1
|
Cui Y, Ma X, Wei J, Chen C, Shakir N, Guirram H, Dai Z, Anderson T, Ferguson D, Qiu S. MET receptor tyrosine kinase promotes the generation of functional synapses in adult cortical circuits. Neural Regen Res 2025; 20:1431-1444. [PMID: 39075910 DOI: 10.4103/nrr.nrr-d-23-01471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/20/2024] [Indexed: 07/31/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202505000-00026/figure1/v/2024-07-28T173839Z/r/image-tiff Loss of synapse and functional connectivity in brain circuits is associated with aging and neurodegeneration, however, few molecular mechanisms are known to intrinsically promote synaptogenesis or enhance synapse function. We have previously shown that MET receptor tyrosine kinase in the developing cortical circuits promotes dendritic growth and dendritic spine morphogenesis. To investigate whether enhancing MET in adult cortex has synapse regenerating potential, we created a knockin mouse line, in which the human MET gene expression and signaling can be turned on in adult (10-12 months) cortical neurons through doxycycline-containing chow. We found that similar to the developing brain, turning on MET signaling in the adult cortex activates small GTPases and increases spine density in prefrontal projection neurons. These findings are further corroborated by increased synaptic activity and transient generation of immature silent synapses. Prolonged MET signaling resulted in an increased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/N-methyl-D-aspartate (AMPA/NMDA) receptor current ratio, indicative of enhanced synaptic function and connectivity. Our data reveal that enhancing MET signaling could be an interventional approach to promote synaptogenesis and preserve functional connectivity in the adult brain. These findings may have implications for regenerative therapy in aging and neurodegeneration conditions.
Collapse
Affiliation(s)
- Yuehua Cui
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Xiaokuang Ma
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Jing Wei
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Chang Chen
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Neha Shakir
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Hitesch Guirram
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Zhiyu Dai
- Department of Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Trent Anderson
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Deveroux Ferguson
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Shenfeng Qiu
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
2
|
Sun X, Liu F, Liu H, Guo L, Ma H, Zhu J, Qian Y. Molecular mechanisms and behavioral relevance underlying neural correlates of childhood neglect. J Affect Disord 2024; 367:795-805. [PMID: 39255872 DOI: 10.1016/j.jad.2024.09.020] [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: 05/08/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Childhood neglect is associated with brain changes, yet the molecular mechanisms and behavioral relevance underlying such associations remain elusive. METHODS We calculated fractional amplitude of low-frequency fluctuations (fALFF) using resting-state functional MRI and tested their correlation with childhood neglect across a large sample of 510 healthy young adults. Then, we investigated the spatial relationships of the identified neural correlates of childhood neglect with gene expression, neurotransmitter, and behavioral domain atlases. RESULTS We found that more severe childhood neglect was correlated with higher fALFF in the bilateral anterior cingulate cortex. Remarkably, the identified neural correlates of childhood neglect were spatially correlated with expression of gene categories primarily involving neuron, synapse, ion channel, cognitive and perceptual processes, and physiological response and regulation. Concurrently, there were significant associations between the neural correlates and specific neurotransmitter systems including serotonin and GABA. Finally, neural correlates of childhood neglect were associated with diverse behavioral domains implicating mental disorders, emotion, cognition, and sensory perception. LIMITATIONS The cross-sectional study design cannot unequivocally establish causality. CONCLUSIONS Our findings may not only add to the current knowledge regarding the relationship between childhood neglect and mental health, but also have clinical implications for developing preventive strategies for individuals exposed to childhood neglect who are at risk for mental disorders.
Collapse
Affiliation(s)
- Xuetian Sun
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Fujun Liu
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Hu Liu
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Lixin Guo
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Haining Ma
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China.
| | - Yinfeng Qian
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China.
| |
Collapse
|
3
|
Du Y, Zhang J, Cao D, Yang W, Li J, Li D, Song M, Yang Z, Zhang J, Jiang T, Liu J. Neuro-immune communication at the core of craving-associated brain structural network reconfiguration in methamphetamine users. Neuroimage 2024; 301:120883. [PMID: 39384079 DOI: 10.1016/j.neuroimage.2024.120883] [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: 06/17/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024] Open
Abstract
Methamphetamine (MA) use disorder is a chronic neurotoxic brain disease characterized by a high risk of relapse driven by intense cravings. However, the neurobiological signatures of cravings remain unclear, limiting the effectiveness of various treatment methods. Diffusion MRI (dMRI) scans from 62 MA users and 57 healthy controls (HC) were used in this study. The MA users were longitudinally followed up during their period of long-term abstinence (duration of long-term abstinence: 347.52±99.25 days). We systematically quantified the control ability of each brain region for craving-associated state transitions using network control theory from a causal perspective. Craving-associated structural alterations (CSA) were investigated through multivariate group comparisons and biological relevance analysis. The neural mechanisms underlying CSA were elucidated using transcriptomic and neurochemical analyses. We observed that long-term abstinence-induced structural alterations significantly influenced the state transition energy involved in the cognitive control response to external information, which correlated with changes in craving scores (r ∼ 0.35, P <0.01). Our causal network analysis further supported the crucial role of the prefrontal cortex (PFC) in craving mechanisms. Notably, while the PFC is central to the craving, the CSAs were distributed widely across multiple brain regions (PFDR<0.05), with strong alterations in somatomotor regions (PFDR<0.05) and moderate alterations in high-level association networks (PFDR<0.05). Additionally, transcriptomic, chemical compounds, cell-type analyses, and molecular imaging collectively highlight the influence of neuro-immune communication on human craving modulation. Our results offer an integrative, multi-scale perspective on unraveling the neural underpinnings of craving and suggest that neuro-immune signaling may be a promising target for future human addiction therapeutics.
Collapse
Affiliation(s)
- Yanyao Du
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| | - Jiaqi Zhang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Dan Cao
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Wenhan Yang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| | - Jin Li
- School of Psychology, Capital Normal University, Beijing 100048, PR China
| | - Deying Li
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ming Song
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Zhengyi Yang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jun Zhang
- Hunan Judicial Police Academy, Changsha, Hunan 410138, PR China
| | - Tianzi Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, PR China; Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China; Xiaoxiang Institute for Brain Health and Yongzhou Central Hospital, Yongzhou 425000, Hunan Province, PR China.
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China; Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan 410011, China; Department of Radiology Quality Control Center, Changsha, Hunan 410011, China.
| |
Collapse
|
4
|
Facca M, Del Felice A, Bertoldo A. Multiscale and multimodal signatures of structure-function coupling variability across the human neocortex. Neuroimage 2024; 302:120902. [PMID: 39490561 DOI: 10.1016/j.neuroimage.2024.120902] [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: 07/11/2024] [Revised: 10/01/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
The relationship between the brain's structural wiring and its dynamic activity is thought to vary regionally, implying that the mechanisms underlying structure-function coupling may differ depending on a region's position within the brain's hierarchy. To better bridge the gap between structure and function, it is crucial to identify the factors shaping this regionality, not only in terms of how static functional connectivity aligns with structure, but also regarding the time-domain variability of this interplay. Here we map structure - function coupling and its time-domain variability and relate them to the heterogeneity of the cortex. We show that these two properties split the cortical landscape into two districts anchored to the opposite ends of the brain's hierarchy. By looking at statistical relationships with layer-specific gene transcription, T1w/T2 w ratio, and synaptic density, we show that macro-scale structure-function coupling may be rooted in the brain's microstructure and meso‑scale laminar specialization. Finally, we demonstrate that a lower and more variable alignment of function and structure may bestow the emergence of unique functional dynamics.
Collapse
Affiliation(s)
| | - Alessandra Del Felice
- Padova Neuroscience Center (PNC), Padova, Italy; Department of Neuroscience, University of Padova, Padova, Italy.
| | - Alessandra Bertoldo
- Padova Neuroscience Center (PNC), Padova, Italy; Department of Information Engineering, University of Padova, Italy
| |
Collapse
|
5
|
Morey RA, Zheng Y, Bayly H, Sun D, Garrett ME, Gasperi M, Maihofer AX, Baird CL, Grasby KL, Huggins AA, Haswell CC, Thompson PM, Medland S, Gustavson DE, Panizzon MS, Kremen WS, Nievergelt CM, Ashley-Koch AE, Logue MW. Genomic structural equation modeling reveals latent phenotypes in the human cortex with distinct genetic architecture. Transl Psychiatry 2024; 14:451. [PMID: 39448598 PMCID: PMC11502831 DOI: 10.1038/s41398-024-03152-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024] Open
Abstract
Genetic contributions to human cortical structure manifest pervasive pleiotropy. This pleiotropy may be harnessed to identify unique genetically-informed parcellations of the cortex that are neurobiologically distinct from functional, cytoarchitectural, or other cortical parcellation schemes. We investigated genetic pleiotropy by applying genomic structural equation modeling (SEM) to map the genetic architecture of cortical surface area (SA) and cortical thickness (CT) for 34 brain regions recently reported in the ENIGMA cortical GWAS. Genomic SEM uses the empirical genetic covariance estimated from GWAS summary statistics with LD score regression (LDSC) to discover factors underlying genetic covariance, which we are denoting genetically informed brain networks (GIBNs). Genomic SEM can fit a multivariate GWAS from summary statistics for each of the GIBNs, which can subsequently be used for LD score regression (LDSC). We found the best-fitting model of cortical SA identified 6 GIBNs and CT identified 4 GIBNs, although sensitivity analyses indicated that other structures were plausible. The multivariate GWASs of the GIBNs identified 74 genome-wide significant (GWS) loci (p < 5 × 10-8), including many previously implicated in neuroimaging phenotypes, behavioral traits, and psychiatric conditions. LDSC of GIBN GWASs found that SA-derived GIBNs had a positive genetic correlation with bipolar disorder (BPD), and cannabis use disorder, indicating genetic predisposition to a larger SA in the specific GIBN is associated with greater genetic risk of these disorders. A negative genetic correlation was observed between attention deficit hyperactivity disorder (ADHD) and major depressive disorder (MDD). CT GIBNs displayed a negative genetic correlation with alcohol dependence. Even though we observed model instability in our application of genomic SEM to high-dimensional data, jointly modeling the genetic architecture of complex traits and investigating multivariate genetic links across neuroimaging phenotypes offers new insights into the genetics of cortical structure and relationships to psychopathology.
Collapse
Affiliation(s)
- Rajendra A Morey
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Yuanchao Zheng
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Henry Bayly
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Delin Sun
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Melanie E Garrett
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
- Department of Medicine, Duke Molecular Physiology Institute, Carmichael Building, Duke University Medical Center, Durham, NC, 27701, USA
| | - Marianna Gasperi
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Adam X Maihofer
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - C Lexi Baird
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
| | - Katrina L Grasby
- Psychiatric Genetics, QIMR, Berghofer Medical Research Institute, 4006, Brisbane, QLD, Australia
| | - Ashley A Huggins
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Courtney C Haswell
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute Keck School of Medicine University of Southern California, Los Angeles, CA, 90033, USA
| | - Sarah Medland
- Queensland Institute for Medical Research, Berghofer Medical Research Institute, 4006, Brisbane, QLD, Australia
| | - Daniel E Gustavson
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Matthew S Panizzon
- Stein Institute for Research on Aging, University of California San Diego, La Jolla, CA, 92093, USA
| | - William S Kremen
- Stein Institute for Research on Aging, University of California San Diego, La Jolla, CA, 92093, USA
| | - Caroline M Nievergelt
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Allison E Ashley-Koch
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
- Department of Medicine, Duke Molecular Physiology Institute, Carmichael Building, Duke University Medical Center, Durham, NC, 27701, USA
| | - Mark W Logue
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA.
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, 02118, USA.
