure. However, the potential causal relationship remains unclear. We performed a two-sample Mendelian randomization analysis to determine whether genetic susceptibility of COVID-19 is causally associated with changes in cortical and subcortical areas of the brain. This 2-sample MR (Mendelian Randomization) study is an instrumental variable analysis of data from the COVID-19 Host Genetics Initiative (HGI) meta-analyses round 5 excluding UK Biobank participants (COVID-19 infection, N=1,348,701; COVID-19 severity, N=1,557,411), the Enhancing NeuroImaging Genetics through Meta Analysis (ENIGMA) Global and regional cortical measures, N=33,709; combined hemispheric subcortical volumes, N=38,851), and UK Biobank (left/right subcortical volumes, N=19,629). A replication analysis was performed on summary statistics from different COVID-19 GWAS study (COVID-19 infection, N=80,932; COVID-19 severity, N=72,733). We found that the genetic susceptibility of COVID-19 was not significantly associated with changes in brain structures, including cortical and subcortical brain structure. Similar results were observed for different (1) MR estimates, (2) COVID-19 GWAS summary statistics, and (3) definitions of COVID-19 infection and severity. This study suggests that the genetic susceptibility of COVID-19 is not causally associated with changes in cortical and subcortical brain structure.
Collapse
|
53
|
Miller AP, Gizer IR. Neurogenetic and multi-omic sources of overlap among sensation seeking, alcohol consumption, and alcohol use disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.30.23290733. [PMID: 37333128 PMCID: PMC10274973 DOI: 10.1101/2023.05.30.23290733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Sensation seeking is bidirectionally associated with levels of alcohol consumption in both adult and adolescent samples and shared neurobiological and genetic influences may in part explain this association. Links between sensation seeking and alcohol use disorder (AUD) may primarily manifest via increased alcohol consumption rather than through direct effects on increasing problems and consequences. Here the overlap between sensation seeking, alcohol consumption, and AUD was examined using multivariate modeling approaches for genome-wide association study (GWAS) summary statistics in conjunction with neurobiologically-informed analyses at multiple levels of investigation. Meta-analytic and genomic structural equation modeling (GenomicSEM) approaches were used to conduct GWAS of sensation seeking, alcohol consumption, and AUD. Resulting summary statistics were used in downstream analyses to examine shared brain tissue enrichment of heritability and genome-wide evidence of overlap (e.g., stratified GenomicSEM, RRHO, genetic correlations with neuroimaging phenotypes) and to identify genomic regions likely contributing to observed genetic overlap across traits (e.g., HMAGMA, LAVA). Across approaches, results supported shared neurogenetic architecture between sensation seeking and alcohol consumption characterized by overlapping enrichment of genes expressed in midbrain and striatal tissues and variants associated with increased cortical surface area. Alcohol consumption and AUD evidenced overlap in relation to variants associated with decreased frontocortical thickness. Finally, genetic mediation models provided evidence of alcohol consumption mediating associations between sensation seeking and AUD. This study extends previous research by examining critical sources of neurogenetic and multi-omic overlap among sensation seeking, alcohol consumption, and AUD which may underlie observed phenotypic associations.
Collapse
Affiliation(s)
- Alex P. Miller
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Ian R. Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
54
|
Sacher J, Witte AV. Genetic heart-brain connections. Science 2023; 380:897-898. [PMID: 37262164 DOI: 10.1126/science.adi2392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Multiorgan imaging unveils the intertwined nature of the human heart and brain.
Collapse
Affiliation(s)
- Julia Sacher
- Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - A Veronica Witte
- Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
55
|
Zhao B, Li T, Fan Z, Yang Y, Shu J, Yang X, Wang X, Luo T, Tang J, Xiong D, Wu Z, Li B, Chen J, Shan Y, Tomlinson C, Zhu Z, Li Y, Stein JL, Zhu H. Heart-brain connections: Phenotypic and genetic insights from magnetic resonance images. Science 2023; 380:abn6598. [PMID: 37262162 DOI: 10.1126/science.abn6598] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/11/2023] [Indexed: 06/03/2023]
Abstract
Cardiovascular health interacts with cognitive and mental health in complex ways, yet little is known about the phenotypic and genetic links of heart-brain systems. We quantified heart-brain connections using multiorgan magnetic resonance imaging (MRI) data from more than 40,000 subjects. Heart MRI traits displayed numerous association patterns with brain gray matter morphometry, white matter microstructure, and functional networks. We identified 80 associated genomic loci (P < 6.09 × 10-10) for heart MRI traits, which shared genetic influences with cardiovascular and brain diseases. Genetic correlations were observed between heart MRI traits and brain-related traits and disorders. Mendelian randomization suggests that heart conditions may causally contribute to brain disorders. Our results advance a multiorgan perspective on human health by revealing heart-brain connections and shared genetic influences.
Collapse
Affiliation(s)
- Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yue Yang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Shu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tianyou Luo
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiarui Tang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Di Xiong
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhenyi Wu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Bingxuan Li
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA
| | - Jie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yue Shan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chalmer Tomlinson
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ziliang Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
56
|
García-Marín LM, Reyes-Pérez P, Diaz-Torres S, Medina-Rivera A, Martin NG, Mitchell BL, Rentería ME. Shared molecular genetic factors influence subcortical brain morphometry and Parkinson's disease risk. NPJ Parkinsons Dis 2023; 9:73. [PMID: 37164954 PMCID: PMC10172359 DOI: 10.1038/s41531-023-00515-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/28/2023] [Indexed: 05/12/2023] Open
Abstract
Parkinson's disease (PD) is a late-onset and genetically complex neurodegenerative disorder. Here we sought to identify genes and molecular pathways underlying the associations between PD and the volume of ten brain structures measured through magnetic resonance imaging (MRI) scans. We leveraged genome-wide genetic data from several cohorts, including the International Parkinson's Disease Genomics Consortium (IPDG), the UK Biobank, the Adolescent Brain Cognitive Development (ABCD) study, the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE), the Enhancing Neuroimaging Genetics through Meta-Analyses (ENIGMA), and 23andMe. We observed significant positive genetic correlations between PD and intracranial and subcortical brain volumes. Genome-wide association studies (GWAS) - pairwise analyses identified 210 genomic segments with shared aetiology between PD and at least one of these brain structures. Pathway enrichment results highlight potential links with chronic inflammation, the hypothalamic-pituitary-adrenal pathway, mitophagy, disrupted vesicle-trafficking, calcium-dependent, and autophagic pathways. Investigations for putative causal genetic effects suggest that a larger putamen volume could influence PD risk, independently of the potential causal genetic effects of intracranial volume (ICV) on PD. Our findings suggest that genetic variants influencing larger intracranial and subcortical brain volumes, possibly during earlier stages of life, influence the risk of developing PD later in life.
Collapse
Affiliation(s)
- Luis M García-Marín
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
- Laboratorio Internacional de Investigación del Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México.
| | - Paula Reyes-Pérez
- Laboratorio Internacional de Investigación del Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Santiago Diaz-Torres
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Alejandra Medina-Rivera
- Laboratorio Internacional de Investigación del Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Nicholas G Martin
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Brittany L Mitchell
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Miguel E Rentería
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
57
|
Lee Y, Park JY, Lee JJ, Gim J, Do AR, Jo J, Park J, Kim K, Park K, Jin H, Choi KY, Kang S, Kim H, Kim S, Moon SH, Farrer LA, Lee KH, Won S. Heritability of cognitive abilities and regional brain structures in middle-aged to elderly East Asians. Cereb Cortex 2023; 33:6051-6062. [PMID: 36642501 PMCID: PMC10183741 DOI: 10.1093/cercor/bhac483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 01/17/2023] Open
Abstract
This study examined the single-nucleotide polymorphism heritability and genetic correlations of cognitive abilities and brain structural measures (regional subcortical volume and cortical thickness) in middle-aged and elderly East Asians (Korean) from the Gwangju Alzheimer's and Related Dementias cohort study. Significant heritability was found in memory function, caudate volume, thickness of the entorhinal cortices, pars opercularis, superior frontal gyri, and transverse temporal gyri. There were 3 significant genetic correlations between (i) the caudate volume and the thickness of the entorhinal cortices, (ii) the thickness of the superior frontal gyri and pars opercularis, and (iii) the thickness of the superior frontal and transverse temporal gyri. This is the first study to describe the heritability and genetic correlations of cognitive and neuroanatomical traits in middle-aged to elderly East Asians. Our results support the previous findings showing that genetic factors play a substantial role in the cognitive and neuroanatomical traits in middle to advanced age. Moreover, by demonstrating shared genetic effects on different brain regions, it gives us a genetic insight into understanding cognitive and brain changes with age, such as aging-related cognitive decline, cortical atrophy, and neural compensation.
Collapse
Affiliation(s)
- Younghwa Lee
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Jun Young Park
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Jang Jae Lee
- Gwangju Alzheimer’s Disease & Related Dementia Cohort Research Center, Chosun University, Gwangju, Korea
| | - Jungsoo Gim
- Gwangju Alzheimer’s Disease & Related Dementia Cohort Research Center, Chosun University, Gwangju, Korea
- Department of Biomedical Science, Chosun University, Gwangju, Korea
| | - Ah Ra Do
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Korea
| | - Jinyeon Jo
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Juhong Park
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Kangjin Kim
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Kyungtaek Park
- Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Heejin Jin
- Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Kyu Yeong Choi
- Gwangju Alzheimer’s Disease & Related Dementia Cohort Research Center, Chosun University, Gwangju, Korea
| | - Sarang Kang
- Gwangju Alzheimer’s Disease & Related Dementia Cohort Research Center, Chosun University, Gwangju, Korea
| | - Hoowon Kim
- Gwangju Alzheimer’s Disease & Related Dementia Cohort Research Center, Chosun University, Gwangju, Korea
- Department of Neurology, Chosun University Hospital, Gwangju, Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine, Seoul, Korea
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Seoul, Korea
| | - Lindsay A Farrer
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Kun Ho Lee
- Gwangju Alzheimer’s Disease & Related Dementia Cohort Research Center, Chosun University, Gwangju, Korea
- Department of Biomedical Science, Chosun University, Gwangju, Korea
- Dementia Research Group, Korea Brain Research Institute, Daegu, Korea
| | - Sungho Won
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Korea
- RexSoft Inc., Seoul, Korea
| |
Collapse
|
58
|
Seyedsalehi A, Warrier V, Bethlehem RAI, Perry BI, Burgess S, Murray GK. Educational attainment, structural brain reserve and Alzheimer's disease: a Mendelian randomization analysis. Brain 2023; 146:2059-2074. [PMID: 36310536 PMCID: PMC10151197 DOI: 10.1093/brain/awac392] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/01/2022] [Accepted: 09/19/2022] [Indexed: 11/13/2022] Open
Abstract
Higher educational attainment is observationally associated with lower risk of Alzheimer's disease. However, the biological mechanisms underpinning this association remain unclear. The protective effect of education on Alzheimer's disease may be mediated via increased brain reserve. We used two-sample Mendelian randomization to explore putative causal relationships between educational attainment, structural brain reserve as proxied by MRI phenotypes and Alzheimer's disease. Summary statistics were obtained from genome-wide association studies of educational attainment (n = 1 131 881), late-onset Alzheimer's disease (35 274 cases, 59 163 controls) and 15 measures of grey or white matter macro- or micro-structure derived from structural or diffusion MRI (nmax = 33 211). We conducted univariable Mendelian randomization analyses to investigate bidirectional associations between (i) educational attainment and Alzheimer's disease; (ii) educational attainment and imaging-derived phenotypes; and (iii) imaging-derived phenotypes and Alzheimer's disease. Multivariable Mendelian randomization was used to assess whether brain structure phenotypes mediated the effect of education on Alzheimer's disease risk. Genetically proxied educational attainment was inversely associated with Alzheimer's disease (odds ratio per standard deviation increase in genetically predicted years of schooling = 0.70, 95% confidence interval 0.60, 0.80). There were positive associations between genetically predicted educational attainment and four cortical metrics (standard deviation units change in imaging phenotype per one standard deviation increase in genetically predicted years of schooling): surface area 0.30 (95% confidence interval 0.20, 0.40); volume 0.29 (95% confidence interval 0.20, 0.37); intrinsic curvature 0.18 (95% confidence interval 0.11, 0.25); local gyrification index 0.21 (95% confidence interval 0.11, 0.31)]; and inverse associations with cortical intracellular volume fraction [-0.09 (95% confidence interval -0.15, -0.03)] and white matter hyperintensities volume [-0.14 (95% confidence interval -0.23, -0.05)]. Genetically proxied levels of surface area, cortical volume and intrinsic curvature were positively associated with educational attainment [standard deviation units change in years of schooling per one standard deviation increase in respective genetically predicted imaging phenotype: 0.13 (95% confidence interval 0.10, 0.16); 0.15 (95% confidence interval 0.11, 0.19) and 0.12 (95% confidence interval 0.04, 0.19)]. We found no evidence of associations between genetically predicted imaging-derived phenotypes and Alzheimer's disease. The inverse association of genetically predicted educational attainment with Alzheimer's disease did not attenuate after adjusting for imaging-derived phenotypes in multivariable analyses. Our results provide support for a protective causal effect of educational attainment on Alzheimer's disease risk, as well as potential bidirectional causal relationships between education and brain macro- and micro-structure. However, we did not find evidence that these structural markers affect risk of Alzheimer's disease. The protective effect of education on Alzheimer's disease may be mediated via other measures of brain reserve not included in the present study, or by alternative mechanisms.
Collapse
Affiliation(s)
- Aida Seyedsalehi
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0SR, UK
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford OX3 7JX, UK
| | - Varun Warrier
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK
| | - Richard A I Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Benjamin I Perry
- Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK
- CAMEO, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge CB4 1PX, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK
| | - Graham K Murray
- Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK
- CAMEO, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge CB4 1PX, UK
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| |
Collapse
|
59
|
Mäkinen E, Wikgren J, Pekkala S, Koch LG, Britton SL, Nokia MS, Lensu S. Genotype determining aerobic exercise capacity associates with behavioral plasticity in middle-aged rats. Behav Brain Res 2023; 443:114331. [PMID: 36774999 DOI: 10.1016/j.bbr.2023.114331] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/20/2023] [Accepted: 02/04/2023] [Indexed: 02/12/2023]
Abstract
Good aerobic fitness associates positively with cognitive performance and brain health and conversely, low aerobic fitness predisposes to neurodegenerative diseases. To study how genotype together with exercise, started at older age, affects brain and behavior, we utilized rats that differ in inherited aerobic fitness. Rats bred for Low Capacity for Running (LCR) are shown to display less synaptic plasticity and more inflammation in the hippocampus and perform worse than rats bred for a High Capacity for Running (HCR) in tasks requiring flexible cognition. Here we used middle-aged (∼ 16 months) HCR and LCR rats to study how genotype and sex associate with anxiety and neural information filtering, termed sensory gating. Further, we assessed how inherited aerobic capacity associates with hippocampus-dependent learning, measured with contextual fear conditioning task. In females, we also investigated the effects of voluntary wheel running (5 weeks) on these characteristics. Our results indicate that independent of sex or voluntary running, HCR rats were more anxious in open-field tasks, exhibited lower sensory gating and learned more efficiently in contextual fear conditioning task than LCR rats. Voluntary running did not markedly affect innate behavior but slightly decreased the differences between female LCR and HCR rats in fear learning. In conclusion, inherited fitness seems to determine cognitive and behavioral traits independent of sex. Although the traits proved to be rather resistant to change at adult age, learning was slightly improved following exercise in LCR females, prone to obesity and poor fitness.
Collapse
Affiliation(s)
- Elina Mäkinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Jan Wikgren
- Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo, OH, USA
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Miriam S Nokia
- Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| | - Sanna Lensu
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland; Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
60
|
Jain P, Yates M, de Celis CR, Drineas P, Jahanshad N, Thompson P, Paschou P. Multiomic approach and Mendelian randomization analysis identify causal associations between blood biomarkers and subcortical brain structure volumes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.30.23287968. [PMID: 37066330 PMCID: PMC10104218 DOI: 10.1101/2023.03.30.23287968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Alterations in subcortical brain structure volumes have been found to be associated with several neurodegenerative and psychiatric disorders. At the same time, genome-wide association studies (GWAS) have identified numerous common variants associated with brain structure. In this study, we integrate these findings, aiming to identify proteins, metabolites, or microbes that have a putative causal association with subcortical brain structure volumes via a two-sample Mendelian randomization approach. This method uses genetic variants as instrument variables to identify potentially causal associations between an exposure and an outcome. The exposure data that we analyzed comprised genetic associations for 2,994 plasma proteins, 237 metabolites, and 103 microbial genera. The outcome data included GWAS data for seven subcortical brain structure volumes including accumbens, amygdala, caudate, hippocampus, pallidum, putamen, and thalamus. Eleven proteins and six metabolites were found to have a significant association with subcortical structure volumes. We found causal associations between amygdala volume and granzyme A as well as association between accumbens volume and plasma protease c1 inhibitor. Among metabolites, urate had the strongest association with thalamic volume. No significant associations were detected between the microbial genera and subcortical brain structure volumes. We also observed significant enrichment for biological processes such as proteolysis, regulation of the endoplasmic reticulum apoptotic signaling pathway, and negative regulation of DNA binding. Our findings provide insights to the mechanisms through which brain volumes may be affected in the pathogenesis of neurodevelopmental and psychiatric disorders and point to potential treatment targets for disorders that are associated with subcortical brain structure volumes.
