1
|
Liao CC, Wu SA, Lee CI, Liao KR, Li JM. Investigating causal relationships between gene expression and major depressive disorder via brain bulk-tissue and cell type-specific eQTL: A Mendelian randomization and Bayesian colocalization study. J Affect Disord 2025; 383:167-178. [PMID: 40311809 DOI: 10.1016/j.jad.2025.04.161] [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: 02/19/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is a highly prevalent psychiatric disorder with complex genetic underpinnings. While genome-wide association studies (GWAS) have identified multiple risk loci, pinpointing causal genes within the human brain remains challenging, particularly given the regulatory complexity across different cell types. METHODS We performed summary data-based MR (SMR) and Bayesian colocalization analyses by integrating bulk-tissue eQTL data from 888 individuals with single-cell eQTL datasets from 192 donors representing major brain cell types (excitatory and inhibitory neurons, astrocytes, microglia, oligodendrocytes, OPCs/COPs, endothelial cells, and pericytes). GWAS summary statistics for MDD (170,756 cases and 329,443 controls) were used to assess the causal impact of gene expression. Sensitivity analyses, including the heterogeneity in dependent instruments (HEIDI) test and Steiger filtering, ensured robust inference. RESULTS In bulk tissue analyses, five genes (BTN3A2, SLC12A5, AREL1, GMPPB, and ZNF660) emerged as having robust causal evidence for MDD, displaying consistent MR signals and strong colocalization. Cell type-specific analyses revealed additional candidate genes in excitatory neurons (FLOT1, AL450423.1), astrocytes (AL121821.1), and oligodendrocytes (YLPM1, COP1). CONCLUSION Our integrative approach reveals that causal gene expression profiles differ markedly between bulk-tissue and specific brain cell types, emphasizing cellular heterogeneity in MDD pathogenesis and informing precision therapeutic strategies. These findings underscore the necessity of considering cell type-specific gene regulation when developing therapeutic interventions for MDD.
Collapse
Affiliation(s)
- Chung-Chih Liao
- Department of Integrated Chinese and Western Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
| | - Shih-An Wu
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Chun-I Lee
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; Division of Infertility, Lee Women's Hospital, Taichung 40652, Taiwan; Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Ke-Ru Liao
- Department of Neurology, Yuanlin Christian Hospital, Yuanlin 51052, Taiwan
| | - Jung-Miao Li
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan.
| |
Collapse
|
2
|
Liu Y, Chen C, Zhao Y, Li M, Gao Y, Yan B, Jing Y, Zhang B, Li J. Transcriptional characteristics of human brain alterations in major depressive disorder: A systematic review. Psychoneuroendocrinology 2025; 177:107472. [PMID: 40288014 DOI: 10.1016/j.psyneuen.2025.107472] [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: 11/19/2024] [Revised: 03/05/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
Many patients with major depressive disorder (MDD) experience limited treatment effectiveness due to an incomplete understanding of its neurobiological underpinnings. This review integrates neuroimaging and genetic data to examine structural and functional brain changes in MDD, alongside their genetic bases. A PRISMA-guided systematic review of imaging transcriptomics over the past decade was conducted using PubMed and Web of Science. Studies included MRI scans of both MDD patients and healthy controls, as well as brain-wide gene expression data, excluding those that were purely meta-analytical, lacked spatial correlations, or involved transdiagnostic analyses. Of the 206 studies reviewed, 20 met the inclusion criteria. Consistent patterns across studies reveal that key biological processes-such as synaptic signaling, calcium ion binding, neurodevelopment, immune regulation, and neurotransmitter transport-play a central role in brain alterations associated with MDD. Additionally, our findings suggest that electroconvulsive therapy (ECT) may alleviate symptoms by modulating these shared pathways. This review underscores the link between brain changes in MDD and specific gene expression profiles, offering insights that could inform more targeted therapeutic approaches.
Collapse
Affiliation(s)
- Yuan Liu
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Chengfeng Chen
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongping Zhao
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Meijuan Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Ying Gao
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Bo Yan
- Department of Geriatrics, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin 300052, China
| | - Yifan Jing
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Bin Zhang
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China.
| | - Jie Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China.
| |
Collapse
|
3
|
Liang S, Gao Y, Palaniyappan L, Song XM, Zhang T, Han JF, Tan ZL, Li T. Transcriptional substrates of cortical thickness alterations in anhedonia of major depressive disorder. J Affect Disord 2025; 379:118-126. [PMID: 40044088 DOI: 10.1016/j.jad.2025.03.003] [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: 08/12/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Anhedonia is a core symptom of major depressive disorder (MDD), which has been shown to be associated with abnormalities in cortical morphology. However, the correlation between cortical thickness (CT) changes with anhedonia in MDD and gene expression remains unclear. METHODS We investigated the link between brain-wide gene expression and CT correlates of anhedonia in individuals with MDD, using 7 Tesla neuroimaging and a publicly available transcriptomic dataset. The interest-activity score was used to evaluation MDD with high anhedonia (HA) and low anhedonia (LA). Nineteen patients with HA, nineteen patients with LA, and twenty healthy controls (HC) were enrolled. We investigated CT alterations of anhedonia subgroups relative to HC and related cortical gene expression, enrichment and specific cell types. We further used Neurosynth and von Economo-Koskinas atlas to assess the meta-analytic cognitive functions and cytoarchitectural variation associated with anhedonia-related cortical changes. RESULTS Both patient subgroups exhibited widespread CT reduction, with HA manifesting more pronounced changes. Gene expression related to anhedonia had significant spatial correlations with CT differences. Transcriptional signatures related to anhedonia-associated cortical thinning were connected to mitochondrial dysfunction and enriched in adipogenesis, oxidative phosphorylation, mTORC1 signaling pathways, involving neurons, astrocytes, and oligodendrocytes. These CT alterations were significantly correlated with meta-analytic terms involving somatosensory processing and pain perception. HA had reduced CT within the somatomotor and ventral attention networks, and in agranular cortical regions. LIMITATIONS These include measuring anhedonia using interest-activity score and employing a cross-sectional design. CONCLUSIONS This study sheds light on the molecular basis underlying gene expression associated with anhedonia in MDD, suggesting directions for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Sugai Liang
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China
| | - Yuan Gao
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China; Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Lena Palaniyappan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec H4H1R3, Canada.; Department of Psychiatry, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A5C1, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A5K8, Canada
| | - Xue-Mei Song
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China; Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Tian Zhang
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China
| | - Jin-Fang Han
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China
| | - Zhong-Lin Tan
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China.
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 310000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310063, China.
| |
Collapse
|
4
|
Petrican R, Chopra S, Murgatroyd C, Fornito A. Sex-Differential Markers of Psychiatric Risk and Treatment Response Based on Premature Aging of Functional Brain Network Dynamics and Peripheral Physiology. Biol Psychiatry 2025; 97:1091-1103. [PMID: 39419460 DOI: 10.1016/j.biopsych.2024.10.008] [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: 06/21/2024] [Revised: 09/16/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Aging is a multilevel process of gradual decline that predicts morbidity and mortality. Independent investigations have implicated senescence of brain and peripheral physiology in psychiatric risk, but it is unclear whether these effects stem from unique or shared mechanisms. METHODS To address this question, we analyzed clinical, blood chemistry, and resting-state functional neuroimaging data in a healthy aging cohort (n = 427; ages 36-100 years) and 2 disorder-specific samples including patients with early psychosis (100 patients, 16-35 years) and major depressive disorder (MDD) (104 patients, 20-76 years). RESULTS We identified sex-dependent coupling between blood chemistry markers of metabolic senescence (i.e., homeostatic dysregulation), functional brain network aging, and psychiatric risk. In females, premature aging of frontoparietal and somatomotor networks was linked to greater homeostatic dysregulation. It also predicted the severity and treatment resistance of mood symptoms (depression/anxiety [all 3 samples], anhedonia [MDD]) and social withdrawal/behavioral inhibition (avoidant personality disorder [healthy aging], negative symptoms [early psychosis]). In males, premature aging of the default mode, cingulo-opercular, and visual networks was linked to reduced homeostatic dysregulation and predicted the severity and treatment resistance of symptoms relevant to hostility/aggression (antisocial personality disorder [healthy aging], mania/positive symptoms [early psychosis]), impaired thought processes (early psychosis, MDD), and somatic problems (healthy aging, MDD). CONCLUSIONS Our findings identify sexually dimorphic relationships between brain dynamics, peripheral physiology, and risk for psychiatric illness, suggesting that the specificity of putative risk biomarkers and precision therapeutics may be improved by considering sex and other relevant personal characteristics.
Collapse
Affiliation(s)
- Raluca Petrican
- Institute of Population Health, Department of Psychology, University of Liverpool, Liverpool, United Kingdom.
| | - Sidhant Chopra
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christopher Murgatroyd
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
5
|
Byeon H. Unveiling the invisible: How cutting-edge neuroimaging transforms adolescent depression diagnosis. World J Psychiatry 2025; 15:102953. [DOI: 10.5498/wjp.v15.i5.102953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 04/30/2025] Open
Abstract
Yu et al's study has advanced the understanding of the neural mechanisms underlying major depressive disorder (MDD) in adolescents, emphasizing the significant role of the amygdala. While traditional diagnostic methods have limitations in objectivity and accuracy, this research demonstrates a notable advancement through the integration of machine learning techniques with neuroimaging data. Utilizing resting-state functional magnetic resonance imaging (fMRI), the study investigated functional connectivity (FC) in adolescents with MDD, identifying notable reductions in regions such as the left inferior temporal gyrus and right lingual gyrus, alongside increased connectivity in Vermis-10. The application of support vector machines (SVM) to resting-state fMRI (rs-fMRI) data achieved an accuracy of 83.91%, sensitivity of 79.55%, and specificity of 88.37%, with an area under the curve of 0.6765. These results demonstrate how SVM analysis of rs-fMRI data represents a significant improvement in diagnostic precision, with reduced FC in the right lingual gyrus emerging as a particularly critical marker. These findings underscore the critical role of the amygdala in MDD pathophysiology and highlight the potential of rs-fMRI and SVM as tools for identifying reliable neuroimaging biomarkers.
Collapse
Affiliation(s)
- Haewon Byeon
- Worker's Care and Digital Health Lab, Department of Future Technology, Korea University of Technology and Education, Cheonan 31253, South Korea
| |
Collapse
|
6
|
Hu J, Cui B, Wang Z, Wang J, Xu X, Lu J. Transcriptomic and glucose metabolism of connectome dynamics variability in temporal lobe epilepsy revealed by simultaneous PET-fMRI. Neurobiol Dis 2025; 212:106967. [PMID: 40398518 DOI: 10.1016/j.nbd.2025.106967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 05/02/2025] [Accepted: 05/18/2025] [Indexed: 05/23/2025] Open
Abstract
Temporal lobe epilepsy (TLE) is associated to genetic predisposition, metabolic abnormalities, and disruptions in brain connectivity. However, the relationships between genetic factors, metabolic processes, and brain network dynamics are not yet fully understood. Simultaneous positron emission tomography and function magnetic resonance imaging (PET/fMRI) data were collected from 66 patients with TLE and 38 healthy controls (HCs). We compared differences in brain network dynamics between TLE patients and HCs using the multilayer network model constructed from extensive temporal features extracted from fMRI. Postmortem whole brain gene expression data were then utilized to identify genes associated with alterations in TLE connectome dynamics, with subsequent enrichment analysis for functional annotation, cellular, and disease associations. Mediation analysis further explored the interrelations among gene expression, glucose metabolism as measured by PET, and brain network dynamics as measured by fMRI. Compared with HCs, individuals with TLE exhibited increased module variability primarily in the default mode network and reduced module variability in the attention network. These case-control differences were validated through split-half analyses and remained unaffected by medication or lateralization. These aberrant module variability patterns were associated with gene expression profiles predominantly related to inhibitory neurons, postsynaptic cell components, MAPK signaling pathway, and these genes were significantly enriched relative to established epilepsy-related gene sets. Moreover, we observed that the effect of gene expression profile on the alterations in TLE connectome dynamics was significantly mediated by changes in glucose metabolism. These findings highlight that alterations in brain network dynamics in TLE are associated with transcriptomic signatures, and that glucose metabolic changes partially mediate this relationship, furthering insights into the biological basis of the disorder.
Collapse
Affiliation(s)
- Jie Hu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Bixiao Cui
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenming Wang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jingjuan Wang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoyin Xu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.
| |
Collapse
|
7
|
Poggi G, Treccani G, von der Bey M, Tanti A, Schmeisser MJ, Müller M. Canonical and non-canonical roles of oligodendrocyte precursor cells in mental disorders. NPJ MENTAL HEALTH RESEARCH 2025; 4:19. [PMID: 40374740 DOI: 10.1038/s44184-025-00133-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025]
Abstract
Psychiatric research has shifted from a neuroncentric view to understanding mental disorders as disturbances of heterogeneous brain networks. Oligodendrocyte precursor cells (OPCs)- actively involved in the modulation of neuronal functions - are altered in psychiatric patients, but the extent and related consequences are unclear. This review explores canonical and non-canonical OPC-related pathways in schizophrenia, bipolar disorder, post-traumatic stress disorder, and depression in humans, highlighting potential mechanisms shared across diagnostic entities.
