1
|
Li Y, Zhou G, Peng J, Liu L, Zhang F, Iturria-Medina Y, Yao D, Biswal BB, Wang P. White matter dysfunction in Alzheimer's disease is associated with disease-related transcriptomic signatures. Commun Biol 2025; 8:820. [PMID: 40437109 PMCID: PMC12120127 DOI: 10.1038/s42003-025-08177-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 05/06/2025] [Indexed: 06/01/2025] Open
Abstract
While anatomical white matter (WM) alterations in Alzheimer's disease (AD) are well-established, functional WM dysregulation remains rarely investigated. The current study examines WM functional connectivity and network properties alterations in AD and mild cognitive impairment (MCI) and further describes their spatially correlated genes. AD and MCI shared decreased functional connectivity, clustering coefficient, and local efficiency within WM regions involved in impaired sensory-motor, visual-spatial, language, or memory functions. AD-specific dysfunction (i.e., AD vs. MCI and cognitively unimpaired participants) was predominantly located in WM, including anterior and posterior limb of internal capsule, corona radiata, and left tapetum. This WM dysfunction spatially correlates with specific genes, which are enriched in multiple biological processes related to synaptic function and development, and are mostly active in neurons and astrocytes. These findings may contribute to understanding molecular, cellular, and functional signatures associated with WM damage in AD.
Collapse
Affiliation(s)
- Yilu Li
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Guanyu Zhou
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinzhong Peng
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Liu
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Fanyu Zhang
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
| | - Dezhong Yao
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Bharat B Biswal
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA.
| | - Pan Wang
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
2
|
Liu X, Niu J, Liao W, Du L. Alterations in gray matter volume and associated transcriptomics after electroconvulsive therapy in major depressive disorder. Psychol Med 2025; 55:e118. [PMID: 40254982 DOI: 10.1017/s0033291725000868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
BACKGROUND The antidepressant mechanism of electroconvulsive therapy (ECT) remains not clearly understood. This study aimed to detect the changes in gray matter volume (GMV) in patients with major depressive disorder (MDD) caused by ECT and exploratorily analyzed the potential functional mechanisms. METHODS A total of 24 patients with MDD who underwent eight ECT sessions were included in the study. Clinical symptom assessments and MRI scans were conducted and compared. Using whole-brain micro-array measurements provided by the Allen Human Brain Atlas (AHBA), regional gene expression profiles were calculated. The differential gene PLS1 was obtained through Partial Least Squares (PLS) regression analysis, and PLS1 was divided into positive contribution (PLS1+) and negative contribution (PLS1-) genes. Through gene function enrichment analysis, the functional pathways and cell types of PLS1 enrichment were identified. RESULTS Gray matter volume (GMV) in the somatosensory and motor cortices, occipital cortex, prefrontal cortex, and insula showed an increasing trend after ECT, while GMV in the temporal cortex, posterior cingulate cortex, and orbitofrontal cortex decreased. PLS1 genes were enriched in synapse- and cell-related biological processes and cellular components (such as 'pre- and post-synapse', 'synapse organization' etc.). A large number of genes in the PLS1+ list were involved in neurons (inhibitory and excitatory), whereas PLS1- genes were significantly involved in Astrocytes (Astro) and Microglia (Micro). CONCLUSIONS This study established a link between treatment-induced GMV changes and specific functional pathways and cell types, which suggests that ECT may exert its effects through synapse-associated functional and affect neurons and glial cells.
Collapse
Affiliation(s)
- Xiaoxue Liu
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), The First Affiliated Hospital of Chongqing Medical University
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Jinpeng Niu
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Wei Liao
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Lian Du
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), The First Affiliated Hospital of Chongqing Medical University
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Liu Q, Du X, Zhang Y, Ding H, Qin W, Zhang Q. Associations between morphometric similarity network and brain gene expression in type 2 diabetes mellitus. Neuroscience 2025; 572:84-92. [PMID: 39884418 DOI: 10.1016/j.neuroscience.2025.01.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Abnormal functional and structural connectivity in brain networks is commonly observed in patients with type 2 diabetes mellitus (T2DM) and is often associated with cognitive impairment. In this study, we employed the Morphometric Similarity Network (MSN) method, which is based on seven morphometric features derived from structural and diffusion magnetic resonance imaging, to investigate structural differences in the brains of T2DM patients by quantifying structural similarities between brain regions. Globally, morphometric similarity (MS) was significantly reduced in T2DM patients. Regionally, MS was decreased in the left sensorimotor network and the right salience/ventral attention network, while it was increased in the bilateral visual network. Notably, the increased MS in the bilateral visual network was negatively correlated with memory function in T2DM patients. Furthermore, using the Allen Human Brain Atlas (AHBA; http://human.brain-map.org), which provides transcriptome data from postmortem adult brains, we linked MSN changes to regional gene expression patterns. Transcription-neuroimaging association analyses identified 298 genes whose expression was significantly spatially correlated with T2DM-related MSN abnormalities. Many of these genes are involved in biological processes such as central nervous system development and neurotransmitter transmission, offering valuable molecular and cellular insights into MS abnormalities and cognitive decline in T2DM. These findings shed light on the neural and genetic mechanisms underlying T2DM-related brain changes and cognitive impairment, providing new perspectives for future research and potential therapeutic approaches.
Collapse
Affiliation(s)
- Qiaohui Liu
- Department of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Du
- Department of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yang Zhang
- Department of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hao Ding
- Department of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wen Qin
- Department of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Quan Zhang
- Department of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
4
|
Ji Y, Liu N, Yang Y, Wang M, Cheng J, Zhu W, Qiu S, Geng Z, Cui G, Yu Y, Liao W, Zhang H, Gao B, Xu X, Han T, Yao Z, Zhang Q, Qin W, Liu F, Liang M, Wang S, Xu Q, Xu J, Fu J, Zhang P, Li W, Shi D, Wang C, Lui S, Yan Z, Chen F, Zhang J, Shen W, Miao Y, Wang D, Gao JH, Zhang X, Xu K, Zuo XN, Zhang L, Ye Z, Li MJ, Xian J, Zhang B, Yu C. Cross-ancestry and sex-stratified genome-wide association analyses of amygdala and subnucleus volumes. Nat Genet 2025; 57:839-850. [PMID: 40097784 DOI: 10.1038/s41588-025-02136-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
The amygdala is a small but critical multi-nucleus structure for emotion, cognition and neuropsychiatric disorders. Although genetic associations with amygdala volumetric traits have been investigated in sex-combined European populations, cross-ancestry and sex-stratified analyses are lacking. Here we conducted cross-ancestry and sex-stratified genome-wide association analyses for 21 amygdala volumetric traits in 6,923 Chinese and 48,634 European individuals. We identified 191 variant-trait associations (P < 2.38 × 10-9), including 47 new associations (12 new loci) in sex-combined univariate analyses and seven additional new loci in sex-combined and sex-stratified multivariate analyses. We identified 12 ancestry-specific and two sex-specific associations. The identified genetic variants include 16 fine-mapped causal variants and regulate amygdala and fetal brain gene expression. The variants were enriched for brain development and colocalized with mood, cognition and neuropsychiatric disorders. These results indicate that cross-ancestry and sex-stratified genetic association analyses may provide insight into the genetic architectures of amygdala and subnucleus volumes.
Collapse
Affiliation(s)
- Yuan Ji
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Nana Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yunjun Yang
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China
- Biomedical Institute, Henan Academy of Sciences, Zhengzhou, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shijun Qiu
- Department of Medical Imaging, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Zuojun Geng
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guangbin Cui
- Functional and Molecular Imaging Key Lab of Shaanxi Province & Department of Radiology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- Molecular Imaging Research Center of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Zhang
- Department of Radiology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Bo Gao
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Radiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Quan Zhang
- Department of Radiology, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Wen Qin
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Liang
- School of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, China
| | - Sijia Wang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Xu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiayuan Xu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jilian Fu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wei Li
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Dapeng Shi
- Department of Radiology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China
| | - Caihong Wang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Su Lui
- Department of Radiology, Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Zhihan Yan
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feng Chen
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Jing Zhang
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
| | - Wen Shen
- Department of Radiology, Tianjin First Center Hospital, Tianjin, China
| | - Yanwei Miao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xiaochu Zhang
- Division of Life Science and Medicine, University of Science & Technology of China, Hefei, China
| | - Kai Xu
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xi-Nian Zuo
- Developmental Population Neuroscience Research Center at IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Longjiang Zhang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Mulin Jun Li
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China.
- School of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
5
|
Koike S, Tanaka SC, Hayashi T. Beyond case-control study in neuroimaging for psychiatric disorders: Harmonizing and utilizing the brain images from multiple sites. Neurosci Biobehav Rev 2025; 171:106063. [PMID: 40020797 DOI: 10.1016/j.neubiorev.2025.106063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/15/2025] [Accepted: 02/09/2025] [Indexed: 03/03/2025]
Abstract
Recent magnetic resonance imaging (MRI) research has advanced our understanding of brain pathophysiology in psychiatric disorders. This progress necessitates re-evaluation of the diagnostic system for psychiatric disorders based on MRI-based biomarkers, with implications for precise clinical diagnosis and optimal therapeutics. To achieve this goal, large-scale multi-site studies are essential to develop a standardized MRI database, with the analysis of several thousands of images and the incorporation of new data. A critical challenge in these studies is to minimize sampling and measurement biases in MRI studies to accurately capture the diversity of disease-derived biomarkers. Various techniques have been employed to consolidate datasets from multiple sites in case-control studies. Traveling subject harmonization stands out as a powerful tool that can differentiate measurement bias from sample variety and sampling bias. A non-linear statistical model for a normative trajectory across the lifespan also strengthens the database to mitigate sampling bias from known factors such as age and sex. These approaches can enhance the alterations between psychiatric disorders and integrate new data and follow-up scans into existing life-course trajectory, enhancing the reliability of machine learning classification and subtyping. Although this approach has been developed using T1-weighted structural image features, future research may extend this framework to other modalities and measures. The required sample size and methodological establishment are needed for future investigations, leading to novel insights into the brain pathophysiology of psychiatric disorders and the development of optimal therapeutics for bedside clinical applications. Sharing big data and their findings also need to be considered.
Collapse
Affiliation(s)
- Shinsuke Koike
- University of Tokyo Institute for Diversity and Adaptation of Human Mind, The University of Tokyo, Tokyo 153-8902, Japan; Center for Evolutionary Cognitive Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo 153-8902, Japan; The International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo 113-8654, Japan.
| | - Saori C Tanaka
- Brain Information Communication Research Laboratory Group, Advanced Telecommunications Research Institute International, Kyoto 619-0288 Japan; Division of Information Science, Nara Institute of Science and Technology, Nara 630-0192, Japan
| | - Takuya Hayashi
- Laboratory for Brain Connectomics Imaging, RIKEN Center for Biosystems Dynamics Research, Hyogo 351-0198, Japan; Department of Brain Connectomics, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| |
Collapse
|
6
|
Islam SR, Xie Z, He W, Zhi D. Vision Transformer Autoencoders for Unsupervised Representation Learning: Capturing Local and Non-Local Features in Brain Imaging to Reveal Genetic Associations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.24.25324549. [PMID: 40196251 PMCID: PMC11974795 DOI: 10.1101/2025.03.24.25324549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The discovery of genetic loci associated with brain architecture can provide deeper insights into neuroscience and improved personalized medicine outcomes. Previously, we designed the Unsupervised Deep learning-derived Imaging Phenotypes (UDIPs) approach to extract endophenotypes from brain imaging using a convolutional (CNN) autoencoder, and conducted brain imaging GWAS on UK Biobank (UKBB). In this work, we leverage a vision transformer (ViT) model due to a different inductive bias and its ability to potentially capture unique patterns through its pairwise attention mechanism. Our approach based on 128 endophenotypes derived from average pooling discovered 10 loci previously unreported by CNN-based UDIP model, 3 of which were not found in the GWAS catalog to have had any associations with brain structure. Our interpretation results demonstrate the ViT's capability in capturing non-local patterns such as left-right hemisphere symmetry within brain MRI data, by leveraging its attention mechanism and positional embeddings. Our results highlight the advantages of transformer-based architectures in feature extraction and representation for genetic discovery.
Collapse
Affiliation(s)
- Samia R Islam
- The University of Texas Health Science Center at Houston, D. Bradley McWilliams School of Biomedical Informatics
| | - Ziqian Xie
- The University of Texas Health Science Center at Houston, D. Bradley McWilliams School of Biomedical Informatics
| | - Wei He
- The University of Texas Health Science Center at Houston, D. Bradley McWilliams School of Biomedical Informatics
| | - Degui Zhi
- The University of Texas Health Science Center at Houston, D. Bradley McWilliams School of Biomedical Informatics
| |
Collapse
|
7
|
Korologou-Linden R, Xu B, Coulthard E, Walton E, Wearn A, Hemani G, White T, Cecil C, Sharp T, Tiemeier H, Banaschewski T, Bokde A, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Paillère Martinot ML, Artiges E, Nees F, Orfanos DP, Paus T, Poustka L, Millenet S, Fröhner JH, Smolka M, Walter H, Winterer J, Whelan R, Schumann G, Howe LD, Ben-Shlomo Y, Davies NM, Anderson EL. Genetics impact risk of Alzheimer's disease through mechanisms modulating structural brain morphology in late life. J Neurol Neurosurg Psychiatry 2025; 96:350-360. [PMID: 38663994 PMCID: PMC7616849 DOI: 10.1136/jnnp-2023-332969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/11/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Alzheimer's disease (AD)-related neuropathological changes can occur decades before clinical symptoms. We aimed to investigate whether neurodevelopment and/or neurodegeneration affects the risk of AD, through reducing structural brain reserve and/or increasing brain atrophy, respectively. METHODS We used bidirectional two-sample Mendelian randomisation to estimate the effects between genetic liability to AD and global and regional cortical thickness, estimated total intracranial volume, volume of subcortical structures and total white matter in 37 680 participants aged 8-81 years across 5 independent cohorts (Adolescent Brain Cognitive Development, Generation R, IMAGEN, Avon Longitudinal Study of Parents and Children and UK Biobank). We also examined the effects of global and regional cortical thickness and subcortical volumes from the Enhancing NeuroImaging Genetics through Meta-Analysis (ENIGMA) Consortium on AD risk in up to 37 741 participants. RESULTS Our findings show that AD risk alleles have an age-dependent effect on a range of cortical and subcortical brain measures that starts in mid-life, in non-clinical populations. Evidence for such effects across childhood and young adulthood is weak. Some of the identified structures are not typically implicated in AD, such as those in the striatum (eg, thalamus), with consistent effects from childhood to late adulthood. There was little evidence to suggest brain morphology alters AD risk. CONCLUSIONS Genetic liability to AD is likely to affect risk of AD primarily through mechanisms affecting indicators of brain morphology in later life, rather than structural brain reserve. Future studies with repeated measures are required for a better understanding and certainty of the mechanisms at play.
Collapse
Affiliation(s)
- Roxanna Korologou-Linden
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Bing Xu
- The Generation R Study Group, Erasmus MC University Medical Center, Rotterdam, UK
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Elizabeth Coulthard
- Bristol Medical School, University of Bristol, Bristol, UK
- North Bristol NHS Trust, Bristol, UK
| | - Esther Walton
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Department of Psychology, University of Bath, Bath, UK
| | - Alfie Wearn
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Gibran Hemani
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Tonya White
- The Generation R Study Group, Erasmus MC University Medical Center, Rotterdam, UK
- Department of Radiology and Nuclear Medicine, Erasmus University School of Medicine, Rotterdam, UK
| | - Charlotte Cecil
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamsin Sharp
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Biostatistics and Health Informatics Department, King's College London, Boston, UK
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Social and Behavioral Sciences, Harvard T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Heidelberg University, Heidelberg, Germany
| | - Arun Bokde
- Psychiatry, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Kings College London, Centre for Population Neuroscience and Precision Medicine (PONS), London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, University of Mannheim, Mannheim, Germany
- Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | | | | | | | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Berlin Institute of Health, Berlin, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299, Paris, France
- Centre Borelli, Cachan, France
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299, Paris, France
- Centre Borelli, Cachan, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299, Paris, France
- Centre Borelli, Cachan, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Heidelberg University, Heidelberg, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, University of Mannheim, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, Kiel University, Kiel, Germany
| | | | - Tomáš Paus
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Montreal, Montreal, Quebec, Canada
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, Göttingen, Germany
| | - Sabina Millenet
- Department of Child and Adolescent Psychiatry and Psychotherapy, Heidelberg University, Heidelberg, Germany
| | - Juliane H Fröhner
- Department of Psychiatry, Technische Universität Dresden, Dresden, Germany
| | - Michael Smolka
- Department of Psychiatry, Technische Universität Dresden, Dresden, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charite, Berlin, Germany
| | - Jeanne Winterer
- Department of Psychiatry and Psychotherapy CCM, Berlin Institute of Health, Berlin, Germany
- Department of Education and Psychology, Freie Universität Berlin, Berlin, Germany
| | - Robert Whelan
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland
| | - Gunter Schumann
- Kings College London, Centre for Population Neuroscience and Precision Medicine (PONS), London, UK
- Fudan University, Shanghai, People's Republic of China
- PONS Centre, Dept. of Psychiatry and Clinical Neuroscience, CCM, Berlin, Germany
| | - Laura D Howe
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Neil M Davies
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
- University College London Division of Psychiatry, London, UK
| | - Emma Louise Anderson
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
- University College London Division of Psychiatry, London, UK
| |
Collapse
|
8
|
Wang P, Luan H, Li S, Han X, Sun W, Gong J, Xu C, Chen R, Wei C. Extensive perivascular spaces burden causally affects neurodegenerative diseases and brain structure: A two-sample bidirectional Mendelian randomization study. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111284. [PMID: 39921030 DOI: 10.1016/j.pnpbp.2025.111284] [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: 07/01/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Extensive perivascular spaces (PVS) burden has been reported to be associated with neurodegenerative diseases and brain structure; however, the causal effects has not been determined yet. Therefore, this study aimed to investigate the causal effect of extensive PVS burden on neurodegenerative diseases and brain structure through Mendelian randomization (MR) analysis. METHODS Two-sample bidirectional MR was conducted based on publicly available genome-wide association studies (GWAS) summary statistics. Causal estimates of extensive PVS burden on neurodegenerative diseases and brain structure were primarily assessed using the inverse-variance weighted (IVW) method, supplemented by additional methods, including MR-Egger, weighted median, simple mode, and weighted mode. Sensitivity analyses were performed to assess heterogeneity and pleiotropy. In addition, we explored whether brain structure act as a mediating factor in the pathway from extensive PVS burden to neurodegenerative diseases. RESULTS Our MR study found that extensive PVS burden in white matter (WM-PVS) burden was associated with lower Alzheimer's disease (AD) risk (IVW OR (95 % CI) = 0.963(0.929 to 0.999), P = 0.0428), with no heterogeneity and pleiotropy detected. In addition, following FDR correction, we found bidirectional causal relationships between extensive PVS burden and brain structure. Moreover, our results of the mediated analysis showed that the surface area of parahippocampal, as a mediating variable, plays an important role in the causal relationship between WM-PVS and AD. The mediation effect is 18 %. CONCLUSIONS Our study provides evidence for the causal associations of different extensive PVS burden phenotypes with neurodegenerative diseases and brain structures, improving our understanding of the complex relationships between different brain injuries.