- Biomedical Genetics, Boston University School of Medicine, Boston, MA, 02118-2526, USA.
| |
Collapse
|
6
|
Xu X, Zhao H, Song Y, Cai H, Zhao W, Tang J, Zhu J, Yu Y. Molecular mechanisms underlying the neural correlates of working memory. BMC Biol 2024; 22:238. [PMID: 39428484 PMCID: PMC11492763 DOI: 10.1186/s12915-024-02039-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 10/11/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Working memory (WM), a core component of executive functions, relies on a dedicated brain system that maintains and stores information in the short term. While extensive neuroimaging research has identified a distributed set of neural substrates relevant to WM, their underlying molecular mechanisms remain enigmatic. This study investigated the neural correlates of WM as well as their underlying molecular mechanisms. RESULTS Our voxel-wise analyses of resting-state functional MRI data from 502 healthy young adults showed that better WM performance (higher accuracy and shorter reaction time of the 3-back task) was associated with lower functional connectivity density (FCD) in the left inferior temporal gyrus and higher FCD in the left anterior cingulate cortex. A combination of transcriptome-neuroimaging spatial correlation and the ensemble-based gene category enrichment analysis revealed that the identified neural correlates of WM were associated with expression of diverse gene categories involving important cortical components and their biological processes as well as sodium channels. Cross-region spatial correlation analyses demonstrated significant associations between the neural correlates of WM and a range of neurotransmitters including dopamine, glutamate, serotonin, and acetylcholine. CONCLUSIONS These findings may help to shed light on the molecular mechanisms underlying the neural correlates of WM.
Collapse
Affiliation(s)
- Xiaotao Xu
- 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
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Han Zhao
- 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
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Yu Song
- 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
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Huanhuan Cai
- 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
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Wenming Zhao
- 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
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Jin Tang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230026, China.
| | - 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.
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, 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.
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China.
| |
Collapse
|
7
|
Zhang D, Teng C, Xu Y, Tian L, Cao P, Wang X, Li Z, Guan C, Hu X. Genetic and molecular correlates of cortical thickness alterations in adults with obsessive-compulsive disorder: a transcription-neuroimaging association analysis. Psychol Med 2024; 54:1-10. [PMID: 39363543 PMCID: PMC11496223 DOI: 10.1017/s0033291724001909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/25/2024] [Accepted: 06/11/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Although numerous neuroimaging studies have depicted neural alterations in individuals with obsessive-compulsive disorder (OCD), a psychiatric disorder characterized by intrusive cognitions and repetitive behaviors, the molecular mechanisms connecting brain structural changes and gene expression remain poorly understood. METHODS This study combined the Allen Human Brain Atlas dataset with neuroimaging data from the Meta-Analysis (ENIGMA) consortium and independent cohorts. Later, partial least squares regression and enrichment analysis were performed to probe the correlation between transcription and cortical thickness variation among adults with OCD. RESULTS The cortical map of case-control differences in cortical thickness was spatially correlated with cortical expression of a weighted combination of genes enriched for neurobiologically relevant ontology terms preferentially expressed across different cell types and cortical layers. These genes were specifically expressed in brain tissue, spanning all cortical developmental stages. Protein-protein interaction analysis revealed that these genes coded a network of proteins encompassing various highly interactive hubs. CONCLUSIONS The study findings bridge the gap between neural structure and transcriptome data in OCD, fostering an integrative understanding of the potential biological mechanisms.
Collapse
Affiliation(s)
- Da Zhang
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Changjun Teng
- Department of Medical Psychology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yinhao Xu
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Tian
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ping Cao
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Wang
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zonghong Li
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chengbin Guan
- Department of Medical Psychology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Hu
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Shahriar S, Biswas S, Zhao K, Akcan U, Tuohy MC, Glendinning MD, Kurt A, Wayne CR, Prochilo G, Price MZ, Stuhlmann H, Brekken RA, Menon V, Agalliu D. VEGF-A-mediated venous endothelial cell proliferation results in neoangiogenesis during neuroinflammation. Nat Neurosci 2024; 27:1904-1917. [PMID: 39256571 DOI: 10.1038/s41593-024-01746-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 08/01/2024] [Indexed: 09/12/2024]
Abstract
Newly formed leaky vessels and blood-brain barrier (BBB) damage are present in demyelinating acute and chronic lesions in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). However, the endothelial cell subtypes and signaling pathways contributing to these leaky neovessels are unclear. Here, using single-cell transcriptional profiling and in vivo validation studies, we show that venous endothelial cells express neoangiogenesis gene signatures and show increased proliferation resulting in enlarged veins and higher venous coverage in acute and chronic EAE lesions in female adult mice. These changes correlate with the upregulation of vascular endothelial growth factor A (VEGF-A) signaling. We also confirmed increased expression of neoangiogenic markers in acute and chronic human MS lesions. Treatment with a VEGF-A blocking antibody diminishes the neoangiogenic transcriptomic signatures and vascular proliferation in female adult mice with EAE, but it does not restore BBB function or ameliorate EAE pathology. Our data demonstrate that venous endothelial cells contribute to neoangiogenesis in demyelinating neuroinflammatory conditions.
Collapse
Affiliation(s)
- Sanjid Shahriar
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Wyss Institute for Biologically Inspired Engineering, Boston, MA, USA
| | - Saptarshi Biswas
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kaitao Zhao
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Uğur Akcan
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mary Claire Tuohy
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael D Glendinning
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ali Kurt
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Charlotte R Wayne
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Grace Prochilo
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Maxwell Z Price
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Heidi Stuhlmann
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Rolf A Brekken
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vilas Menon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dritan Agalliu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
9
|
Nabizadeh F. Local molecular and connectomic contributions of tau-related neurodegeneration. GeroScience 2024:10.1007/s11357-024-01339-1. [PMID: 39343862 DOI: 10.1007/s11357-024-01339-1] [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: 06/04/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Neurodegeneration in Alzheimer's disease (AD) is known to be mostly driven by tau neurofibrillary tangles. However, both tau and neurodegeneration exhibit variability in their distribution across the brain and among individuals, and the relationship between tau and neurodegeneration might be influenced by several factors. I aimed to map local molecular and connectivity characteristics that affect the association between tau pathology and neurodegeneration. The current study was conducted on the cross-sectional tau-PET and longitudinal T1-weighted MRI scan data of 186 participants from the ADNI dataset including 71 cognitively unimpaired (CU) and 115 mild cognitive impairment (MCI) individuals. Furthermore, the normative molecular profile of a region was defined using neurotransmitter receptor densities, gene expression, T1w/T2w ratio (myelination), FDG-PET (glycolytic index, glucose metabolism, and oxygen metabolism), and synaptic density. I found that the excitatory-inhibitory (E:I) ratio, myelination, synaptic density, glycolytic index, and functional connectivity are linked with deviation in the relationship between tau and neurodegeneration. Furthermore, there was spatial similarity between tau pathology and glycolytic index, synaptic density, and functional connectivity across brain regions. The current study demonstrates that the regional susceptibility to tau-related neurodegeneration is associated with specific molecular and connectomic characteristics of the affected neural systems. I found that the molecular and connectivity architecture of the human brain is linked to the different effects of tau pathology on downstream neurodegeneration.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Alzheimer's Disease Institute, Tehran, Iran.
| |
Collapse
|
10
|
Qu J, Zhu R, Wu Y, Xu G, Wang D. Abnormal structural‒functional coupling patterning in progressive supranuclear palsy is associated with diverse gradients and histological features. Commun Biol 2024; 7:1195. [PMID: 39341965 PMCID: PMC11439051 DOI: 10.1038/s42003-024-06877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
The anatomy of the brain supports inherent processes, fostering mental abilities and eventually facilitating adaptive behavior. Recent studies have shown that progressive supranuclear palsy (PSP) is accompanied by alterations in functional and structural networks. However, how the structure and function of PSP coordinates change is not clear, and the relationships between structural‒functional coupling (SFC) and the gradient of hierarchical structure and cellular histology remain largely unknown. Here, we use neuroimaging data from two independent cohorts and a public histological dataset to investigate the relationships among the cellular histology, hierarchical structure, and SFC of PSP patients. We find that the SFC of the entire cortex in PSP is severely disrupted, with higher coupling in the visual network (VN). Moreover, coupling differences in PSP follow a macroscopic organizational principle from unimodal to transmodal gradients. Finally, we elucidate greater laminar differentiation in VN regions sensitive to SFC changes in PSP, which is related mainly to the higher cellular density and smaller size of the internal-granular layer. In conclusion, our findings provide an interpretable framework for understanding SFC changes in PSP and provide new insights into the consistency of structural and functional changes in PSP regarding hierarchical structure and cellular histology.
Collapse
Affiliation(s)
- Junyu Qu
- Department of Radiology, Qilu Hospital of Shandong University; Qilu Medical Imaging Institute of Shandong University, Jinan, China
| | - Rui Zhu
- Department of Radiology, Qilu Hospital of Shandong University; Qilu Medical Imaging Institute of Shandong University, Jinan, China
| | - Yongsheng Wu
- Department of Radiology, Qilu Hospital of Shandong University; Qilu Medical Imaging Institute of Shandong University, Jinan, China
| | - Guihua Xu
- Department of Radiology, Qilu Hospital of Shandong University; Qilu Medical Imaging Institute of Shandong University, Jinan, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University; Qilu Medical Imaging Institute of Shandong University, Jinan, China.
- Research Institute of Shandong University: Magnetic Field-free Medicine & Functional Imaging, Jinan, China.
- Shandong Key Laboratory: Magnetic Field-free Medicine & Functional Imaging (MF), Jinan, China.
| |
Collapse
|
11
|
Yao W, Hou X, Zhou H, You S, Lv T, Chen H, Yang Z, Chen C, Bai F. Associations between the multitrajectory neuroplasticity of neuronavigated rTMS-mediated angular gyrus networks and brain gene expression in AD spectrum patients with sleep disorders. Alzheimers Dement 2024. [PMID: 39324544 DOI: 10.1002/alz.14255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/18/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION The multifactorial influence of repetitive transcranial magnetic stimulation (rTMS) on neuroplasticity in neural networks is associated with improvements in cognitive dysfunction and sleep disorders. The mechanisms of rTMS and the transcriptional-neuronal correlation in Alzheimer's disease (AD) patients with sleep disorders have not been fully elucidated. METHODS Forty-six elderly participants with cognitive impairment (23 patients with low sleep quality and 23 patients with high sleep quality) underwent 4-week periods of neuronavigated rTMS of the angular gyrus and neuroimaging tests, and gene expression data for six post mortem brains were collected from another database. Transcription-neuroimaging association analysis was used to evaluate the effects on cognitive dysfunction and the underlying biological mechanisms involved. RESULTS Distinct variable neuroplasticity in the anterior and posterior angular gyrus networks was detected in the low sleep quality group. These interactions were associated with multiple gene pathways, and the comprehensive effects were associated with improvements in episodic memory. DISCUSSION Multitrajectory neuroplasticity is associated with complex biological mechanisms in AD-spectrum patients with sleep disorders. HIGHLIGHTS This was the first transcription-neuroimaging study to demonstrate that multitrajectory neuroplasticity in neural circuits was induced via neuronavigated rTMS, which was associated with complex gene expression in AD-spectrum patients with sleep disorders. The interactions between sleep quality and neuronavigated rTMS were coupled with multiple gene pathways and improvements in episodic memory. The present strategy for integrating neuroimaging, rTMS intervention, and genetic data provide a new approach to comprehending the biological mechanisms involved in AD.