Collapse
Affiliation(s)
- Pritesh Jain
- Department of Biological Sciences, Purdue University
| | - Madison Yates
- Department of Biological Sciences, Purdue University
| | | | | | - Neda Jahanshad
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California
| | - Paul Thompson
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California
| | | |
Collapse
|
61
|
Pishva E, van den Hove DLA, Laroche V, Lvovs A, Roy A, Ortega G, Burrage J, Veidebaum T, Kanarik M, Mill J, Lesch KP, Harro J. Genome-wide DNA methylation analysis of aggressive behaviour: a longitudinal population-based study. J Child Psychol Psychiatry 2023. [PMID: 36929374 DOI: 10.1111/jcpp.13782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/25/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Human aggression is influenced by an interplay between genetic predisposition and experience across the life span. This interaction is thought to occur through epigenetic mechanisms, inducing differential gene expression, thereby moderating neuronal cell and circuit function, and thus shaping aggressive behaviour. METHODS Genome-wide DNA methylation (DNAm) levels were measured in peripheral blood obtained from 95 individuals participating in the Estonian Children Personality Behaviours and Health Study (ECPBHS) at 15 and 25 years of age. We examined the association between aggressive behaviour, as measured by Life History of Aggression (LHA) total score and DNAm levels both assessed at age 25. We further examined the pleiotropic effect of genetic variants regulating LHA-associated differentially methylated positions (DMPs) and multiple traits related to aggressive behaviours. Lastly, we tested whether the DNA methylomic loci identified in association with LHA at age 25 were also present at age 15. RESULTS We found one differentially methylated position (DMP) (cg17815886; p = 1.12 × 10-8 ) and five differentially methylated regions (DMRs) associated with LHA after multiple testing adjustments. The DMP annotated to the PDLIM5 gene, and DMRs resided in the vicinity of four protein-encoding genes (TRIM10, GTF2H4, SLC45A4, B3GALT4) and a long intergenic non-coding RNA (LINC02068). We observed evidence for the colocalization of genetic variants associated with top DMPs and general cognitive function, educational attainment and cholesterol levels. Notably, a subset of the DMPs associated with LHA at age 25 also displayed altered DNAm patterns at age 15 with high accuracy in predicting aggression. CONCLUSIONS Our findings highlight the potential role of DNAm in the development of aggressive behaviours. We observed pleiotropic genetic variants associated with identified DMPs, and various traits previously established to be relevant in shaping aggression in humans. The concordance of DNAm signatures in adolescents and young adults may have predictive value for inappropriate and maladaptive aggression later in life.
Collapse
Affiliation(s)
- Ehsan Pishva
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.,College of Medicine and Health, University of Exeter Medical School, Exeter, UK
| | - Daniel L A van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.,Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Valentin Laroche
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Aneth Lvovs
- School of Natural Sciences and Health, Tallinn University, Tallinn, Estonia.,Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Arunima Roy
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Gabriela Ortega
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Joe Burrage
- College of Medicine and Health, University of Exeter Medical School, Exeter, UK
| | | | - Margus Kanarik
- Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Jonathan Mill
- College of Medicine and Health, University of Exeter Medical School, Exeter, UK
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.,Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Jaanus Harro
- Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Tartu, Estonia
| |
Collapse
|
62
|
Ou YN, Wu BS, Ge YJ, Zhang Y, Jiang YC, Kuo K, Yang L, Tan L, Feng JF, Cheng W, Yu JT. The genetic architecture of human amygdala volumes and their overlap with common brain disorders. Transl Psychiatry 2023; 13:90. [PMID: 36906575 PMCID: PMC10008562 DOI: 10.1038/s41398-023-02387-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/13/2023] Open
Abstract
The amygdala is a crucial interconnecting structure in the brain that performs several regulatory functions, yet its genetic architectures and involvement in brain disorders remain largely unknown. We carried out the first multivariate genome-wide association study (GWAS) of amygdala subfield volumes in 27,866 UK Biobank individuals. The whole amygdala was segmented into nine nuclei groups using Bayesian amygdala segmentation. The post-GWAS analysis allowed us to identify causal genetic variants in phenotypes at the SNP, locus, and gene levels, as well as genetic overlap with brain health-related traits. We further generalized our GWAS in Adolescent Brain Cognitive Development (ABCD) cohort. The multivariate GWAS identified 98 independent significant variants within 32 genomic loci associated (P < 5 × 10-8) with amygdala volume and its nine nuclei. The univariate GWAS identified significant hits for eight of the ten volumes, tagging 14 independent genomic loci. Overall, 13 of the 14 loci identified in the univariate GWAS were replicated in the multivariate GWAS. The generalization in ABCD cohort supported the GWAS results with the 12q23.2 (RNA gene RP11-210L7.1) being discovered. All of these imaging phenotypes are heritable, with heritability ranging from 15% to 27%. Gene-based analyses revealed pathways relating to cell differentiation/development and ion transporter/homeostasis, with the astrocytes found to be significantly enriched. Pleiotropy analyses revealed shared variants with neurological and psychiatric disorders under the conjFDR threshold of 0.05. These findings advance our understanding of the complex genetic architectures of amygdala and their relevance in neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Bang-Sheng Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yi Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Yu-Chao Jiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Kevin Kuo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Liu Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.,Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China.,MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.,Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Wei Cheng
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China. .,Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China. .,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China. .,Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China.
| |
Collapse
|
63
|
Mao Q, Lin X, Yin Q, Liu P, Zhang Y, Qu S, Xu J, Cheng W, Luo X, Kang L, Taximaimaiti R, Zheng C, Zhang H, Wang X, Ren H, Cao Y, Lin J, Luo X. A significant, functional and replicable risk KTN1 variant block for schizophrenia. Sci Rep 2023; 13:3890. [PMID: 36890161 PMCID: PMC9995530 DOI: 10.1038/s41598-023-27448-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/02/2023] [Indexed: 03/10/2023] Open
Abstract
Cortical and subcortical structural alteration has been extensively reported in schizophrenia, including the unusual expansion of gray matter volumes (GMVs) of basal ganglia (BG), especially putamen. Previous genome-wide association studies pinpointed kinectin 1 gene (KTN1) as the most significant gene regulating the GMV of putamen. In this study, the role of KTN1 variants in risk and pathogenesis of schizophrenia was explored. A dense set of SNPs (n = 849) covering entire KTN1 was analyzed in three independent European- or African-American samples (n = 6704) and one mixed European and Asian Psychiatric Genomics Consortium sample (n = 56,418 cases vs. 78,818 controls), to identify replicable SNP-schizophrenia associations. The regulatory effects of schizophrenia-associated variants on the KTN1 mRNA expression in 16 cortical or subcortical regions in two European cohorts (n = 138 and 210, respectively), the total intracranial volume (ICV) in 46 European cohorts (n = 18,713), the GMVs of seven subcortical structures in 50 European cohorts (n = 38,258), and the surface areas (SA) and thickness (TH) of whole cortex and 34 cortical regions in 50 European cohorts (n = 33,992) and eight non-European cohorts (n = 2944) were carefully explored. We found that across entire KTN1, only 26 SNPs within the same block (r2 > 0.85) were associated with schizophrenia across ≥ 2 independent samples (7.5 × 10-5 ≤ p ≤ 0.048). The schizophrenia-risk alleles, which increased significantly risk for schizophrenia in Europeans (q < 0.05), were all minor alleles (f < 0.5), consistently increased (1) the KTN1 mRNA expression in 12 brain regions significantly (5.9 × 10-12 ≤ p ≤ 0.050; q < 0.05), (2) the ICV significantly (6.1 × 10-4 ≤ p ≤ 0.008; q < 0.05), (3) the SA of whole (9.6 × 10-3 ≤ p ≤ 0.047) and two regional cortices potentially (2.5 × 10-3 ≤ p ≤ 0.042; q > 0.05), and (4) the TH of eight regional cortices potentially (0.006 ≤ p ≤ 0.050; q > 0.05), and consistently decreased (1) the BG GMVs significantly (1.8 × 10-19 ≤ p ≤ 0.050; q < 0.05), especially putamen GMV (1.8 × 10-19 ≤ p ≤ 1.0 × 10-4; q < 0.05, (2) the SA of four regional cortices potentially (0.010 ≤ p ≤ 0.048), and (3) the TH of four regional cortices potentially (0.015 ≤ p ≤ 0.049) in Europeans. We concluded that we identified a significant, functional, and robust risk variant block covering entire KTN1 that might play a critical role in the risk and pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Qiao Mao
- Department of Psychosomatic Medicine, People's Hospital of Deyang City, Deyang, 618000, Sichuan, China
| | - Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Fujian Provincial Cancer Hospital, the Teaching Hospital of Fujian Medical University, Fuzhou, 350014, Fujian, China
| | - Qin Yin
- Department of Respiratory and Critical Care Medicine, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430000, Hubei, China
| | - Ping Liu
- Department of Psychosomatic Medicine, People's Hospital of Deyang City, Deyang, 618000, Sichuan, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin, 300222, China
| | - Shihao Qu
- Zhuhai Center for Maternal and Child Health Care, Zhuhai, Guangdong, 519001, China
| | - Jianying Xu
- Zhuhai Center for Maternal and Child Health Care, Zhuhai, Guangdong, 519001, China
| | - Wenhong Cheng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xinqun Luo
- Department of Neurosurgery, The First Hospital, Fujian Medical University, Fuzhou, 350004, Fujian, China
| | - Longli Kang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research On High Altitude Diseases of Tibet Autonomous Region, Xizang Minzu University School of Medicine, Xiangyang, 712082, Shaanxi, China
| | - Reyisha Taximaimaiti
- Department of Neurology, Shanghai Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Chengchou Zheng
- Minqing Psychiatric Hospital, Minqing, 350800, Fujian, China
| | - Huihao Zhang
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350001, China
| | - Xiaoping Wang
- Department of Neurology, The 1st People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, USA
| | - Honggang Ren
- Department of Internal Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuping Cao
- Department of Psychiatry, Second Xiangya Hospital, Central South University, China National Clinical Research Center On Mental Disorders, China National Technology Institute On Mental Disorders, Changsha, 410011, Hunan, China.
| | - Jie Lin
- Fujian Center for Disease Control and Prevention, Fuzhou, 350012, Fujian, China.
- Fujian Institute of Preventive Medicine, Fuzhou, 350012, Fujian, China.
| | - Xingguang Luo
- Beijing Huilongguan Hospital, Peking University Huilongguan School of Clinical Medicine, Beijing, 100096, China.
| |
Collapse
|
64
|
Kang L, Wang W, Zhang N, Yao L, Tu N, Feng H, Zong X, Bai H, Li R, Wang G, Bu L, Wang F, Liu Z. Anhedonia and dysregulation of an angular gyrus-centred and dynamic functional network in adolescent-onset depression. J Affect Disord 2023; 324:82-91. [PMID: 36581179 DOI: 10.1016/j.jad.2022.12.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Anhedonia is an important aspect of adolescent-onset major depressive disorder (MDD) and is associated with increased risk of suicidal behaviors and poor treatment outcomes. However, the neural circuitry underlying this deficit has not been well defined. This study aims to identify the relationships between anhedonia and changes in static and dynamic functional connectivity (FC) in adolescent-onset MDD patients compared with healthy control subjects (HCs) and adult-onset MDD patients. METHODS A total of 157 participants completed the Snaith-Hamilton Pleasure Scale (SHAPS) to assess hedonic capacity. Resting-state functional imaging scans were analysed using graph theoretical analysis, network-based statistics (NBS) and sliding window correlation analysis to explore the potential patterns of neural network brain disruptions in adolescent-onset MDD. Pearson correlations and support vector machines regression (SVR) were used to explore correlations and predict network measures with SHAPS scores. RESULTS Compared with those with adult-onset MDD, adolescent-onset MDD patients showed decreased FC in 7 nodes and 6 connections, with the right angular gyrus (AG), left AG and left paracentral lobule having the largest number of connected edges (P = 0.0396, NBS-corrected). Their average FC and SHAPS scores were positively correlated (r = 0.309, P = 0.035). Regarding dynamic FC, compared with HCs, adolescent-onset MDD patients showed a tendency towards a decreased frequency in moderate-intensity brain FC states (P = 0.014), which was significantly and positively correlated with SHAPS scores (r = 0.425, P = 0.003). SVR also revealed AG-centred FC and dynamic FC could predict SHAPS scores (MSE = 27.233, P = 0.001). CONCLUSIONS These findings provide distinct evidence on the physiological mechanisms of adolescent-onset MDD and anhedonia.
Collapse
Affiliation(s)
- Lijun Kang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Nan Zhang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lihua Yao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning Tu
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongyan Feng
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaofen Zong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hanping Bai
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruiting Li
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lihong Bu
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China; Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China.
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
65
|
Haddad E, Pizzagalli F, Zhu AH, Bhatt RR, Islam T, Ba Gari I, Dixon D, Thomopoulos SI, Thompson PM, Jahanshad N. Multisite test-retest reliability and compatibility of brain metrics derived from FreeSurfer versions 7.1, 6.0, and 5.3. Hum Brain Mapp 2023; 44:1515-1532. [PMID: 36437735 PMCID: PMC9921222 DOI: 10.1002/hbm.26147] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/29/2022] Open
Abstract
Automatic neuroimaging processing tools provide convenient and systematic methods for extracting features from brain magnetic resonance imaging scans. One tool, FreeSurfer, provides an easy-to-use pipeline to extract cortical and subcortical morphometric measures. There have been over 25 stable releases of FreeSurfer, with different versions used across published works. The reliability and compatibility of regional morphometric metrics derived from the most recent version releases have yet to be empirically assessed. Here, we used test-retest data from three public data sets to determine within-version reliability and between-version compatibility across 42 regional outputs from FreeSurfer versions 7.1, 6.0, and 5.3. Cortical thickness from v7.1 was less compatible with that of older versions, particularly along the cingulate gyrus, where the lowest version compatibility was observed (intraclass correlation coefficient 0.37-0.61). Surface area of the temporal pole, frontal pole, and medial orbitofrontal cortex, also showed low to moderate version compatibility. We confirm low compatibility between v6.0 and v5.3 of pallidum and putamen volumes, while those from v7.1 were compatible with v6.0. Replication in an independent sample showed largely similar results for measures of surface area and subcortical volumes, but had lower overall regional thickness reliability and compatibility. Batch effect correction may adjust for some inter-version effects when most sites are run with one version, but results vary when more sites are run with different versions. Age associations in a quality controlled independent sample (N = 106) revealed version differences in results of downstream statistical analysis. We provide a reference to highlight the regional metrics that may yield recent version-related inconsistencies in published findings. An interactive viewer is provided at http://data.brainescience.org/Freesurfer_Reliability/.
Collapse
Affiliation(s)
- Elizabeth Haddad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Fabrizio Pizzagalli
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA.,Department of Neurosciences, University of Turin, Turin, Italy
| | - Alyssa H Zhu
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Ravi R Bhatt
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Tasfiya Islam
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Iyad Ba Gari
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Daniel Dixon
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| |
Collapse
|
66
|
Zhao B, Li Y, Fan Z, Wu Z, Shu J, Yang X, Yang Y, Wang X, Li B, Wang X, Copana C, Yang Y, Lin J, Li Y, Stein JL, O'Brien JM, Li T, Zhu H. Eye-brain connections revealed by multimodal retinal and brain imaging genetics in the UK Biobank. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.16.23286035. [PMID: 36824893 PMCID: PMC9949187 DOI: 10.1101/2023.02.16.23286035] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
As an anatomical extension of the brain, the retina of the eye is synaptically connected to the visual cortex, establishing physiological connections between the eye and the brain. Despite the unique opportunity retinal structures offer for assessing brain disorders, less is known about their relationship to brain structure and function. Here we present a systematic cross-organ genetic architecture analysis of eye-brain connections using retina and brain imaging endophenotypes. Novel phenotypic and genetic links were identified between retinal imaging biomarkers and brain structure and function measures derived from multimodal magnetic resonance imaging (MRI), many of which were involved in the visual pathways, including the primary visual cortex. In 65 genomic regions, retinal imaging biomarkers shared genetic influences with brain diseases and complex traits, 18 showing more genetic overlaps with brain MRI traits. Mendelian randomization suggests that retinal structures have bidirectional genetic causal links with neurological and neuropsychiatric disorders, such as Alzheimer's disease. Overall, cross-organ imaging genetics reveals a genetic basis for eye-brain connections, suggesting that the retinal images can elucidate genetic risk factors for brain disorders and disease-related changes in intracranial structure and function.
Collapse
Affiliation(s)
- Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yujue Li
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhenyi Wu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Juan Shu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yilin Yang
- Department of Computer and Information Science and Electrical and Systems Engineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxuan Li
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA
| | - Xiyao Wang
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA
| | - Carlos Copana
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yue Yang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jinjie Lin
- Yale School of Management, Yale University, New Haven, CT 06511, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason L. Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joan M. O'Brien
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Diseases, PA, 19104, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
67
|
Miller AP, Gizer IR. Dual-systems models of the genetic architecture of impulsive personality traits: Neurogenetic evidence of distinct but related factors. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.10.23285725. [PMID: 36824800 PMCID: PMC9949186 DOI: 10.1101/2023.02.10.23285725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Background Dual-systems models provide a parsimonious framework for understanding the interplay between cortical and subcortical brain regions relevant to impulsive personality traits (IPTs) and their associations with psychiatric disorders. Despite recent developments in multivariate analysis of genome-wide association studies (GWAS), molecular genetic investigations of these models have not been conducted. Methods Using extant IPT GWAS, we conducted confirmatory genomic structural equation models (GenomicSEM) to empirically evaluate dual-systems models of the genetic architecture of IPTs. Genetic correlations between results of multivariate GWAS of dual-systems factors and GWAS of relevant cortical and subcortical neuroimaging phenotypes (regional/structural volume, cortical surface area, cortical thickness) were calculated and compared. Results Evaluation of GenomicSEM model fit indices for dual-systems models suggested that these models highlight important sources of shared and unique genetic variance between top-down and bottom-up constructs. Specifically, a dual-systems genomic model consisting of sensation seeking and lack of self-control factors demonstrated distinct but related sources of genetic influences ( r g =.60). Genetic correlation analyses provided evidence of differential associations between dual-systems factors and cortical neuroimaging phenotypes (e.g., lack of self-control negatively associated with cortical thickness, sensation seeking positively associated with cortical surface area). However, no significant associations were observed for subcortical phenotypes inconsistent with hypothesized functional localization of dual-systems constructs. Conclusions Dual-systems models of the genetic architecture of IPTs tested here demonstrate evidence of shared and unique genetic influences and associations with relevant neuroimaging phenotypes. These findings emphasize potential advantages in utilizing dual-systems models to study genetic influences for IPTs and transdiagnostic associations with psychiatric disorders.