Collapse
Affiliation(s)
- Pin Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Heya Luan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shaoqi Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiaodong Han
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Wenxian Sun
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jin Gong
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Chang Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Runqi Chen
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Cuibai Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China.
| |
Collapse
|
9
|
Zhu M, Chen Y, Zheng J, Zhao P, Xia M, Tang Y, Wang F. Over-integration of visual network in major depressive disorder and its association with gene expression profiles. Transl Psychiatry 2025; 15:86. [PMID: 40097427 PMCID: PMC11914485 DOI: 10.1038/s41398-025-03265-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 01/06/2025] [Accepted: 01/28/2025] [Indexed: 03/19/2025] Open
Abstract
Major depressive disorder (MDD) is a common psychiatric condition associated with aberrant functional connectivity in large-scale brain networks. However, it is unclear how the network dysfunction is characterized by imbalance or derangement of network modular interaction in MDD patients and whether this disruption is associated with gene expression profiles. We included 262 MDD patients and 297 healthy controls, embarking on a comprehensive analysis of intrinsic brain activity using resting-state functional magnetic resonance imaging (R-fMRI). We assessed brain network integration by calculating the Participation Coefficient (PC) and conducted an analysis of intra- and inter-modular connections to reveal the dysconnectivity patterns underlying abnormal PC manifestations. Besides, we explored the potential relationship between the above graph theory measures and clinical symptoms severity in MDD. Finally, we sought to uncover the association between aberrant graph theory measures and postmortem gene expression data sourced from the Allen Human Brain Atlas (AHBA). Relative to the controls, alterations in systemic functional connectivity were observed in MDD patients. Specifically, increased PC within the bilateral visual network (VIS) was found, accompanied by elevated functional connectivities (FCs) between VIS and both higher-order networks and Limbic network (Limbic), contrasted by diminished FCs within the VIS and between the VIS and the sensorimotor network (SMN). The clinical correlations indicated positive associations between inter-VIS FCs and depression symptom, whereas negative correlations were noted between intra-VIS FCs with depression symptom and cognitive disfunction. The transcriptional profiles explained 21-23.5% variance of the altered brain network system dysconnectivity pattern, with the most correlated genes enriched in trans-synaptic signaling and ion transport regulation. These results highlight the modular connectome dysfunctions characteristic of MDD and its linkage with gene expression profiles and clinical symptomatology, providing insight into the neurobiological underpinnings and holding potential implications for clinical management and therapeutic interventions in MDD.
Collapse
Affiliation(s)
- Mingrui Zhu
- Department of Neurology, Liaoning Provincial People's Hospital, Shenyang, Liaoning, China
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yifan Chen
- School of Public Health, Southeast University, Nanjing, China
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Junjie Zheng
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Pengfei Zhao
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Mingrui Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China.
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, P. R. China.
| | - Yanqing Tang
- Department of psychaitry, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Fei Wang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China.
- Department of Mental Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
10
|
Goovaerts S, Naqvi S, Hoskens H, Herrick N, Yuan M, Shriver MD, Shaffer JR, Walsh S, Weinberg SM, Wysocka J, Claes P. Enhanced insights into the genetic architecture of 3D cranial vault shape using pleiotropy-informed GWAS. Commun Biol 2025; 8:439. [PMID: 40087503 PMCID: PMC11909261 DOI: 10.1038/s42003-025-07875-6] [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: 06/11/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Large-scale GWAS studies have uncovered hundreds of genomic loci linked to facial and brain shape variation, but only tens associated with cranial vault shape, a largely overlooked aspect of the craniofacial complex. Surrounding the neocortex, the cranial vault plays a central role during craniofacial development and understanding its genetics are pivotal for understanding craniofacial conditions. Experimental biology and prior genetic studies have generated a wealth of knowledge that presents opportunities to aid further genetic discovery efforts. Here, we use the conditional FDR method to leverage GWAS data of facial shape, brain shape, and bone mineral density to enhance SNP discovery for cranial vault shape. This approach identified 120 independent genomic loci at 1% FDR, nearly tripling the number discovered through unconditioned analysis and implicating crucial craniofacial transcription factors and signaling pathways. These results significantly advance our genetic understanding of cranial vault shape and craniofacial development more broadly.
Collapse
Affiliation(s)
- Seppe Goovaerts
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
| | - Sahin Naqvi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Genetics and Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Hanne Hoskens
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
- Department of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research, Institute, University of Calgary, Calgary, AB, Canada
| | - Noah Herrick
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, USA
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Meng Yuan
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Mark D Shriver
- Department of Anthropology, Pennsylvania State University, State College, PA, USA
| | - John R Shaffer
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Susan Walsh
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, USA
| | - Seth M Weinberg
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anthropology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter Claes
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium.
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| |
Collapse
|
11
|
Dong Y, Zhang P, Zhong J, Wang J, Xu Y, Huang H, Liu X, Sun W. Modifiable lifestyle factors influencing neurological and psychiatric disorders mediated by structural brain reserve: An observational and Mendelian randomization study. J Affect Disord 2025; 372:440-450. [PMID: 39672473 DOI: 10.1016/j.jad.2024.12.038] [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: 06/18/2024] [Revised: 09/27/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Modifiable lifestyle factors are implicated as risk factors for neurological and psychiatric disorders, but whether these associations are causal remains uncertain. We aimed to evaluate associations and ascertain causal relationships between modifiable lifestyle factors, neurological and psychiatric disorder risk, and brain structural magnetic resonance imaging (MRI) markers. METHODS We analyzed data from over 50,000 UK Biobank participants with self-reported lifestyle factors, including alcohol consumption, smoking, physical activity, diet, sleep, electronic device use, and sexual factors. Primary outcomes were stroke, all-cause dementia, Parkinson's disease (PD), Major depression disorder (MDD), Anxiety Disorders (ANX), and Bipolar Disorder (BIP), alongside MRI markers. Summary statistics were obtained from genome-wide association studies and Mendelian randomization (MR) analyses investigated bidirectional associations between lifestyle factors, neurological/psychiatric disorders, and MRI markers, with mediation assessed using multivariable Mendelian randomization (MVMR). RESULTS Cross-sectional analyses identified lifestyle factors were associated with neurological and psychiatric disorders and brain morphology. MR confirmed causal relationships, including lifetime smoking index on Stroke, PD, MDD, ANX and BIP; play computer games on BIP; leisure screen time on Stroke and MDD; automobile speeding propensity on MDD; sexual factors on MDD and BIP; sleep characteristics on BIP and MDD. Brain structure mediated several lifestyle-disorder associations, such as daytime dozing and dementia, lifetime smoking and PD and age first had sexual intercourse and PD. CONCLUSION Our results provide support for a causal effect of multiple lifestyle measures on the risk of neurological and psychiatric disorders, with brain structural morphology serving as a potential biological mediator in their associations.
Collapse
Affiliation(s)
- Yiran Dong
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Pan Zhang
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Jinghui Zhong
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Jinjing Wang
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yingjie Xu
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Hongmei Huang
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xinfeng Liu
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Wen Sun
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
12
|
İş Ö, Min Y, Wang X, Oatman SR, Abraham Daniel A, Ertekin‐Taner N. Multi Layered Omics Approaches Reveal Glia Specific Alterations in Alzheimer's Disease: A Systematic Review and Future Prospects. Glia 2025; 73:539-573. [PMID: 39652363 PMCID: PMC11784841 DOI: 10.1002/glia.24652] [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: 08/12/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 02/01/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative dementia with multi-layered complexity in its molecular etiology. Multiple omics-based approaches, such as genomics, epigenomics, transcriptomics, proteomics, metabolomics, and lipidomics are enabling researchers to dissect this molecular complexity, and to uncover a plethora of alterations yielding insights into the pathophysiology of this disease. These approaches reveal multi-omics alterations essentially in all cell types of the brain, including glia. In this systematic review, we screen the literature for human studies implementing any omics approach within the last 10 years, to discover AD-associated molecular perturbations in brain glial cells. The findings from over 200 AD-related studies are reviewed under four different glial cell categories: microglia, oligodendrocytes, astrocytes and brain vascular cells. Under each category, we summarize the shared and unique molecular alterations identified in glial cells through complementary omics approaches. We discuss the implications of these findings for the development, progression and ultimately treatment of this complex disease as well as directions for future omics studies in glia cells.
Collapse
Affiliation(s)
- Özkan İş
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Yuhao Min
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Xue Wang
- Department of Quantitative Health SciencesMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Nilüfer Ertekin‐Taner
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Department of NeurologyMayo ClinicJacksonvilleFloridaUSA
| |
Collapse
|
13
|
Garijo D, Yang Q, Vargas H, Gadewar SP, Low K, Ratnakar V, Osorio M, Zhu AH, McMahon A, Gil Y, Jahanshad N. NeuroDISK: An AI Approach to Automate Continuous Inquiry-Driven Discoveries in Neuroimaging Genetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638360. [PMID: 40027637 PMCID: PMC11870421 DOI: 10.1101/2025.02.14.638360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Collaborative and multi-site neuroimaging studies have greatly accelerated the rate at which new and existing data can be aggregated to answer a neuroscientific question. New research initiatives are continuously collecting more data, allowing opportunities to refine previous published findings through continuous and dynamic updates. Yet, we lack a practical framework for researchers to systematically, automatically, and continuously update published findings. We developed NeuroDISK, an automated artificial intelligence based framework that: 1) performs automated and inquiry-driven analyses, and 2) continuously updates these analyses as new data becomes available. NeuroDISK was evaluated using published results from the ENIGMA consortium's work on the genetic architecture of the cerebral cortex. We incorporate both meta-analysis and meta-regression options to showcase our framework on the effect of specific genotypes and moderators on select brain regions. Initial NeuroDISK meta-analysis results replicate the original publication, and we show result updates after adding new data. The NeuroDISK framework can be generalized for users to define question(s), run corresponding workflow(s) and access results interactively and continuously.
Collapse
Affiliation(s)
- Daniel Garijo
- Information Sciences Institute, University of Southern California, Marina del Rey, California, USA
- Ontology Engineering Group, Universidad Politécnica de Madrid, Madrid, Spain
| | - Qifan Yang
- Laboratory of Brain eScience, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, United States
| | - Hernán Vargas
- Information Sciences Institute, University of Southern California, Marina del Rey, California, USA
| | - Shruti P. Gadewar
- Laboratory of Brain eScience, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, United States
| | - Kevin Low
- Laboratory of Brain eScience, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, United States
| | - Varun Ratnakar
- Information Sciences Institute, University of Southern California, Marina del Rey, California, USA
| | - Maximiliano Osorio
- Information Sciences Institute, University of Southern California, Marina del Rey, California, USA
| | - Alyssa H. Zhu
- Laboratory of Brain eScience, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, United States
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Marina del Rey, California, USA
| | - Agnes McMahon
- Laboratory of Brain eScience, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, United States
| | - Yolanda Gil
- Information Sciences Institute, University of Southern California, Marina del Rey, California, USA
| | - Neda Jahanshad
- Laboratory of Brain eScience, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, United States
- Department of Neurology, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Marina del Rey, California, USA
| |
Collapse
|
14
|
Constantino-Pettit A, Gilbert K, Boone K, Luking K, Geselowitz B, Tillman R, Whalen D, Luby J, Barch DM, Vogel A. Associations of Child Amygdala Development With Borderline Personality Symptoms During Adolescence. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2025:S2451-9022(25)00033-3. [PMID: 39884355 DOI: 10.1016/j.bpsc.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND The current understanding of the neural correlates of borderline personality disorder (BPD) is limited, but some evidence suggests that alterations in limbic structures play a role in adult BPD. The developmental course of structural neural differences in BPD is unknown. Whether there is specificity for structural alterations in BPD compared to other psychiatric presentations, such as major depressive disorder (MDD), remains unexplored. In the current study, we examined childhood trajectories of 2 limbic regions that have been implicated in BPD, hippocampal and amygdala volume, as they relate to adolescent BPD symptoms compared to MDD symptoms. METHODS Participants (n = 175; 85 [48.6%] female) were from a 17-year longitudinal study of preschool depression. Participants completed up to 5 magnetic resonance imaging scans from late childhood through adolescence. General linear models were used to examine the relationship between gray matter volume intercepts/slopes and BPD symptoms to understand the influence of the developmental trajectory of brain regions on BPD. Separate models were used to examine the relationship between MDD symptoms and volume intercepts to assess diagnostic specificity. RESULTS Lower childhood amygdala volume (intercept; age 13 centered) across scans was associated with higher adolescent BPD symptoms (β = -0.25, adjusted p = .015). There was no relationship between the slope of amygdala volume and BPD symptoms. There was no relationship between hippocampal volume and BPD or any relationship between amygdala or hippocampal volume and MDD symptoms during adolescence. CONCLUSIONS Our findings add evidence that supports the role of alterations in amygdala structure in BPD development. Decreased amygdala volume as early as age 13 may be an early indicator of the development of BPD during adolescence.
Collapse
Affiliation(s)
- Anna Constantino-Pettit
- Department of Psychiatry, Washington University in St. Louis, School of Medicine, St. Louis, Missouri.
| | - Kirsten Gilbert
- Department of Psychiatry, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Kiran Boone
- Department of Psychology, University of Houston, Houston, Texas
| | - Katherine Luking
- Department of Psychology, St. Louis University, St. Louis, Missouri
| | - Benjamin Geselowitz
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rebecca Tillman
- Department of Psychiatry, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Diana Whalen
- Department of Psychiatry, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Joan Luby
- Department of Psychiatry, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Deanna M Barch
- Department of Psychiatry, Washington University in St. Louis, School of Medicine, St. Louis, Missouri; Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri
| | - Alecia Vogel
- Department of Psychiatry, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| |
Collapse
|
15
|
Ma X, Feng N, Palaniyappan L, Cao L, Gu Z, Kang J, Yuan L, Ouyang L, Wang Y, Li C, Jin K, Chen X, Feng J, He Y, Luo Q. Neuroimaging stratification reveals the striatal vulnerability to stress as a risk for schizophrenia. Transl Psychiatry 2025; 15:18. [PMID: 39843416 PMCID: PMC11754660 DOI: 10.1038/s41398-025-03237-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
The striatum, a core brain structure relevant for schizophrenia, exhibits heterogeneous volumetric changes in this illness. Due to this heterogeneity, its role in the risk of developing schizophrenia following exposure to environmental stress remains poorly understood. Using the putamen (a subnucleus of the striatum) as an indicator for convergent genetic risk of schizophrenia, 63 unaffected first-degree relatives of patients (22.08 ± 4.80 years) with schizophrenia (UFR-SZ) were stratified into two groups. Compared with healthy controls (HC; n = 59), voxel-based and brain-wide volumetric changes and their associations with stressful life events (SLE) were tested. These stratified associations were validated using two large population-based cohorts (the ABCD study; n = 1680, 11.92 ± 0.62 years; and UK Biobank, n = 20547, 55.38 ± 7.43 years). Transcriptomic analysis of brain tissues was used to identify the biological processes associated with the brain mediation effects on the SLE-psychosis relationship. The stratified UFR-SZ subgroup with smaller right putamen had a smaller volume in the left caudate when compared to HC; this caudate volume was associated with both a higher level of SLE and more psychotic symptoms. This caudate-SLE association was replicated in two independent large-scale cohorts, when individuals were stratified by both a higher polygenic burden for schizophrenia and smaller right putamen. In UFR-SZ, the caudate cluster mediated the relationship between SLE and more psychotic symptoms. This mediation was associated with the genes enriched in both glutamatergic synapses and response to oxidative stress. The stratified association between the striatum and stress highlights the differential vulnerability to stress, contributing to the complexity of the gene-by-environment etiology of schizophrenia.
Collapse
Affiliation(s)
- Xiaoqian Ma
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- China National Technology Institute on Mental Disorders, Changsha, Hunan, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China
- Institute of Mental Health, Central South University, Changsha, Hunan, China
| | - Nana Feng
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, PR China
| | - Lena Palaniyappan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
- Robarts Research Institute, London, ON, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Luolong Cao
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, PR China
| | - Zixin Gu
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, PR China
| | - Jujiao Kang
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, PR China
| | - Liu Yuan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Lijun Ouyang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Yujue Wang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Chunwang Li
- Department of Radiology, Hunan Children's Hospital, Changsha, China
| | - Ke Jin
- Department of Radiology, Hunan Children's Hospital, Changsha, China
| | - Xiaogang Chen
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Jianfeng Feng
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, PR China
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Ying He
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
- China National Technology Institute on Mental Disorders, Changsha, Hunan, China.
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China.
- Institute of Mental Health, Central South University, Changsha, Hunan, China.
| | - Qiang Luo
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, PR China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenome Institute, Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, 200032, PR China.
- MOE-Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
16
|
Xie W, Zheng J, Kong C, Luo W, Lin X, Zhou Y. Revealing potential drug targets in schizophrenia through proteome-wide Mendelian randomization genetic insights. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111208. [PMID: 39615872 DOI: 10.1016/j.pnpbp.2024.111208] [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: 07/08/2024] [Revised: 11/23/2024] [Accepted: 11/24/2024] [Indexed: 01/29/2025]
Abstract
BACKGROUND Schizophrenia (SCZ) is a severe, chronic mental disorder with no current cure. Identifying novel pharmacological targets is crucial for developing more effective treatments. METHODS We performed two-sample Mendelian randomization (MR) analyses to estimate the associations between cerebrospinal fluid (CSF) containing 154 proteins and plasma containing 734 proteins and risk of SCZ. Bidirectional MR analysis, steiger filtering, bayesian colocalization, phenotypic scanning, and validation analysis were examined to validate the assumptions of MR. For proteins significantly associated with SCZ identified by MR, we explored their potential impact on brain structures, including cortical surface area (SA), thickness (TH), and the volume of subcortical structures. RESULTS MR analysis identified 13 protein-SCZ pairs at Bonferroni significance (P < 5.63 × 10-5). Notably, the genetically proxied protein level of neuromedin B (NMB) was associated with an increased risk for SCZ (odds ratio [OR] = 1.41; 95 % CI, 1.27 to 1.58; P = 6.68 × 10-10). Bayesian colocalization suggested that NMB shares genetic variations with SCZ. Further, NMB interacts with target proteins of current SCZ drugs and was validated in the UK Biobank. The genetically proxied NMB was positively associated with an increase in the surface area (SA) of the parahippocampal gyrus (β = 8.93 mm2, 95 % CI, 1.58 to 16.3, P = .02). Additionally, an increase in the genetically proxied SA of the parahippocampal gyrus was inversely associated with the risk of SCZ (OR = 0.996, 95 % CI, 0.993 to 0.999, P = .04). CONCLUSIONS The findings suggest that NMB may represent a promising target for pharmacological intervention in SCZ. This warrants further investigation into the specific constituents involved, which could have potential for follow-up studies.
Collapse
Affiliation(s)
- Wenhuo Xie
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jiaping Zheng
- Department of Rehabilitation Medicine, School of Health, Fujian Medical University, Fuzhou, China
| | - Chenghua Kong
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Wei Luo
- Department of Rehabilitation Medicine, School of Health, Fujian Medical University, Fuzhou, China
| | - Xiaoxia Lin
- Department of Pediatrics, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Yu Zhou
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
17
|
van der Es T, Soheili-Nezhad S, Roth Mota N, Franke B, Buitelaar J, Sprooten E. Exploring the genetic architecture of brain structure and ADHD using polygenic neuroimaging-derived scores. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e32987. [PMID: 39016115 DOI: 10.1002/ajmg.b.32987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 04/24/2024] [Accepted: 05/11/2024] [Indexed: 07/18/2024]
Abstract
Genome-wide association studies (GWAS) have provided valuable insights into the genetic basis of neuropsychiatric disorders and highlighted their complexity. Careful consideration of the polygenicity and complex genetic architecture could aid in the understanding of the underlying brain mechanisms. We introduce an innovative approach to polygenic scoring, utilizing imaging-derived phenotypes (IDPs) to predict a clinical phenotype. We leveraged IDP GWAS data from the UK Biobank, to create polygenic imaging-derived scores (PIDSs). As a proof-of-concept, we assessed genetic variations in brain structure between individuals with ADHD and unaffected controls across three NeuroIMAGE waves (n = 954). Out of the 94 PIDS, 72 exhibited significant associations with their corresponding IDPs in an independent sample. Notably, several global measures, including cerebellum white matter, cerebellum cortex, and cerebral white matter, displayed substantial variance explained for their respective IDPs, ranging from 3% to 5.7%. Conversely, the associations between each IDP and the clinical ADHD phenotype were relatively weak. These findings highlight the growing power of GWAS in structural neuroimaging traits, enabling the construction of polygenic scores that accurately reflect the underlying polygenic architecture. However, to establish robust connections between PIDS and behavioral or clinical traits such as ADHD, larger samples are needed. Our novel approach to polygenic risk scoring offers a valuable tool for researchers in the field of psychiatric genetics.