Collapse
Affiliation(s)
- Weina Yao
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinle Hou
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Huijuan Zhou
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shengqi You
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tingyu Lv
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haifeng Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhiyuan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chang Chen
- School of Elderly Care Services and Management, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Bai
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Geriatric Medicine Center, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Geriatric Medicine, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
12
|
Hu S, Li C, Wang Y, Wei T, Wang X, Dong T, Yang Y, Ding Y, Qiu B, Yang W. Structural lesions and transcriptomic specializations shape gradient perturbations in Wilson disease. Brain Commun 2024; 6:fcae329. [PMID: 39372139 PMCID: PMC11450269 DOI: 10.1093/braincomms/fcae329] [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: 07/07/2023] [Revised: 05/27/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
Functional dysregulations in multiple regions are caused by excessive copper deposition in the brain in Wilson disease (WD) patients. The genetic mechanism of WD is thought to involve the abnormal expression of ATP7B in the liver, whereas the biological and molecular processes involved in functional dysregulation within the brain remain unexplored. The objective of this study was to unravel the underpinnings of functional gradient perturbations underlying structural lesions and transcriptomic specializations in WD. In this study, we included 105 WD patients and 93 healthy controls who underwent structural and functional MRI assessments. We used the diffusion mapping embedding model to derive the functional connectome gradient and further employed gray matter volume to uncover structure-function decoupling for WD. Then, we used Neurosynth, clinical data, and whole-brain gene expression data to examine the meta-analytic cognitive function, clinical phenotypes, and transcriptomic specializations related to WD gradient alterations. Compared with controls, WD patients exhibited global topographic changes in the principal pramary-to-transmodal gradient. Meta-analytic terms and clinical characteristics were correlated with these gradient alterations in motor-related processing, higher-order cognition, neurological symptoms, and age. Spatial correlations revealed structure-function decoupling in multiple networks, especially in subcortical and visual networks. Within the cortex, the spatial association between gradient alterations and gene expression profiles has revealed transcriptomic specilizations in WD that display properties indicative of ion homeostasis, neural development, and motor control. Furthermore, for the first time, we characterized the role of the ATP7B gene in impacting subcortical function. The transcriptomic specializations of WD were also associated with other neurological and psychiatric disorders. Finally, we revealed that structural lesions and gradient perturbations may share similar transcriptomic specializations in WD. In conclusion, these findings bridged functional gradient perturbations to structural lesions and gene expression profiles in WD patients, possibly promoting our understanding of the neurobiological mechanisms underlying the emergence of complex neurological and psychiatric phenotypes.
Collapse
Affiliation(s)
- Sheng Hu
- Department of Electronic Engineering and Information Science, Medical Imaging Center, University of Science and Technology of China, Hefei, Anhui, 230026, China
- Institute of Advanced Technology, University of Science and Technology of China, Hefei, Anhui, 230094, China
- School of Medical Information Engineering, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230012, China
| | - Chuanfu Li
- Medical Imaging Center, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| | - Yanming Wang
- Department of Electronic Engineering and Information Science, Medical Imaging Center, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Taohua Wei
- Department of Neurology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
- Key Laboratory of Xinan Medicine of the Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| | - Xiaoxiao Wang
- Department of Electronic Engineering and Information Science, Medical Imaging Center, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Ting Dong
- Department of Neurology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
- Key Laboratory of Xinan Medicine of the Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| | - Yulong Yang
- Department of Neurology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
- Key Laboratory of Xinan Medicine of the Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| | - Yufeng Ding
- Department of Neurology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
- Key Laboratory of Xinan Medicine of the Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| | - Bensheng Qiu
- Department of Electronic Engineering and Information Science, Medical Imaging Center, University of Science and Technology of China, Hefei, Anhui, 230026, China
- Institute of Advanced Technology, University of Science and Technology of China, Hefei, Anhui, 230094, China
| | - Wenming Yang
- Department of Neurology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
- Key Laboratory of Xinan Medicine of the Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| |
Collapse
|
13
|
Liu S, Chen J, Guan L, Xu L, Cai H, Wang J, Zhu DM, Zhu J, Yu Y. The brain, rapid eye movement sleep, and major depressive disorder: A multimodal neuroimaging study. Prog Neuropsychopharmacol Biol Psychiatry 2024; 136:111151. [PMID: 39326695 DOI: 10.1016/j.pnpbp.2024.111151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Evidence has established the prominent involvement of rapid eye movement (REM) sleep disturbance in major depressive disorder (MDD). However, the neural correlates of REM sleep in MDD and their clinical significance are less clear. METHODS Cross-sectional and longitudinal polysomnography and resting-state functional MRI data were collected from 131 MDD patients and 71 healthy controls to measure REM sleep and voxel-mirrored homotopic connectivity (VMHC). Correlation and mediation analyses were performed to examine the associations between REM sleep, VMHC, and clinical variables. Moreover, we conducted spatial correlations between the neural correlates of REM sleep and a multimodal collection of reference brain maps to facilitate genetic, structural and functional annotations. RESULTS MDD patients exhibited REM sleep abnormalities manifesting as higher REM sleep latency and lower REM sleep duration, which were correlated with decreased VMHC of the precentral gyrus and inferior parietal lobe and mediated their associations with more severe anxiety symptoms. Longitudinal data showed that VMHC increase of the inferior parietal lobe was related to improvement of depression symptoms in MDD patients. Spatial correlation analyses revealed that the neural correlates of REM sleep in MDD were linked to gene categories primarily involving cellular metabolic process, signal pathway, and ion channel activity as well as linked to cortical microstructure, metabolism, electrophysiology, and cannabinoid receptor. CONCLUSION These findings may add important context to the growing literature on the complex interplay between sleep and MDD, and more broadly may inform future treatment for depression via regulating sleep.
Collapse
Affiliation(s)
- Siyu Liu
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Jingyao Chen
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Lianzi Guan
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei 230032, China; Department of Sleep Disorders, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230022, China; Hefei Fourth People's Hospital, Hefei 230022, China; Anhui Mental Health Center, Hefei 230022, China
| | - Li Xu
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei 230032, China; Department of Sleep Disorders, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230022, China; Hefei Fourth People's Hospital, Hefei 230022, China; Anhui Mental Health Center, Hefei 230022, China
| | - Huanhuan Cai
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Jie Wang
- 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Dao-Min Zhu
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei 230032, China; Department of Sleep Disorders, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230022, China; Hefei Fourth People's Hospital, Hefei 230022, China; Anhui Mental Health Center, Hefei 230022, China.
| | - 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, 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; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China.
| |
Collapse
|
14
|
Khan AF, Iturria-Medina Y. Beyond the usual suspects: multi-factorial computational models in the search for neurodegenerative disease mechanisms. Transl Psychiatry 2024; 14:386. [PMID: 39313512 PMCID: PMC11420368 DOI: 10.1038/s41398-024-03073-w] [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: 02/01/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
From Alzheimer's disease to amyotrophic lateral sclerosis, the molecular cascades underlying neurodegenerative disorders remain poorly understood. The clinical view of neurodegeneration is confounded by symptomatic heterogeneity and mixed pathology in almost every patient. While the underlying physiological alterations originate, proliferate, and propagate potentially decades before symptomatic onset, the complexity and inaccessibility of the living brain limit direct observation over a patient's lifespan. Consequently, there is a critical need for robust computational methods to support the search for causal mechanisms of neurodegeneration by distinguishing pathogenic processes from consequential alterations, and inter-individual variability from intra-individual progression. Recently, promising advances have been made by data-driven spatiotemporal modeling of the brain, based on in vivo neuroimaging and biospecimen markers. These methods include disease progression models comparing the temporal evolution of various biomarkers, causal models linking interacting biological processes, network propagation models reproducing the spatial spreading of pathology, and biophysical models spanning cellular- to network-scale phenomena. In this review, we discuss various computational approaches for integrating cross-sectional, longitudinal, and multi-modal data, primarily from large observational neuroimaging studies, to understand (i) the temporal ordering of physiological alterations, i(i) their spatial relationships to the brain's molecular and cellular architecture, (iii) mechanistic interactions between biological processes, and (iv) the macroscopic effects of microscopic factors. We consider the extents to which computational models can evaluate mechanistic hypotheses, explore applications such as improving treatment selection, and discuss how model-informed insights can lay the groundwork for a pathobiological redefinition of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ahmed Faraz Khan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada.
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada.
| |
Collapse
|
15
|
Nho K, Risacher SL, Apostolova LG, Bice PJ, Brosch JR, Deardorff R, Faber K, Farlow MR, Foroud T, Gao S, Rosewood T, Kim JP, Nudelman K, Yu M, Aisen P, Sperling R, Hooli B, Shcherbinin S, Svaldi D, Jack CR, Jagust WJ, Landau S, Vasanthakumar A, Waring JF, Doré V, Laws SM, Masters CL, Porter T, Rowe CC, Villemagne VL, Dumitrescu L, Hohman TJ, Libby JB, Mormino E, Buckley RF, Johnson K, Yang HS, Petersen RC, Ramanan VK, Ertekin-Taner N, Vemuri P, Cohen AD, Fan KH, Kamboh MI, Lopez OL, Bennett DA, Ali M, Benzinger T, Cruchaga C, Hobbs D, De Jager PL, Fujita M, Jadhav V, Lamb BT, Tsai AP, Castanho I, Mill J, Weiner MW, Saykin AJ. CYP1B1-RMDN2 Alzheimer's disease endophenotype locus identified for cerebral tau PET. Nat Commun 2024; 15:8251. [PMID: 39304655 PMCID: PMC11415491 DOI: 10.1038/s41467-024-52298-2] [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: 11/14/2023] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Determining the genetic architecture of Alzheimer's disease pathologies can enhance mechanistic understanding and inform precision medicine strategies. Here, we perform a genome-wide association study of cortical tau quantified by positron emission tomography in 3046 participants from 12 independent studies. The CYP1B1-RMDN2 locus is associated with tau deposition. The most significant signal is at rs2113389, explaining 4.3% of the variation in cortical tau, while APOE4 rs429358 accounts for 3.6%. rs2113389 is associated with higher tau and faster cognitive decline. Additive effects, but no interactions, are observed between rs2113389 and diagnosis, APOE4, and amyloid beta positivity. CYP1B1 expression is upregulated in AD. rs2113389 is associated with higher CYP1B1 expression and methylation levels. Mouse model studies provide additional functional evidence for a relationship between CYP1B1 and tau deposition but not amyloid beta. These results provide insight into the genetic basis of cerebral tau deposition and support novel pathways for therapeutic development in AD.
Collapse
Affiliation(s)
- Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of BioHealth Informatics, Indiana University, Indianapolis, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Liana G Apostolova
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Paula J Bice
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Jared R Brosch
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Rachael Deardorff
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Kelley Faber
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, USA
| | - Martin R Farlow
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Tatiana Foroud
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, USA
| | - Thea Rosewood
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Jun Pyo Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Kelly Nudelman
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, USA
| | - Meichen Yu
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Paul Aisen
- Department of Neurology, Keck School of Medicine, University of Southern California, San Diego, USA
| | - Reisa Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | | | | | | | | - William J Jagust
- UC Berkeley Helen Wills Neuroscience Institute, University of California - Berkeley, Berkeley, USA
| | - Susan Landau
- UC Berkeley Helen Wills Neuroscience Institute, University of California - Berkeley, Berkeley, USA
| | | | | | - Vincent Doré
- CSIRO Health and Biosecurity, Melbourne, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Australia
| | - Simon M Laws
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Parkville, Australia
| | - Tenielle Porter
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Australia
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Parkville, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Australia
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Logan Dumitrescu
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, USA
| | - Julia B Libby
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, USA
| | - Elizabeth Mormino
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, USA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Keith Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Center for Alzheimer's Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | | | | | - Nilüfer Ertekin-Taner
- Department of Neurology, Mayo Clinic, Jacksonville, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, USA
| | | | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Kang-Hsien Fan
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Oscar L Lopez
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Medical College, Rush University, Chicago, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University, St. Louis, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, USA
| | - Diana Hobbs
- Department of Radiology, Washington University School of Medicine, St. Louis, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
| | - Vaishnavi Jadhav
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
| | - Bruce T Lamb
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
| | - Andy P Tsai
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, USA
| | - Isabel Castanho
- Department for Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Jonathan Mill
- Department for Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Michael W Weiner
- Departments of Radiology, Medicine, and Psychiatry, University of California-San Francisco, San Francisco, USA
- Department of Veterans Affairs Medical Center, San Francisco, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA.
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA.
| |
Collapse
|
16
|
Kim CY, Park Y, Namgung JY, Park Y, Park BY. The macroscale routing mechanism of structural brain connectivity related to body mass index. Hum Brain Mapp 2024; 45:e70019. [PMID: 39230183 PMCID: PMC11372826 DOI: 10.1002/hbm.70019] [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: 10/20/2023] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
Understanding the brain's mechanisms in individuals with obesity is important for managing body weight. Prior neuroimaging studies extensively investigated alterations in brain structure and function related to body mass index (BMI). However, how the network communication among the large-scale brain networks differs across BMI is underinvestigated. This study used diffusion magnetic resonance imaging of 290 young adults to identify links between BMI and brain network mechanisms. Navigation efficiency, a measure of network routing, was calculated from the structural connectivity computed using diffusion tractography. The sensory and frontoparietal networks indicated positive associations between navigation efficiency and BMI. The neurotransmitter association analysis identified that serotonergic and dopaminergic receptors, as well as opioid and norepinephrine systems, were related to BMI-related alterations in navigation efficiency. The transcriptomic analysis found that genes associated with network routing across BMI overlapped with genes enriched in excitatory and inhibitory neurons, specifically, gene enrichments related to synaptic transmission and neuron projection. Our findings suggest a valuable insight into understanding BMI-related alterations in brain network routing mechanisms and the potential underlying cellular biology, which might be used as a foundation for BMI-based weight management.