Collapse
|
68
|
Shams H, Shao X, Santaniello A, Kirkish G, Harroud A, Ma Q, Isobe N, Schaefer CA, McCauley JL, Cree BAC, Didonna A, Baranzini SE, Patsopoulos NA, Hauser SL, Barcellos LF, Henry RG, Oksenberg JR. Polygenic risk score association with multiple sclerosis susceptibility and phenotype in Europeans. Brain 2023; 146:645-656. [PMID: 35253861 PMCID: PMC10169285 DOI: 10.1093/brain/awac092] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/29/2022] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Polygenic inheritance plays a pivotal role in driving multiple sclerosis susceptibility, an inflammatory demyelinating disease of the CNS. We developed polygenic risk scores (PRS) of multiple sclerosis and assessed associations with both disease status and severity in cohorts of European descent. The largest genome-wide association dataset for multiple sclerosis to date (n = 41 505) was leveraged to generate PRS scores, serving as an informative susceptibility marker, tested in two independent datasets, UK Biobank [area under the curve (AUC) = 0.73, 95% confidence interval (CI): 0.72-0.74, P = 6.41 × 10-146] and Kaiser Permanente in Northern California (KPNC, AUC = 0.8, 95% CI: 0.76-0.82, P = 1.5 × 10-53). Individuals within the top 10% of PRS were at higher than 5-fold increased risk in UK Biobank (95% CI: 4.7-6, P = 2.8 × 10-45) and 15-fold higher risk in KPNC (95% CI: 10.4-24, P = 3.7 × 10-11), relative to the median decile. The cumulative absolute risk of developing multiple sclerosis from age 20 onwards was significantly higher in genetically predisposed individuals according to PRS. Furthermore, inclusion of PRS in clinical risk models increased the risk discrimination by 13% to 26% over models based only on conventional risk factors in UK Biobank and KPNC, respectively. Stratifying disease risk by gene sets representative of curated cellular signalling cascades, nominated promising genetic candidate programmes for functional characterization. These pathways include inflammatory signalling mediation, response to viral infection, oxidative damage, RNA polymerase transcription, and epigenetic regulation of gene expression to be among significant contributors to multiple sclerosis susceptibility. This study also indicates that PRS is a useful measure for estimating susceptibility within related individuals in multicase families. We show a significant association of genetic predisposition with thalamic atrophy within 10 years of disease progression in the UCSF-EPIC cohort (P < 0.001), consistent with a partial overlap between the genetics of susceptibility and end-organ tissue injury. Mendelian randomization analysis suggested an effect of multiple sclerosis susceptibility on thalamic volume, which was further indicated to be through horizontal pleiotropy rather than a causal effect. In summary, this study indicates important, replicable associations of PRS with enhanced risk assessment and radiographic outcomes of tissue injury, potentially informing targeted screening and prevention strategies.
Collapse
Affiliation(s)
- Hengameh Shams
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
- Division of Epidemiology and Biostatistics, School of Public Health, University of California Berkeley, Berkeley, CA 94720, USA
| | - Xiaorong Shao
- Division of Epidemiology and Biostatistics, School of Public Health, University of California Berkeley, Berkeley, CA 94720, USA
| | - Adam Santaniello
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Gina Kirkish
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Adil Harroud
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Qin Ma
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Noriko Isobe
- Department of Neurology, Graduate School of medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | | | - Jacob L McCauley
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Bruce A C Cree
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Alessandro Didonna
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC 27834, USA
| | - Sergio E Baranzini
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Nikolaos A Patsopoulos
- Systems Biology and Computer Science Program, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women’s Hospital, Boston, 02115 MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stephen L Hauser
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Lisa F Barcellos
- Division of Epidemiology and Biostatistics, School of Public Health, University of California Berkeley, Berkeley, CA 94720, USA
| | - Roland G Henry
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jorge R Oksenberg
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
69
|
Zhao Y, Chang C, Zhang J, Zhang Z. Genetic underpinnings of brain structural connectome for young adults. J Am Stat Assoc 2023; 118:1473-1487. [PMID: 37982009 PMCID: PMC10655950 DOI: 10.1080/01621459.2022.2156349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
Abstract
With distinct advantages in power over behavioral phenotypes, brain imaging traits have become emerging endophenotypes to dissect molecular contributions to behaviors and neuropsychiatric illnesses. Among different imaging features, brain structural connectivity (i.e., structural connectome) which summarizes the anatomical connections between different brain regions is one of the most cutting edge while under-investigated traits; and the genetic influence on the structural connectome variation remains highly elusive. Relying on a landmark imaging genetics study for young adults, we develop a biologically plausible brain network response shrinkage model to comprehensively characterize the relationship between high dimensional genetic variants and the structural connectome phenotype. Under a unified Bayesian framework, we accommodate the topology of brain network and biological architecture within the genome; and eventually establish a mechanistic mapping between genetic biomarkers and the associated brain sub-network units. An efficient expectation-maximization algorithm is developed to estimate the model and ensure computing feasibility. In the application to the Human Connectome Project Young Adult (HCP-YA) data, we establish the genetic underpinnings which are highly interpretable under functional annotation and brain tissue eQTL analysis, for the brain white matter tracts connecting the hippocampus and two cerebral hemispheres. We also show the superiority of our method in extensive simulations.
Collapse
Affiliation(s)
- Yize Zhao
- Department of Biostatistics, Yale University
| | - Changgee Chang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania
| | - Jingwen Zhang
- Department of Biostatistics, Boston University, Boston, MA
| | - Zhengwu Zhang
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill
| |
Collapse
|
70
|
Shang MY, Zhang CY, Wu Y, Wang L, Wang C, Li M. Genetic associations between bipolar disorder and brain structural phenotypes. Cereb Cortex 2023:7024717. [PMID: 36734292 DOI: 10.1093/cercor/bhad014] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Patients with bipolar disorder (BD) and their first-degree relatives exhibit alterations in brain volume and cortical structure, whereas the underlying genetic mechanisms remain unclear. In this study, based on the published genome-wide association studies (GWAS), the extent of polygenic overlap between BD and 15 brain structural phenotypes was investigated using linkage disequilibrium score regression and MiXeR tool, and the shared genomic loci were discovered by conjunctional false discovery rate (conjFDR) and expression quantitative trait loci (eQTL) analyses. MiXeR estimated the overall measure of polygenic overlap between BD and brain structural phenotypes as 4-53% on a 0-100% scale (as quantified by the Dice coefficient). Subsequent conjFDR analyses identified 54 independent loci (71 risk single-nucleotide polymorphisms) jointly associated with BD and brain structural phenotypes with a conjFDR < 0.05, among which 33 were novel that had not been reported in the previous BD GWAS. Follow-up eQTL analyses in respective brain regions both confirmed well-known risk genes (e.g. CACNA1C, NEK4, GNL3, MAPK3) and discovered novel risk genes (e.g. LIMK2 and CAMK2N2). This study indicates a substantial shared genetic basis between BD and brain structural phenotypes, and provides novel insights into the developmental origin of BD and related biological mechanisms.
Collapse
Affiliation(s)
- Meng-Yuan Shang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, China.,School of Basic Medical Science, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No. 17 Long-Xin Lu, Kunming, 650201, Yunnan, China
| | - Yong Wu
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, No. 920 Jianshe Road, Wuhan, 430012, Hubei, China
| | - Lu Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No. 17 Long-Xin Lu, Kunming, 650201, Yunnan, China
| | - Chuang Wang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, China.,School of Basic Medical Science, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No. 17 Long-Xin Lu, Kunming, 650201, Yunnan, China
| |
Collapse
|
71
|
Luo X, Lin X, Ide JS, Luo X, Zhang Y, Xu J, Wang L, Chen Y, Cheng W, Zheng J, Wang Z, Yu T, Taximaimaiti R, Jing X, Wang X, Cao Y, Tan Y, Li CSR. Male-specific, replicable and functional roles of genetic variants and cerebral gray matter volumes in ADHD: a gene-wide association study across KTN1 and a region-wide functional validation across brain. Child Adolesc Psychiatry Ment Health 2023; 17:4. [PMID: 36609385 PMCID: PMC9824933 DOI: 10.1186/s13034-022-00536-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/23/2022] [Indexed: 01/07/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is associated with reduction of cortical and subcortical gray matter volumes (GMVs). The kinectin 1 gene (KTN1) has recently been reported to significantly regulate GMVs and ADHD risk. In this study, we aimed to identify sex-specific, replicable risk KTN1 alleles for ADHD and to explore their regulatory effects on mRNA expression and cortical and subcortical GMVs. We examined a total of 1020 KTN1 SNPs in one discovery sample (ABCD cohort: 5573 males and 5082 females) and three independent replication European samples (Samples #1 and #2 each with 802/122 and 472/141 male/female offspring with ADHD; and Sample #3 with 14,154/4945 ADHD and 17,948/16,246 healthy males/females) to identify replicable associations within each sex. We examined the regulatory effects of ADHD-risk alleles on the KTN1 mRNA expression in two European brain cohorts (n = 348), total intracranial volume (TIV) in 46 European cohorts (n = 18,713) and the ABCD cohort, as well as the GMVs of seven subcortical structures in 50 European cohorts (n = 38,258) and of 118 cortical and subcortical regions in the ABCD cohort. We found that four KTN1 variants significantly regulated the risk of ADHD with the same direction of effect in males across discovery and replication samples (0.003 ≤ p ≤ 0.041), but none in females. All four ADHD-risk alleles significantly decreased KTN1 mRNA expression in all brain regions examined (1.2 × 10-5 ≤ p ≤ 0.039). The ADHD-risk alleles significantly increased basal ganglia (2.8 × 10-22 ≤ p ≤ 0.040) and hippocampus (p = 0.010) GMVs but reduced amygdala GMV (p = 0.030) and TIV (0.010 < p ≤ 0.013). The ADHD-risk alleles also significantly reduced some cortical (right superior temporal pole, right rectus) and cerebellar but increased other cortical (0.007 ≤ p ≤ 0.050) GMVs. To conclude, we identified a set of replicable and functional risk KTN1 alleles for ADHD, specifically in males. KTN1 may play a critical role in the pathogenesis of ADHD, and the reduction of specific cortical and subcortical, including amygdalar but not basal ganglia or hippocampal, GMVs may serve as a neural marker of the genetic effects.
Collapse
Affiliation(s)
- Xingguang Luo
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical School of Medicine, Beijing, 100096, China
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Jaime S Ide
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Xinqun Luo
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, Fujian, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin, 300222, China
| | - Jianying Xu
- Zhuhai Center for Maternal and Child Health Care, Zhuhai, 519000, Guangdong, China
| | - Leilei Wang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical School of Medicine, Beijing, 100096, China
| | - Yu Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Wenhong Cheng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Jianming Zheng
- National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University School of Medicine, Shanghai, 200030, China
| | - Zhiren Wang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical School of Medicine, Beijing, 100096, China
| | - Ting Yu
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical School of Medicine, Beijing, 100096, China
| | - Reyisha Taximaimaiti
- Department of Neurology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Xiaozhong Jing
- Department of Neurology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Xiaoping Wang
- Department of Neurology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Yuping Cao
- Department of Psychiatry, Second Xiangya Hospital, Central South University; China National Clinical Research Center On Mental Disorders, China National Technology Institute On Mental Disorders, Changsha, 410011, Hunan, China.
| | - Yunlong Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical School of Medicine, Beijing, 100096, China
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
- Wu Tsai Institute, Yale University, New Haven, CT, 06510, USA
| |
Collapse
|
72
|
Bolhuis K, Mulder RH, de Mol CL, Defina S, Warrier V, White T, Tiemeier H, Muetzel RL, Cecil CAM. Mapping gene by early life stress interactions on child subcortical brain structures: A genome-wide prospective study. JCPP ADVANCES 2022; 2:jcv2.12113. [PMID: 36777645 PMCID: PMC7614163 DOI: 10.1002/jcv2.12113] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022] Open
Abstract
Background Although it is well-established that both genetics and the environment influence brain development, they are typically examined separately. Here, we aimed to prospectively investigate the interactive effects of genetic variants-from a genome-wide approach-and early life stress (ELS) on child subcortical brain structures, and their association with subsequent mental health problems. Method Primary analyses were conducted using data from the Generation R Study (N = 2257), including genotype and cumulative prenatal and postnatal ELS scores (encompassing life events, contextual risk, parental risk, interpersonal risk, direct victimisation). Neuroimaging data were collected at age 10 years, including intracranial and subcortical brain volumes (accumbens, amygdala, caudate, hippocampus, pallidum, putamen, thalamus). Genome-wide association and genome-wide-by-environment interaction analyses (GWEIS, run separately for prenatal/postnatal ELS) were conducted for eight brain outcomes (i.e., 24 genome-wide analyses) in the Generation R Study (discovery). Polygenic scores (PGS) using the resulting weights were calculated in an independent (target) cohort (adolescent brain cognitive development Study; N = 10,751), to validate associations with corresponding subcortical volumes and examine links to later mother-reported internalising and externalising problems. Results One GWEIS-prenatal stress locus was associated with caudate volume (rs139505895, mapping onto PRSS12 and NDST3) and two GWEIS-postnatal stress loci with the accumbens (rs2397823 and rs3130008, mapping onto CUTA, SYNGAP1, and TABP). Functional annotation revealed that these genes play a role in neuronal plasticity and synaptic function, and have been implicated in neuro-developmental phenotypes, for example, intellectual disability, autism, and schizophrenia. None of these associations survived a more stringent correction for multiple testing across all analysis sets. In the validation sample, all PGSgenotype were associated with their respective brain volumes, but no PGSGxE associated with any subcortical volume. None of the PGS associated with internalising or externalising problems. Conclusions This study lends novel suggestive insights into gene-environment interplay on the developing brain as well as pointing to promising candidate loci for future replication and mechanistic studies.
Collapse
Affiliation(s)
- Koen Bolhuis
- Department of Child and Adolescent Psychiatry/PsychologyErasmus MC‐SophiaRotterdamThe Netherlands
| | - Rosa H. Mulder
- Department of PediatricsErasmus MC‐SophiaRotterdamThe Netherlands
| | - Casper Louk de Mol
- Department of NeurologyMS Center ErasMSErasmus MCRotterdamThe Netherlands
| | - Serena Defina
- Department of Child and Adolescent Psychiatry/PsychologyErasmus MC‐SophiaRotterdamThe Netherlands
| | - Varun Warrier
- Department of PsychiatryUniversity of CambridgeCambridgeUK
| | - Tonya White
- Department of Child and Adolescent Psychiatry/PsychologyErasmus MC‐SophiaRotterdamThe Netherlands
- Department of Radiology and Nuclear MedicineErasmus MCRotterdamThe Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry/PsychologyErasmus MC‐SophiaRotterdamThe Netherlands
- Department of Social and Behavioral SciencesHarvard TH Chan School of Public HealthBostonMassachusettsUSA
| | - Ryan L. Muetzel
- Department of Child and Adolescent Psychiatry/PsychologyErasmus MC‐SophiaRotterdamThe Netherlands
| | - Charlotte A. M. Cecil
- Department of Child and Adolescent Psychiatry/PsychologyErasmus MC‐SophiaRotterdamThe Netherlands
- Department of EpidemiologyErasmus MCRotterdamThe Netherlands
- Molecular EpidemiologyDepartment of Biomedical Data SciencesLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
73
|
Han X, Zhang J, Chen S, Yu W, Zhou Y, Gu X. Mapping the current trends and hotspots of vascular cognitive impairment from 2000-2021: A bibliometric analysis. CNS Neurosci Ther 2022; 29:771-782. [PMID: 36415118 PMCID: PMC9928552 DOI: 10.1111/cns.14026] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/03/2022] [Accepted: 10/30/2022] [Indexed: 11/24/2022] Open
Abstract
AIMS To visualize the trends and hotspots in the scientific research related to vascular cognitive impairment (VCI) quantitatively and qualitatively. METHODS Cross-sectional bibliometric analysis of publications that related to VCI was conducted. Publications were found by searching in the Web of Science Core Collection database (WoSCC) - Edition: Science Citation Index Expanded (SCI-Expanded) from January 2000 to December 2021. Publication type was restricted to article and review in the English language. The downloaded data were screened and analyzed in January 2022. RESULTS In total, 16,264 publications were identified, with a steady increase in annual publications. The United States was the leading country in VCI research regarding publication numbers and national influence. National Institute of Aging had the highest influence among all the institutes in the field of VCI. Philip Scheltens was the most active author. The top five active authors' publications focused on pathobiology, neuroimaging standards, risk factors, prevention, and the standard diagnosis of vascular dementia (VaD). A co-cited publication clustering resulted in 19 main clusters, and the prevention, blood-brain barrier, cholesterol, cerebral amyloid angiopathy, and VaD were the top 5 clusters. Moreover, burst keywords detection revealed that the "small vessel disease" is the current hotspot in the field of VCI. CONCLUSIONS This bibliometric analysis mapped the overall research structure of VCI and analyzed the current research trends and hotspots for future studies orientation. Neuroimaging, risk factors detection, and pathobiology are the current trends in VCI research. Small vessel disease and its mechanisms are the current hotspots of VCI research.