Collapse
Affiliation(s)
- Tim van der Es
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | | | - Nina Roth Mota
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, 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
| | - Jan Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Emma Sprooten
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, 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
| |
Collapse
|
18
|
Wang B, Zhang M, Fan F, Yuan C, Wang Z, Tan Y, Tan S. Subcortical and insula functional connectivity aberrations and clinical implications in first-episode schizophrenia. Asian J Psychiatr 2025; 103:104298. [PMID: 39591757 DOI: 10.1016/j.ajp.2024.104298] [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: 08/31/2024] [Revised: 10/18/2024] [Accepted: 10/27/2024] [Indexed: 11/28/2024]
Abstract
INTRODUCTION Schizophrenia is a complex mental disorder whose pathophysiology remains elusive, particularly in the roles of subcortex. This study aims to explore the role of subcortex and insula and their relationship with symptom changes in first-episode schizophrenia (FES) patients by utilizing machine learning algorithms and functional connectivity (FC). METHODS The study encompasses 261 participants, sourced from two independent samples of FES patients and their matched healthy controls (HC). The discovery dataset includes 77 FES patients at baseline (FES0W) and 77 matched HCs, with the patients undergoing a follow-up scan after eight weeks of antipsychotic treatment (FES8W, N = 34). A validation dataset from another region comprises 47 FES patients and 47 matched HCs. RESULTS Significant differences in subcortical FCs were observed between FES and controls, correlating with symptom severity and symptom changes. Machine learning models were developed to diagnose schizophrenia on an individual basis, achieving a balanced accuracy of 79.55 % across diverse centers. CONCLUSIONS These findings suggest that subcortical connectivity patterns offer potential as biomarkers for schizophrenia, enabling personalized treatment strategies and improving prognosis by facilitating early diagnosis.
Collapse
Affiliation(s)
- Bixin Wang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Meng Zhang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Fengmei Fan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Chunyu Yuan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Zhiren Wang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Yunlong Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Shuping Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China.
| |
Collapse
|
19
|
Li P, Liu X, Wu L, Dong L, Zhou J, Song Z. Thyroid Function and Brain Structure: Insight from a Mendelian Randomization Study. Neuroendocrinology 2024; 115:60-71. [PMID: 39653027 DOI: 10.1159/000542955] [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: 08/24/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
INTRODUCTION Thyroid hormones play a critical role in brain development. However, the precise causal associations between thyroid function and structural changes in specific brain regions remain uncertain. METHODS We applied the univariate Mendelian randomization (UVMR) method to assess the causal effects of thyroid function on brain structure. Genome-wide association study (GWAS) data on thyroid-related traits from the ThyroidOmics Consortium including free thyroxine (FT4), free tri-iodothyronine (FT3), thyroid-stimulating hormone (TSH), FT3/FT4 ratio, as well as dichotomized high and low TSH levels were used as exposures. GWAS data on cortical thickness, surface area, and volume of subcortical structures served as outcomes. Inverse variance weighted was the main estimate method. Subsequently, multivariable MR (MVMR) was conducted to validate significant causal associations identified in UVMR. RESULTS UVMR analysis demonstrated a statistically significant inverse association between genetically predicted FT4 and putamen volume (β = -71.91 mm3, 95% confidence interval: -112.11 mm3 to -31.71 mm3, p = 4.54 × 10-4). The findings were robust in sensitivity analysis. MVMR analysis further confirmed a persistent causal relationship between FT4 and putamen volume after adjusting for FT3, TSH, and neuropsychiatric disorders. Functional enrichment analyses indicated the pathways by which FT4 influences putamen volume may be related to the thyroid hormone signaling pathway, sodium-independent organic anion transport, and Rap1 signaling pathway. CONCLUSION MR analysis provides evidence for causal relationships between thyroid function and brain structural alterations, particularly highlighting the impact of FT4 on putamen volume. Further research is warranted to elucidate the underlying mechanisms by which thyroid hormones modulate brain structure.
Collapse
Affiliation(s)
- Ping Li
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China,
| | - Xiao Liu
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Liming Wu
- Department of Urology, Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, China
| | - Liming Dong
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jianbo Zhou
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhihui Song
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
20
|
Mahmoodi M, Ayatollahi Mehrgardi A, Momen M, Serpell JA, Esmailizadeh A. Deciphering the genetic basis of behavioral traits in dogs: Observed-trait GWAS and latent-trait GWAS analysis reveal key genes and variants. Vet J 2024; 308:106251. [PMID: 39368730 DOI: 10.1016/j.tvjl.2024.106251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Dogs exhibit remarkable phenotypic diversity, particularly in behavioral traits, making them an excellent model for studying the genetic basis of complex behaviors. Behavioral traits such as aggression and fear are highly heritable among different dog breeds, but their genetic basis is largely unknown. We used the genome-wide association study (GWAS) to identify candidate genes associated with nine behavioral traits including; stranger-directed aggression (SDA), owner-directed aggression (ODA), dog-directed aggression (DDA), stranger-directed fear (SDF), nonsocial fear (NF), dog-directed fear (DDF), touch sensitivity (TS), separation-related behavior (SRB) and attachment attention-seeking (AAS). The observed behavioral traits were collected from 38,714 to 40,460 individuals across 108 modern dog breeds. We performed a GWAS based on a latent trait extracted using the confirmatory factor analysis (CFA) method with nine observable behavioral traits and compared the results with those from the GWAS of the observed traits. Using both observed-trait and latent-trait GWAS, we identified 41 significant SNPs that were common between both GWAS methods, of which 26 were pleiotropic, as well as 10 SNPs unique to the latent-trait GWAS, and 5 SNPs unique to the observed-trait GWAS discovered. These SNPs were associated with 21 genes in latent-trait GWAS and 22 genes in the observed-trait GWAS, with 19 genes shared by both. According to previous studies, some of the genes from this study have been reported to be related to behavioral and neurological functions in dogs. In the human population, these identified genes play a role in either the formation of the nervous system or are linked to various mental health conditions. Taken together, our findings suggest that latent-trait GWAS for behavioral traits in dogs identifies significant latent genes that are neurologically prioritized.
Collapse
Affiliation(s)
- Maryam Mahmoodi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran.
| | - Ahmad Ayatollahi Mehrgardi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran.
| | - Mehdi Momen
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - James A Serpell
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ali Esmailizadeh
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
21
|
Zhou J, Gong L, Liu X, Chen L, Yang Z. Mendelian randomization in Alzheimer's disease and mild cognitive impairment: Hippocampal volume associations. Neuroscience 2024; 561:30-42. [PMID: 39368607 DOI: 10.1016/j.neuroscience.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
This study investigates the association between cognitive dysfunction and hippocampal volumes in Alzheimer's Disease (AD) and Mild Cognitive Impairment (MCI) using Mendelian randomization. A meta-analysis of 503 healthy controls, 562 MCI patients, and 389 CE patients revealed significant reductions in hippocampal and subregion volumes in MCI and AD compared to controls. While various subregions showed volume reductions, no causal relationship between hippocampal volume and AD was established through Mendelian randomization analysis. In conclusion, significant volume reductions were observed in MCI and AD patients, highlighting the complexity of the relationship between hippocampal volume and cognitive impairment.
Collapse
Affiliation(s)
- Jianguo Zhou
- Department of Radiology, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang 222004, PR China
| | - Lei Gong
- Department of Radiology, The Fourth People's Hospital of Lianyungang, Affiliated Hospital of Nanjing Medical University Kangda, Lianyungang 222000, PR China
| | - Xiaoli Liu
- Department of Rehabilitation, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang 222004, PR China
| | - Liping Chen
- Department of Rehabilitation, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang 222004, PR China
| | - Zhou Yang
- Department of Rehabilitation, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang 222004, PR China.
| |
Collapse
|
22
|
Shi R, Chang X, Banaschewski T, Barker GJ, Bokde ALW, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Martinot17, MLP, Artiges E, Nees F, Orfanos DP, Poustka L, Hohmann S, Holz N, Smolka MN, Vaidya N, Walter H, Whelan R, Schumann G, Lin X, Feng J, IMAGEN Consortium. Gene-environment interactions in the influence of maternal education on adolescent neurodevelopment using ABCD study. SCIENCE ADVANCES 2024; 10:eadp3751. [PMID: 39546599 PMCID: PMC11567010 DOI: 10.1126/sciadv.adp3751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024]
Abstract
Maternal education was strongly correlated with adolescent brain morphology, cognitive performances, and mental health. However, the molecular basis for the effects of maternal education on the structural neurodevelopment remains unknown. Here, we conducted gene-environment-wide interaction study using the Adolescent Brain Cognitive Development cohort. Seven genomic loci with significant gene-environment interactions (G×E) on regional gray matter volumes were identified, with enriched biological functions related to metabolic process, inflammatory process, and synaptic plasticity. Additionally, genetic overlapping results with behavioral and disease-related phenotypes indicated shared biological mechanism between maternal education modified neurodevelopment and related behavioral traits. Finally, by decomposing the multidimensional components of maternal education, we found that socioeconomic status, rather than family environment, played a more important role in modifying the genetic effects on neurodevelopment. In summary, our study provided analytical evidence for G×E effects regarding adolescent neurodevelopment and explored potential biological mechanisms as well as social mechanisms through which maternal education could modify the genetic effects on regional brain development.
Collapse
Affiliation(s)
- Runye Shi
- School of Data Science, Fudan University, Shanghai, China
| | - Xiao Chang
- 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
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Gareth J. Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Arun L. W. Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, 68131 Mannheim, Germany
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, 05405 Burlington, VT, USA
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, UK
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Charité–Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 “Trajectoires développementales en psychiatrie”, Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
| | | | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 “Trajectoires développementales en psychiatrie”, Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein Kiel University, Kiel, Germany
| | | | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie Holz
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Michael N. Smolka
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité–Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Germany
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China
| | - Xiaolei Lin
- School of Data Science, Fudan University, Shanghai, China
| | - Jianfeng Feng
- School of Data Science, Fudan University, 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
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, UK
| | | |
Collapse
|
23
|
Bas-Hoogendam JM. Genetic Vulnerability to Social Anxiety Disorder. Curr Top Behav Neurosci 2024. [PMID: 39543021 DOI: 10.1007/7854_2024_544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Most anxiety disorders 'run within families': people suffering from an anxiety disorder often have family members who are highly anxious as well. In this chapter, we explore recent work devoted to unraveling the complex interplay between genes and environment in the development of anxiety. We review studies focusing on the genetic vulnerability to develop social anxiety disorder (SAD), as SAD is one of the most prevalent anxiety disorders, with an early onset, a chronic course, and associated with significant life-long impairments. More insight into the development of SAD is thus of uttermost importance.First, we will discuss family studies, twin studies, and large-sized population-based registry studies and explain what these studies can reveal about the genetic vulnerability to develop anxiety. Next, we describe the endophenotype approach; in this context, we will summarize results from the Leiden Family Lab study on Social Anxiety Disorder. Subsequently, we review the relationship between the heritable trait 'behavioral inhibition' and the development of SAD, and highlight the relevance of this work for the development and improvement of preventative and therapeutic interventions for socially anxious youth.
Collapse
Affiliation(s)
- Janna Marie Bas-Hoogendam
- Leiden University, Leiden, The Netherlands.
- Leiden University Medical Center, Leiden, The Netherlands.
- Leiden Institute for Brain and Cognition, Leiden, The Netherlands.
| |
Collapse
|
24
|
Yu C, Zhang S, Shang M, Guo L, Han J, Du L. A Multi-Task Deep Feature Selection Method for Brain Imaging Genetics. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:1613-1622. [PMID: 37432805 DOI: 10.1109/tcbb.2023.3294413] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Using brain imaging quantitative traits (QTs) for identifying genetic risk factors is an important research topic in brain imaging genetics. Many efforts have been made for this task via building linear models between imaging QTs and genetic factors such as single nucleotide polymorphisms (SNPs). To the best of our knowledge, linear models could not fully uncover the complicated relationship due to the loci's elusive and diverse influences on imaging QTs. In this paper, we propose a novel multi-task deep feature selection (MTDFS) method for brain imaging genetics. MTDFS first builds a multi-task deep neural network to model the complicated associations between imaging QTs and SNPs. And then designs a multi-task one-to-one layer and imposes a combined penalty to identify SNPs that make significant contributions. MTDFS can not only extract the nonlinear relationship but also arms the deep neural network with feature selection. We compared MTDFS to multi-task linear regression (MTLR) and single-task DFS (DFS) methods on the real neuroimaging genetic data. The experimental results showed that MTDFS performed better than MTLR and DFS on the QT-SNP relationship identification and feature selection. Thus, MTDFS is powerful for identifying risk loci and could be a great supplement to brain imaging genetics.
Collapse
|
25
|
Pawlak M, Kemp J, Bray S, Chenji S, Noel M, Birnie KA, MacMaster FP, Miller JV, Kopala-Sibley DC. Macrostructural Brain Morphology as Moderator of the Relationship Between Pandemic-Related Stress and Internalizing Symptomology During COVID-19 in High-Risk Adolescents. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:1141-1177. [PMID: 39019399 DOI: 10.1016/j.bpsc.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND According to person-by-environment models, individual differences in traits may moderate the association between stressors and the development of psychopathology; however, findings in the literature have been inconsistent and little literature has examined adolescent brain structure as a moderator of the effects of stress on adolescent internalizing symptoms. The COVID-19 pandemic presented a unique opportunity to examine the associations between stress, brain structure, and psychopathology. Given links of cortical morphology with adolescent depression and anxiety, the current study investigated whether cortical morphology moderated the relationship between stress from the COVID-19 pandemic and the development of internalizing symptoms in familial high-risk adolescents. METHODS Prior to the COVID-19 pandemic, 72 adolescents (27 male) completed a measure of depressive and anxiety symptoms and underwent magnetic resonance imaging. T1-weighted images were acquired to assess cortical thickness and surface area. Approximately 6 to 8 months after COVID-19 was declared a global pandemic, adolescents reported their depressive and anxiety symptoms and pandemic-related stress. RESULTS Adjusting for pre-pandemic depressive and anxiety symptoms and stress, increased pandemic-related stress was associated with increased depressive but not anxiety symptoms. This relationship was moderated by cortical thickness and surface area in the anterior cingulate and cortical thickness in the medial orbitofrontal cortex such that increased stress was only associated with increased depressive and anxiety symptoms among adolescents with lower cortical surface area and higher cortical thickness in these regions. CONCLUSIONS Results further our understanding of neural vulnerabilities to the associations between stress and internalizing symptoms in general and during the COVID-19 pandemic in particular.
Collapse
Affiliation(s)
- McKinley Pawlak
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada.
| | - Jennifer Kemp
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
| | - Signe Bray
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Child and Adolescent Imaging Research Program, University of Calgary, Calgary, Alberta, Canada; Department of Radiology, University of Calgary, Calgary, Alberta, Canada; Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Sneha Chenji
- Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada
| | - Melanie Noel
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| | - Kathryn A Birnie
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada; Department of Anesthesiology, Perioperative, and Pain Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Frank P MacMaster
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada; IWK Health, Halifax, Nova Scotia, Canada
| | - Jillian Vinall Miller
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Anesthesiology, Perioperative, and Pain Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daniel C Kopala-Sibley
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada; Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
26
|
Shao X, Li A, Wang Z, Xue G, Zhu B. False recall is associated with larger caudate in males but not in females. Memory 2024; 32:1341-1348. [PMID: 38416016 DOI: 10.1080/09658211.2024.2319314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024]
Abstract
After learning semantically related words, some people are more likely than others to incorrectly recall unstudied but semantically related lures (i.e., Deese-Roediger-McDermott [DRM] false recall). Previous studies have suggested that neural activity in subcortical regions (e.g., the caudate) is involved in false memory, and that there may be sex differences in the neural basis of false memory. However, sex-specific associations between subcortical volumes and false memory are not well understood. This study investigated whether sex modulates the associations between subcortical volumes and DRM false recall in 400 healthy college students. Volumes of subcortical regions including the caudate, accumbens, amygdala, hippocampus, pallidum, putamen and thalamus were obtained from structural magnetic resonance images and measured using FreeSurfer. The results showed that males had lower true and false recall but larger subcortical volumes than females. Interestingly, higher false recall was associated with a larger caudate in males, but not in females. This association was significant after controlling for age and intracranial volume. This study provides new evidence on the neural basis of false recall. It suggests that the caudate plays a role in false recall in young men, and that future studies of the neural correlates of false memory should consider sex differences.