Collapse
Affiliation(s)
- Chae Yeon Kim
- Department of Data Science, Inha University, Incheon, South Korea
| | - Yunseo Park
- Department of Data Science, Inha University, Incheon, South Korea
| | | | - Yeongjun Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, South Korea
| | - Bo-Yong Park
- Department of Brain and Cognitive Engineering, Korea University, Seoul, South Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, South Korea
- Research Center for Small Businesses Ecosystem, Inha University, Incheon, South Korea
| |
Collapse
|
17
|
Hoang N, Sardaripour N, Ramey GD, Schilling K, Liao E, Chen Y, Park JH, Bledsoe X, Landman BA, Gamazon ER, Benton ML, Capra JA, Rubinov M. Integration of estimated regional gene expression with neuroimaging and clinical phenotypes at biobank scale. PLoS Biol 2024; 22:e3002782. [PMID: 39269986 PMCID: PMC11424006 DOI: 10.1371/journal.pbio.3002782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/25/2024] [Accepted: 08/01/2024] [Indexed: 09/15/2024] Open
Abstract
An understanding of human brain individuality requires the integration of data on brain organization across people and brain regions, molecular and systems scales, as well as healthy and clinical states. Here, we help advance this understanding by leveraging methods from computational genomics to integrate large-scale genomic, transcriptomic, neuroimaging, and electronic-health record data sets. We estimated genetically regulated gene expression (gr-expression) of 18,647 genes, across 10 cortical and subcortical regions of 45,549 people from the UK Biobank. First, we showed that patterns of estimated gr-expression reflect known genetic-ancestry relationships, regional identities, as well as inter-regional correlation structure of directly assayed gene expression. Second, we performed transcriptome-wide association studies (TWAS) to discover 1,065 associations between individual variation in gr-expression and gray-matter volumes across people and brain regions. We benchmarked these associations against results from genome-wide association studies (GWAS) of the same sample and found hundreds of novel associations relative to these GWAS. Third, we integrated our results with clinical associations of gr-expression from the Vanderbilt Biobank. This integration allowed us to link genes, via gr-expression, to neuroimaging and clinical phenotypes. Fourth, we identified associations of polygenic gr-expression with structural and functional MRI phenotypes in the Human Connectome Project (HCP), a small neuroimaging-genomic data set with high-quality functional imaging data. Finally, we showed that estimates of gr-expression and magnitudes of TWAS were generally replicable and that the p-values of TWAS were replicable in large samples. Collectively, our results provide a powerful new resource for integrating gr-expression with population genetics of brain organization and disease.
Collapse
Affiliation(s)
- Nhung Hoang
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Neda Sardaripour
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Grace D. Ramey
- Biological and Medical Informatics Division, University of California, San Francisco, California, United States of America
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - Kurt Schilling
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Emily Liao
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Yiting Chen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jee Hyun Park
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Xavier Bledsoe
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Bennett A. Landman
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Eric R. Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Mary Lauren Benton
- Department of Computer Science, Baylor University, Waco, Texas, United States of America
| | - John A. Capra
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Bakar Computational Health Sciences Institute, University of California, San Francisco, California, United States of America
| | - Mikail Rubinov
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Psychology, Vanderbilt University, Nashville, Tennessee, United States of America
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, Virginia, United States of America
| |
Collapse
|
18
|
Huang W, Sun X, Zhang X, Xu R, Qian Y, Zhu J. Neural Correlates of Early-Life Urbanization and Their Spatial Relationships with Gene Expression, Neurotransmitter, and Behavioral Domain Atlases. Mol Neurobiol 2024; 61:6407-6422. [PMID: 38308665 DOI: 10.1007/s12035-024-03962-7] [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: 07/21/2023] [Accepted: 01/15/2024] [Indexed: 02/05/2024]
Abstract
Previous neuroimaging research has established associations between urban exposure during early life and alterations in brain function and structure. However, the molecular mechanisms and behavioral relevance of these associations remain largely unknown. Here, we aimed to address this question using a combined analysis of multimodal data. Initially, we calculated amplitude of low-frequency fluctuations (ALFF) and gray matter volume (GMV) using resting-state functional and structural MRI to investigate their associations with early-life urbanization in a large sample of 511 healthy young adults. Then, we examined the spatial relationships of the identified neural correlates of early-life urbanization with gene expression, neurotransmitter, and behavioral domain atlases. Results showed that higher early-life urbanization scores were correlated with increased ALFF of the right fusiform gyrus and decreased GMV of the left dorsal medial prefrontal cortex and left precuneus. Remarkably, the identified neural correlates of early-life urbanization were spatially correlated with expression of gene categories primarily involving immune system process, signal transduction, and cellular metabolic process. Concurrently, there were significant associations between the neural correlates and specific neurotransmitter systems including dopamine, acetylcholine, and serotonin. Finally, we found that the ALFF correlates were associated with behavioral terms including "perception," "sensory," "cognitive control," and "reasoning." Apart from expanding existing knowledge of early-life urban environmental risk for mental disorders and health in general, our findings may contribute to an emerging framework for integrating social science, neuroscience, genetics, and public policy to respond to the major health challenge of world urbanization.
Collapse
Affiliation(s)
- Weisheng Huang
- 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
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Xuetian Sun
- 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
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Xiaohan Zhang
- 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
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Ruoxuan Xu
- 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
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Yinfeng Qian
- 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.
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China.
| | - 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.
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China.
| |
Collapse
|
19
|
González-Peñas J, Alloza C, Brouwer R, Díaz-Caneja CM, Costas J, González-Lois N, Gallego AG, de Hoyos L, Gurriarán X, Andreu-Bernabeu Á, Romero-García R, Fañanás L, Bobes J, González-Pinto A, Crespo-Facorro B, Martorell L, Arrojo M, Vilella E, Gutiérrez-Zotes A, Perez-Rando M, Moltó MD, Buimer E, van Haren N, Cahn W, O'Donovan M, Kahn RS, Arango C, Pol HH, Janssen J, Schnack H. Accelerated Cortical Thinning in Schizophrenia Is Associated With Rare and Common Predisposing Variation to Schizophrenia and Neurodevelopmental Disorders. Biol Psychiatry 2024; 96:376-389. [PMID: 38521159 DOI: 10.1016/j.biopsych.2024.03.011] [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: 08/17/2023] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Schizophrenia is a highly heritable disorder characterized by increased cortical thinning throughout the life span. Studies have reported a shared genetic basis between schizophrenia and cortical thickness. However, no genes whose expression is related to abnormal cortical thinning in schizophrenia have been identified. METHODS We conducted linear mixed models to estimate the rates of accelerated cortical thinning across 68 regions from the Desikan-Killiany atlas in individuals with schizophrenia compared with healthy control participants from a large longitudinal sample (ncases = 169 and ncontrols = 298, ages 16-70 years). We studied the correlation between gene expression data from the Allen Human Brain Atlas and accelerated thinning estimates across cortical regions. Finally, we explored the functional and genetic underpinnings of the genes that contribute most to accelerated thinning. RESULTS We found a global pattern of accelerated cortical thinning in individuals with schizophrenia compared with healthy control participants. Genes underexpressed in cortical regions that exhibit this accelerated thinning were downregulated in several psychiatric disorders and were enriched for both common and rare disrupting variation for schizophrenia and neurodevelopmental disorders. In contrast, none of these enrichments were observed for baseline cross-sectional cortical thickness differences. CONCLUSIONS Our findings suggest that accelerated cortical thinning, rather than cortical thickness alone, serves as an informative phenotype for neurodevelopmental disruptions in schizophrenia. We highlight the genetic and transcriptomic correlates of this accelerated cortical thinning, emphasizing the need for future longitudinal studies to elucidate the role of genetic variation and the temporal-spatial dynamics of gene expression in brain development and aging in schizophrenia.
Collapse
Affiliation(s)
- Javier González-Peñas
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain; CIBERSAM, Madrid, Spain.
| | - Clara Alloza
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain; CIBERSAM, Madrid, Spain
| | - Rachel Brouwer
- Department of Psychiatry, UMCU Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Covadonga M Díaz-Caneja
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain; CIBERSAM, Madrid, Spain; School of Medicine, Universidad Complutense, Madrid, Spain
| | - Javier Costas
- Instituto de Investigación Sanitària de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde, Santiago de Compostela, Galicia, Spain
| | - Noemí González-Lois
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain
| | - Ana Guil Gallego
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain
| | - Lucía de Hoyos
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain
| | - Xaquín Gurriarán
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain; CIBERSAM, Madrid, Spain
| | - Álvaro Andreu-Bernabeu
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain; CIBERSAM, Madrid, Spain
| | - Rafael Romero-García
- Department of Medical Physiology and Biophysics, Instituto de Biomedicina de Sevilla, HUVR/CSIC/Universidad de Sevilla/CIBERSAM, Instituto de Salud Carlos III, Sevilla, Spain; Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Lourdes Fañanás
- CIBERSAM, Madrid, Spain; Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Julio Bobes
- CIBERSAM, Madrid, Spain; Faculty of Medicine and Health Sciences-Psychiatry, Universidad de Oviedo, Instituto de Investigación Sanitaria del Principado de Asturias, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| | - Ana González-Pinto
- CIBERSAM, Madrid, Spain; BIOARABA Health Research Institute, Organización Sanitaria Integrada Araba, University Hospital, University of the Basque Country, Vitoria, Spain
| | - Benedicto Crespo-Facorro
- CIBERSAM, Madrid, Spain; Hospital Universitario Virgen del Rocío, Department of Psychiatry, Universidad de Sevilla, Sevilla, Spain
| | - Lourdes Martorell
- CIBERSAM, Madrid, Spain; Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-Centres de Recerca de Catalunya, Universitat Rovira i Virgili, Reus, Spain
| | - Manuel Arrojo
- Instituto de Investigación Sanitària de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde, Santiago de Compostela, Galicia, Spain
| | - Elisabet Vilella
- CIBERSAM, Madrid, Spain; Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-Centres de Recerca de Catalunya, Universitat Rovira i Virgili, Reus, Spain
| | - Alfonso Gutiérrez-Zotes
- CIBERSAM, Madrid, Spain; Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-Centres de Recerca de Catalunya, Universitat Rovira i Virgili, Reus, Spain
| | - Marta Perez-Rando
- Fundación Investigación Hospital Clínico de València, Fundación Investigación Hospital Clínico de Valencia, València, Spain; Unidad de Neurobiología, Instituto de Biotecnología y Biomedicina, Universitat de València, València, Spain
| | - María Dolores Moltó
- CIBERSAM, Madrid, Spain; Unidad de Neurobiología, Instituto de Biotecnología y Biomedicina, Universitat de València, València, Spain; Department of Genetics, Universitat de València, Campus of Burjassot, València, Spain
| | - Elizabeth Buimer
- Department of Psychiatry, UMCU Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Neeltje van Haren
- Department of Psychiatry, UMCU Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Wiepke Cahn
- Department of Psychiatry, UMCU Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands; Altrecht Mental Health Institute, Altrecht Science, Utrecht, the Netherlands
| | - Michael O'Donovan
- Medical Research Council for Neuropsychiatric Genetics and Genomics and Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - René S Kahn
- Department of Psychiatry, UMCU Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Celso Arango
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain; CIBERSAM, Madrid, Spain; School of Medicine, Universidad Complutense, Madrid, Spain
| | - Hilleke Hulshoff Pol
- Department of Psychiatry, UMCU Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joost Janssen
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitària Gregorio Marañón, Madrid, Spain; CIBERSAM, Madrid, Spain; Department of Psychiatry, UMCU Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hugo Schnack
- Department of Psychiatry, UMCU Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
20
|
Zhang J, Chen J, Ding G. Universality and language specificity of brain reading networks: A developmental perspective. Dev Sci 2024; 27:e13379. [PMID: 36899475 DOI: 10.1111/desc.13379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/11/2023] [Accepted: 02/03/2023] [Indexed: 03/12/2023]
Abstract
The development of reading networks across different languages and cultures provides an important window to address gene-culture interactions in brain functionality development. Previous meta-analyses have explored the neural correlates of reading in different languages with diverse orthographic transparencies. However, it remains unknown whether the neural topographic relationship of different languages varies when taking development into account. To address this issue, we conducted meta-analyses of neuroimaging studies with approaches of activation likelihood estimation and seed-based effect size mapping and focused on two highly contrasting languages, Chinese and English. The meta-analyses covered 61 studies of Chinese reading and 64 studies of English reading by native speakers. The brain reading networks of child and adult readers were further separately analyzed and compared to explore the developmental effects. The results revealed that the commonalities and differences in reading networks for Chinese and English were inconsistent between children and adults. In addition, the reading networks converged with development, and the effects of writing systems on brain function organizations were more salient in the initial stages of reading. An interesting finding was that the left inferior parietal lobule demonstrated increased effect sizes in adults compared with children in both Chinese and English reading, indicating a common developmental feature of reading mechanisms across the two languages. These findings provide new insights into the functional evolution and cultural modulation of brain reading networks. RESEARCH HIGHLIGHTS: Meta-analyses with approaches of activation likelihood estimation and seed-based effect size mapping were performed to evaluate the developmental characteristics of brain reading networks. The engagement of universal and language-specific reading networks was different between children and adults, and the reading networks converged with increased reading experience. Overall the middle/inferior occipital and inferior/middle frontal gyrus were specific to Chinese and the middle temporal, right inferior frontal gyrus were specific to English. The left inferior parietal lobule was engaged more in adults than children in Chinese and English reading, demonstrating a common developmental feature of reading mechanisms.