Collapse
Affiliation(s)
- Giulia Poggi
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Giulia Treccani
- Department of Systemic Neuroscience Institute of Anatomy and Cell Biology, Philipps Universität Marburg, Marburg, Germany
| | - Martina von der Bey
- Molecular and Translational Neuroscience, Department of Neurology, University Hospital Ulm, Ulm, Germany
| | - Arnaud Tanti
- Inserm, UMR 1253, iBrain, Université de Tours, Tours, France
| | - Michael J Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marianne Müller
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Leibniz Institute for Resilience Research, Mainz, Germany
| |
Collapse
|
8
|
Yao J, Zhou Z, Tong Q, Li L, Wei J, Lu J, Hu S, Bao A, He H. Magnetic resonance imaging of postmortem human brain specimens: methodological considerations and prospects in psychoradiology. PSYCHORADIOLOGY 2025; 5:kkaf012. [PMID: 40395337 PMCID: PMC12090057 DOI: 10.1093/psyrad/kkaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/14/2025] [Accepted: 05/06/2025] [Indexed: 05/22/2025]
Abstract
Ex vivo magnetic resonance imaging (MRI) has revolutionized psychoradiological research by enabling detailed structural and pathological assessments of the brain in conditions ranging from psychiatric disorders to neurodegenerative diseases. By providing high-resolution images of postmortem brain tissue, ex vivo MRI overcomes several limitations inherent in in vivo imaging, offering unparalleled insights into the underlying pathophysiology of mental disorders. This review critically summarizes the state-of-the-art ex vivo MRI methodologies for neuroanatomical mapping and pathological characterization in psychoradiology, while also establishing standardized specimen processing protocols. Furthermore, we explore the prospects of application in ex vivo MRI in schizophrenia, major depressive disorder and bipolar disorder, highlighting its role in understanding neuroanatomical alterations, disease progression, and the validation of in vivo neuroimaging biomarkers.
Collapse
Affiliation(s)
- Junye Yao
- Center for Brain Imaging Science and Technology, Zhejiang University, Hangzhou 310027, China
- Clinical & Technical Support, Philips Healthcare, Shanghai 200072, China
| | - Zihan Zhou
- Center for Brain Imaging Science and Technology, Zhejiang University, Hangzhou 310027, China
- Stanford University Graduate School of Education, Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Qiqi Tong
- Center for Brain Imaging Science and Technology, Zhejiang University, Hangzhou 310027, China
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou 311121, China
| | - Lingyu Li
- Center for Brain Imaging Science and Technology, Zhejiang University, Hangzhou 310027, China
- Polytechnic Institute, Zhejiang University, Hangzhou 310015, China
| | - Jintao Wei
- Center for Brain Imaging Science and Technology, Zhejiang University, Hangzhou 310027, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| | - Jing Lu
- Department of Psychiatry, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shaohua Hu
- Department of Psychiatry, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Aimin Bao
- National Human Brain Bank for Health and Disease, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hongjian He
- Center for Brain Imaging Science and Technology, Zhejiang University, Hangzhou 310027, China
- School of Physics, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China
| |
Collapse
|
9
|
Chase HW, Hafeman DM, Ghane M, Skeba A, Brady T, Aslam HA, Stiffler R, Bonar L, Graur S, Bebko G, Bertocci M, Iyengar S, Phillips ML. Reproducible Effects of Sex and Acquisition Order on Multiple Global Signal Metrics: Implications for Functional Connectivity Studies of Phenotypic Individual Differences Using fMRI. Brain Behav 2025; 15:e70141. [PMID: 40200728 PMCID: PMC11979359 DOI: 10.1002/brb3.70141] [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: 12/06/2023] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 04/10/2025] Open
Abstract
PURPOSE The identification of relationships between individual differences in functional connectivity (FC) and behavior has been the focus of considerable investigation. Although emerging evidence has identified relationships between FC and cognitive performance, relationships between FC and measures of affect, including depressed mood, anhedonia, and anxiety, and decision-making style, including impulsivity and sensation seeking, appear to be more inconsistent across the literature. This may be due to low power, methodological differences across studies, including the use of global signal correction (GSR), or uncontrolled characteristics of the population. METHODS Here, we evaluated measures of FC, regional variance, and global signal (GS) across six functional MRI (fMRI) sequences of different tasks and resting states and their relationship with individual differences in self-reported measures of symptoms of depression, anxiety, impulsivity, reward sensitivity, and sensation seeking, as well as demographic variables and acquisition order, within groups of distressed and healthy young adults (18-25 years old). FINDINGS Adopting a training/testing sample structure to the analysis, we found no evidence of reproducible brain/behavior relationships despite identifying regions and connections that reflect reliable between-scan individual differences. However, summary measures of the GS were reproducibly associated with sex: The most consistent finding was an increase in low frequency variance of the blood-oxygenation-level-dependent (BOLD) signal from all gray matter regions in males relative to females. Post hoc analysis of GS topography yielded sex differences in a number of regions, including cerebellum and putamen. In addition, effects of paradigm acquisition order were observed on GS measures, including an increase in BOLD signal variance across time. In an exploratory analysis, a specific relationship between sex and relative high-frequency within-scanner motion was observed. CONCLUSIONS Together, the findings suggest that FC relationships with affective measures may be inconsistent or modest, but that global phenomena related to state and individual differences can be robust and must be evaluated, particularly in studies of psychiatric disorders such as mood disorders or ADHD, which show sex differences.
Collapse
Affiliation(s)
- Henry W. Chase
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Danella M. Hafeman
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Merage Ghane
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Alexander Skeba
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Tyler Brady
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Haris A. Aslam
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Richelle Stiffler
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Lisa Bonar
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Simona Graur
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Genna Bebko
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Michele Bertocci
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Satish Iyengar
- Department of StatisticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Mary L. Phillips
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
10
|
Li J, Long Z, Ji GJ, Han S, Chen Y, Yao G, Xu Y, Zhang K, Zhang Y, Cheng J, Wang K, Chen H, Liao W. Major depressive disorder on a neuromorphic continuum. Nat Commun 2025; 16:2405. [PMID: 40069198 PMCID: PMC11897166 DOI: 10.1038/s41467-025-57682-0] [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: 09/25/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
The heterogeneity of major depressive disorder (MDD) has hindered clinical translation and neuromarker identification. Biotyping facilitates solving the problems of heterogeneity, by dissecting MDD patients into discrete subgroups. However, interindividual variations suggest that depression may be conceptualized as a "continuum," rather than as a "category." We use a Bayesian model to decompose structural MRI features of MDD patients from a multisite cross-sectional cohort into three latent disease factors (spatial pattern) and continuum factor compositions (individual expression). The disease factors are associated with distinct neurotransmitter receptors/transporters obtained from open PET sources. Increases cortical thickness in sensory and decreases in orbitofrontal cortices (Factor 1) associate with norepinephrine and 5-HT2A density, decreases in the cingulo-opercular network and subcortex (Factor 2) associate with norepinephrine and 5-HTT density, and increases in social and affective brain systems (Factor 3) relate to 5-HTT density. Disease factor patterns can also be used to predict depressive symptom improvement in patients from the longitudinal cohort. Moreover, individual factor expressions in MDD are stable over time in a longitudinal cohort, with differentially expressed disease controls from a transdiagnostic cohort. Collectively, our data-driven disease factors reveal that patients with MDD organize along continuous dimensions that affect distinct sets of regions.
Collapse
Affiliation(s)
- Jiao Li
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
- 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
| | - Zhiliang Long
- School of Psychology, Southwest University, Chongqing, P.R. China
| | - Gong-Jun Ji
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, P.R. China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Yuan Chen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Guanqun Yao
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, P.R. China
| | - Yong Xu
- Department of Clinical Psychology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P.R. China
| | - Kerang Zhang
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, P.R. China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, P.R. China
| | - Huafu Chen
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
- 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
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China.
- 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.
| |
Collapse
|
11
|
Lin M, Xie D, Luo Y, Dong L, Wei Y, Gong Q, Zhu YZ, Gao J. Trilobatin, a Naturally Occurring GPR158 Ligand, Alleviates Depressive-like Behavior by Promoting Mitophagy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5163-5179. [PMID: 39962827 PMCID: PMC11887424 DOI: 10.1021/acs.jafc.4c05431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 03/06/2025]
Abstract
The G-protein-coupled receptor (GPR158), an orphan receptor, is highly expressed in the medial prefrontal cortex (mPFC) and identified as a novel therapeutic target for depression. Trilobatin is a naturally occurring food additive with potent neuroprotective properties. However, its pharmacological effects and molecular mechanisms against depression remain unknown. Therefore, we explored whether trilobatin alleviates depression by targeting GPR158. Our results indicated that trilobatin alleviated chronic unpredictable mild stress (CUMS)-induced depressive-like behavior in mice. Mitophagy contributed to the antidepressant-like effect of trilobatin, as evidenced by the qRT-PCR array. Furthermore, trilobatin up-regulated autophagy-associated protein expression, restored mitochondrial dynamic balance, and inhibited oxidative stress of mPFC in mice after CUMS insult and in corticosterone-induced primary neuron injury. Intriguingly, trilobatin directly bound to GPR158 and decreased its level of protein expression. GPR158 deficiency attenuated depressive-like behavior through promoting mitophagy, while the antidepressant effect of trilobatin was strengthened in GPR158-deficient mice. Our findings highlight that GPR158-mediated mitophagy acts as a crucial pharmacological target for depression and reveal a new-found pharmacological property of trilobatin: serving as a novel naturally occurring ligand of GPR158 to safeguard from depression by oxidative stress by promoting mitophagy.
Collapse
Affiliation(s)
- Mu Lin
- School
of Pharmacy, Faculty of Medicine, Macau
University of Science and Technology, Taipa, Macau SAR 999078, China
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi 563006, China
- Guizhou
Aerospace Hospital, Zunyi 563000, China
| | - Dianyou Xie
- School
of Pharmacy, Faculty of Medicine, Macau
University of Science and Technology, Taipa, Macau SAR 999078, China
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi 563006, China
| | - Yunmei Luo
- School
of Pharmacy, Faculty of Medicine, Macau
University of Science and Technology, Taipa, Macau SAR 999078, China
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi 563006, China
| | - Lan Dong
- School
of Pharmacy, Faculty of Medicine, Macau
University of Science and Technology, Taipa, Macau SAR 999078, China
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi 563006, China
| | - Yu Wei
- Department
of Pharmacy the Affiliated Hospital of Zunyi Medical University, Zunyi 563099, China
| | - Qihai Gong
- School
of Pharmacy, Faculty of Medicine, Macau
University of Science and Technology, Taipa, Macau SAR 999078, China
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi 563006, China
| | - Yi Zhun Zhu
- School
of Pharmacy, Faculty of Medicine, Macau
University of Science and Technology, Taipa, Macau SAR 999078, China
| | - Jianmei Gao
- School
of Pharmacy, Faculty of Medicine, Macau
University of Science and Technology, Taipa, Macau SAR 999078, China
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi 563006, China
| |
Collapse
|
12
|
Li XY, Rao Y, Li GH, He L, Wang Y, He W, Fang P, Pei C, Xi L, Xie H, Lu YR. Single-nucleus RNA sequencing uncovers metabolic dysregulation in the prefrontal cortex of major depressive disorder patients. Sci Rep 2025; 15:7418. [PMID: 40033004 PMCID: PMC11876315 DOI: 10.1038/s41598-025-92030-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
Major depressive disorder (MDD) is a widespread psychiatric condition, recognized as the third leading cause of global disease burden in 2008. In the context of MDD, alterations in synaptic transmission within the prefrontal cortex (PFC) are associated with PFC hypoactivation, a key factor in cognitive function and mood regulation. Given the high energy demands of the central nervous system, these synaptic changes suggest a metabolic imbalance within the PFC of MDD patients. However, the cellular mechanisms underlying this metabolic dysregulation remain not fully elucidated. This study employs single-nucleus RNA sequencing (snRNA-seq) data to predict metabolic alterations in the dorsolateral PFC (DLPFC) of MDD patients. Our analysis revealed cell type-specific metabolic patterns, notably the disruption of oxidative phosphorylation and carbohydrate metabolism in the DLPFC of MDD patients. Gene set enrichment analysis based on human phenotype ontology predicted alterations in serum lactate levels in MDD patients, corroborated by the observed decrease in lactate levels in MDD patients compared to 47 age-matched healthy controls (HCs). This transcriptional analysis offers novel insights into the metabolic disturbances associated with MDD and the energy dynamics underlying DLPFC hypoactivation. These findings are instrumental for comprehending the pathophysiology of MDD and may guide the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xiang-Yao Li
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Yingbo Rao
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Guo-Hao Li
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Luxi He
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yaohan Wang
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wenli He
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ping Fang
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chenyu Pei
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Lun Xi
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Haiyan Xie
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yun-Rong Lu
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- Department of Psychiatry, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
13
|
Shen S, Liang L, Shi T, Shen Z, Yin S, Zhang J, Li W, Mi W, Wang Y, Zhang Y, Yu J. Microglia-Derived Interleukin-6 Triggers Astrocyte Apoptosis in the Hippocampus and Mediates Depression-Like Behavior. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412556. [PMID: 39888279 PMCID: PMC11923973 DOI: 10.1002/advs.202412556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/19/2025] [Indexed: 02/01/2025]
Abstract
In patients with major depressive disorder (MDD) and animal models of depression, key pathological hallmarks include activation of microglia as well as atrophy and loss of astrocytes. Under certain pathological conditions, microglia can inflict damage to neurons and astrocytes. However, the precise mechanisms underlying how activated microglia induced astrocyte atrophy and loss remain enigmatic. In this study, a depression model induced by chronic social defeat stress (CSDS) is utilized. The results show that CSDS induces significant anxiety- and depression-like behaviors, along with notable astrocyte atrophy and apoptosis, microglial activation, and elevated levels of microglial interleukin-6 (IL-6). Subsequent studies demonstrate that IL-6 released from activated microglia promotes astrocyte apoptosis. Furthermore, the knockdown of the P2X7 receptor (P2X7R) in microglia, which is implicated in the stress response, reduces stress-induced microglial activation, IL-6 release, and astrocyte apoptosis. Direct inhibition of microglia by minocycline corroborates these effects. The selective knockdown of IL-6 in microglia and IL-6 receptors in astrocytes effectively mitigates depression-like behaviors and reduces astrocyte atrophy. This study identifies microglial IL-6 as a key factor that contributes to astrocyte apoptosis and depressive symptoms. Consequently, the IL-6/IL-6R pathway has emerged as a promising target for the treatment of depression.