Collapse
Affiliation(s)
- Xuhao Shao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, People's Republic of China
- Institute of Developmental Psychology, Beijing Normal University, Beijing, People's Republic of China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, People's Republic of China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, People's Republic of China
- Collaborative Innovation Center of Assessment for Basic Education Quality, Beijing Normal University, Beijing, People's Republic of China
| | - Ao Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, People's Republic of China
| | - Zehua Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, People's Republic of China
| | - Gui Xue
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, People's Republic of China
| | - Bi Zhu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, People's Republic of China
- Institute of Developmental Psychology, Beijing Normal University, Beijing, People's Republic of China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, People's Republic of China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, People's Republic of China
| |
Collapse
|
27
|
García-Marín LM, Campos AI, Diaz-Torres S, Rabinowitz JA, Ceja Z, Mitchell BL, Grasby KL, Thorp JG, Agartz I, Alhusaini S, Ames D, Amouyel P, Andreassen OA, Arfanakis K, Arias-Vasquez A, Armstrong NJ, Athanasiu L, Bastin ME, Beiser AS, Bennett DA, Bis JC, Boks MPM, Boomsma DI, Brodaty H, Brouwer RM, Buitelaar JK, Burkhardt R, Cahn W, Calhoun VD, Carmichael OT, Chakravarty M, Chen Q, Ching CRK, Cichon S, Crespo-Facorro B, Crivello F, Dale AM, Smith GD, de Geus EJC, De Jager PL, de Zubicaray GI, Debette S, DeCarli C, Depondt C, Desrivières S, Djurovic S, Ehrlich S, Erk S, Espeseth T, Fernández G, Filippi I, Fisher SE, Fleischman DA, Fletcher E, Fornage M, Forstner AJ, Francks C, Franke B, Ge T, Goldman AL, Grabe HJ, Green RC, Grimm O, Groenewold NA, Gruber O, Gudnason V, Håberg AK, Haukvik UK, Heinz A, Hibar DP, Hilal S, Himali JJ, Ho BC, Hoehn DF, Hoekstra PJ, Hofer E, Hoffmann W, Holmes AJ, Homuth G, Hosten N, Ikram MK, Ipser JC, Jack CR, Jahanshad N, Jönsson EG, Kahn RS, Kanai R, Klein M, Knol MJ, Launer LJ, Lawrie SM, Hellard SL, Lee PH, Lemaître H, Li S, Liewald DCM, Lin H, Longstreth WT, Lopez OL, Luciano M, et alGarcía-Marín LM, Campos AI, Diaz-Torres S, Rabinowitz JA, Ceja Z, Mitchell BL, Grasby KL, Thorp JG, Agartz I, Alhusaini S, Ames D, Amouyel P, Andreassen OA, Arfanakis K, Arias-Vasquez A, Armstrong NJ, Athanasiu L, Bastin ME, Beiser AS, Bennett DA, Bis JC, Boks MPM, Boomsma DI, Brodaty H, Brouwer RM, Buitelaar JK, Burkhardt R, Cahn W, Calhoun VD, Carmichael OT, Chakravarty M, Chen Q, Ching CRK, Cichon S, Crespo-Facorro B, Crivello F, Dale AM, Smith GD, de Geus EJC, De Jager PL, de Zubicaray GI, Debette S, DeCarli C, Depondt C, Desrivières S, Djurovic S, Ehrlich S, Erk S, Espeseth T, Fernández G, Filippi I, Fisher SE, Fleischman DA, Fletcher E, Fornage M, Forstner AJ, Francks C, Franke B, Ge T, Goldman AL, Grabe HJ, Green RC, Grimm O, Groenewold NA, Gruber O, Gudnason V, Håberg AK, Haukvik UK, Heinz A, Hibar DP, Hilal S, Himali JJ, Ho BC, Hoehn DF, Hoekstra PJ, Hofer E, Hoffmann W, Holmes AJ, Homuth G, Hosten N, Ikram MK, Ipser JC, Jack CR, Jahanshad N, Jönsson EG, Kahn RS, Kanai R, Klein M, Knol MJ, Launer LJ, Lawrie SM, Hellard SL, Lee PH, Lemaître H, Li S, Liewald DCM, Lin H, Longstreth WT, Lopez OL, Luciano M, Maillard P, Marquand AF, Martin NG, Martinot JL, Mather KA, Mattay VS, McMahon KL, Mecocci P, Melle I, Meyer-Lindenberg A, Mirza-Schreiber N, Milaneschi Y, Mosley TH, Mühleisen TW, Müller-Myhsok B, Maniega SM, Nauck M, Nho K, Niessen WJ, Nöthen MM, Nyquist PA, Oosterlaan J, Pandolfo M, Paus T, Pausova Z, Penninx BWJH, Pike GB, Psaty BM, Pütz B, Reppermund S, Rietschel MD, Risacher SL, Romanczuk-Seiferth N, Romero-Garcia R, Roshchupkin GV, Rotter JI, Sachdev PS, Sämann PG, Saremi A, Sargurupremraj M, Saykin AJ, Schmaal L, Schmidt H, Schmidt R, Schofield PR, Scholz M, Schumann G, Schwarz E, Shen L, Shin J, Sisodiya SM, Smith AV, Smoller JW, Soininen HS, Steen VM, Stein DJ, Stein JL, Thomopoulos SI, Toga AW, Tordesillas-Gutiérrez D, Trollor JN, Valdes-Hernandez MC, van T Ent D, van Bokhoven H, van der Meer D, van der Wee NJA, Vázquez-Bourgon J, Veltman DJ, Vernooij MW, Villringer A, Vinke LN, Völzke H, Walter H, Wardlaw JM, Weinberger DR, Weiner MW, Wen W, Westlye LT, Westman E, White T, Witte AV, Wolf C, Yang J, Zwiers MP, Ikram MA, Seshadri S, Thompson PM, Satizabal CL, Medland SE, Rentería ME. Genomic analysis of intracranial and subcortical brain volumes yields polygenic scores accounting for variation across ancestries. Nat Genet 2024; 56:2333-2344. [PMID: 39433889 DOI: 10.1038/s41588-024-01951-z] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/18/2024] [Indexed: 10/23/2024]
Abstract
Subcortical brain structures are involved in developmental, psychiatric and neurological disorders. Here we performed genome-wide association studies meta-analyses of intracranial and nine subcortical brain volumes (brainstem, caudate nucleus, putamen, hippocampus, globus pallidus, thalamus, nucleus accumbens, amygdala and the ventral diencephalon) in 74,898 participants of European ancestry. We identified 254 independent loci associated with these brain volumes, explaining up to 35% of phenotypic variance. We observed gene expression in specific neural cell types across differentiation time points, including genes involved in intracellular signaling and brain aging-related processes. Polygenic scores for brain volumes showed predictive ability when applied to individuals of diverse ancestries. We observed causal genetic effects of brain volumes with Parkinson's disease and attention-deficit/hyperactivity disorder. Findings implicate specific gene expression patterns in brain development and genetic variants in comorbid neuropsychiatric disorders, which could point to a brain substrate and region of action for risk genes implicated in brain diseases.
Collapse
Affiliation(s)
- Luis M García-Marín
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Adrian I Campos
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Santiago Diaz-Torres
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jill A Rabinowitz
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Zuriel Ceja
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Brittany L Mitchell
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Katrina L Grasby
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Jackson G Thorp
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Ingrid Agartz
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm, Sweden
| | - Saud Alhusaini
- Department of Neurology, Alpert Medical School of Brown University, Providence, RI, USA
- Molecular and Cellular Therapeutics Department, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David Ames
- Academic Unit Psychiatry of Old Age, University of Melbourne, Melbourne, Victoria, Australia
- National Ageing Research Institute, Parkville, Victoria, Australia
| | - Philippe Amouyel
- Universite Lille, U1167-RID-AGE-LabEx DISTALZ-Risk Factors and Molecular Determinants of Aging Diseases, Lille, France
- Institut National de la Santé et de la Recherche Médicale, Lille, France
- Centre Hospitalier Universitaire de Lille Department of Public Health, Lille, France
- Institut Pasteur de Lille UMR1167, Lille, France
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Alejandro Arias-Vasquez
- Departments of Psychiatry and Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicola J Armstrong
- Department of Mathematics and Statistics, Curtin University, Perth, Western Australia, Australia
| | - Lavinia Athanasiu
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- CoE NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Mark E Bastin
- Centre for Clinical Brain Sciences and Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Alexa S Beiser
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Marco P M Boks
- Brain Center University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dorret I Boomsma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU Amsterdam, Amsterdam, The Netherlands
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Rachel M Brouwer
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU Amsterdam, Amsterdam, The Netherlands
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg University, Regensburg, Germany
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Wiepke Cahn
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
- Altrecht Mental Health Institute, Utrecht, The Netherlands
| | - Vince D Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS)-Georgia State, Georgia Tech and Emory University, Atlanta, GA, USA
| | | | - Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Research Centre, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Qiang Chen
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Christopher R K Ching
- Laboratory of Neuro Imaging, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Sven Cichon
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Medical Genetics, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Benedicto Crespo-Facorro
- HU Virgen del Rocio, Instituto de Investigacion Biomedica IBIS-CSIC, Universidad de Sevilla, CIBERSAM, Sevilla, Spain
| | | | - Anders M Dale
- Center for Multimodal Imaging and Genetics, La Jolla, CA, USA
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York City, NY, USA
| | - Greig I de Zubicaray
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Stéphanie Debette
- INSERM U1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
- Department of Neurology, Institute of Neurodegenerative Diseases, Bordeaux University Hospital, Bordeaux, France
| | - Charles DeCarli
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California, Davis, Sacramento, CA, USA
| | - Chantal Depondt
- Department of Neurology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Sylvane Desrivières
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Stefan Ehrlich
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Susanne Erk
- German Center of Mental Health (DZPG), Partner Site Berlin/Potsdam, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Espeseth
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Psychology, Oslo New University College, Oslo, Norway
| | - Guillén Fernández
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Irina Filippi
- INSERM U1299, Paris Saclay University, Gif-sur-Yvette, France
| | - 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
| | - Debra A Fleischman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Evan Fletcher
- Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Myriam Fornage
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Andreas J Forstner
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Clyde Francks
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Barbara Franke
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Robert C Green
- Department of Medicine (Genetics), Mass General Brigham and Harvard Medical School, Boston, MA, USA
| | - Oliver Grimm
- Central Institute of Mental Health, Mannheim, Germany
- Goethe-University Frankfurt, Frankfurt, Germany
| | - Nynke A Groenewold
- Department of Psychiatry and Mental Health, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Asta K Håberg
- Department of Neuromedicine and Movement, NTNU Science, Trondheim, Norway
- MiDT National Research Center, Department of Research, St Olavs Hospital, Trondheim, Norway
| | - Unn K Haukvik
- Norwegian Centre for Mental Health Research (NORMENT), Department of Mental Health and Addiction, University of Oslo, Oslo, Norway
- Centre for Forensic Psychiatry Research, Oslo University Hospital, Oslo, Norway
| | - Andreas Heinz
- German Center of Mental Health (DZPG), Partner Site Berlin/Potsdam, Berlin, Germany
- Centre for Forensic Psychiatry Research, Oslo University Hospital, Oslo, Norway
| | - Derrek P Hibar
- Product Development, Genentech, Inc., South San Francisco, CA, USA
| | - Saima Hilal
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore City, Singapore
| | - Jayandra J Himali
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health Science Center San Antonio, San Antonio, TX, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Beng-Choon Ho
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Pieter J Hoekstra
- Department of Child and Adolescent Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Accare Child Study Center, Groningen, The Netherlands
| | - Edith Hofer
- Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Wolfgang Hoffmann
- German Centre for Neurodegenerative Diseases (DZNE)-Site Rostock/Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Avram J Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Norbert Hosten
- Department of Radiology, University Clinic Greifswald, Greifswald, Germany
| | - M Kamran Ikram
- Departments of Epidemiology and Neurology, Erasmus MC, Rotterdam, The Netherlands
| | - Jonathan C Ipser
- Department of Psychiatry and Mental Health, Neuroscience Institute, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | | | - Neda Jahanshad
- Laboratory of Neuro Imaging, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Erik G Jönsson
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Sciences, Stockholm Region, Stockholm, Sweden
| | - Rene S Kahn
- Altrecht Mental Health Institute, Utrecht, The Netherlands
| | | | - Marieke Klein
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Lenore J Launer
- Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | | | | | - Phil H Lee
- Center for Genomic Medicine, Mass General Brigham, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatry, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hervé Lemaître
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR 5293, CNRS, Université de Bordeaux, Bordeaux, France
| | - Shuo Li
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, USA
| | | | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - W T Longstreth
- Department of Neurology, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Oscar L Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michelle Luciano
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Pauline Maillard
- Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Andre F Marquand
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicholas G Martin
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jean-Luc Martinot
- Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale, INSERM U1299 'Trajectoires développementales Psychiatrie', Ecole Normale Supérieure Paris-Saclay, CNRS UMR 9010, Université Paris Cité, Centre Borelli, Gif sur Yvette, France
| | - Karen A Mather
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Katie L McMahon
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Patrizia Mecocci
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Clinical Geriatrics, NVS Department, Karolinska Institute, Huddinge, Sweden
| | - Ingrid Melle
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nazanin Mirza-Schreiber
- Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany
- Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep and Stress Program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Complex Trait Genetics Program, Amsterdam, The Netherlands
| | | | - Thomas W Mühleisen
- Institute of Neuroscience and Medicine (INM-1), Research Center 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 Hospital Basel and University of Basel, Basel, Switzerland
| | | | - Susana Muñoz Maniega
- Centre for Clinical Brain Sciences and Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wiro J Niessen
- University Medical Center Groningen, Groningen, The Netherlands
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Paul A Nyquist
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- General Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jaap Oosterlaan
- Clinical Neuropsychology Section, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Emma Children's Hospital, University Medical Centers Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Massimo Pandolfo
- Université Libre de Bruxelles, Brussels, Belgium
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Tomas Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| | - Zdenka Pausova
- Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - G Bruce Pike
- Departments of Radiology and Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, Seattle, WA, USA
| | - Benno Pütz
- Translational Psychiatry, Munich, Germany
| | - Simone Reppermund
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Department of Developmental Disability Neuropsychiatry, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Marcella D Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nina Romanczuk-Seiferth
- Department of Psychiatry and Neuroscience, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Psychology, Clinical Psychology and Psychotherapy, MSB Medical School Berlin, Berlin, Germany
| | - Rafael Romero-Garcia
- Departamento de Fisiología Médica y Biofísica, Instituto de Biomedicina de Sevilla (IBiS) HUVR/CSIC/Universidad de Sevilla/CIBERSAM, ISCIII, Sevilla, Spain
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Gennady V Roshchupkin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Neuropsychiatric Institute, The Prince of Wales Hospital, Randwick, New South Wales, Australia
| | | | - Arvin Saremi
- Laboratory of Neuro Imaging, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Muralidharan Sargurupremraj
- INSERM U1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lianne Schmaal
- Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- Orygen, Parkville, Victoria, Australia
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Gottfried Schatz Center for Signaling, Metabolism and Aging, Medical University Graz, Graz, Austria
| | - Reinhold Schmidt
- Department of Neurology, Medical University Graz Austria, Graz, Austria
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Markus Scholz
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Gunter Schumann
- German Center of Mental Health (DZPG), Partner Site Berlin/Potsdam, Berlin, Germany
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Centre for Population Neuroscience and Stratified Medicine (PONS), ISTBI, Fudan University, Shanghai, PR China
- PONS Centre, Department of Psychiatry, CCM, Charite Unversitaetsmedizin Berlin, Berlin, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean Shin
- The Hospital for Sick Children, Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hilkka S Soininen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Vidar M Steen
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Dan J Stein
- SAMRC Research Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Jason L Stein
- Department of Genetics and UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sophia I Thomopoulos
- Laboratory of Neuro Imaging, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Diana Tordesillas-Gutiérrez
- Instituto de Física de Cantabria (CSIC-UC), Santander, Spain
- Department of Radiology, Marqués de Valdecilla University Hospital, Valdecilla Biomedical Research Institute IDIVAL, Santander, Spain
| | - Julian N Trollor
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- The National Centre of Excellence in Intellectual Disability Health, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Maria C Valdes-Hernandez
- Centre for Clinical Brain Sciences and Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Dennis van T Ent
- Department of Biological Psychology and Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Hans van Bokhoven
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dennis van der Meer
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Nic J A van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
| | - Javier Vázquez-Bourgon
- Department of Psychiatry, University Hospital Marqués de Valdecilla-IDIVAL, Santander, Spain
- Departamento de Medicina y Psiquiatría, Universidad de Cantabria, Santander, Spain
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Sevilla, Spain
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human, Cognitive and Brain Sciences, Leipzig, Germany
- Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
| | - Louis N Vinke
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences and Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Centre, University of Edinburgh, Edinburgh, UK
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Michael W Weiner
- University of California, San Francisco, San Francisco, CA, USA
- Northern California Institute for Research and Education (NCIRE), San Francisco, CA, USA
- Veterans Administration Medical Center, San Francisco, CA, USA
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Lars T Westlye
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
| | - Tonya White
- Section on Social and Cognitive Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - A Veronica Witte
- Department of Neurology, Max Planck Institute for Human, Cognitive and Brain Sciences, Leipzig, Germany
- Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
| | | | - Jingyun Yang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Marcel P Zwiers
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sudha Seshadri
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Paul M Thompson
- Laboratory of Neuro Imaging, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Claudia L Satizabal
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, USA
- Department of Population Health Sciences and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Sarah E Medland
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
| | - Miguel E Rentería
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
28
|
Gopinath K, Hoopes A, Alexander DC, Arnold SE, Balbastre Y, Billot B, Casamitjana A, Cheng Y, Chua RYZ, Edlow BL, Fischl B, Gazula H, Hoffmann M, Keene CD, Kim S, Kimberly WT, Laguna S, Larson KE, Van Leemput K, Puonti O, Rodrigues LM, Rosen MS, Tregidgo HFJ, Varadarajan D, Young SI, Dalca AV, Iglesias JE. Synthetic data in generalizable, learning-based neuroimaging. IMAGING NEUROSCIENCE (CAMBRIDGE, MASS.) 2024; 2:1-22. [PMID: 39850547 PMCID: PMC11752692 DOI: 10.1162/imag_a_00337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 01/25/2025]
Abstract
Synthetic data have emerged as an attractive option for developing machine-learning methods in human neuroimaging, particularly in magnetic resonance imaging (MRI)-a modality where image contrast depends enormously on acquisition hardware and parameters. This retrospective paper reviews a family of recently proposed methods, based on synthetic data, for generalizable machine learning in brain MRI analysis. Central to this framework is the concept of domain randomization, which involves training neural networks on a vastly diverse array of synthetically generated images with random contrast properties. This technique has enabled robust, adaptable models that are capable of handling diverse MRI contrasts, resolutions, and pathologies, while working out-of-the-box, without retraining. We have successfully applied this method to tasks such as whole-brain segmentation (SynthSeg), skull-stripping (SynthStrip), registration (SynthMorph, EasyReg), super-resolution, and MR contrast transfer (SynthSR). Beyond these applications, the paper discusses other possible use cases and future work in our methodology. Neural networks trained with synthetic data enable the analysis of clinical MRI, including large retrospective datasets, while greatly alleviating (and sometimes eliminating) the need for substantial labeled datasets, and offer enormous potential as robust tools to address various research goals.
Collapse
Affiliation(s)
- Karthik Gopinath
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Andrew Hoopes
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Massachusetts Institute of Technology, Cambridge, MA, United States
| | | | - Steven E. Arnold
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Yael Balbastre
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Benjamin Billot
- Computer Science & Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | | - You Cheng
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Russ Yue Zhi Chua
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Brian L. Edlow
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Bruce Fischl
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Malte Hoffmann
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - C. Dirk Keene
- University of Washington, Seattle, WA, United States
| | | | - W. Taylor Kimberly
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Kathleen E. Larson
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Koen Van Leemput
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Oula Puonti
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Copenhagen University Hospital, København, Denmark
| | - Livia M. Rodrigues
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Universidade Estadual de Campinas, São Paulo, Brazil
| | - Matthew S. Rosen
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Divya Varadarajan
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sean I. Young
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Adrian V. Dalca
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Juan Eugenio Iglesias
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Massachusetts Institute of Technology, Cambridge, MA, United States
- University College London, London, England
| |
Collapse
|
29
|
Morey RA, Zheng Y, Bayly H, Sun D, Garrett ME, Gasperi M, Maihofer AX, Baird CL, Grasby KL, Huggins AA, Haswell CC, Thompson PM, Medland S, Gustavson DE, Panizzon MS, Kremen WS, Nievergelt CM, Ashley-Koch AE, Logue MW. Genomic structural equation modeling reveals latent phenotypes in the human cortex with distinct genetic architecture. Transl Psychiatry 2024; 14:451. [PMID: 39448598 PMCID: PMC11502831 DOI: 10.1038/s41398-024-03152-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024] Open
Abstract
Genetic contributions to human cortical structure manifest pervasive pleiotropy. This pleiotropy may be harnessed to identify unique genetically-informed parcellations of the cortex that are neurobiologically distinct from functional, cytoarchitectural, or other cortical parcellation schemes. We investigated genetic pleiotropy by applying genomic structural equation modeling (SEM) to map the genetic architecture of cortical surface area (SA) and cortical thickness (CT) for 34 brain regions recently reported in the ENIGMA cortical GWAS. Genomic SEM uses the empirical genetic covariance estimated from GWAS summary statistics with LD score regression (LDSC) to discover factors underlying genetic covariance, which we are denoting genetically informed brain networks (GIBNs). Genomic SEM can fit a multivariate GWAS from summary statistics for each of the GIBNs, which can subsequently be used for LD score regression (LDSC). We found the best-fitting model of cortical SA identified 6 GIBNs and CT identified 4 GIBNs, although sensitivity analyses indicated that other structures were plausible. The multivariate GWASs of the GIBNs identified 74 genome-wide significant (GWS) loci (p < 5 × 10-8), including many previously implicated in neuroimaging phenotypes, behavioral traits, and psychiatric conditions. LDSC of GIBN GWASs found that SA-derived GIBNs had a positive genetic correlation with bipolar disorder (BPD), and cannabis use disorder, indicating genetic predisposition to a larger SA in the specific GIBN is associated with greater genetic risk of these disorders. A negative genetic correlation was observed between attention deficit hyperactivity disorder (ADHD) and major depressive disorder (MDD). CT GIBNs displayed a negative genetic correlation with alcohol dependence. Even though we observed model instability in our application of genomic SEM to high-dimensional data, jointly modeling the genetic architecture of complex traits and investigating multivariate genetic links across neuroimaging phenotypes offers new insights into the genetics of cortical structure and relationships to psychopathology.