Collapse
Affiliation(s)
- Jia Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Jie Chen
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Guosheng Ding
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
21
|
Li J, Jin S, Li Z, Zeng X, Yang Y, Luo Z, Xu X, Cui Z, Liu Y, Wang J. Morphological Brain Networks of White Matter: Mapping, Evaluation, Characterization, and Application. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400061. [PMID: 39005232 PMCID: PMC11425219 DOI: 10.1002/advs.202400061] [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: 01/03/2024] [Revised: 06/27/2024] [Indexed: 07/16/2024]
Abstract
Although white matter (WM) accounts for nearly half of adult brain, its wiring diagram is largely unknown. Here, an approach is developed to construct WM networks by estimating interregional morphological similarity based on structural magnetic resonance imaging. It is found that morphological WM networks showed nontrivial topology, presented good-to-excellent test-retest reliability, accounted for phenotypic interindividual differences in cognition, and are under genetic control. Through integration with multimodal and multiscale data, it is further showed that morphological WM networks are able to predict the patterns of hamodynamic coherence, metabolic synchronization, gene co-expression, and chemoarchitectonic covariance, and associated with structural connectivity. Moreover, the prediction followed WM functional connectomic hierarchy for the hamodynamic coherence, is related to genes enriched in the forebrain neuron development and differentiation for the gene co-expression, and is associated with serotonergic system-related receptors and transporters for the chemoarchitectonic covariance. Finally, applying this approach to multiple sclerosis and neuromyelitis optica spectrum disorders, it is found that both diseases exhibited morphological dysconnectivity, which are correlated with clinical variables of patients and are able to diagnose and differentiate the diseases. Altogether, these findings indicate that morphological WM networks provide a reliable and biologically meaningful means to explore WM architecture in health and disease.
Collapse
Affiliation(s)
- Junle Li
- Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhou510631China
| | - Suhui Jin
- Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhou510631China
| | - Zhen Li
- Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhou510631China
| | - Xiangli Zeng
- Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhou510631China
| | - Yuping Yang
- Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhou510631China
| | - Zhenzhen Luo
- Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhou510631China
| | - Xiaoyu Xu
- State Key Laboratory of Cognitive Neuroscience and LearningBeijing Normal UniversityBeijing100875China
- Chinese Institute for Brain ResearchBeijing102206China
| | - Zaixu Cui
- Chinese Institute for Brain ResearchBeijing102206China
| | - Yaou Liu
- Department of RadiologyBeijing Tiantan HospitalBeijing100070China
| | - Jinhui Wang
- Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhou510631China
- Key Laboratory of BrainCognition and Education SciencesMinistry of EducationGuangzhou510631China
- Center for Studies of Psychological ApplicationSouth China Normal UniversityGuangzhou510631China
- Guangdong Key Laboratory of Mental Health and Cognitive ScienceSouth China Normal UniversityGuangzhou510631China
| |
Collapse
|
22
|
Schleifer CH, Chang SE, Amir CM, O'Hora KP, Fung H, Kang JWD, Kushan-Wells L, Daly E, Di Fabio F, Frascarelli M, Gudbrandsen M, Kates WR, Murphy D, Addington J, Anticevic A, Cadenhead KS, Cannon TD, Cornblatt BA, Keshavan M, Mathalon DH, Perkins DO, Stone WS, Walker E, Woods SW, Uddin LQ, Kumar K, Hoftman GD, Bearden CE. Unique Functional Neuroimaging Signatures of Genetic Versus Clinical High Risk for Psychosis. Biol Psychiatry 2024:S0006-3223(24)01538-5. [PMID: 39181389 DOI: 10.1016/j.biopsych.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND 22q11.2 deletion syndrome (22qDel) is a copy number variant that is associated with psychosis and other neurodevelopmental disorders. Adolescents who are at clinical high risk for psychosis (CHR) are identified based on the presence of subthreshold psychosis symptoms. Whether common neural substrates underlie these distinct high-risk populations is unknown. We compared functional brain measures in 22qDel and CHR cohorts and mapped the results to biological pathways. METHODS We analyzed 2 large multisite cohorts with resting-state functional magnetic resonance imaging data: 1) a 22qDel cohort (n = 164, 47% female) and typically developing (TD) control participants (n = 134, 56% female); and 2) a cohort of CHR individuals (n = 240, 41% female) and TD control participants (n = 149, 46% female) from the NAPLS-2 (North American Prodrome Longitudinal Study-2). We computed global brain connectivity (GBC), local connectivity (LC), and brain signal variability (BSV) across cortical regions and tested case-control differences for 22qDel and CHR separately. Group difference maps were related to published brain maps using autocorrelation-preserving permutation. RESULTS BSV, LC, and GBC were significantly disrupted in individuals with 22qDel compared with TD control participants (false discovery rate-corrected q < .05). Spatial maps of BSV and LC differences were highly correlated with each other, unlike GBC. In the CHR group, only LC was significantly altered versus the control group, with a different spatial pattern than the 22qDel group. Group differences mapped onto biological gradients, with 22qDel effects being strongest in regions with high predicted blood flow and metabolism. CONCLUSIONS 22qDel carriers and CHR individuals exhibited different effects on functional magnetic resonance imaging temporal variability and multiscale functional connectivity. In 22qDel carriers, strong and convergent disruptions in BSV and LC that were not seen in CHR individuals suggest distinct functional brain alterations.
Collapse
Affiliation(s)
- Charles H Schleifer
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California
| | - Sarah E Chang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California
| | - Carolyn M Amir
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California
| | - Kathleen P O'Hora
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California
| | - Hoki Fung
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California
| | - Jee Won D Kang
- Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Leila Kushan-Wells
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California
| | - Eileen Daly
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Fabio Di Fabio
- Department of Human Neurosciences, Sapienza University, Rome, Italy
| | | | - Maria Gudbrandsen
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Research in Psychological Wellbeing, School of Psychology, University of Roehampton, London, United Kingdom
| | - Wendy R Kates
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York
| | - Declan Murphy
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Jean Addington
- Department of Psychiatry, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Alan Anticevic
- Manifest Technologies, New Haven, Connecticut; Department of Psychiatry, Yale University, New Haven, Connecticut
| | - Kristin S Cadenhead
- Department of Psychiatry, University of California, San Diego, San Diego, California
| | - Tyrone D Cannon
- Department of Psychiatry, Yale University, New Haven, Connecticut; Department of Psychology, Yale University, New Haven, Connecticut
| | - Barbara A Cornblatt
- Department of Psychiatry, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Matcheri Keshavan
- Department of Psychiatry, Harvard Medical School at Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Daniel H Mathalon
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco and Veterans Affairs San Francisco Health Care System, San Francisco, California
| | - Diana O Perkins
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - William S Stone
- Department of Psychiatry, Harvard Medical School at Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Elaine Walker
- Department of Psychology, Emory University, Atlanta, Georgia
| | - Scott W Woods
- Department of Psychiatry, Yale University, New Haven, Connecticut
| | - Lucina Q Uddin
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California
| | - Kuldeep Kumar
- Centre de Recherche du CHU Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| | - Gil D Hoftman
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California
| | - Carrie E Bearden
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California; Department of Psychology, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
23
|
Lin L, Chang Z, Zhang Y, Xue K, Xie Y, Wei L, Li X, Zhao Z, Luo Y, Dong H, Liang M, Liu H, Yu C, Qin W, Ding H. Voxel-based texture similarity networks reveal individual variability and correlate with biological ontologies. Neuroimage 2024; 297:120688. [PMID: 38878916 DOI: 10.1016/j.neuroimage.2024.120688] [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: 11/28/2023] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
The human brain is organized as a complex, hierarchical network. However, the structural covariance patterns among brain regions and the underlying biological substrates of such covariance networks remain to be clarified. The present study proposed a novel individualized structural covariance network termed voxel-based texture similarity networks (vTSNs) based on 76 refined voxel-based textural features derived from structural magnetic resonance images. Validated in three independent longitudinal healthy cohorts (40, 23, and 60 healthy participants, respectively) with two common brain atlases, we found that the vTSN could robustly resolve inter-subject variability with high test-retest reliability. In contrast to the regional-based texture similarity networks (rTSNs) that calculate radiomic features based on region-of-interest information, vTSNs had higher inter- and intra-subject variability ratios and test-retest reliability in connectivity strength and network topological properties. Moreover, the Spearman correlation indicated a stronger association of the gene expression similarity network (GESN) with vTSNs than with rTSNs (vTSN: r = 0.600, rTSN: r = 0.433, z = 39.784, P < 0.001). Hierarchical clustering identified 3 vTSN subnets with differential association patterns with 13 coexpression modules, 16 neurotransmitters, 7 electrophysiology, 4 metabolism, and 2 large-scale structural and 4 functional organization maps. Moreover, these subnets had unique biological hierarchical organization from the subcortex-limbic system to the ventral neocortex and then to the dorsal neocortex. Based on 424 unrelated, qualified healthy subjects from the Human Connectome Project, we found that vTSNs could sensitively represent sex differences, especially for connections in the subcortex-limbic system and between the subcortex-limbic system and the ventral neocortex. Moreover, a multivariate variance component model revealed that vTSNs could explain a significant proportion of inter-subject behavioral variance in cognition (80.0 %) and motor functions (63.4 %). Finally, using 494 healthy adults (aged 19-80 years old) from the Southwest University Adult Lifespan Dataset, the Spearman correlation identified a significant association between aging and vTSN strength, especially within the subcortex-limbic system and between the subcortex-limbic system and the dorsal neocortex. In summary, our proposed vTSN is robust in uncovering individual variability and neurobiological brain processes, which can serve as biologically plausible measures for linking biological processes and human behavior.