Collapse
Affiliation(s)
- Shi‐Yu Shen
- Department of Integrative Medicine and NeurobiologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceDepartment of Translational NeuroscienceJing'an District Centre Hospital of ShanghaiInstitutes of Brain ScienceFudan UniversityShanghai200032China
| | - Ling‐Feng Liang
- Department of Integrative Medicine and NeurobiologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Tian‐Le Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceDepartment of Translational NeuroscienceJing'an District Centre Hospital of ShanghaiInstitutes of Brain ScienceFudan UniversityShanghai200032China
| | - Zu‐Qi Shen
- Department of Integrative Medicine and NeurobiologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Shu‐Yuan Yin
- Department of Integrative Medicine and NeurobiologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jia‐Rui Zhang
- Department of Integrative Medicine and NeurobiologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Wei Li
- Department of Integrative Medicine and NeurobiologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Wen‐Li Mi
- Department of Integrative Medicine and NeurobiologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint FunctionFudan UniversityShanghai200433China
| | - Yan‐Qing Wang
- Department of Integrative Medicine and NeurobiologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceDepartment of Translational NeuroscienceJing'an District Centre Hospital of ShanghaiInstitutes of Brain ScienceFudan UniversityShanghai200032China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint FunctionFudan UniversityShanghai200433China
| | - Yu‐Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceDepartment of Translational NeuroscienceJing'an District Centre Hospital of ShanghaiInstitutes of Brain ScienceFudan UniversityShanghai200032China
| | - Jin Yu
- Department of Integrative Medicine and NeurobiologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint FunctionFudan UniversityShanghai200433China
| |
Collapse
|
14
|
Raven F, Medina AV, Schmidt K, He A, Vankampen AA, Balendran V, Aton SJ. Brief sleep disruption alters synaptic structures among hippocampal and neocortical somatostatin-expressing interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.22.604591. [PMID: 39211205 PMCID: PMC11360998 DOI: 10.1101/2024.07.22.604591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Study objectives Brief sleep loss alters cognition and synaptic structures of principal neurons in hippocampus and neocortex. However, while in vivo recording and bioinformatic data suggest that inhibitory interneurons are more strongly affected by sleep loss, it is unclear how sleep and sleep deprivation affect interneurons' synapses. Disruption of the SST+ interneuron population seems to be a critical early sign of neuropathology in Alzheimer's dementia, schizophrenia, and bipolar disorder - and the risk of developing all three is increased by habitual sleep loss. We aimed to test how the synaptic structures of SST+ interneurons in various brain regions are affected by brief sleep disruption. Methods We used Brainbow 3.0 to label SST+ interneurons in the dorsal hippocampus, prefrontal cortex, and visual cortex of male SST-CRE transgenic mice, then compared synaptic structures in labeled neurons after a 6-h period of ad lib sleep, or gentle handling sleep deprivation (SD) starting at lights on. Results Dendritic spine density among SST+ interneurons in both hippocampus and neocortex was altered in a subregion-specific manner, with increased overall and thin spine density in CA1, dramatic increases in spine volume and surface area in CA3, and small but significant changes (primarily decreases) in spine size in CA1, PFC and V1. Conclusions Our suggest that the synaptic connectivity of SST+ interneurons is significantly altered in a brain region-specific manner by a few hours of sleep loss. This suggests a cell type-specific mechanism by which sleep loss disrupts cognition and alters excitatory-inhibitory balance in brain networks.
Collapse
Affiliation(s)
- Frank Raven
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Alexis Vega Medina
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Kailynn Schmidt
- University of New England College of Osteopathic Medicine, Biddeford, ME 04005
| | - Annie He
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Anna A. Vankampen
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Vinodh Balendran
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Sara J. Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| |
Collapse
|
15
|
Xue K, Liu F, Liang S, Guo L, Shan Y, Xu H, Xue J, Jiang Y, Zhang Y, Lu J. Brain connectivity and transcriptomic similarity inform abnormal morphometric similarity patterns in first-episode, treatment-naïve major depressive disorder. J Affect Disord 2025; 370:519-531. [PMID: 39522735 DOI: 10.1016/j.jad.2024.11.021] [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: 06/06/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is associated with disrupted brain structural integration. Morphometric similarity offers a means to capture the coordinated patterns of various structural features. However, it remains unknown whether MDD-related changes can be detected in cortical morphometric similarity through the Morphometric Inverse Divergence (MIND) network. Additionally, the role of brain connectivity in shaping these alterations, and their links to neuroreceptors and gene expression, have yet to be investigated. METHODS Using the T1-weighted MRI data from 71 patients with first-episode, treatment-naïve MDD and 69 healthy controls, we constructed the MIND network for all participants. We then performed between-group comparisons to investigate abnormalities in the network and spatial relationships between the observed patterns of MIND disruption and the patterns of neuroreceptors were estimated. Network-based spreading was utilized to explore the abnormalities constrained by brain connectivity based on structural, functional, and transcriptional connectome architecture and to further identify potential epicenters of MDD. In addition, partial least squares regression was conducted to examine the associations of gene expression profiles with MIND changes in MDD. RESULTS Patients with MDD showed significantly increased MIND in regions associated with sensation and cognition compared with healthy controls, with this altered pattern being influenced by a combination of transcriptional and structural connectivity, and potential epicenters of MDD were identified in the frontal, parietal, and paracentral cortices. Furthermore, the cortical map of case-control differences in MIND was spatially correlated with the cannabinoid CB1 receptor and the brain-wide expression of a weighted combination of genes. These genes were enriched for neurobiologically relevant pathways and preferentially expressed in different cell classes and cortical layers. CONCLUSION These results highlight the abnormal pattern of morphometric similarity observed in MDD, shedding light on the complex interplay between disrupted macroscale coordinated morphology and microscale molecular organization in MDD.
Collapse
Affiliation(s)
- Kaizhong Xue
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China; Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Sixiang Liang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China; Tianjin Anding Hospital, Tianjin 300222, China
| | - Lining Guo
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yi Shan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Huijuan Xu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Jiao Xue
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Yifan Jiang
- School of Nursing, Tianjin Medical University, Tianjin 300070, China
| | - Yong Zhang
- Tianjin Anding Hospital, Tianjin 300222, China.
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China.
| |
Collapse
|
16
|
Tang Q, Peng J, Li Y, Liu L, Wang P, Chen H, Biswal BB. Putative epicenters identified by transcriptome-neuromorphic interactions in attention-deficit/hyperactivity disorder biotypes. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111247. [PMID: 39761817 DOI: 10.1016/j.pnpbp.2025.111247] [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: 09/25/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a heterogenous behavioral disorder with inattention, hyperactivity and impulsivity symptoms, indicating the important implication of identifying biotypes and its epicenters in understanding disease's pathogenesis. The study investigated the neuromorphic heterogeneity relating to transcriptional similarity architecture in ADHD, and further analyzed the epicenters of network-spreading in each ADHD biotype and their correlations with clinical characteristics. Individuals with ADHD could be identified into two discriminative biotypes that exhibited distinct neuromorphic aberrances. As increased regional cortical thickness deviation in ADHD, the first component of partial least squares (PLS1) positively weighted genes were over-expressed, whereas PLS1 negatively weighted genes were under-expressed as its reduction. Both ADHD biotypes exhibited distinct disease epicenters that distributed in cognitive control and attention networks with significantly heterogeneous characteristics, holding promise for advancing our understanding, and ultimately the treatment, of ADHD. Overall, our findings identified two discriminative biotypes and its epicenters in ADHD, promoting the understanding of underlying transcriptome-neuroimaging relationships.
Collapse
Affiliation(s)
- Qin Tang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jinzhong Peng
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yilu Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Lin Liu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Pan Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China.
| | - Huafu Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China.
| | - Bharat B Biswal
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China; Department of Biomedical Engineering, New Jersey Institute of Technology, 11, Newark, NJ, 07102, USA.
| |
Collapse
|
17
|
Zhang H, Sun H, Li J, Lv Z, Tian Y, Lei X. Gene expression is associated with brain function of insomnia disorder, rather than brain structure. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111209. [PMID: 39617164 DOI: 10.1016/j.pnpbp.2024.111209] [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: 06/23/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Previous research has found brain structural and functional abnormalities in patients with insomnia disorder (ID). However, the relationship between brain abnormalities in ID and brain gene expression is unclear. This study explored the relationship between gene expression and brain structural or functional abnormalities in ID, and we validated the reliability of the results with two independent datasets (discover dataset: healthy control (HC) = 129, ID = 264; validation dataset: HC = 160, ID = 115). Brain imaging results show that ID has abnormal resting-state spontaneous activity, regional homogeneity, and widespread gray matter volume reduction compared to HC. The association analysis results with gene expression further revealed that brain function abnormalities in ID were significantly associated with gene expression, but structural abnormalities were not. This study establishes a link between transcriptional changes and brain functional abnormalities in ID, revealing a genetic basis that may involve several biological pathways. Specifically, these pathways include hormonal regulation of the hypothalamic-pituitary-adrenal (HPA) axis, which plays a crucial role in stress response and sleep regulation; ion transport across membranes, vital for neuronal communication; and inhibitory neuronal regulation, essential for maintaining normal brain function. Furthermore, the ID-related genes are enriched for brain tissue and cortical cells, emphasizing their relevance in understanding the biological underpinnings of ID.
Collapse
Affiliation(s)
- Haobo Zhang
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Haonan Sun
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Jiatao Li
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Zhangwei Lv
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Yun Tian
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Xu Lei
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China.
| |
Collapse
|
18
|
Zhang XH, Anderson KM, Dong HM, Chopra S, Dhamala E, Emani PS, Gerstein MB, Margulies DS, Holmes AJ. The cell-type underpinnings of the human functional cortical connectome. Nat Neurosci 2025; 28:150-160. [PMID: 39572742 DOI: 10.1038/s41593-024-01812-2] [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: 07/27/2023] [Accepted: 09/26/2024] [Indexed: 11/27/2024]
Abstract
The functional properties of the human brain arise, in part, from the vast assortment of cell types that pattern the cerebral cortex. The cortical sheet can be broadly divided into distinct networks, which are embedded into processing streams, or gradients, that extend from unimodal systems through higher-order association territories. Here using microarray data from the Allen Human Brain Atlas and single-nucleus RNA-sequencing data from multiple cortical territories, we demonstrate that cell-type distributions are spatially coupled to the functional organization of cortex, as estimated through functional magnetic resonance imaging. Differentially enriched cells follow the spatial topography of both functional gradients and associated large-scale networks. Distinct cellular fingerprints were evident across networks, and a classifier trained on postmortem cell-type distributions was able to predict the functional network allegiance of cortical tissue samples. These data indicate that the in vivo organization of the cortical sheet is reflected in the spatial variability of its cellular composition.
Collapse
Affiliation(s)
- Xi-Han Zhang
- Department of Psychology, Yale University, New Haven, CT, USA.
| | | | - Hao-Ming Dong
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Sidhant Chopra
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Elvisha Dhamala
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Prashant S Emani
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Mark B Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
| | - Daniel S Margulies
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Cognitive Neuroanatomy Lab, Université Paris Cité, INCC UMR 8002, CNRS, Paris, France
| | - Avram J Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
19
|
Snijders GJLJ, Gigase FAJ. Neuroglia in mood disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:287-302. [PMID: 40148049 DOI: 10.1016/b978-0-443-19102-2.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Multiple lines of evidence indicate that mood disorders, such as major depressive and bipolar disorder, are associated with abnormalities in neuroglial cells. This chapter discusses the existing literature investigating the potential role of astrocytes, oligodendrocytes, and microglia in mood pathology. We will describe evidence from in vivo imaging, postmortem, animal models based on (stress) paradigms that mimic depressive-like behavior, and biomarker studies in blood and cerebrospinal fluid in patients with mood disorders. The effect of medication used in the treatment of mood disorders, such as antidepressants and lithium, on glial function is discussed. Lastly, we highlight the most relevant findings about potential deficiencies in glia-glia crosstalk in mood disorders. Overall, decreased astrocyte and oligodendrocyte density and expression and microglial changes in homeostatic functions have frequently been put forward in MDD pathology. Studies of BD report similar findings to some extent; however, the evidence is less well established. Together, these findings are suggestive of reduced glial cell function leading to potential white matter abnormalities, glutamate dysregulation, disrupted neuronal functioning, and neurotransmission. However, more research is required to better understand the exact mechanisms underlying glial cell contributions to mood disorder development.
Collapse
Affiliation(s)
- Gijsje J L J Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Frederieke A J Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
20
|
Parkes L, Kim JZ, Stiso J, Brynildsen JK, Cieslak M, Covitz S, Gur RE, Gur RC, Pasqualetti F, Shinohara RT, Zhou D, Satterthwaite TD, Bassett DS. A network control theory pipeline for studying the dynamics of the structural connectome. Nat Protoc 2024; 19:3721-3749. [PMID: 39075309 PMCID: PMC12039364 DOI: 10.1038/s41596-024-01023-w] [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: 08/23/2023] [Accepted: 05/16/2024] [Indexed: 07/31/2024]
Abstract
Network control theory (NCT) is a simple and powerful tool for studying how network topology informs and constrains the dynamics of a system. Compared to other structure-function coupling approaches, the strength of NCT lies in its capacity to predict the patterns of external control signals that may alter the dynamics of a system in a desired way. An interesting development for NCT in the neuroscience field is its application to study behavior and mental health symptoms. To date, NCT has been validated to study different aspects of the human structural connectome. NCT outputs can be monitored throughout developmental stages to study the effects of connectome topology on neural dynamics and, separately, to test the coherence of empirical datasets with brain function and stimulation. Here, we provide a comprehensive pipeline for applying NCT to structural connectomes by following two procedures. The main procedure focuses on computing the control energy associated with the transitions between specific neural activity states. The second procedure focuses on computing average controllability, which indexes nodes' general capacity to control the dynamics of the system. We provide recommendations for comparing NCT outputs against null network models, and we further support this approach with a Python-based software package called 'network control theory for python'. The procedures in this protocol are appropriate for users with a background in network neuroscience and experience in dynamical systems theory.