Collapse
Affiliation(s)
- Rajendra A Morey
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Yuanchao Zheng
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Henry Bayly
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Delin Sun
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Melanie E Garrett
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
- Department of Medicine, Duke Molecular Physiology Institute, Carmichael Building, Duke University Medical Center, Durham, NC, 27701, USA
| | - Marianna Gasperi
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Adam X Maihofer
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - C Lexi Baird
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
| | - Katrina L Grasby
- Psychiatric Genetics, QIMR, Berghofer Medical Research Institute, 4006, Brisbane, QLD, Australia
| | - Ashley A Huggins
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Courtney C Haswell
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute Keck School of Medicine University of Southern California, Los Angeles, CA, 90033, USA
| | - Sarah Medland
- Queensland Institute for Medical Research, Berghofer Medical Research Institute, 4006, Brisbane, QLD, Australia
| | - Daniel E Gustavson
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Matthew S Panizzon
- Stein Institute for Research on Aging, University of California San Diego, La Jolla, CA, 92093, USA
| | - William S Kremen
- Stein Institute for Research on Aging, University of California San Diego, La Jolla, CA, 92093, USA
| | - Caroline M Nievergelt
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Allison E Ashley-Koch
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
- Department of Medicine, Duke Molecular Physiology Institute, Carmichael Building, Duke University Medical Center, Durham, NC, 27701, USA
| | - Mark W Logue
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA.
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, 02118, USA.
- Biomedical Genetics, Boston University School of Medicine, Boston, MA, 02118-2526, USA.
| |
Collapse
|
30
|
Ning C, Jin M, Cai Y, Fan L, Hu K, Lu Z, Zhang M, Chen C, Li Y, Hu N, Zhang D, Liu Y, Chen S, Jiang Y, He C, Wang Z, Cao Z, Li H, Li G, Ma Q, Geng H, Tian W, Zhang H, Yang X, Huang C, Wei Y, Li B, Zhu Y, Li X, Miao X, Tian J. Genetic architectures of the human hippocampus and those involved in neuropsychiatric traits. BMC Med 2024; 22:456. [PMID: 39394562 PMCID: PMC11470718 DOI: 10.1186/s12916-024-03682-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND The hippocampus, with its complex subfields, is linked to numerous neuropsychiatric traits. While most research has focused on its global structure or a few specific subfields, a comprehensive analysis of hippocampal substructures and their genetic correlations across a wide range of neuropsychiatric traits remains underexplored. Given the hippocampus's high heritability, considering hippocampal and subfield volumes (HASV) as endophenotypes for neuropsychiatric conditions is essential. METHODS We analyzed MRI-derived volumetric data of hippocampal and subfield structures from 41,525 UK Biobank participants. Genome-wide association studies (GWAS) on 24 HASV traits were conducted, followed by genetic correlation, overlap, and Mendelian randomization (MR) analyses with 10 common neuropsychiatric traits. Polygenic risk scores (PRS) based on HASV traits were also evaluated for predicting these traits. RESULTS Our analysis identified 352 independent genetic variants surpassing a significance threshold of 2.1 × 10-9 within the 24 HASV traits, located across 93 chromosomal regions. Notably, the regions 12q14.3, 17q21.31, 12q24.22, 6q21, 9q33.1, 6q25.1, and 2q24.2 were found to influence multiple HASVs. Gene set analysis revealed enrichment of neural differentiation and signaling pathways, as well as protein binding and degradation. Of 240 HASV-neuropsychiatric trait pairs, 75 demonstrated significant genetic correlations (P < 0.05/240), revealing 433 pleiotropic loci. Particularly, genes like ACBD4, ARHGAP27, KANSL1, MAPT, ARL17A, and ARL17B were involved in over 50 HASV-neuropsychiatric pairs. Leveraging Mendelian randomization analysis, we further confirmed that atrophy in the left hippocampus, right hippocampus, right hippocampal body, and right CA1-3 region were associated with an increased risk of developing Parkinson's disease (PD). Furthermore, PRS for all four HASVs were significantly linked to a higher risk of Parkinson's disease (PD), with the highest hazard ratio (HR) of 1.30 (95% CI 1.18-1.43, P = 6.15 × 10⁻⁸) for right hippocampal volume. CONCLUSIONS These findings highlight the extensive distribution of pleiotropic genetic determinants between HASVs and neuropsychiatric traits. Moreover, they suggest a significant potential for effectively managing and intervening in these diseases during their early stages.
Collapse
Affiliation(s)
- Caibo Ning
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China
| | - Meng Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yimin Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China
| | - Linyun Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Kexin Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zequn Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Can Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yanmin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Naifan Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Donghui Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yizhuo Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Shuoni Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yuan Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Chunyi He
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zhuo Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zilong Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Hanting Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Gaoyuan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Qianying Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Hui Geng
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Wen Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Heng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Xiaojun Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chaoqun Huang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yongchang Wei
- Department of Gastrointestinal Oncology, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Bin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China
| | - Xiangpan Li
- Department of Radiation Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China.
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China.
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China.
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
31
|
Bledsoe X, Gamazon ER. NeuroimaGene: an R package for assessing the neurological correlates of genetically regulated gene expression. BMC Bioinformatics 2024; 25:325. [PMID: 39379815 PMCID: PMC11463069 DOI: 10.1186/s12859-024-05936-x] [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: 07/18/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND We present the NeuroimaGene resource as an R package designed to assist researchers in identifying genes and neurologic features relevant to psychiatric and neurological health. While recent studies have identified hundreds of genes as potential components of pathophysiology in neurologic and psychiatric disease, interpreting the physiological consequences of this variation is challenging. The integration of neuroimaging data with molecular findings is a step toward addressing this challenge. In addition to sharing associations with both molecular variation and clinical phenotypes, neuroimaging features are intrinsically informative of cognitive processes. NeuroimaGene provides a tool to understand how disease-associated genes relate to the intermediate structure of the brain. RESULTS We created NeuroimaGene, a user-friendly, open access R package now available for public use. Its primary function is to identify neuroimaging derived brain features that are impacted by genetically regulated expression of user-provided genes or gene sets. This resource can be used to (1) characterize individual genes or gene sets as relevant to the structure and function of the brain, (2) identify the region(s) of the brain or body in which expression of target gene(s) is neurologically relevant, (3) impute the brain features most impacted by user-defined gene sets such as those produced by cohort level gene association studies, and (4) generate publication level, modifiable visual plots of significant findings. We demonstrate the utility of the resource by identifying neurologic correlates of stroke-associated genes derived from pre-existing analyses. CONCLUSIONS Integrating neurologic data as an intermediate phenotype in the pathway from genes to brain-based diagnostic phenotypes increases the interpretability of molecular studies and enriches our understanding of disease pathophysiology. The NeuroimaGene R package is designed to assist in this process and is publicly available for use.
Collapse
Affiliation(s)
- Xavier Bledsoe
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory & Alzheimer's Center, Nashville, TN, USA
| |
Collapse
|
32
|
Li R, Fan YR, Wang YZ, Lu HY, Li PX, Dong Q, Jiang YF, Chen XD, Cui M. Brain Iron in signature regions relating to cognitive aging in older adults: the Taizhou Imaging Study. Alzheimers Res Ther 2024; 16:211. [PMID: 39358805 PMCID: PMC11448274 DOI: 10.1186/s13195-024-01575-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Recent magnetic resonance imaging (MRI) studies have established that brain iron accumulation might accelerate cognitive decline in Alzheimer's disease (AD) patients. Both normal aging and AD are associated with cerebral atrophy in specific regions. However, no studies have investigated aging- and AD-selective iron deposition-related cognitive changes during normal aging. Here, we applied quantitative susceptibility mapping (QSM) to detect iron levels in cortical signature regions and assessed the relationships among iron, atrophy, and cognitive changes in older adults. METHODS In this Taizhou Imaging Study, 770 older adults (mean age 62.0 ± 4.93 years, 57.5% women) underwent brain MRI to measure brain iron and atrophy, of whom 219 underwent neuropsychological tests nearly every 12 months for up to a mean follow-up of 2.68 years. Global cognition was assessed using the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA). Domain-specific cognitive scores were obtained from MoCA subscore components. Regional analyses were performed for cortical regions and 2 signature regions where atrophy affected by aging and AD only: Aging (AG) -specific and AD signature meta-ROIs. The QSM and cortical morphometry means of the above ROIs were also computed. RESULTS Significant associations were found between QSM levels and cognitive scores. In particular, after adjusting for cortical thickness of regions of interest (ROIs), participants in the upper tertile of the cortical and AG-specific signature QSM exhibited worse ZMMSE than did those in the lower tertile [β = -0.104, p = 0.026;β = -0.118, p = 0.021, respectively]. Longitudinal analysis suggested that QSM values in all ROIs might predict decline in ZMoCA and key domains such as attention and visuospatial function (all p < 0.05). Furthermore, iron levels were negatively correlated with classic MRI markers of cortical atrophy (cortical thickness, gray matter volume, and local gyrification index) in total, AG-specific signature and AD signature regions (all p < 0.05). CONCLUSION AG- and AD-selective iron deposition was associated with atrophy and cognitive decline in elderly people, highlighting its potential as a neuroimaging marker for cognitive aging.
Collapse
Affiliation(s)
- Rui Li
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Yi-Ren Fan
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Ying-Zhe Wang
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - He-Yang Lu
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Pei-Xi Li
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Yan-Feng Jiang
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, Human Phenome Institute, Fudan University, 2005 Songhu Road, Shanghai, 200438, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Xing-Dong Chen
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, Human Phenome Institute, Fudan University, 2005 Songhu Road, Shanghai, 200438, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China.
| | - Mei Cui
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.
| |
Collapse
|
33
|
Bahrami S, Nordengen K, Rokicki J, Shadrin AA, Rahman Z, Smeland OB, Jaholkowski PP, Parker N, Parekh P, O'Connell KS, Elvsåshagen T, Toft M, Djurovic S, Dale AM, Westlye LT, Kaufmann T, Andreassen OA. The genetic landscape of basal ganglia and implications for common brain disorders. Nat Commun 2024; 15:8476. [PMID: 39353893 PMCID: PMC11445552 DOI: 10.1038/s41467-024-52583-0] [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: 08/22/2023] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
The basal ganglia are subcortical brain structures involved in motor control, cognition, and emotion regulation. We conducted univariate and multivariate genome-wide association analyses (GWAS) to explore the genetic architecture of basal ganglia volumes using brain scans obtained from 34,794 Europeans with replication in 4,808 white and generalization in 5,220 non-white Europeans. Our multivariate GWAS identified 72 genetic loci associated with basal ganglia volumes with a replication rate of 55.6% at P < 0.05 and 87.5% showed the same direction, revealing a distributed genetic architecture across basal ganglia structures. Of these, 50 loci were novel, including exonic regions of APOE, NBR1 and HLAA. We examined the genetic overlap between basal ganglia volumes and several neurological and psychiatric disorders. The strongest genetic overlap was between basal ganglia and Parkinson's disease, as supported by robust LD-score regression-based genetic correlations. Mendelian randomization indicated genetic liability to larger striatal volume as potentially causal for Parkinson's disease, in addition to a suggestive causal effect of greater genetic liability to Alzheimer's disease on smaller accumbens. Functional analyses implicated neurogenesis, neuron differentiation and development in basal ganglia volumes. These results enhance our understanding of the genetic architecture and molecular associations of basal ganglia structure and their role in brain disorders.
Collapse
Grants
- R01 MH129742 NIMH NIH HHS
- Stiftelsen Kristian Gerhard Jebsen (Kristian Gerhard Jebsen Foundation)
- Norwegian Health Association (22731, 25598), the South-Eastern Norway Regional Health Authority (2013-123, 2017-112, 2019-108, 2014-097, 2015-073, 2016-083), the Research Council of Norway (276082, 323961. 213837, 223273, 248778, 273291, 262656, 229129, 283798, 311993, 324499. 204966, 249795, 273345).
Collapse
Affiliation(s)
- Shahram Bahrami
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway.
| | - Kaja Nordengen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Jaroslav Rokicki
- Centre of Research and Education in Forensic Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Alexey A Shadrin
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Zillur Rahman
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Olav B Smeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Nadine Parker
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pravesh Parekh
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Torbjørn Elvsåshagen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Department of Behavioral Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Mathias Toft
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Srdjan Djurovic
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Lars T Westlye
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, Faculty of Social Sciences, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| | - Ole A Andreassen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway.
- Department of Psychiatry, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
34
|
Yao S, Han JZ, Guo J, Wang X, Qian L, Wu H, Shi W, Zhu RJ, Wang JH, Dong SS, Cui LL, Wang Y, Guo Y, Yang TL. The Causal Relationships Between Gut Microbiota, Brain Volume, and Intelligence: A Two-Step Mendelian Randomization Analysis. Biol Psychiatry 2024; 96:463-472. [PMID: 38432522 DOI: 10.1016/j.biopsych.2024.02.1012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/05/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Growing evidence indicates that dynamic changes in gut microbiome can affect intelligence; however, whether these relationships are causal remains elusive. We aimed to disentangle the poorly understood causal relationship between gut microbiota and intelligence. METHODS We performed a 2-sample Mendelian randomization (MR) analysis using genetic variants from the largest available genome-wide association studies of gut microbiota (N = 18,340) and intelligence (N = 269,867). The inverse-variance weighted method was used to conduct the MR analyses complemented by a range of sensitivity analyses to validate the robustness of the results. Considering the close relationship between brain volume and intelligence, we applied 2-step MR to evaluate whether the identified effect was mediated by regulating brain volume (N = 47,316). RESULTS We found a risk effect of the genus Oxalobacter on intelligence (odds ratio = 0.968 change in intelligence per standard deviation increase in taxa; 95% CI, 0.952-0.985; p = 1.88 × 10-4) and a protective effect of the genus Fusicatenibacter on intelligence (odds ratio = 1.053; 95% CI, 1.024-1.082; p = 3.03 × 10-4). The 2-step MR analysis further showed that the effect of genus Fusicatenibacter on intelligence was partially mediated by regulating brain volume, with a mediated proportion of 33.6% (95% CI, 6.8%-60.4%; p = .014). CONCLUSIONS Our results provide causal evidence indicating the role of the microbiome in intelligence. Our findings may help reshape our understanding of the microbiota-gut-brain axis and development of novel intervention approaches for preventing cognitive impairment.
Collapse
Affiliation(s)
- Shi Yao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ji-Zhou Han
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Guo
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xin Wang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Long Qian
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hao Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wei Shi
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ren-Jie Zhu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jia-Hao Wang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Li-Li Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yan Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
35
|
Shafee R, Moraczewski D, Liu S, Mallard T, Thomas A, Raznahan A. A sex-stratified analysis of the genetic architecture of human brain anatomy. Nat Commun 2024; 15:8041. [PMID: 39271676 PMCID: PMC11399304 DOI: 10.1038/s41467-024-52244-2] [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: 08/28/2023] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Large biobanks have dramatically advanced our understanding of genetic influences on human brain anatomy. However, most studies have combined rather than compared male and female participants. Here we screen for sex differences in the common genetic architecture of over 1000 neuroanatomical phenotypes in the UK Biobank and establish a general concordance between male and female participants in heritability estimates, genetic correlations, and variant-level effects. Notable exceptions include higher mean heritability in the female group for regional volume and surface area phenotypes; between-sex genetic correlations that are significantly below 1 in the insula and parietal cortex; and a common variant with stronger effect in male participants mapping to RBFOX1 - a gene linked to multiple neuropsychiatric disorders more common in men. This work suggests that common variant influences on human brain anatomy are largely consistent between males and females, with a few exceptions that will guide future research in growing datasets.
Collapse
Affiliation(s)
- Rebecca Shafee
- Section on Developmental Neurogenomics, Human Genetics Branch, NIMH Intramural Research Program, NIH, Bethesda, MD, USA.
| | | | - Siyuan Liu
- Section on Developmental Neurogenomics, Human Genetics Branch, NIMH Intramural Research Program, NIH, Bethesda, MD, USA
| | - Travis Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, USA
| | - Adam Thomas
- Data Science and Sharing Team, NIMH, NIH, Bethesda, MD, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, NIMH Intramural Research Program, NIH, Bethesda, MD, USA.
| |
Collapse
|
36
|
Auvergne A, Traut N, Henches L, Troubat L, Frouin A, Boetto C, Kazem S, Julienne H, Toro R, Aschard H. Multitrait Analysis to Decipher the Intertwined Genetic Architecture of Neuroanatomical Phenotypes and Psychiatric Disorders. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024:S2451-9022(24)00266-0. [PMID: 39260564 DOI: 10.1016/j.bpsc.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/28/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND There is increasing evidence of shared genetic factors between psychiatric disorders and brain magnetic resonance imaging (MRI) phenotypes. However, deciphering the joint genetic architecture of these outcomes has proven to be challenging, and new approaches are needed to infer the genetic structures that may underlie those phenotypes. Multivariate analyses are a meaningful approach to reveal links between MRI phenotypes and psychiatric disorders missed by univariate approaches. METHODS First, we conducted univariate and multivariate genome-wide association studies for 9 MRI-derived brain volume phenotypes in 20,000 UK Biobank participants. Next, we performed various complementary enrichment analyses to assess whether and how univariate and multitrait approaches could distinguish disorder-associated and non-disorder-associated variants from 6 psychiatric disorders: bipolar disorder, attention-deficit/hyperactivity disorder, autism, schizophrenia, obsessive-compulsive disorder, and major depressive disorder. Finally, we conducted a clustering analysis of top associated variants based on their MRI multitrait association using an optimized k-medoids approach. RESULTS A univariate MRI genome-wide association study revealed only negligible genetic correlations with psychiatric disorders, while a multitrait genome-wide association study identified multiple new associations and showed significant enrichment for variants related to both attention-deficit/hyperactivity disorder and schizophrenia. Clustering analyses also detected 2 clusters that showed not only enrichment for association with attention-deficit/hyperactivity disorder and schizophrenia but also a consistent direction of effects. Functional annotation analyses of those clusters pointed to multiple potential mechanisms, suggesting in particular a role of neurotrophin pathways in both MRI phenotypes and schizophrenia. CONCLUSIONS Our results show that multitrait association signature can be used to infer genetically driven latent MRI variables associated with psychiatric disorders, thereby opening paths for future biomarker development.