Collapse
Affiliation(s)
- Liyuan Lin
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhongyu Chang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yu Zhang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Kaizhong Xue
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China; 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
| | - Yingying Xie
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Luli Wei
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Li
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhen Zhao
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yun Luo
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Haoyang Dong
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Meng Liang
- School of Medical Imaging, Tianjin Medical University, Tianjin 300070, China
| | - Huaigui Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China; State Key Laboratory of Experimental Hematology, Beijing, China.
| | - Wen Qin
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Hao Ding
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China; School of Medical Imaging, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
24
|
Ma X, Li J, Yang Y, Qiu X, Sheng J, Han N, Wu C, Xu G, Jiang G, Tian J, Weng X, Wang J. Enhanced cerebral blood flow similarity of the somatomotor network in chronic insomnia: Transcriptomic decoding, gut microbial signatures and phenotypic roles. Neuroimage 2024; 297:120762. [PMID: 39089603 DOI: 10.1016/j.neuroimage.2024.120762] [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: 02/29/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
Chronic insomnia (CI) is a complex disease involving multiple factors including genetics, gut microbiota, and brain structure and function. However, there lacks a unified framework to elucidate how these factors interact in CI. By combining data of clinical assessment, sleep behavior recording, cognitive test, multimodal MRI (structural, functional, and perfusion), gene, and gut microbiota, this study demonstrated that enhanced cerebral blood flow (CBF) similarities of the somatomotor network (SMN) acted as a key mediator to link multiple factors in CI. Specifically, we first demonstrated that only CBF but not morphological or functional networks exhibited alterations in patients with CI, characterized by increases within the SMN and between the SMN and higher-order associative networks. Moreover, these findings were highly reproducible and the CBF similarity method was test-retest reliable. Further, we showed that transcriptional profiles explained 60.4 % variance of the pattern of the increased CBF similarities with the most correlated genes enriched in regulation of cellular and protein localization and material transport, and gut microbiota explained 69.7 % inter-individual variance in the increased CBF similarities with the most contributions from Negativicutes and Lactobacillales. Finally, we found that the increased CBF similarities were correlated with clinical variables, accounted for sleep behaviors and cognitive deficits, and contributed the most to the patient-control classification (accuracy = 84.4 %). Altogether, our findings have important implications for understanding the neuropathology of CI and may inform ways of developing new therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Xiaofen Ma
- Department of Nuclear Medicine, Jinan University Affiliated Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Junle Li
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Yuping Yang
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Xiaofan Qiu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Jintao Sheng
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Ningke Han
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Changwen Wu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Guang Xu
- Department of Neurology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Guihua Jiang
- Department of Nuclear Medicine, Jinan University Affiliated Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Junzhang Tian
- Department of Nuclear Medicine, Jinan University Affiliated Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xuchu Weng
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China; Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Center for Studies of Psychological Application, South China Normal University, Guangzhou, China; Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Jinhui Wang
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China; Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Center for Studies of Psychological Application, South China Normal University, Guangzhou, China; Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China.
| |
Collapse
|
25
|
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
|
26
|
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] [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
|
27
|
Peng Y, Chai C, Xue K, Tang J, Wang S, Su Q, Liao C, Zhao G, Wang S, Zhang N, Zhang Z, Lei M, Liu F, Liang M. Unraveling multi-scale neuroimaging biomarkers and molecular foundations for schizophrenia: A combined multivariate pattern analysis and transcriptome-neuroimaging association study. CNS Neurosci Ther 2024; 30:e14906. [PMID: 39118226 PMCID: PMC11310100 DOI: 10.1111/cns.14906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 07/09/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
AIMS Schizophrenia is characterized by alterations in resting-state spontaneous brain activity; however, it remains uncertain whether variations at diverse spatial scales are capable of effectively distinguishing patients from healthy controls. Additionally, the genetic underpinnings of these alterations remain poorly elucidated. We aimed to address these questions in this study to gain better understanding of brain alterations and their underlying genetic factors in schizophrenia. METHODS A cohort of 103 individuals with diagnosed schizophrenia and 110 healthy controls underwent resting-state functional MRI scans. Spontaneous brain activity was assessed using the regional homogeneity (ReHo) metric at four spatial scales: voxel-level (Scale 1) and regional-level (Scales 2-4: 272, 53, 17 regions, respectively). For each spatial scale, multivariate pattern analysis was performed to classify schizophrenia patients from healthy controls, and a transcriptome-neuroimaging association analysis was performed to establish connections between gene expression data and ReHo alterations in schizophrenia. RESULTS The ReHo metrics at all spatial scales effectively discriminated schizophrenia from healthy controls. Scale 2 showed the highest classification accuracy at 84.6%, followed by Scale 1 (83.1%) and Scale 3 (78.5%), while Scale 4 exhibited the lowest accuracy (74.2%). Furthermore, the transcriptome-neuroimaging association analysis showed that there were not only shared but also unique enriched biological processes across the four spatial scales. These related biological processes were mainly linked to immune responses, inflammation, synaptic signaling, ion channels, cellular development, myelination, and transporter activity. CONCLUSIONS This study highlights the potential of multi-scale ReHo as a valuable neuroimaging biomarker in the diagnosis of schizophrenia. By elucidating the complex molecular basis underlying the ReHo alterations of this disorder, this study not only enhances our understanding of its pathophysiology, but also pave the way for future advancements in genetic diagnosis and treatment of schizophrenia.
Collapse
Affiliation(s)
- Yanmin Peng
- School of Medical Imaging and Tianjin Key Laboratory of Functional ImagingTianjin Medical UniversityTianjinChina
| | - Chao Chai
- Department of Radiology and Tianjin Key Laboratory of Functional ImagingTianjin Medical University General HospitalTianjinChina
- Department of Radiology, School of Medicine, Tianjin First Central HospitalNankai UniversityTianjinChina
| | - Kaizhong Xue
- Department of Radiology and Nuclear Medicine, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jie Tang
- Department of Radiology and Tianjin Key Laboratory of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Sijia Wang
- Department of Radiology and Tianjin Key Laboratory of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Qian Su
- Department of Molecular Imaging and Nuclear MedicineTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Chongjian Liao
- School of Medical Imaging and Tianjin Key Laboratory of Functional ImagingTianjin Medical UniversityTianjinChina
| | - Guoshu Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Shaoying Wang
- Department of Radiology and Tianjin Key Laboratory of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Nannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Zhihui Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Minghuan Lei
- Department of Radiology and Tianjin Key Laboratory of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Meng Liang
- School of Medical Imaging and Tianjin Key Laboratory of Functional ImagingTianjin Medical UniversityTianjinChina
| |
Collapse
|
28
|
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
|
29
|
Tu Y, Liu Y, Fan S, Weng J, Li M, Zhang F, Fu Y, Hu J. Relationship between brain white matter damage and grey matter atrophy in hereditary spastic paraplegia types 4 and 5. Eur J Neurol 2024; 31:e16310. [PMID: 38651515 PMCID: PMC11235729 DOI: 10.1111/ene.16310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/11/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND AND PURPOSE White matter (WM) damage is the main target of hereditary spastic paraplegia (HSP), but mounting evidence indicates that genotype-specific grey matter (GM) damage is not uncommon. Our aim was to identify and compare brain GM and WM damage patterns in HSP subtypes and investigate how gene expression contributes to these patterns, and explore the relationship between GM and WM damage. METHODS In this prospective single-centre cohort study from 2019 to 2022, HSP patients and controls underwent magnetic resonance imaging evaluations. The alterations of GM and WM patterns were compared between groups by applying a source-based morphometry approach. Spearman rank correlation was used to explore the associations between gene expression and GM atrophy patterns in HSP subtypes. Mediation analysis was conducted to investigate the interplay between GM and WM damage. RESULTS Twenty-one spastic paraplegia type 4 (SPG4) patients (mean age 50.7 years ± 12.0 SD, 15 men), 21 spastic paraplegia type 5 (SPG5) patients (mean age 29.1 years ± 12.8 SD, 14 men) and 42 controls (sex- and age-matched) were evaluated. Compared to controls, SPG4 and SPG5 showed similar WM damage but different GM atrophy patterns. GM atrophy patterns in SPG4 and SPG5 were correlated with corresponding gene expression (ρ = 0.30, p = 0.008, ρ = 0.40, p < 0.001, respectively). Mediation analysis indicated that GM atrophy patterns were mediated by WM damage in HSP. CONCLUSIONS Grey matter atrophy patterns were distinct between SPG4 and SPG5 and were not only secondary to WM damage but also associated with disease-related gene expression. CLINICAL TRIAL REGISTRATION NO NCT04006418.
Collapse
Affiliation(s)
- Yuqing Tu
- Department of RadiologyFirst Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Ying Liu
- Department of RadiologyFirst Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Shuping Fan
- Department of RadiologyFirst Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Jiaqi Weng
- Department of RadiologyFirst Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Mengcheng Li
- Department of RadiologyFirst Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Fan Zhang
- Department of RadiologyFirst Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Ying Fu
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouFujianChina
| | - Jianping Hu
- Department of RadiologyFirst Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| |
Collapse
|
30
|
Park S, Haak KV, Oldham S, Cho H, Byeon K, Park BY, Thomson P, Chen H, Gao W, Xu T, Valk S, Milham MP, Bernhardt B, Di Martino A, Hong SJ. A shifting role of thalamocortical connectivity in the emergence of cortical functional organization. Nat Neurosci 2024; 27:1609-1619. [PMID: 38858608 DOI: 10.1038/s41593-024-01679-3] [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: 04/26/2023] [Accepted: 05/13/2024] [Indexed: 06/12/2024]
Abstract
The cortical patterning principle has been a long-standing question in neuroscience, yet how this translates to macroscale functional specialization in the human brain remains largely unknown. Here we examine age-dependent differences in resting-state thalamocortical connectivity to investigate its role in the emergence of large-scale functional networks during early life, using a primarily cross-sectional but also longitudinal approach. We show that thalamocortical connectivity during infancy reflects an early differentiation of sensorimotor networks and genetically influenced axonal projection. This pattern changes in childhood, when connectivity is established with the salience network, while decoupling externally and internally oriented functional systems. A developmental simulation using generative network models corroborated these findings, demonstrating that thalamic connectivity contributes to developing key features of the mature brain, such as functional segregation and the sensory-association axis, especially across 12-18 years of age. Our study suggests that the thalamus plays an important role in functional specialization during development, with potential implications for studying conditions with compromised internal and external processing.
Collapse
Affiliation(s)
- Shinwon Park
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Korea
- Autism Center, Child Mind Institute, New York, NY, USA
| | - Koen V Haak
- Department of Cognitive Science and Artificial Intelligence, Tilburg School of Humanities and Digital Sciences, Tilburg University, Tilburg, The Netherlands
- Donders Centre for Cognitive Neuroimaging, Donders Institute, Radboud University, Radboud, The Netherlands
| | - Stuart Oldham
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences and Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Hanbyul Cho
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Korea
| | - Kyoungseob Byeon
- Center for Integrative Developing Brain, Child Mind Institute, New York, NY, USA
| | - Bo-Yong Park
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Korea
- Department of Data Science, Inha University, Incheon, South Korea
| | | | - Haitao Chen
- Department of Biomedical Sciences and Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Bioengineering, University of California at Los Angeles, Los Angeles, CA, USA
| | - Wei Gao
- Department of Biomedical Sciences and Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Ting Xu
- Center for Integrative Developing Brain, Child Mind Institute, New York, NY, USA
| | - Sofie Valk
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute of Neuroscience and Medicine (INM-7), Brain and Behavior, Forschungszentrum, Juelich, Germany
| | - Michael P Milham
- Center for the Developing Brain, Child Mind Institute, New York, NY, USA
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Boris Bernhardt
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | | | - Seok-Jun Hong
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Korea.
- Center for the Developing Brain, Child Mind Institute, New York, NY, USA.
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, South Korea.
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, South Korea.
- Department of MetaBioHealth, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
31
|
Shen T, Vogel JW, Van Deerlin VM, Suh E, Dratch L, Phillips JS, Massimo L, Lee EB, Irwin DJ, McMillan CT. Disparate and shared transcriptomic signatures associated with cortical atrophy in genetic bvFTD. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.25.24310894. [PMID: 39211858 PMCID: PMC11361203 DOI: 10.1101/2024.07.25.24310894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cortical atrophy in behavioral variant frontotemporal degeneration (bvFTD) exhibits spatial heterogeneity across genetic subgroups, potentially driven by distinct biological mechanisms. Using an integrative imaging-transcriptomics approach, we identified disparate and shared transcriptomic signatures associated with cortical thickness in C9orf72 , GRN or MAPT -related bvFTD. Genes associated with cortical thinning in GRN -bvFTD were implicated in neurotransmission, further supported by mapping synaptic density maps to cortical thickness maps. Previously identified genes linked to TDP-43 positive neurons were significantly overlapped with genes associated with C9orf72 -bvFTD and GRN -bvFTD, but not MAPT -bvFTD providing specificity for our associations. C9orf72 -bvFTD and GRN -bvFTD shared genes displaying consistent directionality of correlations with cortical thickness, while MAPT -bvFTD displayed more pronounced differences in transcriptomic signatures with opposing directionality. Overall, we identified disparate and shared genes tied to regional vulnerability with increased biological interpretation including overlap with synaptic density maps and pathologically-specific gene expression, illuminating intricate molecular underpinnings contributing to heterogeneities in bvFTD.