Collapse
Affiliation(s)
- Linden Parkes
- Department of Psychiatry, Rutgers University, Piscataway, NJ, USA.
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Lifespan Informatics and Neuroimaging Center (PennLINC), Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Jason Z Kim
- Department of Physics, Cornell University, Ithaca, NY, USA
| | - Jennifer Stiso
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Julia K Brynildsen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Cieslak
- Lifespan Informatics and Neuroimaging Center (PennLINC), Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn/CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sydney Covitz
- Lifespan Informatics and Neuroimaging Center (PennLINC), Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn/CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Raquel E Gur
- Penn/CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruben C Gur
- Penn/CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fabio Pasqualetti
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, USA
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, Philadelphia, PA, USA
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Endeavor (PennSIVE), Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dale Zhou
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Theodore D Satterthwaite
- Lifespan Informatics and Neuroimaging Center (PennLINC), Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn/CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Dani S Bassett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
21
|
Jahanshad N, Lenzini P, Bijsterbosch J. Current best practices and future opportunities for reproducible findings using large-scale neuroimaging in psychiatry. Neuropsychopharmacology 2024; 50:37-51. [PMID: 39117903 PMCID: PMC11526024 DOI: 10.1038/s41386-024-01938-8] [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: 04/16/2024] [Revised: 06/05/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024]
Abstract
Research into the brain basis of psychopathology is challenging due to the heterogeneity of psychiatric disorders, extensive comorbidities, underdiagnosis or overdiagnosis, multifaceted interactions with genetics and life experiences, and the highly multivariate nature of neural correlates. Therefore, increasingly larger datasets that measure more variables in larger cohorts are needed to gain insights. In this review, we present current "best practice" approaches for using existing databases, collecting and sharing new repositories for big data analyses, and future directions for big data in neuroimaging and psychiatry with an emphasis on contributing to collaborative efforts and the challenges of multi-study data analysis.
Collapse
Affiliation(s)
- Neda Jahanshad
- Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of USC, Marina del Rey, CA, 90292, USA.
| | - Petra Lenzini
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Janine Bijsterbosch
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
22
|
Zhukovsky P, Ironside M, Duda JM, Moser AD, Null KE, Dhaynaut M, Normandin M, Guehl NJ, El Fakhri G, Alexander M, Holsen LM, Misra M, Narendran R, Hoye JM, Morris ED, Esfand SM, Goldstein JM, Pizzagalli DA. Acute Stress Increases Striatal Connectivity With Cortical Regions Enriched for μ and κ Opioid Receptors. Biol Psychiatry 2024; 96:717-726. [PMID: 38395372 PMCID: PMC11339240 DOI: 10.1016/j.biopsych.2024.02.005] [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: 09/05/2023] [Revised: 01/22/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Understanding the neurobiological effects of stress is critical for addressing the etiology of major depressive disorder (MDD). Using a dimensional approach involving individuals with differing degree of MDD risk, we investigated 1) the effects of acute stress on cortico-cortical and subcortical-cortical functional connectivity (FC) and 2) how such effects are related to gene expression and receptor maps. METHODS Across 115 participants (37 control, 39 remitted MDD, 39 current MDD), we evaluated the effects of stress on FC during the Montreal Imaging Stress Task. Using partial least squares regression, we investigated genes whose expression in the Allen Human Brain Atlas was associated with anatomical patterns of stress-related FC change. Finally, we correlated stress-related FC change maps with opioid and GABAA (gamma-aminobutyric acid A) receptor distribution maps derived from positron emission tomography. RESULTS Results revealed robust effects of stress on global cortical connectivity, with increased global FC in frontoparietal and attentional networks and decreased global FC in the medial default mode network. Moreover, robust increases emerged in FC of the caudate, putamen, and amygdala with regions from the ventral attention/salience network, frontoparietal network, and motor networks. Such regions showed preferential expression of genes involved in cell-to-cell signaling (OPRM1, OPRK1, SST, GABRA3, GABRA5), similar to previous genetic MDD studies. CONCLUSIONS Acute stress altered global cortical connectivity and increased striatal connectivity with cortical regions that express genes that have previously been associated with imaging abnormalities in MDD and are rich in μ and κ opioid receptors. These findings point to overlapping circuitry underlying stress response, reward, and MDD.
Collapse
MESH Headings
- Humans
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/metabolism
- Male
- Female
- Adult
- Depressive Disorder, Major/diagnostic imaging
- Depressive Disorder, Major/metabolism
- Depressive Disorder, Major/physiopathology
- Depressive Disorder, Major/genetics
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
- Stress, Psychological/diagnostic imaging
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Magnetic Resonance Imaging
- Cerebral Cortex/diagnostic imaging
- Cerebral Cortex/metabolism
- Cerebral Cortex/physiopathology
- Corpus Striatum/diagnostic imaging
- Corpus Striatum/metabolism
- Young Adult
- Positron-Emission Tomography
- Neural Pathways/diagnostic imaging
- Neural Pathways/physiopathology
- Connectome
- Nerve Net/diagnostic imaging
- Nerve Net/metabolism
- Nerve Net/physiopathology
Collapse
Affiliation(s)
- Peter Zhukovsky
- Center for Depression, Anxiety and Stress Research, Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maria Ironside
- Center for Depression, Anxiety and Stress Research, Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, Massachusetts; Laureate Institute for Brain Research, The University of Tulsa, Tulsa, Oklahoma
| | - Jessica M Duda
- Center for Depression, Anxiety and Stress Research, Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amelia D Moser
- Center for Depression, Anxiety and Stress Research, Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, Massachusetts; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado
| | - Kaylee E Null
- Center for Depression, Anxiety and Stress Research, Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, Massachusetts; Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marc Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nicolas J Guehl
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Madeline Alexander
- Center for Depression, Anxiety and Stress Research, Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, Massachusetts
| | - Laura M Holsen
- Division of Women's Health, Brigham and Women's Hospital, Boston, Massachusetts; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts; Clinical Neuroscience Laboratory of Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Madhusmita Misra
- Division of Pediatric Endocrinology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jocelyn M Hoye
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, Connecticut; Department of Radiology and Biomedical Imaging, Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | - Evan D Morris
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, Connecticut; Department of Radiology and Biomedical Imaging, Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | - Shiba M Esfand
- Center for Depression, Anxiety and Stress Research, Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jill M Goldstein
- Department of Psychology, Yale University, New Haven, Connecticut; Division of Women's Health, Brigham and Women's Hospital, Boston, Massachusetts; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts; Clinical Neuroscience Laboratory of Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, Massachusetts
| | - Diego A Pizzagalli
- Center for Depression, Anxiety and Stress Research, Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
23
|
Ng B, Tasaki S, Greathouse KM, Walker CK, Zhang A, Covitz S, Cieslak M, Weber AJ, Adamson AB, Andrade JP, Poovey EH, Curtis KA, Muhammad HM, Seidlitz J, Satterthwaite T, Bennett DA, Seyfried NT, Vogel J, Gaiteri C, Herskowitz JH. Integration across biophysical scales identifies molecular and cellular correlates of person-to-person variability in human brain connectivity. Nat Neurosci 2024; 27:2240-2252. [PMID: 39482360 PMCID: PMC11537986 DOI: 10.1038/s41593-024-01788-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/16/2024] [Indexed: 11/03/2024]
Abstract
Brain connectivity arises from interactions across biophysical scales, ranging from molecular to cellular to anatomical to network level. To date, there has been little progress toward integrated analysis across these scales. To bridge this gap, from a unique cohort of 98 individuals, we collected antemortem neuroimaging and genetic data, as well as postmortem dendritic spine morphometric, proteomic and gene expression data from the superior frontal and inferior temporal gyri. Through the integration of the molecular and dendritic spine morphology data, we identified hundreds of proteins that explain interindividual differences in functional connectivity and structural covariation. These proteins are enriched for synaptic structures and functions, energy metabolism and RNA processing. By integrating data at the genetic, molecular, subcellular and tissue levels, we link specific biochemical changes at synapses to connectivity between brain regions. These results demonstrate the feasibility of integrating data from vastly different biophysical scales to provide a more comprehensive understanding of brain connectivity.
Collapse
Affiliation(s)
- Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Kelsey M Greathouse
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Courtney K Walker
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ada Zhang
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Sydney Covitz
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Matt Cieslak
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Audrey J Weber
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashley B Adamson
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julia P Andrade
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Emily H Poovey
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kendall A Curtis
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hamad M Muhammad
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jakob Seidlitz
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ted Satterthwaite
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jacob Vogel
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Clinical Science, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Jeremy H Herskowitz
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
24
|
Zhang C, Xu C, Yan H, Liang J, Li X, Tang C, Yu Y, Xie G, Guo W. Correlations between alterations in global brain functional connectivity in patients with major depressive disorder and their genetic characteristics. World J Biol Psychiatry 2024; 25:560-570. [PMID: 39412289 DOI: 10.1080/15622975.2024.2412651] [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: 07/30/2024] [Revised: 09/20/2024] [Accepted: 09/29/2024] [Indexed: 10/23/2024]
Abstract
This study aims to elucidate the neuroimaging changes associated with major depressive disorder (MDD) and their relationship with genetic characteristics. We conducted a global-brain functional connectivity (GFC) and genetic-neuroimaging correlation analysis on 42 MDD patients and 42 healthy controls (HCs), exploring the correlation between GFC abnormalities and clinical variables. Results showed that compared to HCs, MDD patients had significantly decreased GFC values in the bilateral posterior cingulate cortex/precuneus and increased GFC values in the left and right cerebellum Crus I/II. Additionally, a negative correlation was observed between the GFC values of the left cerebellum Crus I/II and subjective support scores, as well as social support revalued scale total scores. We identified genes associated with GFC changes in MDD, which are enriched in biological processes such as synaptic transmission and ion transport. Our findings indicate the presence of abnormal GFC values in severe depression, complementing the pathological research on the condition. Furthermore, this study provides preliminary evidence for the correlation between social support levels and brain functional connectivity, offering insights into the potential association between GFC changes and gene expression in MDD patients.
Collapse
Affiliation(s)
- Chunguo Zhang
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, China
| | - Caixia Xu
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, China
| | - Haohao Yan
- 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, China
| | - Jiaquan Liang
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, China
| | - Xiaoling Li
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, China
| | - Chaohua Tang
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, China
| | - Yang Yu
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, China
| | - Guojun Xie
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, China
| | - Wenbin Guo
- 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, China
| |
Collapse
|
25
|
Lu Y, Zhang X, Hu L, Cheng Q, Zhang Z, Zhang H, Xie Z, Gao Y, Cao D, Chen S, Xu J. Consistent genes associated with structural changes in clinical Alzheimer's disease spectrum. Front Neurosci 2024; 18:1376288. [PMID: 39554844 PMCID: PMC11564164 DOI: 10.3389/fnins.2024.1376288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Background Previous studies have demonstrated widespread brain neurodegeneration in Alzheimer's disease (AD). However, the neurobiological and pathogenic substrates underlying this structural atrophy across the AD spectrum remain largely understood. Methods In this study, we obtained structural MRI data from ADNI datasets, including 83 participants with early-stage cognitive impairments (EMCI), 83 with late-stage mild cognitive impairments (LMCI), 83 with AD, and 83 with normal controls (NC). Our goal was to explore structural atrophy across the full clinical AD spectrum and investigate the genetic mechanism using gene expression data from the Allen Human Brain Atlas. Results As a result, we identified significant volume atrophy in the left thalamus, left cerebellum, and bilateral middle frontal gyrus across the AD spectrum. These structural changes were positively associated with the expression levels of genes such as ABCA7, SORCS1, SORL1, PILRA, PFDN1, PLXNA4, TRIP4, and CD2AP, while they were negatively associated with the expression levels of genes such as CD33, PLCG2, APOE, and ECHDC3 across the clinical AD spectrum. Further gene enrichment analyses revealed that the positively associated genes were mainly involved in the positive regulation of cellular protein localization and the negative regulation of cellular component organization, whereas the negatively associated genes were mainly involved in the positive regulation of iron transport. Conclusion Overall, these results provide a deeper understanding of the biological mechanisms underlying structural changes in prodromal and clinical AD.