Collapse
Affiliation(s)
- Antoine Auvergne
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France.
| | - Nicolas Traut
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Léo Henches
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Lucie Troubat
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Arthur Frouin
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Christophe Boetto
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Sayeh Kazem
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Hanna Julienne
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Roberto Toro
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Hugues Aschard
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
| |
Collapse
|
37
|
Casamitjana A, Mancini M, Robinson E, Peter L, Annunziata R, Althonayan J, Crampsie S, Blackburn E, Billot B, Atzeni A, Puonti O, Balbastre Y, Schmidt P, Hughes J, Augustinack JC, Edlow BL, Zöllei L, Thomas DL, Kliemann D, Bocchetta M, Strand C, Holton JL, Jaunmuktane Z, Iglesias JE. A next-generation, histological atlas of the human brain and its application to automated brain MRI segmentation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.579016. [PMID: 39282320 PMCID: PMC11398399 DOI: 10.1101/2024.02.05.579016] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Magnetic resonance imaging (MRI) is the standard tool to image the human brain in vivo. In this domain, digital brain atlases are essential for subject-specific segmentation of anatomical regions of interest (ROIs) and spatial comparison of neuroanatomy from different subjects in a common coordinate frame. High-resolution, digital atlases derived from histology (e.g., Allen atlas [7], BigBrain [13], Julich [15]), are currently the state of the art and provide exquisite 3D cytoarchitectural maps, but lack probabilistic labels throughout the whole brain. Here we present NextBrain, a next-generation probabilistic atlas of human brain anatomy built from serial 3D histology and corresponding highly granular delineations of five whole brain hemispheres. We developed AI techniques to align and reconstruct ~10,000 histological sections into coherent 3D volumes with joint geometric constraints (no overlap or gaps between sections), as well as to semi-automatically trace the boundaries of 333 distinct anatomical ROIs on all these sections. Comprehensive delineation on multiple cases enabled us to build the first probabilistic histological atlas of the whole human brain. Further, we created a companion Bayesian tool for automated segmentation of the 333 ROIs in any in vivo or ex vivo brain MRI scan using the NextBrain atlas. We showcase two applications of the atlas: automated segmentation of ultra-high-resolution ex vivo MRI and volumetric analysis of Alzheimer's disease and healthy brain ageing based on ~4,000 publicly available in vivo MRI scans. We publicly release: the raw and aligned data (including an online visualisation tool); the probabilistic atlas; the segmentation tool; and ground truth delineations for a 100 μm isotropic ex vivo hemisphere (that we use for quantitative evaluation of our segmentation method in this paper). By enabling researchers worldwide to analyse brain MRI scans at a superior level of granularity without manual effort or highly specific neuroanatomical knowledge, NextBrain holds promise to increase the specificity of MRI findings and ultimately accelerate our quest to understand the human brain in health and disease.
Collapse
Affiliation(s)
- Adrià Casamitjana
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- Research Institute of Computer Vision and Robotics, University of Girona, Girona, Spain
| | - Matteo Mancini
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Italian National Institute of Health, Rome, Italy
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, United Kingdom
| | - Eleanor Robinson
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Loïc Peter
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Roberto Annunziata
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Juri Althonayan
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Shauna Crampsie
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Emily Blackburn
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Benjamin Billot
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Alessia Atzeni
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Oula Puonti
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Yaël Balbastre
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Peter Schmidt
- Advanced Research Computing Centre, University College London, London, United Kingdom
| | - James Hughes
- Advanced Research Computing Centre, University College London, London, United Kingdom
| | - Jean C Augustinack
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Brian L Edlow
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lilla Zöllei
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - David L Thomas
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Dorit Kliemann
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, United States
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- Centre for Cognitive and Clinical Neuroscience, Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, United Kingdom
| | - Catherine Strand
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Janice L Holton
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Zane Jaunmuktane
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Juan Eugenio Iglesias
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, United States
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
38
|
Zhou D, Wang W, Gu J, Lu Q. Causal effects of sepsis on structural changes in cerebral cortex: A Mendelian randomization investigation. Medicine (Baltimore) 2024; 103:e39404. [PMID: 39252275 PMCID: PMC11383497 DOI: 10.1097/md.0000000000039404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/01/2024] [Indexed: 09/11/2024] Open
Abstract
Previous research has shown a strong correlation between sepsis and brain structure. However, whether this relationship represents a causality remains elusive. In this study, we employed Mendelian randomization (MR) to probe the associations of genetically predicted sepsis and sepsis-related death with structural changes in specific brain regions. Genome-wide association study (GWAS) data for sepsis phenotypes (sepsis and sepsis-related death) were obtained from the IEU OpenGWAS. Correspondingly, GWAS data for brain structural traits (volume of the subcortical structure, cortical thickness, and surface area) were derived from the ENIGMA consortium. Inverse variance weighted was mainly utilized to assess the causal effects, while weighted median and MR-Egger regression served as complementary methods. Sensitivity analyses were implemented with Cochran Q test, MR-Egger regression, and MR-PRESSO. In addition, a reverse MR analysis was carried out to assess the possibility of reverse causation. We identified that genetic liability to sepsis was normally significantly associated with a reduced surface area of the postcentral gyrus (β = -35.5280, SE = 13.7465, P = .0096). The genetic liability to sepsis-related death showed a suggestive positive correlation with the surface area of fusiform gyrus (β = 11.0920, SE = 3.6412, P = .0023) and posterior cingulate gyrus (β = 3.6530, SE = 1.6684, P = .0286), While it presented a suggestive negative correlation with surface area of the caudal middle frontal gyrus (β = -11.4586, SE = 5.1501, P = .0261) and frontal pole (β = -1.0024, SE = 0.4329, P = .0206). We also indicated a possible bidirectional causal association between genetic liability to sepsis-related death and the thickness of the transverse temporal gyrus. Sensitivity analyses verified the robustness of the above associations. These findings suggested that genetically determined liability to sepsis might influence the specific brain structure in a causal way, offering new perspectives to investigate the mechanism of sepsis-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Dengfeng Zhou
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| | - Weina Wang
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| | - Jiaying Gu
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| | - Qiaofa Lu
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| |
Collapse
|
39
|
Liu M, Wang L, Zhang Y, Dong H, Wang C, Chen Y, Qian Q, Zhang N, Wang S, Zhao G, Zhang Z, Lei M, Wang S, Zhao Q, Liu F. Investigating the shared genetic architecture between depression and subcortical volumes. Nat Commun 2024; 15:7647. [PMID: 39223129 PMCID: PMC11368965 DOI: 10.1038/s41467-024-52121-y] [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: 01/22/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Depression, a widespread and highly heritable mental health condition, profoundly affects millions of individuals worldwide. Neuroimaging studies have consistently revealed volumetric abnormalities in subcortical structures associated with depression. However, the genetic underpinnings shared between depression and subcortical volumes remain inadequately understood. Here, we investigate the extent of polygenic overlap using the bivariate causal mixture model (MiXeR), leveraging summary statistics from the largest genome-wide association studies for depression (N = 674,452) and 14 subcortical volumetric phenotypes (N = 33,224). Additionally, we identify shared genomic loci through conditional/conjunctional FDR analyses. MiXeR shows that subcortical volumetric traits share a substantial proportion of genetic variants with depression, with 44 distinct shared loci identified by subsequent conjunctional FDR analysis. These shared loci are predominantly located in intronic regions (58.7%) and non-coding RNA intronic regions (25.4%). The 269 protein-coding genes mapped by these shared loci exhibit specific developmental trajectories, with the expression level of 55 genes linked to both depression and subcortical volumes, and 30 genes linked to cognitive abilities and behavioral symptoms. These findings highlight a shared genetic architecture between depression and subcortical volumetric phenotypes, enriching our understanding of the neurobiological underpinnings of depression.
Collapse
Affiliation(s)
- Mengge Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Wang
- Department of Geriatrics and Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yujie Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Haoyang Dong
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Caihong Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yayuan Chen
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qian Qian
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Nannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaoying Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Guoshu Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhihui Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Minghuan Lei
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Sijia Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| | - Qiyu Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
40
|
Guo J, He C, Song H, Gao H, Yao S, Dong SS, Yang TL. Unveiling Promising Neuroimaging Biomarkers for Schizophrenia Through Clinical and Genetic Perspectives. Neurosci Bull 2024; 40:1333-1352. [PMID: 38703276 PMCID: PMC11365900 DOI: 10.1007/s12264-024-01214-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/08/2024] [Indexed: 05/06/2024] Open
Abstract
Schizophrenia is a complex and serious brain disorder. Neuroscientists have become increasingly interested in using magnetic resonance-based brain imaging-derived phenotypes (IDPs) to investigate the etiology of psychiatric disorders. IDPs capture valuable clinical advantages and hold biological significance in identifying brain abnormalities. In this review, we aim to discuss current and prospective approaches to identify potential biomarkers for schizophrenia using clinical multimodal neuroimaging and imaging genetics. We first described IDPs through their phenotypic classification and neuroimaging genomics. Secondly, we discussed the applications of multimodal neuroimaging by clinical evidence in observational studies and randomized controlled trials. Thirdly, considering the genetic evidence of IDPs, we discussed how can utilize neuroimaging data as an intermediate phenotype to make association inferences by polygenic risk scores and Mendelian randomization. Finally, we discussed machine learning as an optimum approach for validating biomarkers. Together, future research efforts focused on neuroimaging biomarkers aim to enhance our understanding of schizophrenia.
Collapse
Affiliation(s)
- Jing Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Changyi He
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Huimiao Song
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Huiwu Gao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shi Yao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
41
|
Hoang N, Sardaripour N, Ramey GD, Schilling K, Liao E, Chen Y, Park JH, Bledsoe X, Landman BA, Gamazon ER, Benton ML, Capra JA, Rubinov M. Integration of estimated regional gene expression with neuroimaging and clinical phenotypes at biobank scale. PLoS Biol 2024; 22:e3002782. [PMID: 39269986 PMCID: PMC11424006 DOI: 10.1371/journal.pbio.3002782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/25/2024] [Accepted: 08/01/2024] [Indexed: 09/15/2024] Open
Abstract
An understanding of human brain individuality requires the integration of data on brain organization across people and brain regions, molecular and systems scales, as well as healthy and clinical states. Here, we help advance this understanding by leveraging methods from computational genomics to integrate large-scale genomic, transcriptomic, neuroimaging, and electronic-health record data sets. We estimated genetically regulated gene expression (gr-expression) of 18,647 genes, across 10 cortical and subcortical regions of 45,549 people from the UK Biobank. First, we showed that patterns of estimated gr-expression reflect known genetic-ancestry relationships, regional identities, as well as inter-regional correlation structure of directly assayed gene expression. Second, we performed transcriptome-wide association studies (TWAS) to discover 1,065 associations between individual variation in gr-expression and gray-matter volumes across people and brain regions. We benchmarked these associations against results from genome-wide association studies (GWAS) of the same sample and found hundreds of novel associations relative to these GWAS. Third, we integrated our results with clinical associations of gr-expression from the Vanderbilt Biobank. This integration allowed us to link genes, via gr-expression, to neuroimaging and clinical phenotypes. Fourth, we identified associations of polygenic gr-expression with structural and functional MRI phenotypes in the Human Connectome Project (HCP), a small neuroimaging-genomic data set with high-quality functional imaging data. Finally, we showed that estimates of gr-expression and magnitudes of TWAS were generally replicable and that the p-values of TWAS were replicable in large samples. Collectively, our results provide a powerful new resource for integrating gr-expression with population genetics of brain organization and disease.
Collapse
Affiliation(s)
- Nhung Hoang
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Neda Sardaripour
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Grace D. Ramey
- Biological and Medical Informatics Division, University of California, San Francisco, California, United States of America
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - Kurt Schilling
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Emily Liao
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Yiting Chen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jee Hyun Park
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Xavier Bledsoe
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Bennett A. Landman
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Eric R. Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Mary Lauren Benton
- Department of Computer Science, Baylor University, Waco, Texas, United States of America
| | - John A. Capra
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Bakar Computational Health Sciences Institute, University of California, San Francisco, California, United States of America
| | - Mikail Rubinov
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Psychology, Vanderbilt University, Nashville, Tennessee, United States of America
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, Virginia, United States of America
| |
Collapse
|
42
|
García-Marín LM, Campos AI, Diaz-Torres S, Rabinowitz JA, Ceja Z, Mitchell BL, Grasby KL, Thorp JG, Agartz I, Alhusaini S, Ames D, Amouyel P, Andreassen OA, Arfanakis K, Vasquez AA, Armstrong NJ, Athanasiu L, Bastin ME, Beiser AS, Bennett DA, Bis JC, Boks MP, Boomsma DI, Brodaty H, Brouwer RM, Buitelaar JK, Burkhardt R, Cahn W, Calhoun VD, Carmichael OT, Chakravarty M, Chen Q, Ching CRK, Cichon S, Crespo-Facorro B, Crivello F, Dale AM, Smith GD, de Geus EJ, De Jager PL, de Zubicaray GI, Debette S, DeCarli C, Depondt C, Desrivières S, Djurovic S, Ehrlich S, Erk S, Espeseth T, Fernández G, Filippi I, Fisher SE, Fleischman DA, Fletcher E, Fornage M, Forstner AJ, Francks C, Franke B, Ge T, Goldman AL, Grabe HJ, Green RC, Grimm O, Groenewold NA, Gruber O, Gudnason V, Håberg AK, Haukvik UK, Heinz A, Hibar DP, Hilal S, Himali JJ, Ho BC, Hoehn DF, Hoekstra PJ, Hofer E, Hoffmann W, Holmes AJ, Homuth G, Hosten N, Ikram MK, Ipser JC, Jack CR, Jahanshad N, Jönsson EG, Kahn RS, Kanai R, Klein M, Knol MJ, Launer LJ, Lawrie SM, Hellard SL, Lee PH, Lemaître H, Li S, Liewald DC, Lin H, Longstreth WT, Lopez OL, Luciano M, et alGarcía-Marín LM, Campos AI, Diaz-Torres S, Rabinowitz JA, Ceja Z, Mitchell BL, Grasby KL, Thorp JG, Agartz I, Alhusaini S, Ames D, Amouyel P, Andreassen OA, Arfanakis K, Vasquez AA, Armstrong NJ, Athanasiu L, Bastin ME, Beiser AS, Bennett DA, Bis JC, Boks MP, Boomsma DI, Brodaty H, Brouwer RM, Buitelaar JK, Burkhardt R, Cahn W, Calhoun VD, Carmichael OT, Chakravarty M, Chen Q, Ching CRK, Cichon S, Crespo-Facorro B, Crivello F, Dale AM, Smith GD, de Geus EJ, De Jager PL, de Zubicaray GI, Debette S, DeCarli C, Depondt C, Desrivières S, Djurovic S, Ehrlich S, Erk S, Espeseth T, Fernández G, Filippi I, Fisher SE, Fleischman DA, Fletcher E, Fornage M, Forstner AJ, Francks C, Franke B, Ge T, Goldman AL, Grabe HJ, Green RC, Grimm O, Groenewold NA, Gruber O, Gudnason V, Håberg AK, Haukvik UK, Heinz A, Hibar DP, Hilal S, Himali JJ, Ho BC, Hoehn DF, Hoekstra PJ, Hofer E, Hoffmann W, Holmes AJ, Homuth G, Hosten N, Ikram MK, Ipser JC, Jack CR, Jahanshad N, Jönsson EG, Kahn RS, Kanai R, Klein M, Knol MJ, Launer LJ, Lawrie SM, Hellard SL, Lee PH, Lemaître H, Li S, Liewald DC, Lin H, Longstreth WT, Lopez OL, Luciano M, Maillard P, Marquand AF, Martin NG, Martinot JL, Mather KA, Mattay VS, McMahon KL, Mecocci P, Melle I, Meyer-Lindenberg A, Mirza-Schreiber N, Milaneschi Y, Mosley TH, Mühleisen TW, Müller-Myhsok B, Muñoz Maniega S, Nauck M, Nho K, Niessen WJ, Nöthen MM, Nyquist PA, Oosterlaan J, Pandolfo M, Paus T, Pausova Z, Penninx BW, Pike GB, Psaty BM, Pütz B, Reppermund S, Rietschel MD, Risacher SL, Romanczuk-Seiferth N, Romero-Garcia R, Roshchupkin GV, Rotter JI, Sachdev PS, Sämann PG, Saremi A, Sargurupremraj M, Saykin AJ, Schmaal L, Schmidt H, Schmidt R, Schofield PR, Scholz M, Schumann G, Schwarz E, Shen L, Shin J, Sisodiya SM, Smith AV, Smoller JW, Soininen HS, Steen VM, Stein DJ, Stein JL, Thomopoulos SI, Toga AW, Tordesillas-Gutiérrez D, Trollor JN, Valdes-Hernandez MC, van 't Ent D, van Bokhoven H, van der Meer D, van der Wee NJ, Vázquez-Bourgon J, Veltman DJ, Vernooij MW, Villringer A, Vinke LN, Völzke H, Walter H, Wardlaw JM, Weinberger DR, Weiner MW, Wen W, Westlye LT, Westman E, White T, Witte AV, Wolf C, Yang J, Zwiers MP, Ikram MA, Seshadri S, Thompson PM, Satizabal CL, Medland SE, Rentería ME. Genomic analysis of intracranial and subcortical brain volumes yields polygenic scores accounting for variation across ancestries. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.13.24311922. [PMID: 39371125 PMCID: PMC11451674 DOI: 10.1101/2024.08.13.24311922] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Subcortical brain structures are involved in developmental, psychiatric and neurological disorders. We performed GWAS meta-analyses of intracranial and nine subcortical brain volumes (brainstem, caudate nucleus, putamen, hippocampus, globus pallidus, thalamus, nucleus accumbens, amygdala and, for the first time, the ventral diencephalon) in 74,898 participants of European ancestry. We identified 254 independent loci associated with these brain volumes, explaining up to 35% of phenotypic variance. We observed gene expression in specific neural cell types across differentiation time points, including genes involved in intracellular signalling and brain ageing-related processes. Polygenic scores for brain volumes showed predictive ability when applied to individuals of diverse ancestries. We observed causal genetic effects of brain volumes with Parkinson's disease and ADHD. Findings implicate specific gene expression patterns in brain development and genetic variants in comorbid neuropsychiatric disorders, which could point to a brain substrate and region of action for risk genes implicated in brain diseases.