Collapse
|
32
|
Jiang Y, Zhou Y, Xie Y, Zhou J, Cai M, Tang J, Liu F, Ma J, Liu H. Functional magnetic resonance imaging alternations in suicide attempts individuals and their association with gene expression. Neuroimage Clin 2024; 43:103645. [PMID: 39059208 PMCID: PMC11326948 DOI: 10.1016/j.nicl.2024.103645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/29/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Functional Magnetic Resonance Imaging (fMRI) has shown brain activity alterations in individuals with a history of attempted suicide (SA) who are diagnosed with depression disorder (DD) or bipolar disorder (BD). However, patterns of spontaneous brain activity and their genetic correlations need further investigation. METHODS A voxel-based meta-analysis of 19 studies including 26 datasets, involving 742 patients with a history of SA and 978 controls (both nonsuicidal patients and healthy controls) was conducted. We examined fMRI changes in SA patients and analyzed the association between these changes and gene expression profiles using data from the Allen Human Brain Atlas by partial least squares regression analysis. RESULTS SA patients demonstrated increased spontaneous brain activity in several brain regions including the bilateral inferior temporal gyrus, hippocampus, fusiform gyrus, and right insula, and decreased activity in areas like the bilateral paracentral lobule and inferior frontal gyrus. Additionally, 5,077 genes were identified, exhibiting expression patterns associated with SA-related fMRI alterations. Functional enrichment analyses demonstrated that these SA-related genes were enriched for biological functions including glutamatergic synapse and mitochondrial structure. Concurrently, specific expression analyses showed that these genes were specifically expressed in the brain tissue, in neurons cells, and during early developmental periods. CONCLUSION Our findings suggest a neurobiological basis for fMRI abnormalities in SA patients with DD or BD, potentially guiding future genetic and therapeutic research.
Collapse
Affiliation(s)
- Yurong Jiang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yujing Zhou
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, 116000 Dalian, Liaoning, China
| | - Yingying Xie
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Junzi Zhou
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Mengjing Cai
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jie Tang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Juanwei Ma
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Huaigui Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
33
|
Almeida FC, Pereira AI, Mendes-Pinto C, Lopes J, Moura J, Sousa JM, Videira G, Samões R, Oliveira TG. MR Imaging Findings in Anti-Leucine-Rich Glioma Inactivated Protein 1 Encephalitis: A Systematic Review and Meta-analysis. AJNR Am J Neuroradiol 2024; 45:977-986. [PMID: 38871367 DOI: 10.3174/ajnr.a8256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/14/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Antibodies against leucine-rich glioma inactivated protein 1 (LGI1) constitute a common form of autoimmune encephalitis. On MR imaging, it may show T2 FLAIR hyperintensities of the medial temporal lobe (T2 FLAIR-MTL), involve the basal ganglia, or be unremarkable. PURPOSE We performed a systematic review and meta-analysis to obtain prevalence estimates of abnormal findings on MR imaging in anti-LGI1 encephalitis. A human brain map of the LGI1 microarray gene expression was derived from the Allen Human Brain Atlas. DATA SOURCES PubMed and Web of Science were searched with the terms "LGI1" and "encephalitis" from inception to April 7, 2022. STUDY SELECTION Thirty-one research publications, encompassing case series and retrospective cohort and case-control studies, with >10 patients with anti-LGI1 encephalitis and MR imaging data were included. DATA ANALYSIS Pooled prevalence estimates were calculated using Freeman-Tukey double-arcsine transformation. Meta-analysis used DerSimonian and Laird random effects models. DATA SYNTHESIS Of 1318 patients in 30 studies, T2 FLAIR-MTL hyperintensities were present in 54% (95% CI, 0.48-0.60; I2 = 76%). Of 394 patients in 13 studies, 27% showed bilateral (95% CI, 0.19-0.36; I2 = 71%) and 24% unilateral T2 FLAIR-MTL abnormalities (95% CI, 0.17-0.32; I2 = 61%). Of 612 patients in 15 studies, basal ganglia abnormalities were present in 10% (95% CI, 0.06-0.15; I2 = 67%). LGI1 expression was highest in the amygdala, hippocampus, and caudate nucleus. LIMITATIONS Only part of the spectrum of MR imaging abnormalities in anti-LGI1 encephalitis could be included in a meta-analysis. MR imaging findings were not the main outcomes in most studies, limiting available information. I2 values ranged from 62% to 76%, representing moderate-to-large heterogeneity. CONCLUSIONS T2 FLAIR-MTL hyperintensities were present in around one-half of patients with anti-LGI1. The prevalence of unilateral and bilateral presentations was similar, suggesting unilaterality should raise the suspicion of this disease in the appropriate clinical context. Around 10% of patients showed basal ganglia abnormalities, indicating that special attention should be given to this region. LGI1 regional expression coincided with the most frequently reported abnormal findings on MR imaging. Regional specificity might be partially determined by expression levels of the target protein.
Collapse
Affiliation(s)
- Francisco C Almeida
- From the Department of Neuroradiology (F.C.A., A.I.P., C.M.-P.), Centro Hospitalar Universitário de Santo António, Porto, Portugal
- Life and Health Sciences Research Institute (F.C.A., T.G.O.), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory (F.C.A., T.G.O.), Braga/Guimarães, Portugal
| | - Ana I Pereira
- From the Department of Neuroradiology (F.C.A., A.I.P., C.M.-P.), Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Catarina Mendes-Pinto
- From the Department of Neuroradiology (F.C.A., A.I.P., C.M.-P.), Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Joana Lopes
- Department of Neurology (J.L., J.M., G.V., R.S.), Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - João Moura
- Department of Neurology (J.L., J.M., G.V., R.S.), Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - José Maria Sousa
- Department of Neuroradiology (J.M.S.), Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Gonçalo Videira
- Department of Neurology (J.L., J.M., G.V., R.S.), Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Raquel Samões
- Department of Neurology (J.L., J.M., G.V., R.S.), Centro Hospitalar Universitário de Santo António, Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine (R.S.), Instituto de Ciências Biomédicas de Abel Salazar da Universidade do Porto, Porto, Portugal
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (F.C.A., T.G.O.), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory (F.C.A., T.G.O.), Braga/Guimarães, Portugal
- Department of Neuroradiology (T.G.O.), Hospital de Braga, Braga, Portugal
| |
Collapse
|
34
|
Xie H, Wang Y, Zhu F, Zhang F, Wu B, Zhao Z, Gan R, Gong Q, Jia Z. Genes associated with cortical thickness alterations in behavioral addiction. Cereb Cortex 2024; 34:bhae298. [PMID: 39051658 DOI: 10.1093/cercor/bhae298] [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: 05/13/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024] Open
Abstract
Behavioral addiction (BA) is a conceptually new addictive phenotype characterized by compulsive reward-seeking behaviors despite adverse consequences. Currently, its underlying neurogenetic mechanism remains unclear. Here, this study aimed to investigate the association between cortical thickness (CTh) and genetic phenotypes in BA. We conducted a systematic search in five databases and extracted gene expression data from the Allen Human Brain Atlas. Meta-analysis of 10 studies (343 addicted individuals and 355 controls) revealed that the BA group showed thinner CTh in the precuneus, postcentral gyrus, orbital-frontal cortex, and dorsolateral prefrontal cortex (P < 0.005). Meta-regression showed that the CTh in the precuneus and postcentral gyrus were negatively associated with the addiction severity (P < 0.0005). More importantly, the CTh phenotype of BA was spatially correlated with the expression of 12 genes (false discovery rate [FDR] < 0.05), and the dopamine D2 receptor had the highest correlation (rho = 0.55). Gene enrichment analysis further revealed that the 12 genes were involved in the biological processes of behavior regulation and response to stimulus (FDR < 0.05). In conclusion, our findings demonstrated the thinner CTh in cognitive control-related brain areas in BA, which could be associated with the expression of genes involving dopamine metabolism and behavior regulation.
Collapse
Affiliation(s)
- Hongsheng Xie
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Yuanyuan Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Fei Zhu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Feifei Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Baolin Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Ziru Zhao
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Ruoqiu Gan
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Qiyong Gong
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, 699 Jinyuan Xi Road, Jimei District, 361021 Xiamen, Fujian, China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| |
Collapse
|
35
|
Vogel JW, Alexander-Bloch AF, Wagstyl K, Bertolero MA, Markello RD, Pines A, Sydnor VJ, Diaz-Papkovich A, Hansen JY, Evans AC, Bernhardt B, Misic B, Satterthwaite TD, Seidlitz J. Deciphering the functional specialization of whole-brain spatiomolecular gradients in the adult brain. Proc Natl Acad Sci U S A 2024; 121:e2219137121. [PMID: 38861593 PMCID: PMC11194492 DOI: 10.1073/pnas.2219137121] [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: 11/08/2022] [Accepted: 04/27/2024] [Indexed: 06/13/2024] Open
Abstract
Cortical arealization arises during neurodevelopment from the confluence of molecular gradients representing patterned expression of morphogens and transcription factors. However, whether similar gradients are maintained in the adult brain remains unknown. Here, we uncover three axes of topographic variation in gene expression in the adult human brain that specifically capture previously identified rostral-caudal, dorsal-ventral, and medial-lateral axes of early developmental patterning. The interaction of these spatiomolecular gradients i) accurately reconstructs the position of brain tissue samples, ii) delineates known functional territories, and iii) can model the topographical variation of diverse cortical features. The spatiomolecular gradients are distinct from canonical cortical axes differentiating the primary sensory cortex from the association cortex, but radiate in parallel with the axes traversed by local field potentials along the cortex. We replicate all three molecular gradients in three independent human datasets as well as two nonhuman primate datasets and find that each gradient shows a distinct developmental trajectory across the lifespan. The gradients are composed of several well-known transcription factors (e.g., PAX6 and SIX3), and a small set of genes shared across gradients are strongly enriched for multiple diseases. Together, these results provide insight into the developmental sculpting of functionally distinct brain regions, governed by three robust transcriptomic axes embedded within brain parenchyma.