Collapse
Affiliation(s)
- Xu Han
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jian Zhang
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Sifan Chen
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weifeng Yu
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yan Zhou
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiyao Gu
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
74
|
Strelnikov D, Alijanpourotaghsara A, Piroska M, Szalontai L, Forgo B, Jokkel Z, Persely A, Hernyes A, Kozak LR, Szabo A, Maurovich-Horvat P, Tarnoki DL, Tarnoki AD. Heritability of Subcortical Grey Matter Structures. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1687. [PMID: 36422226 PMCID: PMC9696305 DOI: 10.3390/medicina58111687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 02/03/2024]
Abstract
Background and Objectives: Subcortical grey matter structures play essential roles in cognitive, affective, social, and motoric functions in humans. Their volume changes with age, and decreased volumes have been linked with many neuropsychiatric disorders. The aim of our study was to examine the heritability of six subcortical brain volumes (the amygdala, caudate nucleus, pallidum, putamen, thalamus, and nucleus accumbens) and four general brain volumes (the total intra-cranial volume and the grey matter, white matter, and cerebrospinal fluid (CSF) volume) in twins. Materials and Methods: A total of 118 healthy adult twins from the Hungarian Twin Registry (86 monozygotic and 32 dizygotic; median age 50 ± 27 years) underwent brain magnetic resonance imaging. Two automated volumetry pipelines, Computational Anatomy Toolbox 12 (CAT12) and volBrain, were used to calculate the subcortical and general brain volumes from three-dimensional T1-weighted images. Age- and sex-adjusted monozygotic and dizygotic intra-pair correlations were calculated, and the univariate ACE model was applied. Pearson's correlation test was used to compare the results obtained by the two pipelines. Results: The age- and sex-adjusted heritability estimates, using CAT12 for the amygdala, caudate nucleus, pallidum, putamen, and nucleus accumbens, were between 0.75 and 0.95. The thalamus volume was more strongly influenced by common environmental factors (C = 0.45-0.73). The heritability estimates, using volBrain, were between 0.69 and 0.92 for the nucleus accumbens, pallidum, putamen, right amygdala, and caudate nucleus. The left amygdala and thalamus were more strongly influenced by common environmental factors (C = 0.72-0.85). A strong correlation between CAT12 and volBrain (r = 0.74-0.94) was obtained for all volumes. Conclusions: The majority of examined subcortical volumes appeared to be strongly heritable. The thalamus was more strongly influenced by common environmental factors when investigated with both segmentation methods. Our results underline the importance of identifying the relevant genes responsible for variations in the subcortical structure volume and associated diseases.
Collapse
Affiliation(s)
- David Strelnikov
- Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary
| | | | - Marton Piroska
- Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary
| | - Laszlo Szalontai
- Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary
| | - Bianka Forgo
- Department of Radiology, Faculty of Medicine and Health, Örebro University, 702 81 Örebro, Sweden
| | - Zsofia Jokkel
- Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary
| | - Alíz Persely
- Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary
| | - Anita Hernyes
- Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary
| | | | - Adam Szabo
- Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary
| | | | | | | |
Collapse
|
75
|
Tian M, Xu F, Xia Q, Tang Y, Zhang Z, Lin X, Meng H, Feng L, Liu S. Morphological development of the human fetal striatum during the second trimester. Cereb Cortex 2022; 32:5072-5082. [PMID: 35078212 DOI: 10.1093/cercor/bhab532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/24/2021] [Accepted: 12/25/2021] [Indexed: 12/27/2022] Open
Abstract
The morphological development of the fetal striatum during the second trimester has remained poorly described. We manually segmented the striatum using 7.0-T MR images of the fetal specimens ranging from 14 to 22 gestational weeks. The global development of the striatum was evaluated by volume measurement. The absolute volume (Vabs) of the caudate nucleus (CN) increased linearly with gestational age, while the relative volume (Vrel) showed a quadratic growth. Both Vabs and Vrel of putamen increased linearly. Through shape analysis, the changes of local structure in developing striatum were specifically demonstrated. Except for the CN tail, the lateral and medial parts of the CN grew faster than the middle regions, with a clear rostral-caudal growth gradient as well as a distinct "outside-in" growth gradient. For putamen, the dorsal and ventral regions grew obviously faster than the other regions, with a dorsal-ventral bidirectional developmental pattern. The right CN was larger than the left, whereas there was no significant hemispheric asymmetry in the putamen. By establishing the developmental trajectories, spatial heterochrony, and hemispheric dimorphism of human fetal striatum, these data bring new insight into the fetal striatum development and provide detailed anatomical references for future striatal studies.
Collapse
Affiliation(s)
- Mimi Tian
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Feifei Xu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Qing Xia
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Yuchun Tang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Zhonghe Zhang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Department of Medical Imaging, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Xiangtao Lin
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Department of Medical Imaging, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Haiwei Meng
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Lei Feng
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| | - Shuwei Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Key Laboratory of Mental Disorders, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
76
|
Doust C, Fontanillas P, Eising E, Gordon SD, Wang Z, Alagöz G, Molz B, Pourcain BS, Francks C, Marioni RE, Zhao J, Paracchini S, Talcott JB, Monaco AP, Stein JF, Gruen JR, Olson RK, Willcutt EG, DeFries JC, Pennington BF, Smith SD, Wright MJ, Martin NG, Auton A, Bates TC, Fisher SE, Luciano M. Discovery of 42 genome-wide significant loci associated with dyslexia. Nat Genet 2022; 54:1621-1629. [PMID: 36266505 PMCID: PMC9649434 DOI: 10.1038/s41588-022-01192-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 08/23/2022] [Indexed: 12/11/2022]
Abstract
Reading and writing are crucial life skills but roughly one in ten children are affected by dyslexia, which can persist into adulthood. Family studies of dyslexia suggest heritability up to 70%, yet few convincing genetic markers have been found. Here we performed a genome-wide association study of 51,800 adults self-reporting a dyslexia diagnosis and 1,087,070 controls and identified 42 independent genome-wide significant loci: 15 in genes linked to cognitive ability/educational attainment, and 27 new and potentially more specific to dyslexia. We validated 23 loci (13 new) in independent cohorts of Chinese and European ancestry. Genetic etiology of dyslexia was similar between sexes, and genetic covariance with many traits was found, including ambidexterity, but not neuroanatomical measures of language-related circuitry. Dyslexia polygenic scores explained up to 6% of variance in reading traits, and might in future contribute to earlier identification and remediation of dyslexia.
Collapse
Affiliation(s)
- Catherine Doust
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | | | - Else Eising
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Scott D Gordon
- Genetic Epidemiology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Zhengjun Wang
- School of Psychology, Shaanxi Normal University and Shaanxi Key Research Center of Child Mental and Behavioral Health, Xi'an, China
| | - Gökberk Alagöz
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Barbara Molz
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | | | | | - Beate St Pourcain
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Clyde Francks
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jingjing Zhao
- School of Psychology, Shaanxi Normal University and Shaanxi Key Research Center of Child Mental and Behavioral Health, Xi'an, China
| | | | - Joel B Talcott
- Institute of Health and Neurodevelopment, Aston University, Birmingham, UK
| | | | - John F Stein
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Jeffrey R Gruen
- Departments of Pediatrics and Genetics, Yale Medical School, New Haven, CT, USA
| | - Richard K Olson
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - Erik G Willcutt
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - John C DeFries
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | | | - Shelley D Smith
- Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas G Martin
- Genetic Epidemiology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Timothy C Bates
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Michelle Luciano
- Department of Psychology, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
77
|
Brown AA, Clocksin HE, Abbene EE, Ursery M, Christ SE. The relationship between metabolic control and basal ganglia morphometry and function in individuals with early-treated phenylketonuria. Mol Genet Metab 2022; 137:249-256. [PMID: 36209659 DOI: 10.1016/j.ymgme.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022]
Abstract
Abnormalities of the cortical white matter are the most prominent and widely-reported neurological findings in individuals with early-treated phenylketonuria (ETPKU). Much less is known regarding the effects of ETPKU on gray matter structures in the brain such as the basal ganglia. Previous findings on basal ganglia in ETPKU have been mixed. The current study was designed to further elucidate the effects of ETPKU and elevated phe levels on the morphometry of basal ganglia structures (i.e., putamen, caudate nucleus, nucleus accumbens, and globus pallidus). High resolution magnetic resonance imaging (MRI) data was collected from a sample of 37 adults with ETPKU and a demographically-matched comparison group of 33 individuals without PKU. No overall group differences (ETPKU vs. non-PKU) in basal ganglia volumes were observed. However, within the ETPKU group, poorer metabolic control (as reflected by higher blood phenylalanine levels) was associated with larger putamen volume. Vertex-wise shape analysis revealed that the volume increase was accompanied by shape changes in the middle left putamen. Consistent with this area's role in motor control, a significant correlation between left putamen volume and motor performance was also observed. Additional research is needed to fully understand the cellular level processes underlying this effect as well as to better understand the clinical impact of these morphometric changes and their potential relation to treatment response.
Collapse
Affiliation(s)
- Alexander A Brown
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Hayley E Clocksin
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Emily E Abbene
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Mikayla Ursery
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Shawn E Christ
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
78
|
Lahti J, Tuominen S, Yang Q, Pergola G, Ahmad S, Amin N, Armstrong NJ, Beiser A, Bey K, Bis JC, Boerwinkle E, Bressler J, Campbell A, Campbell H, Chen Q, Corley J, Cox SR, Davies G, De Jager PL, Derks EM, Faul JD, Fitzpatrick AL, Fohner AE, Ford I, Fornage M, Gerring Z, Grabe HJ, Grodstein F, Gudnason V, Simonsick E, Holliday EG, Joshi PK, Kajantie E, Kaprio J, Karell P, Kleineidam L, Knol MJ, Kochan NA, Kwok JB, Leber M, Lam M, Lee T, Li S, Loukola A, Luck T, Marioni RE, Mather KA, Medland S, Mirza SS, Nalls MA, Nho K, O'Donnell A, Oldmeadow C, Painter J, Pattie A, Reppermund S, Risacher SL, Rose RJ, Sadashivaiah V, Scholz M, Satizabal CL, Schofield PW, Schraut KE, Scott RJ, Simino J, Smith AV, Smith JA, Stott DJ, Surakka I, Teumer A, Thalamuthu A, Trompet S, Turner ST, van der Lee SJ, Villringer A, Völker U, Wilson RS, Wittfeld K, Vuoksimaa E, Xia R, Yaffe K, Yu L, Zare H, Zhao W, Ames D, Attia J, Bennett DA, Brodaty H, Chasman DI, Goldman AL, Hayward C, Ikram MA, Jukema JW, Kardia SLR, Lencz T, Loeffler M, Mattay VS, Palotie A, Psaty BM, Ramirez A, Ridker PM, Riedel-Heller SG, Sachdev PS, Saykin AJ, Scherer M, Schofield PR, Sidney S, Starr JM, Trollor J, Ulrich W, Wagner M, Weir DR, Wilson JF, Wright MJ, Weinberger DR, Debette S, Eriksson JG, Mosley TH, Launer LJ, van Duijn CM, Deary IJ, Seshadri S, Räikkönen K. Genome-wide meta-analyses reveal novel loci for verbal short-term memory and learning. Mol Psychiatry 2022; 27:4419-4431. [PMID: 35974141 PMCID: PMC9734053 DOI: 10.1038/s41380-022-01710-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 12/14/2022]
Abstract
Understanding the genomic basis of memory processes may help in combating neurodegenerative disorders. Hence, we examined the associations of common genetic variants with verbal short-term memory and verbal learning in adults without dementia or stroke (N = 53,637). We identified novel loci in the intronic region of CDH18, and at 13q21 and 3p21.1, as well as an expected signal in the APOE/APOC1/TOMM40 region. These results replicated in an independent sample. Functional and bioinformatic analyses supported many of these loci and further implicated POC1. We showed that polygenic score for verbal learning associated with brain activation in right parieto-occipital region during working memory task. Finally, we showed genetic correlations of these memory traits with several neurocognitive and health outcomes. Our findings suggest a role of several genomic loci in verbal memory processes.
Collapse
Affiliation(s)
- Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland.
- Turku Institute of Advanced Studies, University of Turku, Turku, Finland.
| | - Samuli Tuominen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Qiong Yang
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Giulio Pergola
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Basic Medical Science, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Nicola J Armstrong
- Department of Mathematics and Statistics, Murdoch University, Murdoch, WA, Australia
| | - Alexa Beiser
- Department of Biostatistics, Boston University, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Katharina Bey
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Qiang Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Janie Corley
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Simon R Cox
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Gail Davies
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA
| | - Eske M Derks
- Translational Neurogenomics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Annette L Fitzpatrick
- Department of Family Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Alison E Fohner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Institute of Public Health Genetics, University of Washington, Seattle, WA, USA
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, UK
| | - Myriam Fornage
- McGovern Medical School, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zachary Gerring
- Translational Neurogenomics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases, Greifswald, Germany
| | - Francine Grodstein
- Channing Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Harvard School of Public Health, Boston, MA, USA
| | - Vilmundur Gudnason
- Icelandic Heart Assocation, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Eleanor Simonsick
- Translational Gerontology Branch, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Elizabeth G Holliday
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- Institute of Social and Preventive Medicine, University of Lausanne, Lausanne, Switzerland
| | - Eero Kajantie
- National Institute for Health and Welfare, Helsinki and Oulu, Oulu, Finland
- Hospital for Children and Adolescents, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Pauliina Karell
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases, Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Nicole A Kochan
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - John B Kwok
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Markus Leber
- Department of Psychiatry, University of Cologne, Cologne, Germany
| | - Max Lam
- Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY, USA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Teresa Lee
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Shuo Li
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Anu Loukola
- Helsinki Biobank, University of Helsinki Central Hospital, Helsinki, Finland
| | - Tobias Luck
- Department of Economic and Social Sciences & Institute of Social Medicine, Rehabilitation Sciences and Healthcare Research, University of Applied Sciences Nordhausen, Nordhausen, Germany
- University of Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, Leipzig, Germany
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Karen A Mather
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Sunnybrook Health Sciences Centre, University of Toronto, Randwick, NSW, Australia
| | - Sarah Medland
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Saira S Mirza
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adrienne O'Donnell
- Department of Biostatistics, Boston University, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Christopher Oldmeadow
- Clinical Research Design, IT and Statistical Support Unit, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Jodie Painter
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Alison Pattie
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Simone Reppermund
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - 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
| | - Richard J Rose
- Department of Psychological & Brain Sciences, Indiana University Bloomington, Bloomington, IN, USA
| | - Vijay Sadashivaiah
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Claudia L Satizabal
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Peter W Schofield
- Neuropsychiatry Service, Hunter New England Local Health District, Charlestown, NSW, Australia
| | - Katharina E Schraut
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Jeannette Simino
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Albert V Smith
- Icelandic Heart Assocation, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
- Institute of Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI, USA
| | - David J Stott
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Ida Surakka
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Stella Trompet
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sven J van der Lee
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Arno Villringer
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Day Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Robert S Wilson
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases, Greifswald, Germany
| | - Eero Vuoksimaa
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Rui Xia
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kristine Yaffe
- Department of Psychiatry, University of California, San Francisco, CA, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Habil Zare
- Department of Cell Systems & Anatomy, The University of Texas Health Science Center, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas, San Antonio, TX, USA
- University of Texas Health Sciences Center, Houston, NA, US
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - David Ames
- National Ageing Research Institute, Parkville, Melbourne, VIC, Australia
- University of Melbourne, Academic Unit for Psychiatry of Old Age, St George's Hospital, Melbourne, VIC, Australia
| | - John Attia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
- Clinical Research Design, IT and Statistical Support Unit, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Dementia Collaborative Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aaron L Goldman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Todd Lencz
- Hofstra Northwell School of Medicine, Hempstead, NY, USA
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Venkata S Mattay
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Food and Drug Administration, Washington, DC, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology and Department of Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Heath Research Institute, Seattle, WA, USA
| | - Alfredo Ramirez
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
- Department of Psychiatry, University of Cologne, Cologne, Germany
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Steffi G Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Leipzig, Germany
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Martin Scherer
- Institute of Primary Medical Care, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter R Schofield
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- Neuroscience Research Australia, Randwick, NSW, Australia
| | - Stephen Sidney
- Kaiser Permanente Northern California, Division of Research, Oakland, CA, USA
| | - John M Starr
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - Julian Trollor
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Developmental Disability Neuropsychiatry, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - William Ulrich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Michael Wagner
- German Center for Neurodegenerative Diseases, Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Margaret J Wright
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephanie Debette
- Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, University of Bordeaux, Bordeaux, France
- Bordeaux University Hospital (CHU Bordeaux), Department of Neurology, Bordeaux, France
| | - Johan G Eriksson
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, and Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Helsinki, Singapore
| | - Thomas H Mosley
- Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Public Health, Oxford University, Oxford, UK
| | - Ian J Deary
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Sudha Seshadri
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Katri Räikkönen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
79
|
Mavromatis LA, Rosoff DB, Cupertino RB, Garavan H, Mackey S, Lohoff FW. Association Between Brain Structure and Alcohol Use Behaviors in Adults: A Mendelian Randomization and Multiomics Study. JAMA Psychiatry 2022; 79:869-878. [PMID: 35947372 PMCID: PMC9366661 DOI: 10.1001/jamapsychiatry.2022.2196] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/15/2022] [Indexed: 11/27/2022]
Abstract
Importance Past studies have identified associations between brain macrostructure and alcohol use behaviors. However, identifying directional associations between these phenotypes is difficult due to the limitations of observational studies. Objective To use mendelian randomization (MR) to identify directional associations between brain structure and alcohol use and elucidate the transcriptomic and cellular underpinnings of identified associations. Design, Setting, and Participants The main source data comprised summary statistics from population-based and case-control genome-wide association studies (GWAS) of neuroimaging, behavioral, and clinical phenotypes (N = 763 874). Using these data, bidirectional and multivariable MR was performed analyzing associations between brain macrostructure and alcohol use. Downstream transcriptome-wide association studies (TWAS) and cell-type enrichment analyses investigated the biology underlying identified associations. The study approach was data driven and did not test any a priori hypotheses. Data were analyzed August 2021 to May 2022. Main Outcomes and Measures Brain structure phenotypes (global cortical thickness [GCT] and global cortical surface area [GCSA] in 33 709 individuals and left-right subcortical volumes in 19 629 individuals) and alcohol use behaviors (alcoholic drinks per week [DPW] in 537 349 individuals, binge drinking frequency in 143 685 individuals, and alcohol use disorder in 8845 individuals vs 20 657 control individuals [total of 29 502]). Results The main bidirectional MR analyses were performed in samples totaling 763 874 individuals, among whom more than 94% were of European ancestry, 52% to 54% were female, and the mean cohort ages were 40 to 63 years. Negative associations were identified between genetically predicted GCT and binge drinking (β, -2.52; 95% CI, -4.13 to -0.91) and DPW (β, -0.88; 95% CI, -1.37 to -0.40) at a false discovery rate (FDR) of 0.05. These associations remained significant in multivariable MR models that accounted for neuropsychiatric phenotypes, substance use, trauma, and neurodegeneration. TWAS of GCT and alcohol use behaviors identified 5 genes at the 17q21.31 locus oppositely associated with GCT and binge drinking or DPW (FDR = 0.05). Cell-type enrichment analyses implicated glutamatergic cortical neurons in alcohol use behaviors. Conclusions and Relevance The findings in this study show that the associations between GCT and alcohol use may reflect a predispositional influence of GCT and that 17q21.31 genes and glutamatergic cortical neurons may play a role in this association. While replication studies are needed, these findings should enhance the understanding of associations between brain structure and alcohol use.