Collapse
Affiliation(s)
- Yingqi Lu
- Department of Rehabilitation Medicine, The People’s Hospital of Baoan Shenzhen, Shenzhen, China
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaodong Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Children’s Hospital, Shenzhen, China
| | - Liyu Hu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qinxiu Cheng
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhewei Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Haoran Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhuoran Xie
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yiheng Gao
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dezhi Cao
- Shenzhen Children’s Hospital, Shenzhen, China
| | - Shangjie Chen
- Department of Rehabilitation Medicine, The People’s Hospital of Baoan Shenzhen, Shenzhen, China
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jinping Xu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
26
|
Long H, Chen Z, Xu X, Zhou Q, Fang Z, Lv M, Yang XH, Xiao J, Sun H, Fan M. Elucidating genetic and molecular basis of altered higher-order brain structure-function coupling in major depressive disorder. Neuroimage 2024; 297:120722. [PMID: 38971483 DOI: 10.1016/j.neuroimage.2024.120722] [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: 03/25/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024] Open
Abstract
Previous studies have shown that major depressive disorder (MDD) patients exhibit structural and functional impairments, but few studies have investigated changes in higher-order coupling between structure and function. Here, we systematically investigated the effect of MDD on higher-order coupling between structural connectivity (SC) and functional connectivity (FC). Each brain region was mapped into embedding vector by the node2vec algorithm. We used support vector machine (SVM) with the brain region embedding vector to distinguish MDD patients from health controls (HCs) and identify the most discriminative brain regions. Our study revealed that MDD patients had decreased higher-order coupling in connections between the most discriminative brain regions and local connections in rich-club organization and increased higher-order coupling in connections between the ventral attentional network and limbic network compared with HCs. Interestingly, transcriptome-neuroimaging association analysis demonstrated the correlations between regional rSC-FC coupling variations between MDD patients and HCs and α/β-hydrolase domain-containing 6 (ABHD6), β 1,3-N-acetylglucosaminyltransferase-9(β3GNT9), transmembrane protein 45B (TMEM45B), the correlation between regional dSC-FC coupling variations and retinoic acid early transcript 1E antisense RNA 1(RAET1E-AS1), and the correlations between regional iSC-FC coupling variations and ABHD6, β3GNT9, katanin-like 2 protein (KATNAL2). In addition, correlation analysis with neurotransmitter receptor/transporter maps found that the rSC-FC and iSC-FC coupling variations were both correlated with neuroendocrine transporter (NET) expression, and the dSC-FC coupling variations were correlated with metabotropic glutamate receptor 5 (mGluR5). Further mediation analysis explored the relationship between genes, neurotransmitter receptor/transporter and MDD related higher-order coupling variations. These findings indicate that specific genetic and molecular factors underpin the observed disparities in higher-order SC-FC coupling between MDD patients and HCs. Our study confirmed that higher-order coupling between SC and FC plays an important role in diagnosing MDD. The identification of new biological evidence for MDD etiology holds promise for the development of innovative antidepressant therapies.
Collapse
Affiliation(s)
- Haixia Long
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Zihao Chen
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Xinli Xu
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Qianwei Zhou
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Zhaolin Fang
- Network Information Center, Zhejiang University of Technology, Hangzhou 310023, China
| | - Mingqi Lv
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Xu-Hua Yang
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Jie Xiao
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Hui Sun
- College of Electrical Engineering, Sichuan University, Chengdu 610065, China.
| | - Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, China.
| |
Collapse
|
27
|
Hu X, Cheng B, Tang Y, Long T, Huang Y, Li P, Song Y, Song X, Li K, Yin Y, Chen X. Gray matter volume and corresponding covariance connectivity are biomarkers for major depressive disorder. Brain Res 2024; 1837:148986. [PMID: 38714227 DOI: 10.1016/j.brainres.2024.148986] [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: 10/13/2023] [Revised: 04/06/2024] [Accepted: 05/04/2024] [Indexed: 05/09/2024]
Abstract
The major depressive disorder (MDD) is a common and severe mental disorder. To identify a reliable biomarker for MDD is important for early diagnosis and prevention. Given easy access and high reproducibility, the structural magnetic resonance imaging (sMRI) is an ideal method to identify the biomarker for depression. In this study, sMRI data of first episode, treatment-naïve 66 MDD patients and 54 sex-, age-, and education-matched healthy controls (HC) were used to identify the differences in gray matter volume (GMV), group-level, individual-level covariance connections. Finally, the abnormal GMV and individual covariance connections were applied to classify MDD from HC. MDD patients showed higher GMV in middle occipital gyrus (MOG) and precuneus (PCun), and higher structural covariance connections between MOG and PCun. In addition, the Allen Human Brain Atlas (AHBA) was applied and revealed the genetic basis for the changes of gray matter volume. Importantly, we reported that GMV in MOG, PCun and structural covariance connectivity between MOG and PCun are able to discriminate MDD from HC. Our results revealed structural underpinnings for MDD, which may contribute towards early discriminating for depression.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Bochao Cheng
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yuying Tang
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Tong Long
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Huang
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Pei Li
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Song
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Xiyang Song
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Kun Li
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yijie Yin
- School of Sociality and Psychology, Southwest Minzu University, Chengdu 610041, China
| | - Xijian Chen
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
28
|
Liu W, Su JP, Zeng LL, Shen H, Hu DW. Gene expression and brain imaging association study reveals gene signatures in major depressive disorder. Brain Commun 2024; 6:fcae258. [PMID: 39185029 PMCID: PMC11342243 DOI: 10.1093/braincomms/fcae258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 06/03/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024] Open
Abstract
Major depressive disorder is often characterized by changes in the structure and function of the brain, which are influenced by modifications in gene expression profiles. How the depression-related genes work together within the scope of time and space to cause pathological changes remains unclear. By integrating the brain-wide gene expression data and imaging data in major depressive disorder, we identified gene signatures of major depressive disorder and explored their temporal-spatial expression specificity, network properties, function annotations and sex differences systematically. Based on correlation analysis with permutation testing, we found 345 depression-related genes significantly correlated with functional and structural alteration of brain images in major depressive disorder and separated them by directional effects. The genes with negative effect for grey matter density and positive effect for functional indices are enriched in downregulated genes in the post-mortem brain samples of patients with depression and risk genes identified by genome-wide association studies than genes with positive effect for grey matter density and negative effect for functional indices and control genes, confirming their potential association with major depressive disorder. By introducing a parameter of dispersion measure on the gene expression data of developing human brains, we revealed higher spatial specificity and lower temporal specificity of depression-related genes than control genes. Meanwhile, we found depression-related genes tend to be more highly expressed in females than males, which may contribute to the difference in incidence rate between male and female patients. In general, we found the genes with negative effect have lower network degree, more specialized function, higher spatial specificity, lower temporal specificity and more sex differences than genes with positive effect, indicating they may play different roles in the occurrence and development of major depressive disorder. These findings can enhance the understanding of molecular mechanisms underlying major depressive disorder and help develop tailored diagnostic and treatment strategies for patients of depression of different sex.
Collapse
Affiliation(s)
- Wei Liu
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| | - Jian-Po Su
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| | - Ling-Li Zeng
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| | - Hui Shen
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| | - De-Wen Hu
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| |
Collapse
|
29
|
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
|
30
|
Sun H, Bai T, Zhang X, Fan X, Zhang K, Zhang J, Hu Q, Xu J, Tian Y, Wang K. Molecular mechanisms underlying structural plasticity of electroconvulsive therapy in major depressive disorder. Brain Imaging Behav 2024; 18:930-941. [PMID: 38664360 DOI: 10.1007/s11682-024-00884-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2024] [Indexed: 08/31/2024]
Abstract
Although previous studies reported structural changes associated with electroconvulsive therapy (ECT) in major depressive disorder (MDD), the underlying molecular basis of ECT remains largely unknown. Here, we combined two independent structural MRI datasets of MDD patients receiving ECT and transcriptomic gene expression data from Allen Human Brain Atlas to reveal the molecular basis of ECT for MDD. We performed partial least square regression to explore whether/how gray matter volume (GMV) alterations were associated with gene expression level. Functional enrichment analysis was conducted using Metascape to explore ontological pathways of the associated genes. Finally, these genes were further assigned to seven cell types to determine which cell types contribute most to the structural changes in MDD patients after ECT. We found significantly increased GMV in bilateral hippocampus in MDD patients after ECT. Transcriptome-neuroimaging association analyses showed that expression levels of 726 genes were positively correlated with the increased GMV in MDD after ECT. These genes were mainly involved in synaptic signaling, calcium ion binding and cell-cell signaling, and mostly belonged to excitatory and inhibitory neurons. Moreover, we found that the MDD risk genes of CNR1, HTR1A, MAOA, PDE1A, and SST as well as ECT related genes of BDNF, DRD2, APOE, P2RX7, and TBC1D14 showed significantly positive associations with increased GMV. Overall, our findings provide biological and molecular mechanisms underlying structural plasticity induced by ECT in MDD and the identified genes may facilitate future therapy for MDD.
Collapse
Affiliation(s)
- Hui Sun
- College of Electrical Engineering, Sichuan University, Chengdu, China
| | - Tongjian Bai
- Department of Neurology, the First Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaodong Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xinxin Fan
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kai Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jiang Zhang
- College of Electrical Engineering, Sichuan University, Chengdu, China
| | - Qingmao Hu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jinping Xu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Yanghua Tian
- Department of Neurology, the First Hospital of Anhui Medical University, Hefei, 230022, China.
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230022, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230022, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230022, China.
- Department of Neurology, the Second Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Kai Wang
- Department of Neurology, the First Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230022, China
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230022, China
- Anhui Province clinical research center for neurological disease, Hefei, 230022, China
| |
Collapse
|
31
|
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
|
32
|
Yang M, Wang Z, Cao X, Zhu J, Chen Y. Susceptibility or resilience to childhood peer abuse can be explained by cortical thickness in brain regions involved in emotional regulation. Psychiatry Res Neuroimaging 2024; 342:111829. [PMID: 38875765 DOI: 10.1016/j.pscychresns.2024.111829] [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: 01/16/2024] [Revised: 04/08/2024] [Accepted: 05/14/2024] [Indexed: 06/16/2024]
Abstract
Experiencing peer abuse in childhood can damage mental health, but some people exhibit resilience against these negative outcomes. However, it remains uncertain which specific changes in brain structures are associated with this type of resilience. We categorized 217 participants into three groups: resilience group, susceptibility group, and healthy control group, based on their experiences of peer abuse and mental health problems. They underwent MRI scans to measure cortical thickness in various brain regions of the prefrontal cortex. We employed covariance analysis to compare cortical thickness among these groups. Individuals who resilient to anxiety exhibited smaller cortical thickness in the bilateral inferior frontal gyrus (IFG), and with larger thickness in the right medial orbitofrontal cortex (mOFC), while those resilient to stress was associated with smaller thickness in both the bilateral IFG and bilateral middle frontal gyrus (MFG). These findings deepen our understanding of the neural mechanisms underlying resilience and offer insight into improving individual resilience.
Collapse
Affiliation(s)
- Mengchun Yang
- Center for Early Environment and Brain Development, School of Education, Guangzhou University, Guangzhou, China; Department of Psychology, Guangzhou University; Guangzhou, China
| | - Zhengxinyue Wang
- Center for Cognition and Brain Disorders of Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Xinyu Cao
- Center for Cognition and Brain Disorders of Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Jianjun Zhu
- Center for Early Environment and Brain Development, School of Education, Guangzhou University, Guangzhou, China; Department of Psychology, Guangzhou University; Guangzhou, China
| | - Yuanyuan Chen
- Center for Early Environment and Brain Development, School of Education, Guangzhou University, Guangzhou, China; Department of Psychology, Guangzhou University; Guangzhou, China.
| |
Collapse
|
33
|
Cai Q, Meng L, Wang L, Ren J, Yang J, Ming D. Transcriptomic Study of Neural Regulation in the Medial Prefrontal Cortex of Depressive Mice with Low-Intensity Focused Ultrasound. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039959 DOI: 10.1109/embc53108.2024.10782705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Exploring new effective treatments for depression holds important social significance and clinical value. Low-intensity focused ultrasound stimulation (LIFUS) has been proven to have significant neuroprotective effects on depression. However, the specific genes regulated by LIFUS in depressive mice remain unclear. We established a depression mice model using chronic restraint stress (CRS) and applied LIFUS to the medial prefrontal cortex (mPFC) following CRS. We explored the impact of modeling and treatment on gene expression changes through transcriptomics, revealing that the therapeutic effect of LIFUS may be associated with the improvement of the activity of calcium signaling pathway and synaptic plasticity. The study provides preclinical evidence and a theoretical basis for applying LIFUS for depression treatment.
Collapse
|
34
|
Thng G, Shen X, Stolicyn A, Adams MJ, Yeung HW, Batziou V, Conole ELS, Buchanan CR, Lawrie SM, Bastin ME, McIntosh AM, Deary IJ, Tucker-Drob EM, Cox SR, Smith KM, Romaniuk L, Whalley HC. A comprehensive hierarchical comparison of structural connectomes in Major Depressive Disorder cases v. controls in two large population samples. Psychol Med 2024; 54:2515-2526. [PMID: 38497116 DOI: 10.1017/s0033291724000643] [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] [Indexed: 03/19/2024]
Abstract
BACKGROUND The brain can be represented as a network, with nodes as brain regions and edges as region-to-region connections. Nodes with the most connections (hubs) are central to efficient brain function. Current findings on structural differences in Major Depressive Disorder (MDD) identified using network approaches remain inconsistent, potentially due to small sample sizes. It is still uncertain at what level of the connectome hierarchy differences may exist, and whether they are concentrated in hubs, disrupting fundamental brain connectivity. METHODS We utilized two large cohorts, UK Biobank (UKB, N = 5104) and Generation Scotland (GS, N = 725), to investigate MDD case-control differences in brain network properties. Network analysis was done across four hierarchical levels: (1) global, (2) tier (nodes grouped into four tiers based on degree) and rich club (between-hub connections), (3) nodal, and (4) connection. RESULTS In UKB, reductions in network efficiency were observed in MDD cases globally (d = -0.076, pFDR = 0.033), across all tiers (d = -0.069 to -0.079, pFDR = 0.020), and in hubs (d = -0.080 to -0.113, pFDR = 0.013-0.035). No differences in rich club organization and region-to-region connections were identified. The effect sizes and direction for these associations were generally consistent in GS, albeit not significant in our lower-N replication sample. CONCLUSION Our results suggest that the brain's fundamental rich club structure is similar in MDD cases and controls, but subtle topological differences exist across the brain. Consistent with recent large-scale neuroimaging findings, our findings offer a connectomic perspective on a similar scale and support the idea that minimal differences exist between MDD cases and controls.