Collapse
Affiliation(s)
- Luis M García-Marín
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Adrian I Campos
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Santiago Diaz-Torres
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Jill A Rabinowitz
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Zuriel Ceja
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Brittany L Mitchell
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Katrina L Grasby
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jackson G Thorp
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Ingrid Agartz
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, 0319, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, 0407, Norway
- Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm, SE-11364, Sweden
| | - Saud Alhusaini
- Department of Neurology, Alpert Medical School of Brown University, Providence, RI, 02903, USA
- Molecular & Cellular Therapeutics Department, Royal College of Surgeons in Ireland, Dublin, D15, Ireland
| | - David Ames
- Academic Unit Psychiatry of Old Age, University of Melbourne, Kew, VIC, 3101, Australia
- National Ageing Research Institute, Parkville, VIC, 3052, Australia
| | - Philippe Amouyel
- Universite Lille, U1167 - RID-AGE - LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, Lille, F-59000, France
- Institut National de la Sante et de la Recherche Medicale, U1167, Lille, F-59000, France
- Centre Hospitalier Universitaire de Lille, Department of Public Health, Lille, F-59000, Franch
- Institut Pasteur de Lille UMR1167, Lille, F-59000, France
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, 0319, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, 0407, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, 0407, Norway
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, 60616, USA
| | - Alejandro Arias Vasquez
- Departments of Psychiatry and Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Nicola J Armstrong
- Department of Mathematics and Statistics, Curtin University, Perth, Australia
| | - Lavinia Athanasiu
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, 0319, Norway
- CoE NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway, Oslo, 0455, Norway
| | - Mark E Bastin
- Centre for Clinical Brain Sciences and Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - Alexa S Beiser
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, 02118, USA
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, 02118, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, 98195-9458, USA
| | - Marco Pm Boks
- Brain Center University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | | | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Rachel M Brouwer
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neurocience, VU Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6525 EN, The Netherlands
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg University, Regensburg, 93053, Germany
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, 04103, Germany
| | - Wiepke Cahn
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, 3584CX, The Netherlands
- Altrecht Mental Health Institute, Utrecht, 3512PG, The Netherlands
| | - Vince D Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), {Georgia State, Georgia Tech, Emory}, Atlanta, GA, 30303, USA
| | | | - Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Research Centre, Montreal, QC, H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, QC, H3A 1A1, Canada
| | - Qiang Chen
- Lieber Institute for Brain Development, Baltimore, MD, 21205, USA
| | - Christopher R K Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, 90292, USA
| | - Sven Cichon
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, 52428, Germany
- Department of Biomedicine, University of Basel, Basel, CH-4031, Switzerland
- Medical Genetics, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, 4031, Switzerland
| | - Benedicto Crespo-Facorro
- HU Virgen del Rocio, Instituto de Investigacion biomedica IBIS-CSIC, Universidad de Sevilla, CIBERSAM, Sevilla, 41013, Spain
| | - Fabrice Crivello
- CNRS, IMN, UMR 5293, University of Bordeaux, Bordeaux, 33076, France
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, La Jolla, CA, 92093, USA
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, United Kingdom
- Population Health Sciences, University of Bristol, Bristol, BS8 BN, United Kingdom
| | - Eco Jc de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, 1081 BT, The Netherlands
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10538, USA
| | - Greig I de Zubicaray
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, 4059, Australia
| | - Stéphanie Debette
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, 4059, Australia
- Department of Neurology, Institute of Neurodegenerative Diseases, Bordeaux University Hospital, Bordeaux, F-33000, France
| | - Charles DeCarli
- Imaging of Dementia and Aging Laboratory, Department of Neurology, University of California, Davis, Sacramento, CA, 95817, USA
| | - Chantal Depondt
- Department of Neurology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, 1070, Belgium
| | - Sylvane Desrivières
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, United Kingdom
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, 0450, Norway
- Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
| | - Stefan Ehrlich
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, 01307, Germany
| | - Susanne Erk
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, 11017, Germany
| | - Thomas Espeseth
- Department of Psychology, University of Oslo, Oslo, 0373, Norway
- Department of Psychology, Oslo New University College, Oslo, 0456, Norway
| | - Guillén Fernández
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Irina Filippi
- INSERM U1299, Paris Saclay University, Gif-sur-Yvette, 91190, France
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, 6525 XD, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, 6500 HE, The Netherlands
| | - Debra A Fleischman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Evan Fletcher
- Department of Neurology, University of California Davis, Davis, CA, 95616, USA
| | - Myriam Fornage
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Andreas J Forstner
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, 52428, Germany
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, 53127, Germany
| | - Clyde Francks
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, 6525 XD, The Netherlands
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Barbara Franke
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6525 EN, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Aaron L Goldman
- Lieber Institute for Brain Development, Baltimore, MD, 21205, USA
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, 17475, Germany
| | - Robert C Green
- Department of Medicine (Genetics), Mass General Brigham and Harvard Medical School, Boston, MA, 02115, USA
| | - Oliver Grimm
- Central Institute of Mental Health, Mannheim, 68159, Germany
- Goethe-University Frankfurt, Frankfurt am Main, 60528, Germany
| | - Nynke A Groenewold
- Department of Psychiatry and Mental Health, Neuroscience Institute, University of Cape Town, Cape Town, 7925, South Africa
| | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, D-69115, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Asta K Håberg
- Department of Neuromedicine and Movement, NTNU Science, Trondheim, 7030, Norway
- MiDT National Research Center, Department of Research, St Olavs Hospital, Trondheim, 7006, Norway
| | - Unn K Haukvik
- Norwegian Centre for Mental Health Research (NORMENT), Department of Mental Health and Addiction, University of Oslo, Oslo, 0450, Norway
- Centre for Forensic Psychiatry Research, Oslo University Hospital, Oslo, 0455, Norway
| | - Andreas Heinz
- Centre for Forensic Psychiatry Research, Oslo University Hospital, Oslo, 0455, Norway
| | - Derrek P Hibar
- Product Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Saima Hilal
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, 117549, Singapore
| | - Jayandra J Himali
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, 02118, USA
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, 02118, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, 78229-3900, USA
- Department of Population Health Sciences, UT Health Science Center San Antonio, San Antonio, TX, 78229, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Beng-Choon Ho
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52246, USA
| | - David F Hoehn
- Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Pieter J Hoekstra
- Department of Child and Adolescent Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, 9713 GZ, The Netherlands
- Accare Child Study Center, Groningen, 9723 HE, The Netherlands
| | - Edith Hofer
- Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, 8036, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, 8036, Austria
| | - Wolfgang Hoffmann
- German Centre for Neurodegenerative Diseases (DZNE) - site Rostock/Greifswald, Greifswald, 17489, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, 17495, Germany
| | - Avram J Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, 08854, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, 17475, Germany
| | - Norbert Hosten
- Department of Radiology, University Clinic Greifswald, Greifswald, 17475, Germany
| | - M Kamran Ikram
- Departments of Epidemiology and Neurology, Erasmus MC, Rotterdam, 3015 CN , The Netherlands
| | - Jonathan C Ipser
- Department of Psychiatry and Mental Health, Neuroscience Institute, Groote Schuur Hospital, University of Cape Town, Cape Town, 7925, South Africa
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, 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, CA, 90292, USA
| | - Erik G Jönsson
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, 0319, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Sciences, Stockholm Region, Stockholm, SE-11364, Sweden
| | - Rene S Kahn
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, 3584CX, The Netherlands
| | | | - Marieke Klein
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015 GD, The Netherlands
| | - Lenore J Launer
- Intramural Research Program, National Institute on Aging, Baltimore, MD, 21224, USA
| | - Stephen M Lawrie
- Division of Psychiatry, University of Edinburgh, Edinburgh, EH10 5HF, United Kingdom
| | | | - Phil H Lee
- Center for Genomic Medicine, Mass General Brigham, Boston, MA, 02114, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Stanley Center for Psychiatry, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Hervé Lemaître
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR 5293, CNRS, Université de Bordeaux, Bordeaux, 33076, France
| | - Shuo Li
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, 02118, USA
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, 02118, USA
| | | | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - W T Longstreth
- Department of Neurology, University of Washington, Seattle, WA, 98104-2420, USA
- Department of Epidemiology, University of Washington, Seattle, WA, 98195-9458, USA
| | - Oscar L Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Michelle Luciano
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, United Kingdom
| | - Pauline Maillard
- Department of Neurology, University of California Davis, Davis, CA, 95616, USA
| | - Andre F Marquand
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Nicholas G Martin
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Jean-Luc Martinot
- Université Paris-Saclay; Institut National de la Santé et de la Recherche Médicale, INSERM U1299 "Trajectoires développementales Psychiatrie", Ecole Normale Supérieure Paris-Saclay, CNRS UMR 9010, Université Paris Cité, Centre Borelli, Gif sur Yvette, 911
| | - Karen A Mather
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Venkata S Mattay
- Lieber Institute for Brain Development, Baltimore, MD, 21205, USA
| | - Katie L McMahon
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, 4001, Australia
| | - Patrizia Mecocci
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, 06132, Italy
- Clinical Geriatrics, NVS Department, Karolinska Institute, Huddinge, 14152, Sweden
| | - Ingrid Melle
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, 0319, Norway
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68159, Germany
| | - Nazanin Mirza-Schreiber
- Institute of Neurogenomics,Helmholtz Munich, 85764, Neuherberg, Germany
- Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Munich, 85764, Neuherberg, Germany
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, 1081 HJ, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, 1081 BT, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress program, Amsterdam, 1081 BT, The Netherlands
- Amsterdam Neuroscience, Complex Trait Genetics program, Amsterdam, 1081 HV, The Netherlands
| | | | - Thomas W Mühleisen
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, 52428, Germany
- Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, D-40225, Germany
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, CH-4031, Switzerland
| | - Bertram Müller-Myhsok
- Statistics Genetics Group, Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Susana Muñoz Maniega
- Centre for Clinical Brain Sciences and Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, 17489, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17489, Germany
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Wiro J Niessen
- University Medical Center Groningen, Groningen, 9713GZ, The Netherlands
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, 53127, Germany
| | - Paul A Nyquist
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
- General internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Jaap Oosterlaan
- Clinical Neuropsychology section, Vrije Universiteit Amsterdam, Amsterdam, 1081 BT, The Netherlands
- Emma Children's Hospital, University Medical Centers Amsterdam, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, 1100 DD, The Netherlands
| | - Massimo Pandolfo
- Université Libre de Bruxelles, Brussels, 1070, Belgium
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Tomas Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1C5, Canada
- Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, QC, H3T 1C5, Canada
| | - Zdenka Pausova
- Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Physiology, University of Toronto, Toronto, M5G 0A4, Canada
| | - Brenda Wjh Penninx
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, 1081 HJ, The Netherlands
| | - G Bruce Pike
- Departments of Radiology and Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, 98195-9458, USA
- Department of Epidemiology, University of Washington, Seattle, WA, 98195-9458, USA
- Department of Health Systems and Population Health, Seattle, WA, 98195-9458, USA
| | - Benno Pütz
- Translational Psychiatry, Munich, 80804, Germany
| | - Simone Reppermund
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- Department of Developmental Disability Neuropsychiatry, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Marcella D Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, 68159, Germany
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer's Disease Research Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nina Romanczuk-Seiferth
- Department of Psychiatry and Neuroscience, Charité - Universitätsmedizin Berlin, Berlin, 10117, Germany
- Department of Psychology, Clinical Psychology and Psychotherapy, MSB Medical School Berlin, Berlin, 14197, Germany
| | - Rafael Romero-Garcia
- Instituto de Biomedicina de Sevilla (IBiS) HUVR/CSIC/Universidad de Sevilla/ CIBERSAM, ISCIII, Dpto. de Fisiología Médica y Biofísica, Sevilla, 41013, Spain
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0SZ, United Kingdom
| | - Gennady V Roshchupkin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015 GD, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, 3015 GD, The Netherlands
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- Neuropsychiatric Institute, The Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | | | - Arvin Saremi
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, 90292, USA
| | - Muralidharan Sargurupremraj
- INSERM U1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, F-33000, France
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, 78229-3900, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Lianne Schmaal
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
- Orygen, Parkville, VIC, 3052, Australia
| | - Helena Schmidt
- Institute of Molecular Biology & Biochemistry, Gottfried Schatz Center for Signaling, Metabolism & Aging, Medical University Graz, Graz, 8010, Austria
| | - Reinhold Schmidt
- Department of Neurology, Medical University Graz Austria, Graz, 8023, Austria
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, 2031, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Markus Scholz
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, 04103, Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, 04107, Germany
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS), ISTBI, Fudan University, Shanghai, 200031, P.R. China
- PONS Centre, Department of Psychiatry, CCM, Charite Unversitaetsmedizin Berlin, Berlin, 10017, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68159, Germany
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jean Shin
- The Hospital for Sick Children, Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, M5G 0A4, Canada
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
- Chalfont Centre for Epilepsy, Chalfont St Peter, SL9 0RJ, United Kingdom
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, 201, Iceland
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Hilkka S Soininen
- Department of Neurology, Institute of Clinical Mediciine, University of Eastern Finland, Kuopio, 70100, Finland
| | - Vidar M Steen
- Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, N-5021, Norway
| | - Dan J Stein
- SAMRC Research Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, 7925, South Africa
| | - Jason L Stein
- Department of Genetics & UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7250, 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, CA, 90292, USA
| | - Arthur W Toga
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, 90292, USA
| | - Diana Tordesillas-Gutiérrez
- Instituto de Física de Cantabria (CSIC-UC), Santander, E-39005, Spain
- Department of Radiology, Marqués de Valdecilla University Hospital, Valdecilla Biomedical Research Institute IDIVAL, Santander, 39011, Spain
| | - Julian N Trollor
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- The National Centre of Excellence in Intellectual Disability Health,, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Maria C Valdes-Hernandez
- Centre for Clinical Brain Sciences and Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - Dennis van 't Ent
- Department of Biological Psychology & Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, 1081 BT, The Netherlands
| | - Hans van Bokhoven
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6525 EN, The Netherlands
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Dennis van der Meer
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, 0319, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, 6200MD, The Netherlands
| | - Nic Ja van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Javier Vázquez-Bourgon
- Department of Psychiatry, University Hospital Marqués de Valdecilla - IDIVAL, Santander, 39008, Spain
- Departamento de Medicina y Psiquiatría, Universidad de Cantabria, Santander, 39008, Spain
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Sevilla, 41013, Spain
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, 1081 HJ, The Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015 GD, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, 3015 GD, The Netherlands
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human, Cognitive and Brain Sciences, Leipzig, 04103, Germany
- Cognitive Neurology, University of Leipzig Medical Center, Leipzig, 04103, Germany
| | - Louis N Vinke
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, 17495, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, 11017, Germany
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences and Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
- UK Dementia Research Institute Centre, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Michael W Weiner
- University of California San Francisco, San Francisco, CA, 94121, USA
- Northern California Institute for Research & Education (NCIRE), San Francisco, CA, 94121, USA
- Veterans Administration Medical Center, San Francisco, CA, 94121, USA
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Lars T Westlye
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, 0319, Norway
- Department of Psychology, University of Oslo, Oslo, 0373, Norway
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, 14183, Sweden
| | - Tonya White
- Section on Social and Cognitive Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD, 20892-1276, USA
| | - A Veronica Witte
- Department of Neurology, Max Planck Institute for Human, Cognitive and Brain Sciences, Leipzig, 04103, Germany
- Cognitive Neurology, University of Leipzig Medical Center, Leipzig, 04103, Germany
| | | | - Jingyun Yang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Marcel P Zwiers
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, 6525 EN, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3015 GD, The Netherlands
| | - Sudha Seshadri
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, 02118, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, 78229-3900, 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, CA, 90292, USA
| | - Claudia L Satizabal
- Framingham Heart Study, Chobanian and Avedisian Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Population Health Sciences and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, 78229, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Sarah E Medland
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, 4059, Australia
- School of Psychology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Miguel E Rentería
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
43
|
Zhu J, Chen X, Lu B, Li XY, Wang ZH, Cao LP, Chen GM, Chen JS, Chen T, Chen TL, Cheng YQ, Chu ZS, Cui SX, Cui XL, Deng ZY, Gong QY, Guo WB, He CC, Hu ZJY, Huang Q, Ji XL, Jia FN, Kuang L, Li BJ, Li F, Li HX, Li T, Lian T, Liao YF, Liu XY, Liu YS, Liu ZN, Long YC, Lu JP, Qiu J, Shan XX, Si TM, Sun PF, Wang CY, Wang HN, Wang X, Wang Y, Wang YW, Wu XP, Wu XR, Wu YK, Xie CM, Xie GR, Xie P, Xu XF, Xue ZP, Yang H, Yu H, Yuan ML, Yuan YG, Zhang AX, Zhao JP, Zhang KR, Zhang W, Zhang ZJ, Yan CG, Yu Y. Transcriptomic decoding of regional cortical vulnerability to major depressive disorder. Commun Biol 2024; 7:960. [PMID: 39117859 PMCID: PMC11310478 DOI: 10.1038/s42003-024-06665-w] [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: 01/17/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Previous studies in small samples have identified inconsistent cortical abnormalities in major depressive disorder (MDD). Despite genetic influences on MDD and the brain, it is unclear how genetic risk for MDD is translated into spatially patterned cortical vulnerability. Here, we initially examined voxel-wise differences in cortical function and structure using the largest multi-modal MRI data from 1660 MDD patients and 1341 controls. Combined with the Allen Human Brain Atlas, we then adopted transcription-neuroimaging spatial correlation and the newly developed ensemble-based gene category enrichment analysis to identify gene categories with expression related to cortical changes in MDD. Results showed that patients had relatively circumscribed impairments in local functional properties and broadly distributed disruptions in global functional connectivity, consistently characterized by hyper-function in associative areas and hypo-function in primary regions. Moreover, the local functional alterations were correlated with genes enriched for biological functions related to MDD in general (e.g., endoplasmic reticulum stress, mitogen-activated protein kinase, histone acetylation, and DNA methylation); and the global functional connectivity changes were associated with not only MDD-general, but also brain-relevant genes (e.g., neuron, synapse, axon, glial cell, and neurotransmitters). Our findings may provide important insights into the transcriptomic signatures of regional cortical vulnerability to MDD.
Collapse
Affiliation(s)
- Jiajia Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Research Center of Clinical Medical Imaging, Anhui Province, Hefei, 230032, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - Xiao Chen
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bin Lu
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue-Ying Li
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zi-Han Wang
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li-Ping Cao
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
| | - Guan-Mao Chen
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 250024, China
| | - Jian-Shan Chen
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
| | - Tao Chen
- Department of Radiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Tao-Lin Chen
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610044, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, 610052, China
| | - Yu-Qi Cheng
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Zhao-Song Chu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Shi-Xian Cui
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 101408, China
- Sino-Danish Center for Education and Research, Graduate University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Xi-Long Cui
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Zhao-Yu Deng
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qi-Yong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610044, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, 610052, China
| | - Wen-Bin Guo
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Can-Can He
- Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing, Jiangsu, 210009, China
| | - Zheng-Jia-Yi Hu
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 101408, China
- Sino-Danish Center for Education and Research, Graduate University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Qian Huang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Xin-Lei Ji
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Feng-Nan Jia
- Department of Clinical Psychology, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, 215003, China
| | - Li Kuang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Bao-Juan Li
- Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Feng Li
- Beijing Anding Hospital, Capital Medical University, Beijing, 100120, China
| | - Hui-Xian Li
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310063, China
- Mental Health Center and Psychiatric Laboratory, West China Hospital of Sichuan University, Chengdu, Sichuan, 610044, China
| | - Tao Lian
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yi-Fan Liao
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiao-Yun Liu
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yan-Song Liu
- Department of Clinical Psychology, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, 215003, China
| | - Zhe-Ning Liu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Yi-Cheng Long
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Jian-Ping Lu
- Shenzhen Kangning Hospital Shenzhen, Guangzhou, 518020, China
| | - Jiang Qiu
- Faculty of Psychology, Southwest University, Chongqing, 400715, China
| | - Xiao-Xiao Shan
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Tian-Mei Si
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital) & Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, 100191, China
| | - Peng-Feng Sun
- Xi'an Central Hospital, Xi'an, Shaanxi, 710004, China
| | - Chuan-Yue Wang
- Beijing Anding Hospital, Capital Medical University, Beijing, 100120, China
| | - Hua-Ning Wang
- Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiang Wang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Ying Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 250024, China
| | - Yu-Wei Wang
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Ping Wu
- Xi'an Central Hospital, Xi'an, Shaanxi, 710004, China
| | - Xin-Ran Wu
- Faculty of Psychology, Southwest University, Chongqing, 400715, China
| | - Yan-Kun Wu
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital) & Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, 100191, China
| | - Chun-Ming Xie
- Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing, Jiangsu, 210009, China
| | - Guang-Rong Xie
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Peng Xie
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400000, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Xiu-Feng Xu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Zhen-Peng Xue
- Shenzhen Kangning Hospital Shenzhen, Guangzhou, 518020, China
| | - Hong Yang
- Department of Radiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Hua Yu
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310063, China
- Mental Health Center and Psychiatric Laboratory, West China Hospital of Sichuan University, Chengdu, Sichuan, 610044, China
| | - Min-Lan Yuan
- West China Hospital of Sichuan University, Chengdu, Sichuan, 610044, China
| | - Yong-Gui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Ai-Xia Zhang
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jing-Ping Zhao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Ke-Rang Zhang
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Wei Zhang
- West China Hospital of Sichuan University, Chengdu, Sichuan, 610044, China
| | - Zi-Jing Zhang
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chao-Gan Yan
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences, Beijing, 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 101408, China
- Sino-Danish Center for Education and Research, Graduate University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Research Center of Clinical Medical Imaging, Anhui Province, Hefei, 230032, China.