Collapse
Affiliation(s)
- Lucas A. Mavromatis
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Daniel B. Rosoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
- National Institutes of Health–Oxford-Cambridge Scholars Program; Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Renata B. Cupertino
- Department of Psychiatry, University of Vermont College of Medicine, Burlington
| | - Hugh Garavan
- Department of Psychiatry, University of Vermont College of Medicine, Burlington
| | - Scott Mackey
- Department of Psychiatry, University of Vermont College of Medicine, Burlington
| | - Falk W. Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
80
|
Qi S, Sui J, Pearlson G, Bustillo J, Perrone-Bizzozero NI, Kochunov P, Turner JA, Fu Z, Shao W, Jiang R, Yang X, Liu J, Du Y, Chen J, Zhang D, Calhoun VD. Derivation and utility of schizophrenia polygenic risk associated multimodal MRI frontotemporal network. Nat Commun 2022; 13:4929. [PMID: 35995794 PMCID: PMC9395379 DOI: 10.1038/s41467-022-32513-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 08/03/2022] [Indexed: 12/23/2022] Open
Abstract
Schizophrenia is a highly heritable psychiatric disorder characterized by widespread functional and structural brain abnormalities. However, previous association studies between MRI and polygenic risk were mostly ROI-based single modality analyses, rather than identifying brain-based multimodal predictive biomarkers. Based on schizophrenia polygenic risk scores (PRS) from healthy white people within the UK Biobank dataset (N = 22,459), we discovered a robust PRS-associated brain pattern with smaller gray matter volume and decreased functional activation in frontotemporal cortex, which distinguished schizophrenia from controls with >83% accuracy, and predicted cognition and symptoms across 4 independent schizophrenia cohorts. Further multi-disease comparisons demonstrated that these identified frontotemporal alterations were most severe in schizophrenia and schizo-affective patients, milder in bipolar disorder, and indistinguishable from controls in autism, depression and attention-deficit hyperactivity disorder. These findings indicate the potential of the identified PRS-associated multimodal frontotemporal network to serve as a trans-diagnostic gene intermediated brain biomarker specific to schizophrenia.
Collapse
Affiliation(s)
- Shile Qi
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Jing Sui
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China.
| | - Godfrey Pearlson
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Juan Bustillo
- Departments of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Nora I Perrone-Bizzozero
- Departments of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Peter Kochunov
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jessica A Turner
- Department of Psychology, Georgia State University, Atlanta, GA, USA
| | - Zening Fu
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), [Georgia State University, Georgia Institute of Technology, Emory University], Atlanta, GA, USA
| | - Wei Shao
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Rongtao Jiang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Xiao Yang
- Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Jingyu Liu
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), [Georgia State University, Georgia Institute of Technology, Emory University], Atlanta, GA, USA
| | - Yuhui Du
- School of Computer & Information Technology, Shanxi University, Taiyuan, China
| | - Jiayu Chen
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), [Georgia State University, Georgia Institute of Technology, Emory University], Atlanta, GA, USA.
| | - Daoqiang Zhang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Vince D Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), [Georgia State University, Georgia Institute of Technology, Emory University], Atlanta, GA, USA
| |
Collapse
|
81
|
Makowski C, Wang H, Chen CH. Clinical opportunity awaits at the intersection of genomics and brain imaging. J Psychiatry Neurosci 2022; 47:E293-E298. [PMID: 35948342 PMCID: PMC9377545 DOI: 10.1503/jpn.220075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
| | | | - Chi-Hua Chen
- From the Center for Multimodal Imaging and Genetics, Department of Radiology, University of California San Diego, San Diego, Cali., USA
| |
Collapse
|
82
|
Wingo TS, Liu Y, Gerasimov ES, Vattathil SM, Wynne ME, Liu J, Lori A, Faundez V, Bennett DA, Seyfried NT, Levey AI, Wingo AP. Shared mechanisms across the major psychiatric and neurodegenerative diseases. Nat Commun 2022; 13:4314. [PMID: 35882878 PMCID: PMC9325708 DOI: 10.1038/s41467-022-31873-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 07/07/2022] [Indexed: 12/14/2022] Open
Abstract
Several common psychiatric and neurodegenerative diseases share epidemiologic risk; however, whether they share pathophysiology is unclear and is the focus of our investigation. Using 25 GWAS results and LD score regression, we find eight significant genetic correlations between psychiatric and neurodegenerative diseases. We integrate the GWAS results with human brain transcriptomes (n = 888) and proteomes (n = 722) to identify cis- and trans- transcripts and proteins that are consistent with a pleiotropic or causal role in each disease, referred to as causal proteins for brevity. Within each disease group, we find many distinct and shared causal proteins. Remarkably, 30% (13 of 42) of the neurodegenerative disease causal proteins are shared with psychiatric disorders. Furthermore, we find 2.6-fold more protein-protein interactions among the psychiatric and neurodegenerative causal proteins than expected by chance. Together, our findings suggest these psychiatric and neurodegenerative diseases have shared genetic and molecular pathophysiology, which has important ramifications for early treatment and therapeutic development.
Collapse
Affiliation(s)
- Thomas S Wingo
- Goizueta Alzheimer's Disease Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| | - Yue Liu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Selina M Vattathil
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Meghan E Wynne
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jiaqi Liu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Adriana Lori
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA
| | - Victor Faundez
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Goizueta Alzheimer's Disease Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Aliza P Wingo
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA.
- Veterans Affairs Atlanta Health Care System, Decatur, GA, USA.
| |
Collapse
|
83
|
Kirchler M, Konigorski S, Norden M, Meltendorf C, Kloft M, Schurmann C, Lippert C. transferGWAS: GWAS of images using deep transfer learning. Bioinformatics 2022; 38:3621-3628. [PMID: 35640976 DOI: 10.1093/bioinformatics/btac369] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/05/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Medical images can provide rich information about diseases and their biology. However, investigating their association with genetic variation requires non-standard methods. We propose transferGWAS, a novel approach to perform genome-wide association studies directly on full medical images. First, we learn semantically meaningful representations of the images based on a transfer learning task, during which a deep neural network is trained on independent but similar data. Then, we perform genetic association tests with these representations. RESULTS We validate the type I error rates and power of transferGWAS in simulation studies of synthetic images. Then we apply transferGWAS in a genome-wide association study of retinal fundus images from the UK Biobank. This first-of-a-kind GWAS of full imaging data yielded 60 genomic regions associated with retinal fundus images, of which 7 are novel candidate loci for eye-related traits and diseases. AVAILABILITY AND IMPLEMENTATION Our method is implemented in Python and available at https://github.com/mkirchler/transferGWAS/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Matthias Kirchler
- Digital Health-Machine Learning Research Group, Digital Health Center, Hasso Plattner Institute, University of Potsdam, 14482 Potsdam, Germany.,Department of Computer Science, TU Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Stefan Konigorski
- Digital Health-Machine Learning Research Group, Digital Health Center, Hasso Plattner Institute, University of Potsdam, 14482 Potsdam, Germany.,Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Matthias Norden
- Digital Health & Personalized Medicine Research Group, Digital Health Center, Hasso Plattner Institute, University of Potsdam, 14482 Potsdam, Germany.,Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Christian Meltendorf
- Department of Electrical Engineering - Mechatronics - Optometry, Beuth University of Applied Sciences Berlin, 13353 Berlin, Germany
| | - Marius Kloft
- Department of Computer Science, TU Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Claudia Schurmann
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Digital Health & Personalized Medicine Research Group, Digital Health Center, Hasso Plattner Institute, University of Potsdam, 14482 Potsdam, Germany
| | - Christoph Lippert
- Digital Health-Machine Learning Research Group, Digital Health Center, Hasso Plattner Institute, University of Potsdam, 14482 Potsdam, Germany.,Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
84
|
Li CS, Chen Y, Ide JS. Gray matter volumetric correlates of attention deficit and hyperactivity traits in emerging adolescents. Sci Rep 2022; 12:11367. [PMID: 35790754 PMCID: PMC9256746 DOI: 10.1038/s41598-022-15124-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 06/20/2022] [Indexed: 11/08/2022] Open
Abstract
Previous research has demonstrated reduction in cortical and subcortical, including basal ganglia (BG), gray matter volumes (GMV) in individuals with attention deficit hyperactivity disorder (ADHD), a neurodevelopmental condition that is more prevalent in males than in females. However, the volumetric deficits vary across studies. Whether volumetric reductions are more significant in males than females; to what extent these neural markers are heritable and relate to cognitive dysfunction in ADHD remain unclear. To address these questions, we followed published routines and performed voxel-based morphometry analysis of a data set (n = 11,502; 5,464 girls, 9-10 years) curated from the Adolescent Brain Cognition Development project, a population-based study of typically developing children. Of the sample, 634 and 2,826 were identified as monozygotic twins and dizygotic twins/siblings, respectively. In linear regressions, a cluster in the hypothalamus showed larger GMV, and bilateral caudate and putamen, lateral orbitofrontal and occipital cortex showed smaller GMVs, in correlation with higher ADHD scores in girls and boys combined. When examined separately, boys relative to girls showed more widespread (including BG) and stronger associations between GMV deficits and ADHD scores. ADHD traits and the volumetric correlates demonstrated heritability estimates (a2) between 0.59 and 0.79, replicating prior findings of the genetic basis of ADHD. Further, ADHD traits and the volumetric correlates (except for the hypothalamus) were each negatively and positively correlated with N-back performance. Together, these findings confirm volumetric deficits in children with more prominent ADHD traits. Highly heritable in both girls and boys and potentially more significant in boys than in girls, the structural deficits underlie diminished capacity in working memory and potentially other cognitive deficits in ADHD.
Collapse
Affiliation(s)
- Clara S Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
- Smith College, Northampton, MA, 06492, USA
| | - Yu Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA.
| | - Jaime S Ide
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA.
| |
Collapse
|
85
|
Wang Z, Rodriguez‐Moreno DV, Cycowicz YM, Amsel LV, Cheslack‐Postava K, He X, Ryan M, Geronazzo‐Alman L, Musa GJ, Bisaga A, Hoven CW. Shapes of subcortical structures in adolescents with and without familial history of substance use disorder. Hum Brain Mapp 2022; 43:2759-2770. [PMID: 35393707 PMCID: PMC9120549 DOI: 10.1002/hbm.25804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/09/2022] Open
Abstract
It has been suggested that intergenerational transmission of risk for substance use disorder (SUD) manifests in the brain anatomy of substance naïve adolescents. While volume and shapes of subcortical structures (SSS) have been shown to be heritable, these structures, especially the pallidum, putamen, nucleus accumbens, and hippocampus, have also been associated with substance use disorders. However, it is not clear if those anatomical differences precede substance use or are the result of that use. Therefore, we examined if volume and SSS of adolescents with a family history (FH+) of SUD differed from adolescents without such a history (FH-). Because risk for SUD is associated with anxiety and impulsivity, we also examined correlations between these psychological characteristics and volume/SSS. Using structural MRI and FSL software, we segmented subcortical structures and obtained indices of SSS and volumes of 64 FH+ and 58 FH- adolescents. We examined group differences in volume and SSS, and the correlations between volume/SSS and trait anxiety and impulsivity. FH+ adolescents had a significant inward deformation in the shape of the right anterior hippocampus compared to FH- adolescents, while the volume of this structure did not differ between groups. Neither shape nor volume of the other subcortical structures differed between groups. In the FH+ adolescents, the left hippocampus shape was positively correlated with both trait anxiety and impulsivity, while in FH- adolescents a negative correlation pattern of SSS was seen in the hippocampus. SSS appears to capture local anatomical features that traditional volumetric analysis does not. The inward shape deformation in the right anterior hippocampus in FH+ adolescents may be related to the known increased risk for behavioral dysregulation leading to SUD in FH+ offspring. Hippocampus shape also exhibits opposite patterns of correlation with anxiety and impulsivity scores across the FH+ and FH- adolescents. These novel findings may reveal neural correlates, not captured by traditional volumetric analysis, of familial transmission of increased vulnerability to SUD.
Collapse
Affiliation(s)
- Zhishun Wang
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
| | - Diana V. Rodriguez‐Moreno
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
| | - Yael M. Cycowicz
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
| | - Lawrence V. Amsel
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
| | - Keely Cheslack‐Postava
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
| | - Xiaofu He
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
| | - Megan Ryan
- New York State Psychiatric InstituteNew YorkNew YorkUSA
| | - Lupo Geronazzo‐Alman
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
| | - George J. Musa
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
- Department of EpidemiologyMailman School of Public health, Columbia UniversityNew YorkNew YorkUSA
| | - Adam Bisaga
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
| | - Christina W. Hoven
- The Department of PsychiatryVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkNew YorkUSA
- New York State Psychiatric InstituteNew YorkNew YorkUSA
- Department of EpidemiologyMailman School of Public health, Columbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
86
|
Luo X, Fang W, Lin X, Guo X, Chen Y, Tan Y, Wang L, Jing X, Wang X, Zhang Y, Yu T, Ide J, Cao Y, Yang L, Li CSR. Sex-different interrelationships of rs945270, cerebral gray matter volumes, and attention deficit hyperactivity disorder: a region-wide study across brain. Transl Psychiatry 2022; 12:225. [PMID: 35654767 PMCID: PMC9163172 DOI: 10.1038/s41398-022-02007-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/28/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022] Open
Abstract
Previous genome-wide association studies (GWAS) reported that the allele C of rs945270 of the kinectin 1 gene (KTN1) most significantly increased the gray matter volume (GMV) of the putamen and modestly regulated the risk for attention deficit hyperactivity disorder (ADHD). On the other hand, ADHD is known to be associated with a reduction in subcortical and cortical GMVs. Here, we examined the interrelationships of the GMVs, rs945270 alleles, and ADHD symptom scores in the same cohort of children. With data of rs945270 genotypes, GMVs of 118 brain regions, and ADHD symptom scores of 3372 boys and 3129 girls of the Adolescent Brain Cognition Development project, we employed linear regression analyses to examine the pairwise correlations adjusted for the third of the three traits and other relevant covariates, and examine their mediation effects. We found that the major allele C of rs945270 modestly increased risk for ADHD in males only when controlling for the confounding effects of the GMV of any one of the 118 cerebral regions (0.026 ≤ p ≤ 0.059: Top two: left and right putamen). This allele also significantly increased putamen GMV in males alone (left p = 2.8 × 10-5, and right p = 9.4 × 10-5; α = 2.1 × 10-4) and modestly increased other subcortical and cortical GMVs in both sexes (α < p < 0.05), whether or not adjusted for ADHD symptom scores. Both subcortical and cortical GMVs were significantly or suggestively reduced in ADHD when adjusted for rs945270 alleles, each more significantly in females (3.6 × 10-7 ≤ p < α; Top two: left pallidum and putamen) and males (3.5 × 10-6 ≤ p < α), respectively. Finally, the left and right putamen GMVs reduced 14.0% and 11.7% of the risk effects of allele C on ADHD, and allele C strengthened 4.5% (left) and 12.2% (right) of the protective effects of putamen GMVs on ADHD risk, respectively. We concluded that the rs945270-GMVs-ADHD relationships were sex-different. In males, the major allele C of rs945270 increased risk for ADHD, which was compromised by putamen GMVs; this allele also but only significantly increased putamen GMVs that then significantly protected against ADHD risk. In females, the top two GMVs significantly decreasing ADHD risk were left pallidum and putamen GMVs. Basal ganglia the left putamen in particular play the most critical role in the pathogenesis of ADHD.