Collapse
Affiliation(s)
- Gladi Thng
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Xueyi Shen
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Aleks Stolicyn
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Mark J Adams
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Hon Wah Yeung
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Venia Batziou
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Eleanor L S Conole
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
| | - Colin R Buchanan
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence Collaboration (SINAPSE), Edinburgh, UK
| | - Stephen M Lawrie
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Mark E Bastin
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence Collaboration (SINAPSE), Edinburgh, UK
| | - Andrew M McIntosh
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Generation Scotland, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ian J Deary
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Elliot M Tucker-Drob
- Department of Psychology, University of Texas, Austin, TX, USA
- Population Research Center and Center on Aging and Population Sciences, University of Texas, Austin, TX, USA
| | - Simon R Cox
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence Collaboration (SINAPSE), Edinburgh, UK
| | - Keith M Smith
- Department of Computer and Information Sciences, University of Strathclyde, Glasgow, UK
| | - Liana Romaniuk
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Heather C Whalley
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Generation Scotland, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
35
|
Zygouris NC. Differences in Children and Adolescents with Depression before and after a Remediation Program: An Event-Related Potential Study. Brain Sci 2024; 14:660. [PMID: 39061401 PMCID: PMC11275103 DOI: 10.3390/brainsci14070660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/19/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Depression is clinically diagnosed when a defined constellation of symptoms manifests over a specific duration with notable severity. According to the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5), Major Depressive Disorder (MDD) is characterized by the presence of five or more symptoms persisting for at least two weeks. As a profound mental health condition affecting millions globally, depression presents a considerable challenge for researchers and clinicians alike. In pediatric and adolescent populations, depression can precipitate adverse outcomes, including substance abuse, academic difficulties, risky sexual behaviors, physical health problems, impaired social relationships, and a markedly elevated risk of suicide-up to thirty times higher than the general population. This paper details a study that evaluated the efficacy of Cognitive Behavioral Therapy (CBT) alone vs. CBT combined with selective serotonin reuptake inhibitors (SSRIs) in a treatment program. The study cohort comprised sixteen (16) children and adolescents diagnosed with depression (eight males and eight females) and sixteen (16) typically developing peers (eight males and eight females) aged from 9 to 15 years (Mean age = 11.94, standard deviation = 2.02). Initial assessments employed Event-Related Potentials (ERPs), the Children's Depression Inventory (CDI), and reaction time measurements. The results reveal that participants with depression exhibit cognitive deficits in attention and memory, as evidenced by prolonged P300 latencies. Following intervention with either CBT alone or CBT combined with medication, the depressed participants demonstrated significant improvements, evidenced by lower CDI scores, reduced P300 latencies, and faster reaction times, both compared to their pre-treatment status and relative to the control group.
Collapse
Affiliation(s)
- Nikolaos C Zygouris
- Digital Neuropsychological Assessment Laboratory, Department of Informatics and Telecommunications, University of Thessaly, 35100 Lamia, Greece
| |
Collapse
|
36
|
Zhukovsky P, Tio ES, Coughlan G, Bennett DA, Wang Y, Hohman TJ, Pizzagalli DA, Mulsant BH, Voineskos AN, Felsky D. Genetic influences on brain and cognitive health and their interactions with cardiovascular conditions and depression. Nat Commun 2024; 15:5207. [PMID: 38890310 PMCID: PMC11189393 DOI: 10.1038/s41467-024-49430-7] [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: 05/08/2023] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Approximately 40% of dementia cases could be prevented or delayed by modifiable risk factors related to lifestyle and environment. These risk factors, such as depression and vascular disease, do not affect all individuals in the same way, likely due to inter-individual differences in genetics. However, the precise nature of how genetic risk profiles interact with modifiable risk factors to affect brain health is poorly understood. Here we combine multiple data resources, including genotyping and postmortem gene expression, to map the genetic landscape of brain structure and identify 367 loci associated with cortical thickness and 13 loci associated with white matter hyperintensities (P < 5×10-8), with several loci also showing a significant association with cognitive function. We show that among 220 unique genetic loci associated with cortical thickness in our genome-wide association studies (GWAS), 95 also showed evidence of interaction with depression or cardiovascular conditions. Polygenic risk scores based on our GWAS of inferior frontal thickness also interacted with hypertension in predicting executive function in the Canadian Longitudinal Study on Aging. These findings advance our understanding of the genetic underpinning of brain structure and show that genetic risk for brain and cognitive health is in part moderated by treatable mid-life factors.
Collapse
Grants
- P30 AG072975 NIA NIH HHS
- U01 AG046152 NIA NIH HHS
- U01 AG061356 NIA NIH HHS
- R01 AG017917 NIA NIH HHS
- P30 AG010161 NIA NIH HHS
- R01 AG059716 NIA NIH HHS
- Wellcome Trust
- R01 AG015819 NIA NIH HHS
- Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- D.F. is supported by the generous contributions from the Michael and Sonja Koerner Foundation and the Krembil Family Foundation. D.F. is also supported in part by the Centre for Addiction and Mental Health (CAMH) Discovery Fund and CIHR.
- PZ was funded by the Canadian Institute of Health Research Postdoctoral Fellowship.
- Over the past 3 years, D.A.P has received consulting fees from Albright Stonebridge Group, Boehringer Ingelheim, Compass Pathways, Engrail Therapeutics, Neumora Therapeutics (formerly BlackThorn Therapeutics), Neurocrine Biosciences, Neuroscience Software, Otsuka, Sunovion, and Takeda; he has received honoraria from the Psychonomic Society and American Psychological Association (for editorial work) and from Alkermes; he has received research funding from the Brain and Behavior Research Foundation, the Dana Foundation, Millennium Pharmaceuticals, Wellcome Leap MCPsych, and NIMH; he has received stock options from Compass Pathways, Engrail Therapeutics, Neumora Therapeutics, and Neuroscience Software. No funding from these entities was used to support the current work, and all views expressed are solely those of the authors.
- U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)
- A.N.V. currently receives funding from CIHR, the NIH, the National Sciences and Engineering Research Council (NSERC), the CAMH Foundation, and the University of Toronto. E.S.T. was funded by the Ontario Graduate Scholarship.
Collapse
Affiliation(s)
- Peter Zhukovsky
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Earvin S Tio
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Gillian Coughlan
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - David A Bennett
- Department of Neurological Sciences, RUSH Medical College, Chicago, IL, 60612, USA
| | - Yanling Wang
- Department of Neurological Sciences, RUSH Medical College, Chicago, IL, 60612, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Diego A Pizzagalli
- Department of Psychiatry, Harvard Medical School and Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, MA, 02478, USA
| | - Benoit H Mulsant
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada
| | - Aristotle N Voineskos
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada.
| | - Daniel Felsky
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada.
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Rotman Research Institute, Baycrest Hospital, Toronto, ON, M6A 2E1, Canada.
| |
Collapse
|
37
|
Xiao H, Tang D, Zheng C, Yang Z, Zhao W, Guo S. Atypical dynamic network reconfiguration and genetic mechanisms in patients with major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110957. [PMID: 38365102 DOI: 10.1016/j.pnpbp.2024.110957] [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: 08/06/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Brain dynamics underlie complex forms of flexible cognition or the ability to shift between different mental modes. However, the precise dynamic reconfiguration based on multi-layer network analysis and the genetic mechanisms of major depressive disorder (MDD) remains unclear. METHODS Resting-state functional magnetic resonance imaging (fMRI) data were acquired from the REST-meta-MDD consortium, including 555 patients with MDD and 536 healthy controls (HC). A time-varying multi-layer network was constructed, and dynamic modular characteristics were used to investigate the network reconfiguration. Additionally, partial least squares regression analysis was performed using transcriptional data provided by the Allen Human Brain Atlas (AHBA) to identify genes associated with atypical dynamic network reconfiguration in MDD. RESULTS In comparison to HC, patients with MDD exhibited lower global and local recruitment coefficients. The local reduction was particularly evident in the salience and subcortical networks. Spatial transcriptome correlation analysis revealed an association between gene expression profiles and atypical dynamic network reconfiguration observed in MDD. Further functional enrichment analyses indicated that positively weighted reconfiguration-related genes were primarily associated with metabolic and biosynthetic pathways. Additionally, negatively enriched genes were predominantly related to programmed cell death-related terms. CONCLUSIONS Our findings offer robust evidence of the atypical dynamic reconfiguration in patients with MDD from a novel perspective. These results offer valuable insights for further exploration into the mechanisms underlying MDD.
Collapse
Affiliation(s)
- Hairong Xiao
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, China
| | - Dier Tang
- School of Mathematics, Jilin University, Changchun 130015, China
| | - Chuchu Zheng
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, China
| | - Zeyu Yang
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, China
| | - Wei Zhao
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, China; Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, China
| | - Shuixia Guo
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, China; Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, China.
| |
Collapse
|
38
|
Zhang S, Larsen B, Sydnor VJ, Zeng T, An L, Yan X, Kong R, Kong X, Gur RC, Gur RE, Moore TM, Wolf DH, Holmes AJ, Xie Y, Zhou JH, Fortier MV, Tan AP, Gluckman P, Chong YS, Meaney MJ, Deco G, Satterthwaite TD, Yeo BTT. In vivo whole-cortex marker of excitation-inhibition ratio indexes cortical maturation and cognitive ability in youth. Proc Natl Acad Sci U S A 2024; 121:e2318641121. [PMID: 38814872 PMCID: PMC11161789 DOI: 10.1073/pnas.2318641121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 04/04/2024] [Indexed: 06/01/2024] Open
Abstract
A balanced excitation-inhibition ratio (E/I ratio) is critical for healthy brain function. Normative development of cortex-wide E/I ratio remains unknown. Here, we noninvasively estimate a putative marker of whole-cortex E/I ratio by fitting a large-scale biophysically plausible circuit model to resting-state functional MRI (fMRI) data. We first confirm that our model generates realistic brain dynamics in the Human Connectome Project. Next, we show that the estimated E/I ratio marker is sensitive to the gamma-aminobutyric acid (GABA) agonist benzodiazepine alprazolam during fMRI. Alprazolam-induced E/I changes are spatially consistent with positron emission tomography measurement of benzodiazepine receptor density. We then investigate the relationship between the E/I ratio marker and neurodevelopment. We find that the E/I ratio marker declines heterogeneously across the cerebral cortex during youth, with the greatest reduction occurring in sensorimotor systems relative to association systems. Importantly, among children with the same chronological age, a lower E/I ratio marker (especially in the association cortex) is linked to better cognitive performance. This result is replicated across North American (8.2 to 23.0 y old) and Asian (7.2 to 7.9 y old) cohorts, suggesting that a more mature E/I ratio indexes improved cognition during normative development. Overall, our findings open the door to studying how disrupted E/I trajectories may lead to cognitive dysfunction in psychopathology that emerges during youth.
Collapse
Affiliation(s)
- Shaoshi Zhang
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore119077, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
- Department of Pediatrics, University of Minnesota, Minneapolis, MN55455
| | - Valerie J. Sydnor
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
| | - Tianchu Zeng
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Lijun An
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Xiaoxuan Yan
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore119077, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Ru Kong
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Xiaolu Kong
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
- ByteDance, Singapore048583, Singapore
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA19104
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA19104
| | - Tyler M. Moore
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
| | - Daniel H. Wolf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
| | - Avram J. Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ07103
- Wu Tsai Institute, Yale University, New Haven, CT06520
| | - Yapei Xie
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore119077, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Marielle V. Fortier
- Department of Diagnostic and Interventional Imaging, Kandang Kerbau Women’s and Children’s Hospital, Singapore229899, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore138632, Singapore
| | - Ai Peng Tan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore138632, Singapore
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore119074, Singapore
| | - Peter Gluckman
- Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland1142, New Zealand
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore138632, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore119228, Singapore
| | - Michael J. Meaney
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore138632, Singapore
- Department of Neurology and Neurosurgery, McGill University, Montreal, QCH3A1A1, Canada
| | - Gustavo Deco
- Center for Brain and Cognition, Department of Technology and Information, Universitat Pompeu Fabra, Barcelona08002, Spain
- Institució Catalana de la Recerca i Estudis Avançats, Universitat Barcelona, Barcelona08010, Spain
| | - Theodore D. Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
| | - B. T. Thomas Yeo
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore119077, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
- Martinos Center for Biomedical Imaging, Massachusetts General Hopstial, Charlestown, MA02129
| |
Collapse
|
39
|
Dear R, Wagstyl K, Seidlitz J, Markello RD, Arnatkevičiūtė A, Anderson KM, Bethlehem RAI, Raznahan A, Bullmore ET, Vértes PE. Cortical gene expression architecture links healthy neurodevelopment to the imaging, transcriptomics and genetics of autism and schizophrenia. Nat Neurosci 2024; 27:1075-1086. [PMID: 38649755 PMCID: PMC11156586 DOI: 10.1038/s41593-024-01624-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 03/18/2024] [Indexed: 04/25/2024]
Abstract
Human brain organization involves the coordinated expression of thousands of genes. For example, the first principal component (C1) of cortical transcription identifies a hierarchy from sensorimotor to association regions. In this study, optimized processing of the Allen Human Brain Atlas revealed two new components of cortical gene expression architecture, C2 and C3, which are distinctively enriched for neuronal, metabolic and immune processes, specific cell types and cytoarchitectonics, and genetic variants associated with intelligence. Using additional datasets (PsychENCODE, Allen Cell Atlas and BrainSpan), we found that C1-C3 represent generalizable transcriptional programs that are coordinated within cells and differentially phased during fetal and postnatal development. Autism spectrum disorder and schizophrenia were specifically associated with C1/C2 and C3, respectively, across neuroimaging, differential expression and genome-wide association studies. Evidence converged especially in support of C3 as a normative transcriptional program for adolescent brain development, which can lead to atypical supragranular cortical connectivity in people at high genetic risk for schizophrenia.