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China.
| |
Collapse
|
44
|
Tesfaye M, Spindola LM, Stavrum AK, Shadrin A, Melle I, Andreassen OA, Le Hellard S. Sex effects on DNA methylation affect discovery in epigenome-wide association study of schizophrenia. Mol Psychiatry 2024; 29:2467-2477. [PMID: 38503926 PMCID: PMC11412896 DOI: 10.1038/s41380-024-02513-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024]
Abstract
Sex differences in the epidemiology and clinical characteristics of schizophrenia are well-known; however, the molecular mechanisms underlying these differences remain unclear. Further, the potential advantages of sex-stratified meta-analyses of epigenome-wide association studies (EWAS) of schizophrenia have not been investigated. Here, we performed sex-stratified EWAS meta-analyses to investigate whether sex stratification improves discovery, and to identify differentially methylated regions (DMRs) in schizophrenia. Peripheral blood-derived DNA methylation data from 1519 cases of schizophrenia (male n = 989, female n = 530) and 1723 controls (male n = 997, female n = 726) from three publicly available datasets, and the TOP cohort were meta-analyzed to compare sex-specific, sex-stratified, and sex-adjusted EWAS. The predictive power of each model was assessed by polymethylation score (PMS). The number of schizophrenia-associated differentially methylated positions identified was higher for the sex-stratified model than for the sex-adjusted one. We identified 20 schizophrenia-associated DMRs in the sex-stratified analysis. PMS from sex-stratified analysis outperformed that from sex-adjusted analysis in predicting schizophrenia. Notably, PMSs from the sex-stratified and female-only analyses, but not those from sex-adjusted or the male-only analyses, significantly predicted schizophrenia in males. The findings suggest that sex-stratified EWAS meta-analyses improve the identification of schizophrenia-associated epigenetic changes and highlight an interaction between sex and schizophrenia status on DNA methylation. Sex-specific DNA methylation may have potential implications for precision psychiatry and the development of stratified treatments for schizophrenia.
Collapse
Affiliation(s)
- Markos Tesfaye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway.
| | - Leticia M Spindola
- NORMENT, 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
- Bergen Center for Brain Plasticity, Haukeland University Hospital, Bergen, Norway
| | - Anne-Kristin Stavrum
- NORMENT, 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
| | - Alexey Shadrin
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ingrid Melle
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Stephanie Le Hellard
- NORMENT, 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.
- Bergen Center for Brain Plasticity, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
45
|
Bandeira CE, Grevet EH, Vitola ES, da Silva BS, Cupertino RB, Picon FA, Ito LT, Tavares MEDA, Rovaris DL, Grimm O, Bau CHD. Exploring Neuroimaging Association Scores in adulthood ADHD and middle-age trajectories. J Psychiatr Res 2024; 176:348-353. [PMID: 38936238 DOI: 10.1016/j.jpsychires.2024.06.025] [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: 12/17/2023] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Attention-Deficit/Hyperactivity Disorder (ADHD) is a neurodevelopmental disorder associated with brain differences in children, but not in adults. A combined evaluation of the regional brain differences could improve statistical power and, consequently, allow the detection of possible effects in adults. Thus, our aim is to verify whether Neuroimaging Association Scores (NAS) are associated with adulthood ADHD and clinical trajectories of the disorder in midlife. Clinical and neuroimaging data were collected for 121 subjects with ADHD (mean age: 47.1 ± 10.5; 43% male) and 82 controls (mean age: 38.2 ± 9.0; 54.9% male). Cases were assessed seven and thirteen years after baseline diagnosis, and their clinical trajectories were classified as stable if they fulfilled ADHD diagnosis in all assessments or unstable if they presented remission and recurrence of symptoms. Neuroimaging data were acquired in the last clinical assessment (thirteen years after baseline) and NAS were calculated as a weighted sum of the associations previously reported by meta-analyses for three types of structural brain modalities: cortical thickness, cortical surface area, and subcortical volume. The NAS for cortical surface area was higher in cases compared to controls. No association was found for NAS and number of symptoms of ADHD or clinical trajectories. The fact that differences were restricted to ADHD diagnostic status suggests a susceptibility effect that is not extended to subtle aspects of the disorder. Our results also suggest that evaluating overall effects may have advantages especially when applied to adult ADHD samples.
Collapse
Affiliation(s)
- Cibele Edom Bandeira
- ADHD Outpatient Program, Clinical Research Center, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Instituto de Ciencias Biomedicas da Universidade de São Paulo, São Paulo, Brazil
| | - Eugenio Horacio Grevet
- ADHD Outpatient Program, Clinical Research Center, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Psychiatry, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo Schneider Vitola
- ADHD Outpatient Program, Clinical Research Center, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Instituto de Ciencias Biomedicas da Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Santos da Silva
- ADHD Outpatient Program, Clinical Research Center, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | | | - Felipe Almeida Picon
- ADHD Outpatient Program, Clinical Research Center, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lucas Toshio Ito
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil; Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria Eduarda de Araujo Tavares
- ADHD Outpatient Program, Clinical Research Center, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Diego Luiz Rovaris
- ADHD Outpatient Program, Clinical Research Center, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Instituto de Ciencias Biomedicas da Universidade de São Paulo, São Paulo, Brazil
| | - Oliver Grimm
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Claiton Henrique Dotto Bau
- ADHD Outpatient Program, Clinical Research Center, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
46
|
Liu A, Wang J, Jin T, Jiang Z, Huang S, Li S, Ying Z, Jiang H. Identifying the genetic association between the cerebral cortex and fibromyalgia. Cereb Cortex 2024; 34:bhae318. [PMID: 39106177 DOI: 10.1093/cercor/bhae318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/08/2024] [Indexed: 08/09/2024] Open
Abstract
Fibromyalgia (FM) is a central sensitization syndrome that is strongly associated with the cerebral cortex. This study used bidirectional two-sample Mendelian randomization (MR) analysis to investigate the bidirectional causality between FM and the cortical surface area and cortical thickness of 34 brain regions. Inverse variance weighted (IVW) was used as the primary method for this study, and sensitivity analyses further supported the results. The forward MR analysis revealed that genetically determined thinner cortical thickness in the parstriangularis (OR = 0.0567 mm, PIVW = 0.0463), caudal middle frontal (OR = 0.0346 mm, PIVW = 0.0433), and rostral middle frontal (OR = 0.0285 mm, PIVW = 0.0463) was associated with FM. Additionally, a reduced genetically determined cortical surface area in the pericalcarine (OR = 0.9988 mm2, PIVW = 0.0085) was associated with an increased risk of FM. Conversely, reverse MR indicated that FM was associated with cortical thickness in the caudal middle frontal region (β = -0.0035 mm, PIVW = 0.0265), fusiform region (β = 0.0024 mm, SE = 0.0012, PIVW = 0.0440), the cortical surface area in the supramarginal (β = -9.3938 mm2, PIVW = 0.0132), and postcentral regions (β = -6.3137 mm2, PIVW = 0.0360). Reduced cortical thickness in the caudal middle frontal gyrus is shown to have a significant relationship with FM prevalence in a bidirectional causal analysis.
Collapse
Affiliation(s)
- Aihui Liu
- Department of Rheumatology and Immunology, Center for General Practice Medicine, Hangzhou Medical College, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang 310000, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 31000, China
- Rheumatology and Immunology Research Institute, Hangzhou Medical College, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 310000, China
| | - Jing Wang
- Department of Rheumatology and Immunology, Center for General Practice Medicine, Hangzhou Medical College, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang 310000, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 31000, China
- Rheumatology and Immunology Research Institute, Hangzhou Medical College, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 310000, China
| | - Tianyu Jin
- China Rehabilitation Research center, No. 10, Jiaomen North Road, Fengtai District, Beijing 100068, China
| | - Zhaoyu Jiang
- Department of Rheumatology and Immunology, Center for General Practice Medicine, Hangzhou Medical College, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang 310000, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 31000, China
- Rheumatology and Immunology Research Institute, Hangzhou Medical College, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 310000, China
| | - Shan Huang
- Department of Rheumatology and Immunology, Center for General Practice Medicine, Hangzhou Medical College, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang 310000, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 31000, China
- Rheumatology and Immunology Research Institute, Hangzhou Medical College, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 310000, China
| | - Shinan Li
- Department of Rheumatology and Immunology, Center for General Practice Medicine, Hangzhou Medical College, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang 310000, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 31000, China
- Rheumatology and Immunology Research Institute, Hangzhou Medical College, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 310000, China
| | - Zhenhua Ying
- Department of Rheumatology and Immunology, Center for General Practice Medicine, Hangzhou Medical College, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang 310000, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 31000, China
- Rheumatology and Immunology Research Institute, Hangzhou Medical College, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 310000, China
| | - Hongyang Jiang
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 31000, China
- Rheumatology and Immunology Research Institute, Hangzhou Medical College, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang Province 310000, China
- Department of Radiology, Center for Rehabilitation Medicine, Hangzhou Medical College, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, No. 158 Shangtang Road, Gongshu District, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
47
|
Qu J, Qu Y, Zhu R, Wu Y, Xu G, Wang D. Transcriptional expression patterns of the cortical morphometric similarity network in progressive supranuclear palsy. CNS Neurosci Ther 2024; 30:e14901. [PMID: 39097922 PMCID: PMC11298202 DOI: 10.1111/cns.14901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/09/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND It has been demonstrated that progressive supranuclear palsy (PSP) correlates with structural abnormalities in several distinct regions of the brain. However, whether there are changes in the morphological similarity network (MSN) and the relationship between changes in brain structure and gene expression remain largely unknown. METHODS We used two independent cohorts (discovery dataset: PSP: 51, healthy controls (HC): 82; replication dataset: PSP: 53, HC: 55) for MSN analysis and comparing the longitudinal changes in the MSN of PSP. Then, we applied partial least squares regression to determine the relationships between changes in MSN and spatial transcriptional features and identified specific genes associated with MSN differences in PSP. We further investigated the biological processes enriched in PSP-associated genes and the cellular characteristics of these genes, and finally, we performed an exploratory analysis of the relationship between MSN changes and neurotransmitter receptors. RESULTS We found that the MSN in PSP patients was mainly decreased in the frontal and temporal cortex but increased in the occipital cortical region. This difference is replicable. In longitudinal studies, MSN differences are mainly manifested in the frontal and parietal regions. Furthermore, the expression pattern associated with MSN changes in PSP involves genes implicated in astrocytes and excitatory and inhibitory neurons and is functionally enriched in neuron-specific biological processes related to synaptic signaling. Finally, we found that the changes in MSN were mainly negatively correlated with the levels of serotonin, norepinephrine, and opioid receptors. CONCLUSIONS These results have enhanced our understanding of the microscale genetic and cellular mechanisms responsible for large-scale morphological abnormalities in PSP patients, suggesting potential targets for future therapeutic trials.
Collapse
Affiliation(s)
- Junyu Qu
- Department of RadiologyQilu Hospital of Shandong University, Qilu Medical Imaging Institute of Shandong UniversityJinanChina
| | - Yancai Qu
- Department of NeurosurgeryTraditional Chinese Medicine Hospital of Muping DistrictYantaiChina
| | - Rui Zhu
- Department of RadiologyQilu Hospital of Shandong University, Qilu Medical Imaging Institute of Shandong UniversityJinanChina
| | - Yongsheng Wu
- Department of RadiologyQilu Hospital of Shandong University, Qilu Medical Imaging Institute of Shandong UniversityJinanChina
| | - Guihua Xu
- Department of RadiologyQilu Hospital of Shandong University, Qilu Medical Imaging Institute of Shandong UniversityJinanChina
| | - Dawei Wang
- Department of RadiologyQilu Hospital of Shandong University, Qilu Medical Imaging Institute of Shandong UniversityJinanChina
- Magnetic Field‐free Medicine & Functional ImagingResearch Institute of Shandong UniversityJinanChina
- Magnetic Field‐free Medicine & Functional Imaging (MF)Shandong Key LaboratoryJinanChina
| |
Collapse
|
48
|
Zhao S, Huang Y, Shi S, Chen W, Chen R, Wang Z, Wang D. Causal effects of hypertensive disorders of pregnancy on structural changes in specific brain regions: a Mendelian randomization study. Cereb Cortex 2024; 34:bhae282. [PMID: 38984704 DOI: 10.1093/cercor/bhae282] [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: 02/20/2024] [Revised: 06/16/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
This study utilized Mendelian randomization to explore the impact of hypertensive disorders of pregnancy and their subtypes on brain structures, using genome-wide association study data from the FinnGen consortium for hypertensive disorders of pregnancy exposure and brain structure data from the ENIGMA consortium as outcomes. The inverse-variance weighted method, along with Cochran's Q test, Mendelian randomization-Egger regression, Mendelian randomization-PRESSO global test, and the leave-one-out approach, were applied to infer causality and assess heterogeneity and pleiotropy. Findings indicate hypertensive disorders of pregnancy are associated with structural brain alterations, including reduced cortical thickness in areas like the insula, isthmus cingulate gyrus, superior temporal gyrus, temporal pole, and transverse temporal gyrus, and an increased surface area in the superior frontal gyrus. Specific associations were found for hypertensive disorders of pregnancy subtypes: chronic hypertension with superimposed preeclampsia increased cortical thickness in the supramarginal gyrus; preeclampsia/eclampsia led to thinner cortex in the lingual gyrus and larger hippocampal volume and superior parietal lobule surface area. Chronic hypertension was associated with reduced cortical thickness in the caudal and rostral anterior cingulate and increased surface area of the cuneus and thickness of the pars orbitalis cortex. Gestational hypertension showed no significant brain region changes. These insights clarify hypertensive disorders of pregnancies' neurological and cognitive effects by identifying affected brain regions.
Collapse
Affiliation(s)
- Shanshan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Yihong Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Shaole Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Wei Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Run Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Zilian Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Dongyu Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, 58 Zhongshan Road II, Guangzhou 510080, China
| |
Collapse
|
49
|
Ohi K, Tanaka Y, Otowa T, Shimada M, Kaiya H, Nishimura F, Sasaki T, Tanii H, Shioiri T, Hara T. Discrimination between healthy participants and people with panic disorder based on polygenic scores for psychiatric disorders and for intermediate phenotypes using machine learning. Aust N Z J Psychiatry 2024; 58:603-614. [PMID: 38581251 DOI: 10.1177/00048674241242936] [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] [Indexed: 04/08/2024]
Abstract
OBJECTIVE Panic disorder is a modestly heritable condition. Currently, diagnosis is based only on clinical symptoms; identifying objective biomarkers and a more reliable diagnostic procedure is desirable. We investigated whether people with panic disorder can be reliably diagnosed utilizing combinations of multiple polygenic scores for psychiatric disorders and their intermediate phenotypes, compared with single polygenic score approaches, by applying specific machine learning techniques. METHODS Polygenic scores for 48 psychiatric disorders and intermediate phenotypes based on large-scale genome-wide association studies (n = 7556-1,131,881) were calculated for people with panic disorder (n = 718) and healthy controls (n = 1717). Discrimination between people with panic disorder and healthy controls was based on the 48 polygenic scores using five methods for classification: logistic regression, neural networks, quadratic discriminant analysis, random forests and a support vector machine. Differences in discrimination accuracy (area under the curve) due to an increased number of polygenic score combinations and differences in the accuracy across five classifiers were investigated. RESULTS All five classifiers performed relatively well for distinguishing people with panic disorder from healthy controls by increasing the number of polygenic scores. Of the 48 polygenic scores, the polygenic score for anxiety UK Biobank was the most useful for discrimination by the classifiers. In combinations of two or three polygenic scores, the polygenic score for anxiety UK Biobank was included as one of polygenic scores in all classifiers. When all 48 polygenic scores were used in combination, the greatest areas under the curve significantly differed among the five classifiers. Support vector machine and logistic regression had higher accuracy than quadratic discriminant analysis and random forests. For each classifier, the greatest area under the curve was 0.600 ± 0.030 for logistic regression (polygenic score combinations N = 14), 0.591 ± 0.039 for neural networks (N = 9), 0.603 ± 0.033 for quadratic discriminant analysis (N = 10), 0.572 ± 0.039 for random forests (N = 25) and 0.617 ± 0.041 for support vector machine (N = 11). The greatest areas under the curve at the best polygenic score combination significantly differed among the five classifiers. Random forests had the lowest accuracy among classifiers. Support vector machine had higher accuracy than neural networks. CONCLUSIONS These findings suggest that increasing the number of polygenic score combinations up to approximately 10 effectively improved the discrimination accuracy and that support vector machine exhibited greater accuracy among classifiers. However, the discrimination accuracy for panic disorder, when based solely on polygenic score combinations, was found to be modest.
Collapse
Affiliation(s)
- Kazutaka Ohi
- Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan
- Department of General Internal Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Yuta Tanaka
- Department of Intelligence Science and Engineering, Gifu University Graduate School of Natural Science and Technology, Gifu, Japan
| | - Takeshi Otowa
- Department of Psychiatry, East Medical Center, Nagoya City University, Nagoya, Japan
| | - Mihoko Shimada
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Hisanobu Kaiya
- Panic Disorder Research Center, Warakukai Medical Corporation, Tokyo, Japan
| | - Fumichika Nishimura
- Center for Research on Counseling and Support Services, The University of Tokyo, Tokyo, Japan
| | - Tsukasa Sasaki
- Department of Physical and Health Education, Graduate School of Education, The University of Tokyo, Tokyo, Japan
| | - Hisashi Tanii
- Center for Physical and Mental Health, Mie University, Mie, Japan
- Graduate School of Medicine, Department of Health Promotion and Disease Prevention, Mie University, Mie, Japan
| | - Toshiki Shioiri
- Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takeshi Hara
- Department of Intelligence Science and Engineering, Gifu University Graduate School of Natural Science and Technology, Gifu, Japan
| |
Collapse
|
50
|
Yang HH, Han KM, Kim A, Kang Y, Tae WS, Han MR, Ham BJ. Neuroimaging and epigenetic analysis reveal novel epigenetic loci in major depressive disorder. Psychol Med 2024; 54:2585-2598. [PMID: 38721773 DOI: 10.1017/s0033291724000709] [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] [Indexed: 10/10/2024]
Abstract
BACKGROUND Epigenetic modifications, such as DNA methylation, contribute to the pathophysiology of major depressive disorder (MDD). This study aimed to identify novel MDD-associated epigenetic loci using DNA methylation profiles and explore the correlations between epigenetic loci and cortical thickness changes in patients with MDD. METHODS A total of 350 patients with MDD and 161 healthy controls (HCs) were included in the epigenome-wide association studies (EWAS). We analyzed methylation, copy number alteration (CNA), and gene network profiles in the MDD group. A total of 234 patients with MDD and 135 HCs were included in neuroimaging methylation analysis. Pearson's partial correlation analysis was used to estimate the correlation between cortical thickness of brain regions and DNA methylation levels of the loci. RESULTS In total, 2018 differentially methylated probes (DMPs) and 351 differentially methylated regions (DMRs) were identified. DMP-related genes were enriched in two networks involved in the central nervous system. In neuroimaging analysis, patients with MDD showed cortical thinning in the prefrontal regions and cortical thickening in several occipital regions. Cortical thickness of the left ventrolateral prefrontal cortex (VLPFC, i.e. pars triangularis) was negatively correlated with eight DMPs associated with six genes (EML6, ZFP64, CLSTN3, KCNMA1, TAOK2, and NT5E). CONCLUSION Through combining DNA methylation and neuroimaging analyses, negative correlations were identified between the cortical thickness of the left VLPFC and DNA methylation levels of eight DMPs. Our findings could improve our understanding of the pathophysiology of MDD.
Collapse
Affiliation(s)
- Hyun-Ho Yang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Kyu-Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Brain Convergence Research Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Aram Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Youbin Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo-Suk Tae
- Brain Convergence Research Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Mi-Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Brain Convergence Research Center, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|