Collapse
Affiliation(s)
- Xingguang Luo
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical School of Medicine, Beijing, 100096, China
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Wenhua Fang
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xiaoyun Guo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
- Shanghai Mental Health Center, Shanghai, 200030, China
| | - Yu Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Yunlong Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical School of Medicine, Beijing, 100096, China
| | - Leilei Wang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical School of Medicine, Beijing, 100096, China
| | - Xiaozhong Jing
- Department of Neurology, Shanghai Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Xiaoping Wang
- Department of Neurology, Shanghai Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin, 300222, China
| | - Ting Yu
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical School of Medicine, Beijing, 100096, China
| | - Jaime Ide
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Yuping Cao
- Department of Psychiatry, the Second Xiangya Hospital, Central South University; The China National Clinical Research Center for Mental Health Disorders; National Technology Institute of Psychiatry; Key Laboratory of Psychiatry and Mental Health of Hunan Province, Changsha, 410017, China.
| | - Lingli Yang
- Department of Neurology, Shanghai Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
- Wu Tsai Institute, Yale University, New Haven, CT, 06510, USA
| |
Collapse
|
87
|
Le Grand Q, Satizabal CL, Sargurupremraj M, Mishra A, Soumaré A, Laurent A, Crivello F, Tsuchida A, Shin J, Macalli M, Singh B, Beiser AS, DeCarli C, Fletcher E, Paus T, Lathrop M, Adams HHH, Bis JC, Seshadri S, Tzourio C, Mazoyer B, Debette S. Genomic Studies Across the Lifespan Point to Early Mechanisms Determining Subcortical Volumes. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:616-628. [PMID: 34700051 PMCID: PMC9395126 DOI: 10.1016/j.bpsc.2021.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/28/2021] [Accepted: 10/14/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Subcortical brain structures play a key role in pathological processes of age-related neurodegenerative disorders. Mounting evidence also suggests that early-life factors may have an impact on the development of common late-life neurological diseases, including genetic factors that can influence both brain maturation and neurodegeneration. METHODS Using large population-based brain imaging datasets across the lifespan (N ≤ 40,628), we aimed to 1) estimate the heritability of subcortical volumes in young (18-35 years), middle (35-65 years), and older (65+ years) age, and their genetic correlation across age groups; 2) identify whether genetic loci associated with subcortical volumes in older persons also show associations in early adulthood, and explore underlying genes using transcriptome-wide association studies; and 3) explore their association with neurological phenotypes. RESULTS Heritability of subcortical volumes consistently decreased with increasing age. Genetic risk scores for smaller caudate nucleus, putamen, and hippocampus volume in older adults were associated with smaller volumes in young adults. Individually, 10 loci associated with subcortical volumes in older adults also showed associations in young adults. Within these loci, transcriptome-wide association studies showed that expression of several genes in brain tissues (especially MYLK2 and TUFM) was associated with subcortical volumes in both age groups. One risk variant for smaller caudate nucleus volume (TUFM locus) was associated with lower cognitive performance. Genetically predicted Alzheimer's disease was associated with smaller subcortical volumes in middle and older age. CONCLUSIONS Our findings provide novel insights into the genetic determinants of subcortical volumes across the lifespan. More studies are needed to decipher the underlying biology and clinical impact.
Collapse
Affiliation(s)
- Quentin Le Grand
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, Texas; Department of Population Health Sciences, UT Health San Antonio, San Antonio, Texas; Framingham Heart Study, Framingham, Massachusetts; Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Muralidharan Sargurupremraj
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, Texas
| | - Aniket Mishra
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France
| | - Aicha Soumaré
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France
| | - Alexandre Laurent
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France
| | - Fabrice Crivello
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France
| | - Ami Tsuchida
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France
| | - Jean Shin
- Department of Physiology, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Department of Nutritional Sciences, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Mélissa Macalli
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France
| | - Baljeet Singh
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California Davis, Davis, California
| | - Alexa S Beiser
- Framingham Heart Study, Framingham, Massachusetts; Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Charles DeCarli
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California Davis, Davis, California
| | - Evan Fletcher
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California Davis, Davis, California
| | - Tomas Paus
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Psychology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, Centre Hospitalier Universitaire Sainte-Justine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Mark Lathrop
- McGill Genome Center, McGill University, Montreal, Quebec, Canada
| | - Hieab H H Adams
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, Texas; Department of Population Health Sciences, UT Health San Antonio, San Antonio, Texas; Framingham Heart Study, Framingham, Massachusetts; Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Christophe Tzourio
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France; Bordeaux University Hospital, Department of Medical Informatics, Bordeaux, France
| | - Bernard Mazoyer
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional imaging group, Bordeaux, France; Bordeaux University Hospital, Department of Neuroradiology, Bordeaux, France
| | - Stéphanie Debette
- University of Bordeaux, INSERM, Bordeaux Population Health Center, UMR1219, Bordeaux, France; Bordeaux University Hospital, Department of Neurology, Institute of Neurodegenerative Diseases, Bordeaux, France.
| |
Collapse
|
88
|
Rubbi L, Zhang H, Feng J, He C, Kurnia P, Ratan P, Tammana A, House S, Thompson M, Farrell C, Snir S, Stahler D, Ostrander EA, vonHoldt BM, Pellegrini M. The effects of age, sex, weight, and breed on canid methylomes. Epigenetics 2022; 17:1497-1512. [PMID: 35502722 PMCID: PMC9586589 DOI: 10.1080/15592294.2022.2069385] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Unlike genomes, which are static throughout the lifespan of an organism, DNA methylomes are dynamic. To study these dynamics, we developed quantitative models that measure the effect of multiple factors on DNA methylomes including, age, sex, weight, and genetics. We conducted our study in canids, which prove to be an ideal species to assess epigenetic moderators due to their extreme variability in size and well-characterized genetic structure. We collected buccal swabs from 217 canids (207 domestic dogs and 10 grey wolves) and used targeted bisulphite sequencing to measure methylomes. We also measured genotypes at over one thousand single nucleotide polymorphisms (SNPs). As expected, we found that DNA methylomes are strongly associated with age, enabling the construction of epigenetic clocks. However, we also identify novel associations between methylomes and sex, weight, and sterilization status, leading to accurate models that predict these factors. Methylomes are also affected by genetics, and we observe multiple associations between SNP loci and methylated CpGs. Finally, we show that several factors moderate the relationship between epigenetic ages and real ages, such as body weight, which increases epigenetic ageing. In conclusion, we demonstrate that the plasticity of DNA methylomes is impacted by myriad genetics and physiological factors, and that DNA methylation biomarkers are accurate predictors of age, sex and sterilization status.
Collapse
Affiliation(s)
- Liudmilla Rubbi
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Haoxuan Zhang
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Junxi Feng
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Christopher He
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Patrick Kurnia
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Prashansa Ratan
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Aakash Tammana
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Sabina House
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael Thompson
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Colin Farrell
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Sagi Snir
- Department Evolutionary and Environmental Biology, University of Haifa, Israel
| | - Daniel Stahler
- Yellowstone Center for Resources, Yellowstone National Park, Wyo, USA
| | - Elaine A Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, CA, USA
| | - Bridgett M vonHoldt
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA
| | - Matteo Pellegrini
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
89
|
Fan CC, Loughnan R, Makowski C, Pecheva D, Chen CH, Hagler DJ, Thompson WK, Parker N, van der Meer D, Frei O, Andreassen OA, Dale AM. Multivariate genome-wide association study on tissue-sensitive diffusion metrics highlights pathways that shape the human brain. Nat Commun 2022; 13:2423. [PMID: 35505052 PMCID: PMC9065144 DOI: 10.1038/s41467-022-30110-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 04/12/2022] [Indexed: 11/12/2022] Open
Abstract
The molecular determinants of tissue composition of the human brain remain largely unknown. Recent genome-wide association studies (GWAS) on this topic have had limited success due to methodological constraints. Here, we apply advanced whole-brain analyses on multi-shell diffusion imaging data and multivariate GWAS to two large scale imaging genetic datasets (UK Biobank and the Adolescent Brain Cognitive Development study) to identify and validate genetic association signals. We discover 503 unique genetic loci that have impact on multiple regions of human brain. Among them, more than 79% are validated in either of two large-scale independent imaging datasets. Key molecular pathways involved in axonal growth, astrocyte-mediated neuroinflammation, and synaptogenesis during development are found to significantly impact the measured variations in tissue-specific imaging features. Our results shed new light on the biological determinants of brain tissue composition and their potential overlap with the genetic basis of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Chun Chieh Fan
- Population Neuroscience and Genetics Lab, University of California, San Diego, La Jolla, CA, USA. .,Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, USA. .,Department of Radiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA.
| | - Robert Loughnan
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA
| | - Carolina Makowski
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, USA.,Department of Radiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Diliana Pecheva
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, USA.,Department of Radiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Chi-Hua Chen
- Department of Radiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Donald J Hagler
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, USA.,Department of Radiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Wesley K Thompson
- Population Neuroscience and Genetics Lab, University of California, San Diego, La Jolla, CA, USA.,Department of Radiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Nadine Parker
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dennis van der Meer
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Oleksandr Frei
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Centre for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, USA.,Department of Radiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA.,Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA.,Department of Neuroscience, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
90
|
Cheon EJ, Bearden CE, Sun D, Ching CRK, Andreassen OA, Schmaal L, Veltman DJ, Thomopoulos SI, Kochunov P, Jahanshad N, Thompson PM, Turner JA, van Erp TG. Cross disorder comparisons of brain structure in schizophrenia, bipolar disorder, major depressive disorder, and 22q11.2 deletion syndrome: A review of ENIGMA findings. Psychiatry Clin Neurosci 2022; 76:140-161. [PMID: 35119167 PMCID: PMC9098675 DOI: 10.1111/pcn.13337] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/29/2021] [Accepted: 01/21/2022] [Indexed: 12/25/2022]
Abstract
This review compares the main brain abnormalities in schizophrenia (SZ), bipolar disorder (BD), major depressive disorder (MDD), and 22q11.2 Deletion Syndrome (22q11DS) determined by ENIGMA (Enhancing Neuro Imaging Genetics through Meta Analysis) consortium investigations. We obtained ranked effect sizes for subcortical volumes, regional cortical thickness, cortical surface area, and diffusion tensor imaging abnormalities, comparing each of these disorders relative to healthy controls. In addition, the studies report on significant associations between brain imaging metrics and disorder-related factors such as symptom severity and treatments. Visual comparison of effect size profiles shows that effect sizes are generally in the same direction and scale in severity with the disorders (in the order SZ > BD > MDD). The effect sizes for 22q11DS, a rare genetic syndrome that increases the risk for psychiatric disorders, appear to be much larger than for either of the complex psychiatric disorders. This is consistent with the idea of generally larger effects on the brain of rare compared to common genetic variants. Cortical thickness and surface area effect sizes for 22q11DS with psychosis compared to 22q11DS without psychosis are more similar to those of SZ and BD than those of MDD; a pattern not observed for subcortical brain structures and fractional anisotropy effect sizes. The observed similarities in effect size profiles for cortical measures across the psychiatric disorders mimic those observed for shared genetic variance between these disorders reported based on family and genetic studies and are consistent with shared genetic risk for SZ and BD and structural brain phenotypes.
Collapse
Affiliation(s)
- Eun-Jin Cheon
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, 5251 California Ave, Irvine, CA, 92617, USA
- Department of Psychiatry, Yeungnam University College of Medicine, Yeungnam University Medical Center, Daegu, Republic of Korea
| | - Carrie E. Bearden
- Departments of Psychiatry and Biobehavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles
| | - Daqiang Sun
- Departments of Psychiatry and Biobehavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles
- Department of Mental Health, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Christopher R. K. Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ole A. Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Lianne Schmaal
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Australia
- Orygen, Parkville, Australia
| | - Dick J. Veltman
- Department of Psychiatry, Amsterdam UMC, location VUMC, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Sophia I. Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jessica A. Turner
- Psychology Department and Neuroscience Institute, Georgia State University, Atlant, GA, USA
| | - Theo G.M. van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, 5251 California Ave, Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, 309 Qureshey Research Lab, Irvine, CA, 92697, USA
| |
Collapse
|
91
|
Gillespie NA, Hatton SN, Hagler DJ, Dale AM, Elman JA, McEvoy LK, Eyler LT, Fennema-Notestine C, Logue MW, McKenzie RE, Puckett OK, Tu XM, Whitsel N, Xian H, Reynolds CA, Panizzon MS, Lyons MJ, Neale MC, Kremen WS, Franz C. The Impact of Genes and Environment on Brain Ageing in Males Aged 51 to 72 Years. Front Aging Neurosci 2022; 14:831002. [PMID: 35493948 PMCID: PMC9051484 DOI: 10.3389/fnagi.2022.831002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/15/2022] [Indexed: 01/27/2023] Open
Abstract
Magnetic resonance imaging data are being used in statistical models to predicted brain ageing (PBA) and as biomarkers for neurodegenerative diseases such as Alzheimer's Disease. Despite their increasing application, the genetic and environmental etiology of global PBA indices is unknown. Likewise, the degree to which genetic influences in PBA are longitudinally stable and how PBA changes over time are also unknown. We analyzed data from 734 men from the Vietnam Era Twin Study of Aging with repeated MRI assessments between the ages 51-72 years. Biometrical genetic analyses "twin models" revealed significant and highly correlated estimates of additive genetic heritability ranging from 59 to 75%. Multivariate longitudinal modeling revealed that covariation between PBA at different timepoints could be explained by a single latent factor with 73% heritability. Our results suggest that genetic influences on PBA are detectable in midlife or earlier, are longitudinally very stable, and are largely explained by common genetic influences.
Collapse
Affiliation(s)
- Nathan A. Gillespie
- Virginia Institute for Psychiatric and Behaviour Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, United States,QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia,*Correspondence: Nathan A. Gillespie,
| | - Sean N. Hatton
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States,Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, United States,Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Donald J. Hagler
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
| | - Anders M. Dale
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States,Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, United States,Halıcıoğlu Data Science Institute, University of California, San Diego, La Jolla, CA, United States
| | - Jeremy A. Elman
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States,Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, United States
| | - Linda K. McEvoy
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, United States
| | - Lisa T. Eyler
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States,Mental Illness Research Education and Clinical Center, VA San Diego Healthcare System, San Diego, CA, United States
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States,Department of Radiology, University of California, San Diego, La Jolla, CA, United States
| | - Mark W. Logue
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, United States,Department of Psychiatry and Biomedical Genetics Section, Boston University School of Medicine, Boston, MA, United States,Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - Ruth E. McKenzie
- Department of Psychology, Boston University, Boston, MA, United States,School of Education and Social Policy, Merrimack College, North Andover, MA, United States
| | - Olivia K. Puckett
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States,Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, United States
| | - Xin M. Tu
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, United States,Division of Biostatistics and Bioinformatics, Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, United States
| | - Nathan Whitsel
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States,Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, United States
| | - Hong Xian
- Department of Epidemiology and Biostatistics, Saint. Louis University, St. Louis, MO, United States,Research Service, VA St. Louis Healthcare System, St. Louis, MO, United States
| | - Chandra A. Reynolds
- Department of Psychology, University of California, Riverside, Riverside, CA, United States
| | - Matthew S. Panizzon
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States,Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, United States
| | - Michael J. Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, United States
| | - Michael C. Neale
- Virginia Institute for Psychiatric and Behaviour Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, United States,Department of Biological Psychology, Free University of Amsterdam, Amsterdam, Netherlands
| | - William S. Kremen
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States,Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, United States,Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, La Jolla, CA, United States,William S. Kremen,
| | - Carol Franz
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States,Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, United States,Carol Franz,
| |
Collapse
|
92
|
Genetic and environment effects on structural neuroimaging endophenotype for bipolar disorder: a novel molecular approach. Transl Psychiatry 2022; 12:137. [PMID: 35379780 PMCID: PMC8980067 DOI: 10.1038/s41398-022-01892-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 12/15/2022] Open
Abstract
We investigated gene-environment effects on structural brain endophenotype in bipolar disorder (BD) using a novel method of combining polygenic risk scores with epigenetic signatures since traditional methods of examining the family history and trauma effects have significant limitations. The study enrolled 119 subjects, including 55 BD spectrum (BDS) subjects diagnosed with BD or major depressive disorder (MDD) with subthreshold BD symptoms and 64 non-BDS subjects comprising 32 MDD subjects without BD symptoms and 32 healthy subjects. The blood samples underwent genome-wide genotyping and methylation quantification. We derived polygenic risk score (PRS) and methylation profile score (MPS) as weighted summations of risk single nucleotide polymorphisms and methylation probes, respectively, which were considered as molecular measures of genetic and environmental risks for BD. Linear regression was used to relate PRS, MPS, and their interaction to 44 brain structure measures quantified from magnetic resonance imaging (MRI) on 47 BDS subjects, and the results were compared with those based on family history and childhood trauma. After multiplicity corrections using false discovery rate (FDR), MPS was found to be negatively associated with the volume of the medial geniculate thalamus (FDR = 0.059, partial R2 = 0.208). Family history, trauma scale, and PRS were not associated with any brain measures. PRS and MPS show significant interactions on whole putamen (FDR = 0.09, partial R2 = 0.337). No significant gene-environment interactions were identified for the family history and trauma scale. PRS and MPS generally explained greater proportions of variances of the brain measures (range of partial R2 = [0.008, 0.337]) than the clinical risk factors (range = [0.004, 0.228]).