Collapse
Affiliation(s)
- Richard Dear
- Department of Psychiatry, University of Cambridge, Cambridge, UK.
| | | | - Jakob Seidlitz
- Lifespan Brain Institute, Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Ross D Markello
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Aurina Arnatkevičiūtė
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | | | | | - Armin Raznahan
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| | | | - Petra E Vértes
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
40
|
Xie K, Royer J, Larivière S, Rodriguez-Cruces R, Frässle S, Cabalo DG, Ngo A, DeKraker J, Auer H, Tavakol S, Weng Y, Abdallah C, Arafat T, Horwood L, Frauscher B, Caciagli L, Bernasconi A, Bernasconi N, Zhang Z, Concha L, Bernhardt BC. Atypical connectome topography and signal flow in temporal lobe epilepsy. Prog Neurobiol 2024; 236:102604. [PMID: 38604584 DOI: 10.1016/j.pneurobio.2024.102604] [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: 06/26/2023] [Revised: 12/18/2023] [Accepted: 04/07/2024] [Indexed: 04/13/2024]
Abstract
Temporal lobe epilepsy (TLE) is the most common pharmaco-resistant epilepsy in adults. While primarily associated with mesiotemporal pathology, recent evidence suggests that brain alterations in TLE extend beyond the paralimbic epicenter and impact macroscale function and cognitive functions, particularly memory. Using connectome-wide manifold learning and generative models of effective connectivity, we examined functional topography and directional signal flow patterns between large-scale neural circuits in TLE at rest. Studying a multisite cohort of 95 patients with TLE and 95 healthy controls, we observed atypical functional topographies in the former group, characterized by reduced differentiation between sensory and transmodal association cortices, with most marked effects in bilateral temporo-limbic and ventromedial prefrontal cortices. These findings were consistent across all study sites, present in left and right lateralized patients, and validated in a subgroup of patients with histopathological validation of mesiotemporal sclerosis and post-surgical seizure freedom. Moreover, they were replicated in an independent cohort of 30 TLE patients and 40 healthy controls. Further analyses demonstrated that reduced differentiation related to decreased functional signal flow into and out of temporolimbic cortical systems and other brain networks. Parallel analyses of structural and diffusion-weighted MRI data revealed that topographic alterations were independent of TLE-related cortical thinning but partially mediated by white matter microstructural changes that radiated away from paralimbic circuits. Finally, we found a strong association between the degree of functional alterations and behavioral markers of memory dysfunction. Our work illustrates the complex landscape of macroscale functional imbalances in TLE, which can serve as intermediate markers bridging microstructural changes and cognitive impairment.
Collapse
Affiliation(s)
- Ke Xie
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Jessica Royer
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada; Analytical Neurophysiology Laboratory, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Sara Larivière
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Raul Rodriguez-Cruces
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Stefan Frässle
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Donna Gift Cabalo
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Alexander Ngo
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Jordan DeKraker
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Hans Auer
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Shahin Tavakol
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Yifei Weng
- Department of Medical Imaging, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Chifaou Abdallah
- Analytical Neurophysiology Laboratory, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Thaera Arafat
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Linda Horwood
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada; Analytical Neurophysiology Laboratory, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Birgit Frauscher
- Analytical Neurophysiology Laboratory, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada; Department of Neurology, Duke University School of Medicine and Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC 27705, USA
| | - Lorenzo Caciagli
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurology, Inselspital, Sleep-Wake-Epilepsy-Center, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3 BG, United Kingdom
| | - Andrea Bernasconi
- Neuroimaging of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Neda Bernasconi
- Neuroimaging of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Zhiqiang Zhang
- Department of Medical Imaging, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Luis Concha
- Institute of Neurobiology, Universidad Nacional Autónoma de Mexico (UNAM), Queretaro, Mexico
| | - Boris C Bernhardt
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada.
| |
Collapse
|
41
|
Chen K, Yang J, Li F, Chen J, Chen M, Shao H, He C, Cai D, Zhang X, Wang L, Luo Y, Cheng B, Wang J. Molecular basis underlying default mode network functional abnormalities in postpartum depression with and without anxiety. Hum Brain Mapp 2024; 45:e26657. [PMID: 38544486 PMCID: PMC10973776 DOI: 10.1002/hbm.26657] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/04/2024] [Accepted: 02/27/2024] [Indexed: 11/12/2024] Open
Abstract
Although Postpartum depression (PPD) and PPD with anxiety (PPD-A) have been well characterized as functional disruptions within or between multiple brain systems, however, how to quantitatively delineate brain functional system irregularity and the molecular basis of functional abnormalities in PPD and PPD-A remains unclear. Here, brain sample entropy (SampEn), resting-state functional connectivity (RSFC), transcriptomic and neurotransmitter density data were used to investigate brain functional system irregularity, functional connectivity abnormalities and associated molecular basis for PPD and PPD-A. PPD-A exhibited higher SampEn in medial prefrontal cortex (MPFC) and posterior cingulate cortex (PPC) than healthy postnatal women (HPW) and PPD while PPD showed lower SampEn in PPC compared to HPW and PPD-A. The functional connectivity analysis with MPFC and PPC as seed areas revealed decreased functional couplings between PCC and paracentral lobule and between MPFC and angular gyrus in PPD compared to both PPD-A and HPW. Moreover, abnormal SampEn and functional connectivity were associated with estrogenic level and clinical symptoms load. Importantly, spatial association analyses between functional changes and transcriptome and neurotransmitter density maps revealed that these functional changes were primarily associated with synaptic signaling, neuron projection, neurotransmitter level regulation, amino acid metabolism, cyclic adenosine monophosphate (cAMP) signaling pathways, and neurotransmitters of 5-hydroxytryptamine (5-HT), norepinephrine, glutamate, dopamine and so on. These results reveal abnormal brain entropy and functional connectivities primarily in default mode network (DMN) and link these changes to transcriptome and neurotransmitters to establish the molecular basis for PPD and PPD-A for the first time. Our findings highlight the important role of DMN in neuropathology of PPD and PPD-A.
Collapse
Affiliation(s)
- Kexuan Chen
- Faculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
- Medical SchoolKunming University of Science and TechnologyKunmingChina
| | - Jia Yang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Fang Li
- Medical SchoolKunming University of Science and TechnologyKunmingChina
| | - Jin Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Meiling Chen
- Department of Clinical Psychology, the First People's Hospital of Yunnan ProvinceThe Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Heng Shao
- Department of Geriatrics, the First People's Hospital of Yunnan ProvinceThe Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Chongjun He
- People's Hospital of Lijiangthe Affiliated Hospital of Kunming University of Science and TechnologyLijiangChina
| | - Defang Cai
- The Second People's Hospital of Yuxithe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Xing Zhang
- The Second People's Hospital of Yuxithe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Libo Wang
- The Second People's Hospital of Yuxithe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Yuejia Luo
- Medical SchoolKunming University of Science and TechnologyKunmingChina
- Center for Brain Disorders and Cognitive Sciences, School of PsychologyShenzhen UniversityShenzhenChina
- The State Key Lab of Cognitive and Learning, Faculty of PsychologyBeijing Normal UniversityBeijingChina
| | - Bochao Cheng
- Department of RadiologyWest China Second University Hospital of Sichuan UniversityChengduChina
| | - Jiaojian Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| |
Collapse
|
42
|
Zhang S, Larsen B, Sydnor VJ, Zeng T, An L, Yan X, Kong R, Kong X, Gur RC, Gur RE, Moore TM, Wolf DH, Holmes AJ, Xie Y, Zhou JH, Fortier MV, Tan AP, Gluckman P, Chong YS, Meaney MJ, Deco G, Satterthwaite TD, Yeo BT. In-vivo whole-cortex marker of excitation-inhibition ratio indexes cortical maturation and cognitive ability in youth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.22.546023. [PMID: 38586012 PMCID: PMC10996460 DOI: 10.1101/2023.06.22.546023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
A balanced excitation-inhibition ratio (E/I ratio) is critical for healthy brain function. Normative development of cortex-wide E/I ratio remains unknown. Here we non-invasively estimate a putative marker of whole-cortex E/I ratio by fitting a large-scale biophysically-plausible circuit model to resting-state functional MRI (fMRI) data. We first confirm that our model generates realistic brain dynamics in the Human Connectome Project. Next, we show that the estimated E/I ratio marker is sensitive to the GABA-agonist benzodiazepine alprazolam during fMRI. Alprazolam-induced E/I changes are spatially consistent with positron emission tomography measurement of benzodiazepine receptor density. We then investigate the relationship between the E/I ratio marker and neurodevelopment. We find that the E/I ratio marker declines heterogeneously across the cerebral cortex during youth, with the greatest reduction occurring in sensorimotor systems relative to association systems. Importantly, among children with the same chronological age, a lower E/I ratio marker (especially in association cortex) is linked to better cognitive performance. This result is replicated across North American (8.2 to 23.0 years old) and Asian (7.2 to 7.9 years old) cohorts, suggesting that a more mature E/I ratio indexes improved cognition during normative development. Overall, our findings open the door to studying how disrupted E/I trajectories may lead to cognitive dysfunction in psychopathology that emerges during youth.
Collapse
Affiliation(s)
- Shaoshi Zhang
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Valerie J. Sydnor
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tianchu Zeng
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Lijun An
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Xiaoxuan Yan
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Ru Kong
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Xiaolu Kong
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
- ByteDance, Singapore
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tyler M. Moore
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel H. Wolf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Avram J Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, United States
- Wu Tsai Institute, Yale University, New Haven, CT, United States
| | - Yapei Xie
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Marielle V Fortier
- Department of Diagnostic and Interventional Imaging, KK Women’s and Children’s Hospital, Singapore
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ai Peng Tan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Peter Gluckman
- UK Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Michael J Meaney
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Gustavo Deco
- Center for Brain and Cognition, Department of Technology and Information, Universitat Pompeu Fabra, Barcelona, Spain
- Institució Catalana de la Recerca i Estudis Avançats, Universitat Barcelona, Barcelona, Spain
| | - Theodore D. Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - B.T. Thomas Yeo
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
- Martinos Center for Biomedical Imaging, Massachusetts General Hopstial, Charlestown, MA, USA
| |
Collapse
|
43
|
Zhao L, Liu J, Zhao W, Chen J, Fan J, Ge T, Tu Y. Morphological and genetic decoding shows heterogeneous patterns of brain aging in chronic musculoskeletal pain. NATURE MENTAL HEALTH 2024; 2:435-449. [DOI: 10.1038/s44220-024-00223-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 02/29/2024] [Indexed: 04/02/2025]
|
44
|
Yu X, Chen K, Ma Y, Bai T, Zhu S, Cai D, Zhang X, Wang K, Tian Y, Wang J. Molecular basis underlying changes of brain entropy and functional connectivity in major depressive disorders after electroconvulsive therapy. CNS Neurosci Ther 2024; 30:e14690. [PMID: 38529527 PMCID: PMC10964037 DOI: 10.1111/cns.14690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/03/2024] [Accepted: 02/23/2024] [Indexed: 03/27/2024] Open
Abstract
INTRODUCTION Electroconvulsive therapy (ECT) is widely used for treatment-resistant depression. However, it is unclear whether/how ECT can be targeted to affect brain regions and circuits in the brain to dynamically regulate mood and cognition. METHODS This study used brain entropy (BEN) to measure the irregular levels of brain systems in 46 major depressive disorder (MDD) patients before and after ECT treatment. Functional connectivity (FC) was further adopted to reveal changes of functional couplings. Moreover, transcriptomic and neurotransmitter receptor data were used to reveal genetic and molecular basis of the changes of BEN and functional connectivities. RESULTS Compared to pretreatment, the BEN in the posterior cerebellar lobe (PCL) significantly decreased and FC between the PCL and the right temporal pole (TP) significantly increased in MDD patients after treatment. Moreover, we found that these changes of BEN and FC were closely associated with genes' expression profiles involved in MAPK signaling pathway, GABAergic synapse, and dopaminergic synapse and were significantly correlated with the receptor/transporter density of 5-HT, norepinephrine, glutamate, etc. CONCLUSION: These findings suggest that loops in the cerebellum and TP are crucial for ECT regulation of mood and cognition, which provides new evidence for the antidepressant effects of ECT and the potential molecular mechanism leading to cognitive impairment.
Collapse
Affiliation(s)
- Xiaohui Yu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Kexuan Chen
- Medical SchoolKunming University of Science and TechnologyKunmingChina
| | - Yingzi Ma
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Tongjian Bai
- Department of NeurologyThe First Hospital of Anhui Medical UniversityHefeiChina
| | - Shunli Zhu
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Defang Cai
- The Second People's Hospital of YuxiThe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Xing Zhang
- The Second People's Hospital of YuxiThe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Kai Wang
- Department of NeurologyThe First Hospital of Anhui Medical UniversityHefeiChina
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric DisordersHefeiChina
- School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental HealthHefeiChina
- Anhui Province Clinical Research Center for Neurological DiseaseHefeiChina
| | - Yanghua Tian
- Department of NeurologyThe First Hospital of Anhui Medical UniversityHefeiChina
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric DisordersHefeiChina
- School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental HealthHefeiChina
- Anhui Province Clinical Research Center for Neurological DiseaseHefeiChina
- Institute of Artificial IntelligenceHefei Comprehensive National Science CenterHefeiChina
| | - Jiaojian Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| |
Collapse
|
45
|
Li J, Long Z, Sheng W, Du L, Qiu J, Chen H, Liao W. Transcriptomic Similarity Informs Neuromorphic Deviations in Depression Biotypes. Biol Psychiatry 2024; 95:414-425. [PMID: 37573006 DOI: 10.1016/j.biopsych.2023.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) is complicated by population heterogeneity, motivating the investigation of biotypes through imaging-derived phenotypes. However, neuromorphic heterogeneity in MDD remains unclear, and how the correlated gene expression (CGE) connectome constrains these neuromorphic anomalies in MDD biotypes has not yet been studied. METHODS Here, we related cortical thickness deviations in MDD biotypes to a pattern of CGE connectome. Cortical thickness was estimated from 3-dimensional T1-weighted magnetic resonance images in 2 independent cohorts (discovery cohort: N = 425; replication cohort: N = 217). The transcriptional activity was measured according to Allen Human Brain Atlas. A density peak-based clustering algorithm was used to identify MDD biotypes. RESULTS We found that patients with MDD were clustered into 2 replicated biotypes based on single-patient regional deviations from healthy control participants across 2 datasets. Biotype 1 mainly exhibited cortical thinning across the brain, whereas biotype 2 mainly showed cortical thickening in the brain. Using brainwide gene expression data, we found that deviations of transcriptionally connected neighbors predicted regional deviation for both biotypes. Furthermore, putative CGE-informed epicenters of biotype 1 were concentrated on the cognitive control circuit, whereas biotype 2 epicenters were located in the social perception circuit. The patterns of epicenter likelihood were separately associated with depression- and anxiety-response maps, suggesting that epicenters of MDD biotypes may be associated with clinical efficacies. CONCLUSIONS Our findings linked the CGE connectome and neuromorphic deviations to identify distinct epicenters in MDD biotypes, providing insight into how microscale gene expressions informed MDD biotypes.
Collapse
Affiliation(s)
- Jiao Li
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory 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
| | - Zhiliang Long
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Southwest University, Chongqing, P.R. China
| | - Wei Sheng
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory 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, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Jiang Qiu
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Southwest University, Chongqing, P.R. China
| | - Huafu Chen
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory 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
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory 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.
| |
Collapse
|
46
|
Moodie JE, Harris SE, Harris MA, Buchanan CR, Davies G, Taylor A, Redmond P, Liewald DCM, Valdés Hernández MDC, Shenkin S, Russ TC, Muñoz Maniega S, Luciano M, Corley J, Stolicyn A, Shen X, Steele D, Waiter G, Sandu A, Bastin ME, Wardlaw JM, McIntosh A, Whalley H, Tucker‐Drob EM, Deary IJ, Cox SR. General and specific patterns of cortical gene expression as spatial correlates of complex cognitive functioning. Hum Brain Mapp 2024; 45:e26641. [PMID: 38488470 PMCID: PMC10941541 DOI: 10.1002/hbm.26641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/29/2024] [Accepted: 02/18/2024] [Indexed: 03/18/2024] Open
Abstract
Gene expression varies across the brain. This spatial patterning denotes specialised support for particular brain functions. However, the way that a given gene's expression fluctuates across the brain may be governed by general rules. Quantifying patterns of spatial covariation across genes would offer insights into the molecular characteristics of brain areas supporting, for example, complex cognitive functions. Here, we use principal component analysis to separate general and unique gene regulatory associations with cortical substrates of cognition. We find that the region-to-region variation in cortical expression profiles of 8235 genes covaries across two major principal components: gene ontology analysis suggests these dimensions are characterised by downregulation and upregulation of cell-signalling/modification and transcription factors. We validate these patterns out-of-sample and across different data processing choices. Brain regions more strongly implicated in general cognitive functioning (g; 3 cohorts, total meta-analytic N = 39,519) tend to be more balanced between downregulation and upregulation of both major components (indicated by regional component scores). We then identify a further 29 genes as candidate cortical spatial correlates of g, beyond the patterning of the two major components (|β| range = 0.18 to 0.53). Many of these genes have been previously associated with clinical neurodegenerative and psychiatric disorders, or with other health-related phenotypes. The results provide insights into the cortical organisation of gene expression and its association with individual differences in cognitive functioning.
Collapse
Affiliation(s)
- Joanna E. Moodie
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
| | - Sarah E. Harris
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
| | - Mathew A. Harris
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
| | - Colin R. Buchanan
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
| | - Gail Davies
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
| | - Adele Taylor
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
| | - Paul Redmond
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
| | - David C. M. Liewald
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
| | - Maria del C. Valdés Hernández
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
| | - Susan Shenkin
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
- Ageing and Health Research Group, Usher InstituteUniversity of EdinburghUK
| | - Tom C. Russ
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
- Alzheimer Scotland Dementia Research CentreUniversity of EdinburghUK
| | - Susana Muñoz Maniega
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
| | - Michelle Luciano
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
| | - Janie Corley
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
| | - Aleks Stolicyn
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
| | - Xueyi Shen
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
| | - Douglas Steele
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
| | - Gordon Waiter
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
| | - Anca‐Larisa Sandu
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
| | - Mark E. Bastin
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
| | - Joanna M. Wardlaw
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
| | | | | | | | - Ian J. Deary
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
| | - Simon R. Cox
- Lothian Birth Cohorts, Department of PsychologyThe University of EdinburghEdinburghUK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationEdinburghUK
| |
Collapse
|
47
|
Wang Y, Wang Y, Wang H, Ma L, Eickhoff SB, Madsen KH, Chu C, Fan L. Spatio-molecular profiles shape the human cerebellar hierarchy along the sensorimotor-association axis. Cell Rep 2024; 43:113770. [PMID: 38363683 DOI: 10.1016/j.celrep.2024.113770] [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: 09/26/2023] [Revised: 12/27/2023] [Accepted: 01/25/2024] [Indexed: 02/18/2024] Open
Abstract
Cerebellar involvement in both motor and non-motor functions manifests in specific regions of the human cerebellum, revealing the functional heterogeneity within it. One compelling theory places the heterogeneity within the cerebellar functional hierarchy along the sensorimotor-association (SA) axis. Despite extensive neuroimaging studies, evidence for the cerebellar SA axis from different modalities and scales was lacking. Thus, we establish a significant link between the cerebellar SA axis and spatio-molecular profiles. Utilizing the gene set variation analysis, we find the intermediate biological principles the significant genes leveraged to scaffold the cerebellar SA axis. Interestingly, we find these spatio-molecular profiles notably associated with neuropsychiatric dysfunction and recent evolution. Furthermore, cerebello-cerebral interactions at genetic and functional connectivity levels mirror the cerebral cortex and cerebellum's SA axis. These findings can provide a deeper understanding of how the human cerebellar SA axis is shaped and its role in transitioning from sensorimotor to association functions.
Collapse
Affiliation(s)
- Yaping Wang
- Sino-Danish Center, University of Chinese Academy of Sciences, Beijing 100190, China; Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yufan Wang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Haiyan Wang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Liang Ma
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour), Research Centre Jülich, 52425 Jülich, Germany; Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Kristoffer Hougaard Madsen
- Sino-Danish Center, University of Chinese Academy of Sciences, Beijing 100190, China; Department of Applied Mathematics and Computer Science, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital-Amager and Hvidovre, 2650 Hvidovre, Denmark
| | - Congying Chu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.
| | - Lingzhong Fan
- Sino-Danish Center, University of Chinese Academy of Sciences, Beijing 100190, China; Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266000, China.
| |
Collapse
|
48
|
Yu M, Risacher SL, Nho KT, Wen Q, Oblak AL, Unverzagt FW, Apostolova LG, Farlow MR, Brosch JR, Clark DG, Wang S, Deardorff R, Wu YC, Gao S, Sporns O, Saykin AJ. Spatial transcriptomic patterns underlying amyloid-β and tau pathology are associated with cognitive dysfunction in Alzheimer's disease. Cell Rep 2024; 43:113691. [PMID: 38244198 PMCID: PMC10926093 DOI: 10.1016/j.celrep.2024.113691] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/29/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024] Open
Abstract
Amyloid-β (Aβ) and tau proteins accumulate within distinct neuronal systems in Alzheimer's disease (AD). Although it is not clear why certain brain regions are more vulnerable to Aβ and tau pathologies than others, gene expression may play a role. We study the association between brain-wide gene expression profiles and regional vulnerability to Aβ (gene-to-Aβ associations) and tau (gene-to-tau associations) pathologies by leveraging two large independent AD cohorts. We identify AD susceptibility genes and gene modules in a gene co-expression network with expression profiles specifically related to regional vulnerability to Aβ and tau pathologies in AD. In addition, we identify distinct biochemical pathways associated with the gene-to-Aβ and the gene-to-tau associations. These findings may explain the discordance between regional Aβ and tau pathologies. Finally, we propose an analytic framework, linking the identified gene-to-pathology associations to cognitive dysfunction in AD at the individual level, suggesting potential clinical implication of the gene-to-pathology associations.
Collapse
Affiliation(s)
- Meichen Yu
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Network Science Institute, Bloomington, IN, USA.
| | - Shannon L Risacher
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik T Nho
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Network Science Institute, Bloomington, IN, USA
| | - Qiuting Wen
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adrian L Oblak
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Frederick W Unverzagt
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liana G Apostolova
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Martin R Farlow
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jared R Brosch
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David G Clark
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sophia Wang
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rachael Deardorff
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yu-Chien Wu
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Olaf Sporns
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Network Science Institute, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Network Science Institute, Bloomington, IN, USA.
| |
Collapse
|
49
|
Zheng J, Womer FY, Tang L, Guo H, Zhang X, Tang Y, Wang F. Integrative omics analysis reveals epigenomic and transcriptomic signatures underlying brain structural deficits in major depressive disorder. Transl Psychiatry 2024; 14:17. [PMID: 38195555 PMCID: PMC10776753 DOI: 10.1038/s41398-023-02724-8] [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: 10/09/2023] [Revised: 12/11/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024] Open
Abstract
Several lines of evidence support the involvement of transcriptomic and epigenetic mechanisms in the brain structural deficits of major depressive disorder (MDD) separately. However, research in these two areas has remained isolated. In this study, we proposed an integrative strategy that combined neuroimaging, brain-wide gene expression, and peripheral DNA methylation data to investigate the genetic basis of gray matter abnormalities in MDD. The MRI T1-weighted images and Illumina 850 K DNA methylation microarrays were obtained from 269 patients and 416 healthy controls, and brain-wide transcriptomic data were collected from Allen Human Brain Atlas. The between-group differences in gray matter volume (GMV) and differentially methylated CpG positions (DMPs) were examined. The genes with their expression patterns spatially related to GMV changes and genes with DMPs were overlapped and selected. Using principal component regression, the associations between DMPs in overlapped genes and GMV across individual patients were investigated, and the region-specific correlations between methylation status and gene expression were examined. We found significant associations between the decreased GMV and DMPs methylation status in the anterior cingulate cortex, inferior frontal cortex, and fusiform face cortex regions. These DMPs genes were primarily enriched in the neurodevelopmental and synaptic transmission process. There was a significant negative correlation between DNA methylation and gene expression in genes associated with GMV changes of the frontal cortex in MDD. Our findings suggest that GMV abnormalities in MDD may have a transcriptomic and epigenetic basis. This imaging-transcriptomic-epigenetic integrative analysis provides spatial and biological links between cortical morphological deficits and peripheral epigenetic signatures in MDD.
Collapse
Affiliation(s)
- 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
| | - Fay Y Womer
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lili Tang
- 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
| | - Huiling Guo
- 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
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Xizhe Zhang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Yanqing Tang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China.
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, China.
- Department of Gerontology, The First Hospital of China Medical University, Shenyang, China.
- Shengjing Hospital of China Medical University, Shenyang, China.
| | - Fei Wang
- 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
|
50
|
Xiao Y, Zhao L, Zang X, Xue S. Compressed primary-to-transmodal gradient is accompanied with subcortical alterations and linked to neurotransmitters and cellular signatures in major depressive disorder. Hum Brain Mapp 2023; 44:5919-5935. [PMID: 37688552 PMCID: PMC10619397 DOI: 10.1002/hbm.26485] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/18/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
Major depressive disorder (MDD) has been shown to involve widespread changes in low-level sensorimotor and higher-level cognitive functions. Recent research found that a primary-to-transmodal gradient could capture a cortical hierarchical organization ranging from perception and action to cognition in healthy subjects, but a prominent gradient dysfunction in MDD patients. However, whether and how this cortical gradient is linked to subcortical impairments and whether it is reflected in the microscale neurotransmitter systems and cell type-specific transcriptional signatures remain largely unknown. Data were acquired from 323 MDD patients and 328 sex- and age-matched healthy controls derived from the REST-meta-MDD project, and the human brain neurotransmitter systems density maps and gene expression data were drawn from two publicly available datasets. We investigated alterations of the primary-to-transmodal gradient in MDD patients and their correlations with clinical symptoms of depression and anxiety, as well as their paralleled subcortical impairments. The correlations between MDD-related gradient alterations and densities of the neurotransmitter systems and gene expression information were assessed, respectively. The results demonstrated that MDD patients had a compressed primary-to-transmodal gradient accompanied by paralleled alterations in subcortical regions including the caudate, amygdala, and thalamus. The case-control gradient differences were spatially correlated with the densities of the neurotransmitter systems including the serotonin and dopamine receptors, and meanwhile with gene expression enriched in astrocytes, excitatory and inhibitory neuronal cells. These findings mapped the paralleled subcortical impairments in cortical hierarchical organization and also helped us understand the possible molecular and cellular substrates of the co-occurrence of high-level cognitive impairments with low-level sensorimotor abnormalities in MDD.
Collapse
Affiliation(s)
- Yang Xiao
- Center for Cognition and Brain DisordersThe Affiliated Hospital of Hangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Institute of Psychological ScienceHangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive ImpairmentsHangzhouZhejiang ProvincePR China
| | - Lei Zhao
- Center for Cognition and Brain DisordersThe Affiliated Hospital of Hangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Institute of Psychological ScienceHangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive ImpairmentsHangzhouZhejiang ProvincePR China
| | - Xuelian Zang
- Center for Cognition and Brain DisordersThe Affiliated Hospital of Hangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Institute of Psychological ScienceHangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive ImpairmentsHangzhouZhejiang ProvincePR China
| | - Shao‐Wei Xue
- Center for Cognition and Brain DisordersThe Affiliated Hospital of Hangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Institute of Psychological ScienceHangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive ImpairmentsHangzhouZhejiang ProvincePR China
| |
Collapse
|