Collapse
|
93
|
Brouwer RM, Klein M, Grasby KL, Schnack HG, Jahanshad N, Teeuw J, Thomopoulos SI, Sprooten E, Franz CE, Gogtay N, Kremen WS, Panizzon MS, Olde Loohuis LM, Whelan CD, Aghajani M, Alloza C, Alnæs D, Artiges E, Ayesa-Arriola R, Barker GJ, Bastin ME, Blok E, Bøen E, Breukelaar IA, Bright JK, Buimer EEL, Bülow R, Cannon DM, Ciufolini S, Crossley NA, Damatac CG, Dazzan P, de Mol CL, de Zwarte SMC, Desrivières S, Díaz-Caneja CM, Doan NT, Dohm K, Fröhner JH, Goltermann J, Grigis A, Grotegerd D, Han LKM, Harris MA, Hartman CA, Heany SJ, Heindel W, Heslenfeld DJ, Hohmann S, Ittermann B, Jansen PR, Janssen J, Jia T, Jiang J, Jockwitz C, Karali T, Keeser D, Koevoets MGJC, Lenroot RK, Malchow B, Mandl RCW, Medel V, Meinert S, Morgan CA, Mühleisen TW, Nabulsi L, Opel N, de la Foz VOG, Overs BJ, Paillère Martinot ML, Redlich R, Marques TR, Repple J, Roberts G, Roshchupkin GV, Setiaman N, Shumskaya E, Stein F, Sudre G, Takahashi S, Thalamuthu A, Tordesillas-Gutiérrez D, van der Lugt A, van Haren NEM, Wardlaw JM, Wen W, Westeneng HJ, Wittfeld K, Zhu AH, Zugman A, Armstrong NJ, Bonfiglio G, Bralten J, Dalvie S, Davies G, Di Forti M, Ding L, Donohoe G, Forstner AJ, Gonzalez-Peñas J, Guimaraes JPOFT, Homuth G, Hottenga JJ, Knol MJ, Kwok JBJ, Le Hellard S, Mather KA, Milaneschi Y, Morris DW, Nöthen MM, Papiol S, Rietschel M, Santoro ML, Steen VM, Stein JL, Streit F, Tankard RM, Teumer A, van 't Ent D, van der Meer D, van Eijk KR, Vassos E, Vázquez-Bourgon J, Witt SH, Adams HHH, Agartz I, Ames D, Amunts K, Andreassen OA, Arango C, Banaschewski T, Baune BT, Belangero SI, Bokde ALW, Boomsma DI, Bressan RA, Brodaty H, Buitelaar JK, Cahn W, Caspers S, Cichon S, Crespo-Facorro B, Cox SR, Dannlowski U, Elvsåshagen T, Espeseth T, Falkai PG, Fisher SE, Flor H, Fullerton JM, Garavan H, Gowland PA, Grabe HJ, Hahn T, Heinz A, Hillegers M, Hoare J, Hoekstra PJ, Ikram MA, Jackowski AP, Jansen A, Jönsson EG, Kahn RS, Kircher T, Korgaonkar MS, Krug A, Lemaitre H, Malt UF, Martinot JL, McDonald C, Mitchell PB, Muetzel RL, Murray RM, Nees F, Nenadić I, Oosterlaan J, Ophoff RA, Pan PM, Penninx BWJH, Poustka L, Sachdev PS, Salum GA, Schofield PR, Schumann G, Shaw P, Sim K, Smolka MN, Stein DJ, Trollor JN, van den Berg LH, Veldink JH, Walter H, Westlye LT, Whelan R, White T, Wright MJ, Medland SE, Franke B, Thompson PM, Hulshoff Pol HE. Genetic variants associated with longitudinal changes in brain structure across the lifespan. Nat Neurosci 2022; 25:421-432. [PMID: 35383335 PMCID: PMC10040206 DOI: 10.1038/s41593-022-01042-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/28/2022] [Indexed: 02/08/2023]
Abstract
Human brain structure changes throughout the lifespan. Altered brain growth or rates of decline are implicated in a vast range of psychiatric, developmental and neurodegenerative diseases. In this study, we identified common genetic variants that affect rates of brain growth or atrophy in what is, to our knowledge, the first genome-wide association meta-analysis of changes in brain morphology across the lifespan. Longitudinal magnetic resonance imaging data from 15,640 individuals were used to compute rates of change for 15 brain structures. The most robustly identified genes GPR139, DACH1 and APOE are associated with metabolic processes. We demonstrate global genetic overlap with depression, schizophrenia, cognitive functioning, insomnia, height, body mass index and smoking. Gene set findings implicate both early brain development and neurodegenerative processes in the rates of brain changes. Identifying variants involved in structural brain changes may help to determine biological pathways underlying optimal and dysfunctional brain development and aging.
Collapse
Affiliation(s)
- Rachel M Brouwer
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU Amsterdam, Amsterdam, The Netherlands.
| | - Marieke Klein
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Katrina L Grasby
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Hugo G Schnack
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Utrecht Institute of Linguistics OTS, Utrecht University, Utrecht, The Netherlands
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Jalmar Teeuw
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Emma Sprooten
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carol E Franz
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - Nitin Gogtay
- American Psychiatric Association, Washington, DC, USA
| | - William S Kremen
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| | - Matthew S Panizzon
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - Loes M Olde Loohuis
- Center for Neurobehavioral Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Moji Aghajani
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Institute of Education & Child Studies, Section Forensic Family & Youth Care, Leiden University, Leiden, The Netherlands
| | - Clara Alloza
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, IiSGM, CIBERSAM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Dag Alnæs
- NORMENT Centre, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Eric Artiges
- INSERM U1299 Trajectoires Développementales en Psychiatrie, Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, Université Paris Cité, CNRS UMR 9010; Centre Borelli, Gif-sur-Yvette, France
| | - Rosa Ayesa-Arriola
- Valdecilla Biomedical Research Institute (IDIVAL), Marqués de Valdecilla University Hospital (HUMV), School of Medicine, University of Cantabria, Santander, Spain
- CIBERSAM, Biomedical Research Network on Mental Health Area, Santander, Spain
- Universidad de Cantabria, Santander, Spain
| | - Gareth J Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Mark E Bastin
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Elisabet Blok
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Erlend Bøen
- Psychosomatic and CL Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Isabella A Breukelaar
- Brain Dynamics Centre, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Joanna K Bright
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Elizabeth E L Buimer
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Dara M Cannon
- Centre for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Simone Ciufolini
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Nicolas A Crossley
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christienne G Damatac
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Paola Dazzan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Casper L de Mol
- Department of Neurology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sonja M C de Zwarte
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Sylvane Desrivières
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Covadonga M Díaz-Caneja
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, IiSGM, CIBERSAM, School of Medicine, Universidad Complutense, Madrid, Spain
| | | | - Katharina Dohm
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Juliane H Fröhner
- Section of Systems Neuroscience, Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Antoine Grigis
- Université Paris-Saclay, CEA, Neurospin, Gif-sur-Yvette, France
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Laura K M Han
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Mathew A Harris
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Catharina A Hartman
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), Groningen, The Netherlands
| | - Sarah J Heany
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Walter Heindel
- Clinic for Radiology, University Hospital Münster, Münster, Germany
| | - Dirk J Heslenfeld
- Departments of Experimental and Clinical Psychology, Amsterdam, The Netherlands
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | | | - Philip R Jansen
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU Amsterdam, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Human Genetics, VUmc, Amsterdam UMC, Amsterdam, The Netherlands
| | - Joost Janssen
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, IiSGM, CIBERSAM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Tianye Jia
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Science and Technology for Brain-Inspired Intelligence and MoE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology and Neuroscience, SGDP Centre, King's College London, London, UK
| | - Jiyang Jiang
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia
| | - Christiane Jockwitz
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Temmuz Karali
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany
- NeuroImaging Core Unit Munich (NICUM), University Hospital LMU, Munich, Germany
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany
- NeuroImaging Core Unit Munich (NICUM), University Hospital LMU, Munich, Germany
- Munich Center for Neurosciences (MCN) - Brain & Mind, Planegg-Martinsried, Germany
| | - Martijn G J C Koevoets
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Rhoshel K Lenroot
- School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
- School of Psychiatry and Behavioral Sciences, School of Medicine, University of New Mexico, Albuquerque, NM, USA
- Neuroscience Research Australia, Sydney, NSW, Australia
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - René C W Mandl
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Vicente Medel
- Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Catherine A Morgan
- School of Psychology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand - Rangahau Roro Aotearoa, Auckland, New Zealand
| | - Thomas W Mühleisen
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Leila Nabulsi
- Centre for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Nils Opel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Víctor Ortiz-García de la Foz
- Valdecilla Biomedical Research Institute (IDIVAL), Marqués de Valdecilla University Hospital (HUMV), School of Medicine, University of Cantabria, Santander, Spain
- CIBERSAM, Biomedical Research Network on Mental Health Area, Santander, Spain
- Neuroimaging Unit, Technological Facilities, Valdecilla Biomedical Research Institute IDIVAL, Santander, Spain
| | | | - Marie-Laure Paillère Martinot
- INSERM U1299 Trajectoires Développementales en Psychiatrie, Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, Université Paris Cité, CNRS UMR 9010; Centre Borelli, Gif-sur-Yvette, France
- APHP, Sorbonne Université, Pitie-Salpetriere Hospital, Department of Child and Adolescent Psychiatry, Paris, France
| | - Ronny Redlich
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychology, University of Halle, Halle, Germany
| | - Tiago Reis Marques
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Imperial College London, London, UK
| | - Jonathan Repple
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Gloria Roberts
- School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Gennady V Roshchupkin
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nikita Setiaman
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Elena Shumskaya
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Gustavo Sudre
- Social and Behavioral Research Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Shun Takahashi
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia
| | - Diana Tordesillas-Gutiérrez
- Department of Radiology, IDIVAL, Marqués de Valdecilla University Hospital, Santander, Spain
- Advanced Computing and e-Science, Instituto de Física de Cantabria (UC-CSIC), Santander, Spain
| | - Aad van der Lugt
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Neeltje E M van Haren
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joanna M Wardlaw
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences and UK Dementia Research Institute Centre, University of Edinburgh, Edinburgh, UK
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia
| | - Henk-Jan Westeneng
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Katharina Wittfeld
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Alyssa H Zhu
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Andre Zugman
- Laboratory of Integrative Neuroscience (LiNC), Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents (INPD), CNPq, São Paulo, Brazil
| | | | - Gaia Bonfiglio
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU Amsterdam, Amsterdam, The Netherlands
| | - Janita Bralten
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Shareefa Dalvie
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Gail Davies
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Marta Di Forti
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Linda Ding
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Gary Donohoe
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
| | - Andreas J Forstner
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Centre for Human Genetics, Philipps-University Marburg, Marburg, Germany
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Javier Gonzalez-Peñas
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, IiSGM, CIBERSAM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Joao P O F T Guimaraes
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Jouke-Jan Hottenga
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Maria J Knol
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - John B J Kwok
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Stephanie Le Hellard
- NORMENT Centre of Excellence, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Karen A Mather
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia
- Neuroscience Research Australia, Sydney, NSW, Australia
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Derek W Morris
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Sergi Papiol
- CIBERSAM, Biomedical Research Network on Mental Health Area, Santander, Spain
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital LMU, Munich, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marcos L Santoro
- Laboratory of Integrative Neuroscience (LiNC), Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents (INPD), CNPq, São Paulo, Brazil
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Vidar M Steen
- NORMENT Centre of Excellence, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Jason L Stein
- Department of Genetics & UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Fabian Streit
- Department of Genetic Epidemiology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rick M Tankard
- Mathematics and Statistics, Curtin University, Perth, WA, Australia
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Dennis van 't Ent
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Dennis van der Meer
- NORMENT Centre, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Kristel R van Eijk
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Evangelos Vassos
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- NIHR Maudsley Biomedical Research Centre, South London and Maudsley NHS Trust, London, UK
| | - Javier Vázquez-Bourgon
- Valdecilla Biomedical Research Institute (IDIVAL), Marqués de Valdecilla University Hospital (HUMV), School of Medicine, University of Cantabria, Santander, Spain
- CIBERSAM, Biomedical Research Network on Mental Health Area, Santander, Spain
- Universidad de Cantabria, Santander, Spain
| | - Stephanie H Witt
- Department of Genetic Epidemiology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Hieab H H Adams
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
| | - Ingrid Agartz
- NORMENT Centre, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - David Ames
- Academic Unit for Psychiatry of Old Age, University of Melbourne, Parkville, VIC, Australia
- National Ageing Research Institute, Parkville, VIC, Australia
| | - Katrin Amunts
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ole A Andreassen
- NORMENT Centre, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Celso Arango
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, IiSGM, CIBERSAM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Bernhard T Baune
- Department of Psychiatry, University of Melbourne, Melbourne VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Sintia I Belangero
- Laboratory of Integrative Neuroscience (LiNC), Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents (INPD), CNPq, São Paulo, Brazil
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Arun L W Bokde
- Discipline of Psychiatry and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Dorret I Boomsma
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Rodrigo A Bressan
- Laboratory of Integrative Neuroscience (LiNC), Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents (INPD), CNPq, São Paulo, Brazil
- Instituto Ame Sua Mente, São Paulo, Brazil
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Karakter Child and Adolescent Psychiatry University Centre, Nijmegen, The Netherlands
| | - Wiepke Cahn
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Altrecht Science, Altrecht Mental Health Institute, Utrecht, The Netherlands
| | - Svenja Caspers
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Sven Cichon
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Benedicto Crespo-Facorro
- CIBERSAM, Biomedical Research Network on Mental Health Area, Santander, Spain
- Department of Psychiatry, Virgen del Rocio University Hospital, School of Medicine, University of Seville, IBIS, Seville, Spain
| | - Simon R Cox
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Edinburgh, UK
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Torbjørn Elvsåshagen
- NORMENT Centre, Oslo University Hospital, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thomas Espeseth
- Department of Psychology, University of Oslo, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Peter G Falkai
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany
| | - Simon E Fisher
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Janice M Fullerton
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Hugh Garavan
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Penny A Gowland
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, UK
| | - Hans J Grabe
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | | | - Manon Hillegers
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jacqueline Hoare
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- Faculty of Health, Peninsula Medical School, University of Plymouth, Plymouth, UK
| | - Pieter J Hoekstra
- University of Groningen, University Medical Center Groningen, Department of Child and Adolescent Psychiatry & Accare Child Study Center, Groningen, The Netherlands
| | - Mohammad A Ikram
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrea P Jackowski
- Laboratory of Integrative Neuroscience (LiNC), Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents (INPD), CNPq, São Paulo, Brazil
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
- Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany
| | - Erik G Jönsson
- NORMENT Centre, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| | - Rene S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- VISN 2 Mental Illness Research, Education & Clinical Center (MIRECC), James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Mayuresh S Korgaonkar
- Brain Dynamics Centre, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Herve Lemaitre
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, CNRS UMR 5293, Université de Bordeaux, Centre Broca Nouvelle-Aquitaine, Bordeaux, France
| | - Ulrik F Malt
- Unit for Psychosomatic Medicine and C-L Psychiatry, University of Oslo, Oslo, Norway
| | - Jean-Luc Martinot
- INSERM U1299 Trajectoires Développementales en Psychiatrie, Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, Université Paris Cité, CNRS UMR 9010; Centre Borelli, Gif-sur-Yvette, France
| | - Colm McDonald
- Centre for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Philip B Mitchell
- School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Jaap Oosterlaan
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Emma Neuroscience Group, Department of Pediatrics, Amsterdam Reproduction & Development, Amsterdam, The Netherlands
- Vrije Universiteit, Clinical Neuropsychology Section, Amsterdam, The Netherlands
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry, Erasmus Medical Center, Erasmus University, Rotterdam, The Netherlands
| | - Pedro M Pan
- Laboratory of Integrative Neuroscience (LiNC), Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents (INPD), CNPq, São Paulo, Brazil
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry, University Medical Center Goettingen, Göttingen, Germany
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia
- Neuropsychiatric Institute, The Prince of Wales Hospital, Sydney, NSW, Australia
| | - Giovanni A Salum
- National Institute of Developmental Psychiatry for Children and Adolescents (INPD), CNPq, São Paulo, Brazil
- Department of Psychiatry and Legal Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Section on Negative Affect and Social Processes, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Gunter Schumann
- Center for Population Neuroscience and Precision Medicine (PONS), Institute for Science and Technology for Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China
- PONS Centre, Department of Psychiatry and Clinical Neuroscience, CCM, Charite University Medicine, Berlin, Germany
| | - Philip Shaw
- Social and Behavioral Research Branch, National Human Genome Research Institute, Bethesda, MD, USA
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Kang Sim
- West Region, Institute of Mental Health, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Dan J Stein
- SAMRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Julian N Trollor
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia
- Department of Developmental Disability Neuropsychiatry, Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia
| | - Leonard H van den Berg
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Henrik Walter
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute for Health, Berlin, Germany
| | - Lars T Westlye
- NORMENT Centre, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Robert Whelan
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Tonya White
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, QLD, Australia
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Barbara Franke
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Hilleke E Hulshoff Pol
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.
- Department of Psychology, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
94
|
Wang Z, Chen W, Cao Y, Dou Y, Fu Y, Zhang Y, Luo X, Kang L, Liu N, Shi YS, Li CSR, Xu Y, Guo X, Luo X. An independent, replicable, functional and significant risk variant block at intron 3 of CACNA1C for schizophrenia. Aust N Z J Psychiatry 2022; 56:385-397. [PMID: 33938268 DOI: 10.1177/00048674211009595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Genome-wide association studies have identified a significant risk gene, CACNA1C, for schizophrenia. In this study, we comprehensively investigated a large set of CACNA1C single-nucleotide polymorphisms (SNPs) to identify the replicable risk alleles for schizophrenia and explore their biological functions. METHODS One Jewish (1044 cases vs 2052 controls), one European (1350 cases vs 1378 controls) and one exploratory African American samples (98 cases vs 20 controls) were analyzed to identify replicable single-nucleotide polymorphism-schizophrenia associations. The regulatory effects of risk alleles on CACNA1C messenger RNA expression were examined. The most robust risk tagSNP (rs1006737) was meta-analyzed on 17 studies (74,122 cases vs 109,062 controls), and associated with the gray matter volumes of seven subcortical structures in 38,258 Europeans, and the surface areas and thickness of 34 cortical regions in 33,992 Europeans and 2944 non-Europeans. RESULTS Forty-seven replicable risk single-nucleotide polymorphisms, including a 20-single-nucleotide polymorphism haplotype block, were identified in our samples (1.8 × 10-4 ⩽ p ⩽ 0.049). This variant block was consistently associated with schizophrenia across four independent Psychiatric Genomics Consortium cohorts (79,645 cases vs 109,590 controls; 2.5 × 10-17 ⩽ p ⩽ 0.017). This block showed significant expression quantitative trait loci in three independent European brain cohorts (5.1 × 10-12 ⩽ p ⩽ 8.3 × 10-3) and could be tagged by the most significant risk single-nucleotide polymorphism rs1006737. The minor allele A of rs1006737 significantly increased risk for schizophrenia across the Jewish and European samples (p = 0.029 and 0.004, respectively), and this association was highly significant in the meta-analysis (p = 1.62 × 10-42). This allele also significantly altered the CACNA1C messenger RNA expression in five brain regions (5.1 × 10-12 ⩽ p ⩽ 0.05), decreased the gray matter volume of thalamus (p = 0.010), the surface area of isthmus cingulate cortex (p = 0.013) and the thickness of transverse temporal and superior temporal sulcus cortexes (0.005 ⩽ p ⩽ 0.043). CONCLUSION We identified an independent, replicable, functional, and significant risk variant block at CACNA1C for schizophrenia, which could be tagged by the most robust risk marker rs1006737, suggesting an important role of CACNA1C in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Zuxing Wang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Psychosomatic Medicine of Sichuan Provincial Center for Mental Health, The Center of Psychosomatic Medicine of Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenzhong Chen
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuping Cao
- Department of Psychiatry of the Second Xiangya Hospital, Central South University; China National Clinical Research Center on Mental Disorders, China National Technology Institute on Mental Disorders, Changsha, China
| | - Yikai Dou
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yingmei Fu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Zhang
- Department of Psychiatry, Tianjin Mental Health Center, Tianjin, China
| | - Xingqun Luo
- Department of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Longli Kang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Diseases of Tibet Autonomous Region, Xizang Minzu University School of Medicine, Xiangyang, China
| | - Na Liu
- Department of Psychiatry, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yun Stone Shi
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yifeng Xu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyun Guo
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingguang Luo
- Division of Psychiatric Genetics, Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing, China
| |
Collapse
|
95
|
Chambers T, Escott-Price V, Legge S, Baker E, Singh KD, Walters JTR, Caseras X, Anney RJL. Genetic common variants associated with cerebellar volume and their overlap with mental disorders: a study on 33,265 individuals from the UK-Biobank. Mol Psychiatry 2022; 27:2282-2290. [PMID: 35079123 PMCID: PMC9126806 DOI: 10.1038/s41380-022-01443-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 12/16/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022]
Abstract
Interest in the cerebellum is expanding given evidence of its contributions to cognition and emotion, and dysfunction in various psychopathologies. However, research into its genetic architecture and shared influences with liability for mental disorders is lacking. We conducted a genome-wide association study (GWAS) of total cerebellar volume and underlying cerebellar lobe volumes in 33,265 UK-Biobank participants. Total cerebellar volume was heritable (h2SNP = 50.6%), showing moderate genetic homogeneity across lobes (h2SNP from 35.4% to 57.1%; mean genetic correlation between lobes rg ≈ 0.44). We identified 33 GWAS signals associated with total cerebellar volume, of which 6 are known to alter protein-coding gene structure, while a further five mapped to genomic regions known to alter cerebellar tissue gene expression. Use of summary data-based Mendelian randomisation further prioritised genes whose change in expression appears to mediate the SNP-trait association. In total, we highlight 21 unique genes of greatest interest for follow-up analyses. Using LD-regression, we report significant genetic correlations between total cerebellar volume and brainstem, pallidum and thalamus volumes. While the same approach did not result in significant correlations with psychiatric phenotypes, we report enrichment of schizophrenia, bipolar disorder and autism spectrum disorder associated signals within total cerebellar GWAS results via conditional and conjunctional-FDR analysis. Via these methods and GWAS catalogue, we identify which of our cerebellar genomic regions also associate with psychiatric traits. Our results provide important insights into the common allele architecture of cerebellar volume and its overlap with other brain volumes and psychiatric phenotypes.
Collapse
Affiliation(s)
- Tom Chambers
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Valentina Escott-Price
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Sophie Legge
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Emily Baker
- UK Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Krish D Singh
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Xavier Caseras
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK.
| | - Richard J L Anney
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
96
|
Teeuw J, Klein M, Mota NR, Brouwer RM, van ‘t Ent D, Al-Hassaan Z, Franke B, Boomsma DI, Hulshoff Pol HE. Multivariate Genetic Structure of Externalizing Behavior and Structural Brain Development in a Longitudinal Adolescent Twin Sample. Int J Mol Sci 2022; 23:ijms23063176. [PMID: 35328598 PMCID: PMC8949114 DOI: 10.3390/ijms23063176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 12/10/2022] Open
Abstract
Externalizing behavior in its more extreme form is often considered a problem to the individual, their families, teachers, and society as a whole. Several brain structures have been linked to externalizing behavior and such associations may arise if the (co)development of externalizing behavior and brain structures share the same genetic and/or environmental factor(s). We assessed externalizing behavior with the Child Behavior Checklist and Youth Self Report, and the brain volumes and white matter integrity (fractional anisotropy [FA] and mean diffusivity [MD]) with magnetic resonance imaging in the BrainSCALE cohort, which consisted of twins and their older siblings from 112 families measured longitudinally at ages 10, 13, and 18 years for the twins. Genetic covariance modeling based on the classical twin design, extended to also include siblings of twins, showed that genes influence externalizing behavior and changes therein (h2 up to 88%). More pronounced externalizing behavior was associated with higher FA (observed correlation rph up to +0.20) and lower MD (rph up to −0.20), with sizeable genetic correlations (FA ra up to +0.42; MD ra up to −0.33). The cortical gray matter (CGM; rph up to −0.20) and cerebral white matter (CWM; rph up to +0.20) volume were phenotypically but not genetically associated with externalizing behavior. These results suggest a potential mediating role for global brain structures in the display of externalizing behavior during adolescence that are both partially explained by the influence of the same genetic factor.
Collapse
Affiliation(s)
- Jalmar Teeuw
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (R.M.B.); (Z.A.-H.); (H.E.H.P.)
- Correspondence: ; Tel.: +31-(088)-75-53-387
| | - Marieke Klein
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA;
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.R.M.); (B.F.)
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 XZ Nijmegen, The Netherlands
| | - Nina Roth Mota
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.R.M.); (B.F.)
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 XZ Nijmegen, The Netherlands
| | - Rachel M. Brouwer
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (R.M.B.); (Z.A.-H.); (H.E.H.P.)
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Dennis van ‘t Ent
- Department of Biological Psychology, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (D.v.‘t.E.); (D.I.B.)
| | - Zyneb Al-Hassaan
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (R.M.B.); (Z.A.-H.); (H.E.H.P.)
| | - Barbara Franke
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.R.M.); (B.F.)
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 XZ Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Dorret I. Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (D.v.‘t.E.); (D.I.B.)
- Amsterdam Public Health (APH) Research Institute, 1081 BT Amsterdam, The Netherlands
| | - Hilleke E. Hulshoff Pol
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (R.M.B.); (Z.A.-H.); (H.E.H.P.)
- Department of Psychology, Utrecht University, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
97
|
Hou J, Zhu H, Xiao L, Zhao CW, Liao G, Tang Y, Feng L. Alterations in Cortical-Subcortical Metabolism in Temporal Lobe Epilepsy With Impaired Awareness Seizures. Front Aging Neurosci 2022; 14:849774. [PMID: 35360210 PMCID: PMC8961434 DOI: 10.3389/fnagi.2022.849774] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/26/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveThe features of cerebral metabolism associated with loss of consciousness in patients with temporal lobe epilepsy (TLE) have not been fully elucidated. We aim to investigate the alterations in cortical-subcortical metabolism in temporal lobe epilepsy with impaired awareness seizures (IAS).MethodsRegional cerebral metabolism was measured using fluorine-18-fluorodeoxyglucose positron emission tomography (18F-FDG PET) in patients with TLE-IAS and healthy controls. All patients had a comprehensive evaluation to confirm their seizure origin and lateralization. Videos of all seizures were viewed and rated by at least two epileptologists to identify the state of consciousness when a seizure occurred. By synthesizing the seizure history, semeiology, and video EEG of all patients, as long as the patients had one seizure with impaired awareness, she/he will be included. 76 patients with TLE-IAS and 60 age-matched healthy controls were enrolled in this study. Regional cerebral metabolic patterns were analyzed for TLE-IAS and healthy control groups using statistical parametric mapping. Besides, we compared the MRI-negative patients and MRI-positive patients with healthy controls, respectively.ResultsThere were no significant differences in the age and sex of TLE-IAS patients and healthy control. TLE-IAS patients showed extensive bilateral hypermetabolism in the frontoparietal regions, cingulate gyrus, corpus callosum, occipital lobes, basal ganglia, thalamus, brainstem, and cerebellum. The region of metabolic change was more extensive in right TLE-IAS than that of the left, including extensive hypometabolism in the ipsilateral temporal, frontal, parietal, and insular lobes. And contralateral temporal lobe, bilateral frontoparietal regions, occipital lobes, the anterior and posterior regions of the cingulate gyrus, bilateral thalamus, bilateral basal ganglia, brainstem, and bilateral cerebellum showed hypermetabolism. The TLE patients with impaired awareness seizure showed hypermetabolism in the cortical-subcortical network including the arousal system. Additionally, 48 MRI-positive and 28 MRI-negative TLE-IAS patients were included in our study. TLE-IAS patients with MRI-negative and MRI-positive were both showed hypermetabolism in the cingulate gyrus. Hypometabolism in the bilateral temporal lobe was showed in the TLE-IAS with MRI-positive.ConclusionThese findings suggested that the repetitive consciousness impairing ictal events may have an accumulative effect on brain metabolism, resulting in abnormal interictal cortical-subcortical metabolic disturbance in TLE patients with impaired awareness seizure. Understanding these metabolic mechanisms may guide future clinical treatments to prevent seizure-related awareness deficits and improve quality of life in people with TLE.
Collapse
Affiliation(s)
- Jiale Hou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Haoyue Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Xiao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | | | - Guang Liao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yongxiang Tang,
| | - Li Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (XIANGYA), Xiangya Hospital, Central South University, Changsha, China
- Li Feng,
| |
Collapse
|
98
|
Pan X, Zhang M, Tian A, Chen L, Sun Z, Wang L, Chen P. Exploring the genetic correlation between obesity-related traits and regional brain volumes: Evidence from UK Biobank cohort. Neuroimage Clin 2022; 33:102870. [PMID: 34872017 PMCID: PMC8648807 DOI: 10.1016/j.nicl.2021.102870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To determine whether there is a correlation between obesity-related variants and regional brain volumes. METHODS Based on a mixed linear model (MLM), we analyzed the association between 1,498 obesity-related SNPs in the GWAS Catalog and 164 regional brain volumes from 29,420 participants (discovery cohort N = 19,997, validation cohort N = 9,423) in UK Biobank. The statistically significant brain regions in association analysis were classified into 6 major neural networks (dopamine (DA) motive system, central autonomic network (CAN), cognitive emotion regulation, visual object recognition network, auditory object recognition network, and sensorimotor system). We summarized the association between obesity-related variants (metabolically healthy obesity variants, metabolically unhealthy obesity variants, and unclassified obesity-related variants) and neural networks. RESULTS From association analysis, we determined that 17 obesity-related SNPs were associated with 51 regional brain volumes. Several single SNPs (e.g., rs13107325-T (SLC39A8), rs1876829-C (CRHR1), and rs1538170-T (CENPW)) were associated with multiple regional brain volumes. In addition, several single brain regions (e.g., the white matter, the grey matter in the putamen, subcallosal cortex, and insular cortex) were associated with multiple obesity-related variants. The metabolically healthy obesity variants were mainly associated with the regional brain volumes in the DA motive system, sensorimotor system and cognitive emotion regulation neural networks, while metabolically unhealthy obesity variants were mainly associated with regional brain volumes in the CAN and total tissue volumes. In addition, unclassified obesity-related variants were mainly associated with auditory object recognition network and total tissue volumes. The results of MeSH (medical subject headings) enrichment analysis showed that obesity genes associated with brain structure pointed to the functional relatedness with 5-Hydroxytryptamine receptor 4 (5-HT4), growth differentiation factor 5 (GDF5), and high mobility group protein AT-hook 2 (HMGA2 protein). CONCLUSION In summary, we found that obesity-related variants were associated with different brain volume measures. On the basis of the multiple SNPs, we found that metabolically healthy and unhealthy obesity-related SNPs were associated with different brain neural networks. Based on our enrichment analysis, modifications of the 5-HT4 pathway might be a promising therapeutic strategy for obesity.
Collapse
Affiliation(s)
- Xingchen Pan
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China; Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Miaoran Zhang
- Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Aowen Tian
- Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Lanlan Chen
- School of Clinical Medicine, Jilin University, Changchun, 130000, China
| | - Zewen Sun
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| | - Peng Chen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China; Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
99
|
Ohi K, Ishibashi M, Torii K, Hashimoto M, Yano Y, Shioiri T. Differences in subcortical brain volumes among patients with schizophrenia and bipolar disorder and healthy controls. J Psychiatry Neurosci 2022; 47:E77-E85. [PMID: 35232800 PMCID: PMC8896343 DOI: 10.1503/jpn.210144] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/09/2021] [Accepted: 10/10/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Patients with schizophrenia and bipolar disorder have an overlapping polygenic architecture and clinical similarities, although the 2 disorders are distinct diagnoses with clinical dissimilarities. It remains unclear whether there are specific differences in subcortical volumes between schizophrenia and bipolar disorder, and whether the subcortical differences are affected by any clinical characteristics. We investigated differences in subcortical volumes bilaterally among patients with schizophrenia, patients with bipolar disorder and healthy controls. We also investigated the influences of clinical characteristics on specific subcortical volumes in these patient groups. METHODS We collected 3 T T 1-weighted MRI brain scans from 413 participants (157 with schizophrenia, 51 with bipolar disorder and 205 controls) with a single scanner at a single institute. We used FreeSurfer version 6.0 for processing the T 1-weighted images to segment the following subcortical brain volumes: thalamus, caudate, putamen, globus pallidus, hippocampus, amygdala and nucleus accumbens. Differences in the 7 subcortical volumes were investigated among the groups. We also evaluated correlations between subcortical volumes and clinical variables in these patient groups. RESULTS Of 7 subcortical regions, patients with schizophrenia had significantly smaller volumes in the left thalamus (Cohen d = -0.29, p = 5.83 × 10-3), bilateral hippocampi (left, d = -0.36, p = 8.85 × 10-4; right, d = -0.41, p = 1.15 × 10-4) and left amygdala (d = -0.31, p = 4.02 × 10-3) than controls. Compared with controls, patients with bipolar disorder had bilateral reductions only in the hippocampal volumes (left, d = -0.52, p = 1.12 × 10-3; right, d = -0.58, p = 0.30 × 10-4). We also found that patients with schizophrenia had significantly smaller volumes in the bilateral amygdalae (left, d = -0.43, p = 4.22 × 10-3; right, d = -0.45, p = 4.56 × 10-3) than patients with bipolar disorder. We did not find any significant volumetric differences in the other 6 subcortical structures between patient groups (p > 0.05). Smaller left amygdalar volumes were significantly correlated with younger onset age only in patients with schizophrenia (r = 0.22, p = 5.78 × 10-3). LIMITATIONS We did not evaluate the differences in subcortical volumes between patients stratified based on clinical bipolar disorder subtype and a history of psychotic episodes because our sample size of patients with bipolar disorder was limited. CONCLUSION Our findings suggest that volumetric differences in the amygdala between patients with schizophrenia and those with bipolar disorder may be a putative biomarker for distinguishing 2 clinically similar diagnoses.
Collapse
Affiliation(s)
- Kazutaka Ohi
- From the Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan (Ohi, Shioiri); the Department of General Internal Medicine, Kanazawa Medical University, Ishikawa, Japan (Ohi); and the School of Medicine, Gifu University, Gifu, Japan (Ishibashi, Torii, Hashimoto, Yano)
| | | | | | | | | | | |
Collapse
|
100
|
Rabinowitz JA, Campos AI, Ong JS, García-Marín LM, Alcauter S, Mitchell BL, Grasby KL, Cuéllar-Partida G, Gillespie NA, Huhn AS, Martin NG, Thompson PM, Medland SE, Maher BS, Rentería ME. Shared Genetic Etiology between Cortical Brain Morphology and Tobacco, Alcohol, and Cannabis Use. Cereb Cortex 2022; 32:796-807. [PMID: 34379727 PMCID: PMC8841600 DOI: 10.1093/cercor/bhab243] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified genetic variants associated with brain morphology and substance use behaviors (SUB). However, the genetic overlap between brain structure and SUB has not been well characterized. We leveraged GWAS summary data of 71 brain imaging measures and alcohol, tobacco, and cannabis use to investigate their genetic overlap using linkage disequilibrium score regression. We used genomic structural equation modeling to model a "common SUB genetic factor" and investigated its genetic overlap with brain structure. Furthermore, we estimated SUB polygenic risk scores (PRS) and examined whether they predicted brain imaging traits using the Adolescent Behavior and Cognitive Development (ABCD) study. We identified 8 significant negative genetic correlations, including between (1) alcoholic drinks per week and average cortical thickness, and (2) intracranial volume with age of smoking initiation. We observed 5 positive genetic correlations, including those between (1) insula surface area and lifetime cannabis use, and (2) the common SUB genetic factor and pericalcarine surface area. SUB PRS were associated with brain structure variation in ABCD. Our findings highlight a shared genetic etiology between cortical brain morphology and SUB and suggest that genetic variants associated with SUB may be causally related to brain structure differences.
Collapse
Affiliation(s)
- Jill A Rabinowitz
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Adrian I Campos
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jue-Sheng Ong
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Luis M García-Marín
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Sarael Alcauter
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, México
| | - Brittany L Mitchell
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland 4059, Australia
| | - Katrina L Grasby
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Gabriel Cuéllar-Partida
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Nathan A Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Andrew S Huhn
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Baltimore, MD 21205, USA
| | - Nicholas G Martin
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, USA
| | - Sarah E Medland
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Brion S Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Miguel E Rentería
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland 4059, Australia
| |
Collapse
|