Collapse
Affiliation(s)
- Jacob W. Vogel
- Department of Clinical Sciences Malmö, SciLifeLab, Lund University, Lund, Sweden202 13
- Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
| | - Aaron F. Alexander-Bloch
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Penn-Children’s Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA19104
| | - Konrad Wagstyl
- Wellcome Centre for Human Neuroimaging, Institute of Neurology, University College London, LondonWC1N 3AR, United Kingdom
| | - Maxwell A. Bertolero
- Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
| | - Ross D. Markello
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QCH3A 2B4, Canada
| | - Adam Pines
- Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA94305
| | - Valerie J. Sydnor
- Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
| | - Alex Diaz-Papkovich
- Quantitative Life Sciences, McGill University, Montreal, QCH3A 1E3, Canada
- McGill Genome Centre, McGill University, Montreal, QCH3A 0G1, Canada
| | - Justine Y. Hansen
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QCH3A 2B4, Canada
| | - Alan C. Evans
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QCH3A 2B4, Canada
| | - Boris Bernhardt
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QCH3A 2B4, Canada
| | - Bratislav Misic
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QCH3A 2B4, Canada
| | - Theodore D. Satterthwaite
- Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Penn-Children’s Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA19104
| | - Jakob Seidlitz
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Penn-Children’s Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
36
|
Li Z, Ma Q, Deng Y, Rolls ET, Shen C, Li Y, Zhang W, Xiang S, Langley C, Sahakian BJ, Robbins TW, Yu JT, Feng J, Cheng W. Irritable Bowel Syndrome Is Associated With Brain Health by Neuroimaging, Behavioral, Biochemical, and Genetic Analyses. Biol Psychiatry 2024; 95:1122-1132. [PMID: 38199582 DOI: 10.1016/j.biopsych.2023.12.024] [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: 07/10/2023] [Revised: 11/14/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) interacts with psychopathology in a complex way; however, little is known about the underlying brain, biochemical, and genetic mechanisms. METHODS To clarify the phenotypic and genetic associations between IBS and brain health, we performed a comprehensive retrospective cohort study on a large population. Our study included 171,104 participants from the UK Biobank who underwent a thorough assessment of IBS, with the majority also providing neuroimaging, behavioral, biochemical, and genetic information. Multistage linked analyses were conducted, including phenome-wide association analysis, polygenic risk score calculation, and 2-sample Mendelian randomization analysis. RESULTS The phenome-wide association analysis showed that IBS was linked to brain health problems, including anxiety and depression, and poor cognitive performance. Significantly lower brain volumes associated with more severe IBS were found in key areas related to emotional regulation and higher-order cognition, including the medial orbitofrontal cortex/ventromedial prefrontal cortex, anterior insula, anterior and mid-cingulate cortices, dorsolateral prefrontal cortex, and hippocampus. Higher triglycerides, lower high-intensity lipoprotein, and lower platelets were also related (p < 1 × 10-10) to more severe IBS. Finally, Mendelian randomization analyses demonstrated potential causal relationships between IBS and brain health and indicated possible mediating effects of dyslipidemia and inflammation. CONCLUSIONS For the first time, this study provides a comprehensive understanding of the relationship between IBS and brain health phenotypes, integrating perspectives from neuroimaging, behavioral performance, biochemical factors, and genetics, which is of great significance for clinical applications to potentially address brain health impairments in patients with IBS.
Collapse
Affiliation(s)
- Zeyu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qing Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yueting Deng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Edmund T Rolls
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Department of Computer Science, University of Warwick, Coventry, United Kingdom; Oxford Centre for Computational Neuroscience, Oxford, United Kingdom.
| | - Chun Shen
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yuzhu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shitong Xiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Christelle Langley
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Barbara J Sahakian
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Trevor W Robbins
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| | - Jin-Tai Yu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Department of Computer Science, University of Warwick, Coventry, United Kingdom; Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, China.
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, China.
| |
Collapse
|
37
|
Cohen Z, Lau L, Ahmed M, Jack CR, Liu C. Quantitative susceptibility mapping in the brain reflects spatial expression of genes involved in iron homeostasis and myelination. Hum Brain Mapp 2024; 45:e26688. [PMID: 38896001 PMCID: PMC11187871 DOI: 10.1002/hbm.26688] [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: 03/01/2023] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 06/21/2024] Open
Abstract
Quantitative susceptibility mapping (QSM) is an MRI modality used to non-invasively measure iron content in the brain. Iron exhibits a specific anatomically varying pattern of accumulation in the brain across individuals. The highest regions of accumulation are the deep grey nuclei, where iron is stored in paramagnetic molecule ferritin. This form of iron is considered to be what largely contributes to the signal measured by QSM in the deep grey nuclei. It is also known that QSM is affected by diamagnetic myelin contents. Here, we investigate spatial gene expression of iron and myelin related genes, as measured by the Allen Human Brain Atlas, in relation to QSM images of age-matched subjects. We performed multiple linear regressions between gene expression and the average QSM signal within 34 distinct deep grey nuclei regions. Our results show a positive correlation (p < .05, corrected) between expression of ferritin and the QSM signal in deep grey nuclei regions. We repeated the analysis for other genes that encode proteins thought to be involved in the transport and storage of iron in the brain, as well as myelination. In addition to ferritin, our findings demonstrate a positive correlation (p < .05, corrected) between the expression of ferroportin, transferrin, divalent metal transporter 1, several gene markers of myelinating oligodendrocytes, and the QSM signal in deep grey nuclei regions. Our results suggest that the QSM signal reflects both the storage and active transport of iron in the deep grey nuclei regions of the brain.
Collapse
Affiliation(s)
- Zoe Cohen
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Laurance Lau
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Maruf Ahmed
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Clifford R. Jack
- Mayo Foundation for Medical Education and ResearchRochesterMinnesotaUSA
| | - Chunlei Liu
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| |
Collapse
|
38
|
Ganglberger F, Kargl D, Töpfer M, Hernandez-Lallement J, Lawless N, Fernandez-Albert F, Haubensak W, Bühler K. BrainTACO: an explorable multi-scale multi-modal brain transcriptomic and connectivity data resource. Commun Biol 2024; 7:730. [PMID: 38877144 PMCID: PMC11178817 DOI: 10.1038/s42003-024-06355-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/20/2024] [Indexed: 06/16/2024] Open
Abstract
Exploring the relationships between genes and brain circuitry can be accelerated by joint analysis of heterogeneous datasets from 3D imaging data, anatomical data, as well as brain networks at varying scales, resolutions, and modalities. Generating an integrated view, beyond the individual resources' original purpose, requires the fusion of these data to a common space, and a visualization that bridges the gap across scales. However, despite ever expanding datasets, few platforms for integration and exploration of this heterogeneous data exist. To this end, we present the BrainTACO (Brain Transcriptomic And Connectivity Data) resource, a selection of heterogeneous, and multi-scale neurobiological data spatially mapped onto a common, hierarchical reference space, combined via a holistic data integration scheme. To access BrainTACO, we extended BrainTrawler, a web-based visual analytics framework for spatial neurobiological data, with comparative visualizations of multiple resources. This enables gene expression dissection of brain networks with, to the best of our knowledge, an unprecedented coverage and allows for the identification of potential genetic drivers of connectivity in both mice and humans that may contribute to the discovery of dysconnectivity phenotypes. Hence, BrainTACO reduces the need for time-consuming manual data aggregation often required for computational analyses in script-based toolboxes, and supports neuroscientists by directly leveraging the data instead of preparing it.
Collapse
Affiliation(s)
- Florian Ganglberger
- Biomedical Image Informatics, VRVis Research Center, Vienna, Austria
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma, Biberach an der Riss, Germany
| | - Dominic Kargl
- Department of Neuronal Cell Biology, Vienna Medical University, Vienna, Austria
| | - Markus Töpfer
- Biomedical Image Informatics, VRVis Research Center, Vienna, Austria
| | - Julien Hernandez-Lallement
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma, Biberach an der Riss, Germany
| | - Nathan Lawless
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma, Biberach an der Riss, Germany
| | - Francesc Fernandez-Albert
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma, Biberach an der Riss, Germany
| | - Wulf Haubensak
- Department of Neuronal Cell Biology, Vienna Medical University, Vienna, Austria
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Katja Bühler
- Biomedical Image Informatics, VRVis Research Center, Vienna, Austria.
| |
Collapse
|
39
|
Chen G, Wang W, Wu H, Zhao X, Kang X, Ren J, Zhang J, Sun Y, He J, Sun S, Zhong Z, Shang D, Fan M, Cheng J, Zhang D, Su C, Lin J. Disrupted topological properties of structural brain networks present a glutamatergic neuropathophysiology in people with narcolepsy. Sleep 2024; 47:zsae002. [PMID: 38173348 DOI: 10.1093/sleep/zsae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/18/2023] [Indexed: 01/05/2024] Open
Abstract
STUDY OBJECTIVES Growing evidences have documented various abnormalities of the white matter bundles in people with narcolepsy. We sought to evaluate topological properties of brain structural networks, and their association with symptoms and neuropathophysiological features in people with narcolepsy. METHODS Diffusion tensor imaging was conducted for people with narcolepsy (n = 30) and matched healthy controls as well as symptoms assessment. Structural connectivity for each participant was generated to analyze global and regional topological properties and their correlations with narcoleptic features. Further human brain transcriptome was extracted and spatially registered for connectivity vulnerability. Genetic functional enrichment analysis was performed and further clarified using in vivo emission computed tomography data. RESULTS A wide and dramatic decrease in structural connectivities was observed in people with narcolepsy, with descending network degree and global efficiency. These metrics were not only correlated with sleep latency and awakening features, but also reflected alterations of sleep macrostructure in people with narcolepsy. Network-based statistics identified a small hyperenhanced subnetwork of cingulate gyrus that was closely related to rapid eye movement sleep behavior disorder (RBD) in narcolepsy. Further imaging genetics analysis suggested glutamatergic signatures were responsible for the preferential vulnerability of connectivity alterations in people with narcolepsy, while additional PET/SPECT data verified that structural alteration was significantly correlated with metabotropic glutamate receptor 5 (mGlutR5) and N-methyl-D-aspartate receptor (NMDA). CONCLUSIONS People with narcolepsy endured a remarkable decrease in the structural architecture, which was not only closely related to narcolepsy symptoms but also glutamatergic signatures.
Collapse
Affiliation(s)
- Guoyan Chen
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Wen Wang
- Department of Radiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Haoyang Wu
- Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Xianchao Zhao
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Xiaopeng Kang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Jiafeng Ren
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jun Zhang
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yingzhi Sun
- Department of Radiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jiaxiu He
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Shihui Sun
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Zhao Zhong
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Danqing Shang
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Mengmeng Fan
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jinxiang Cheng
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Dan Zhang
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Changjun Su
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jiaji Lin
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
- Department of Radiology, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
40
|
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
|
41
|
Osetrova M, Tkachev A, Mair W, Guijarro Larraz P, Efimova O, Kurochkin I, Stekolshchikova E, Anikanov N, Foo JC, Cazenave-Gassiot A, Mitina A, Ogurtsova P, Guo S, Potashnikova DM, Gulin AA, Vasin AA, Sarycheva A, Vladimirov G, Fedorova M, Kostyukevich Y, Nikolaev E, Wenk MR, Khrameeva EE, Khaitovich P. Lipidome atlas of the adult human brain. Nat Commun 2024; 15:4455. [PMID: 38796479 PMCID: PMC11127996 DOI: 10.1038/s41467-024-48734-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/07/2024] [Indexed: 05/28/2024] Open
Abstract
Lipids are the most abundant but poorly explored components of the human brain. Here, we present a lipidome map of the human brain comprising 75 regions, including 52 neocortical ones. The lipidome composition varies greatly among the brain regions, affecting 93% of the 419 analyzed lipids. These differences reflect the brain's structural characteristics, such as myelin content (345 lipids) and cell type composition (353 lipids), but also functional traits: functional connectivity (76 lipids) and information processing hierarchy (60 lipids). Combining lipid composition and mRNA expression data further enhances functional connectivity association. Biochemically, lipids linked with structural and functional brain features display distinct lipid class distribution, unsaturation extent, and prevalence of omega-3 and omega-6 fatty acid residues. We verified our conclusions by parallel analysis of three adult macaque brains, targeted analysis of 216 lipids, mass spectrometry imaging, and lipidome assessment of sorted murine neurons.
Collapse
Affiliation(s)
- Maria Osetrova
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Anna Tkachev
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Waltraud Mair
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | - Olga Efimova
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Ilia Kurochkin
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | | | - Juat Chin Foo
- Singapore Lipidomics Incubator, Life Sciences Institute and Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore, Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Singapore Lipidomics Incubator, Life Sciences Institute and Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore, Singapore, Singapore
| | | | | | - Song Guo
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Daria M Potashnikova
- Department of Cell Biology and Histology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alexander A Gulin
- N. N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, Moscow, Russia
| | - Alexander A Vasin
- N. N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | | | - Gleb Vladimirov
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | | | - Evgeny Nikolaev
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute and Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore, Singapore, Singapore.
| | | | | |
Collapse
|
42
|
Qin Y, Chen L, Zhu W, Song J, Lin J, Li Y, Zhang J, Song X, Xing T, Guo T, Duan X, Zhang Y, Ruan E, Wang Q, Li B, Yang W, Yin P, Yan XX, Li S, Li XJ, Yang S. TRIM37 is a primate-specific E3 ligase for Huntingtin and accounts for the striatal degeneration in Huntington's disease. SCIENCE ADVANCES 2024; 10:eadl2036. [PMID: 38758800 PMCID: PMC11100560 DOI: 10.1126/sciadv.adl2036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 04/12/2024] [Indexed: 05/19/2024]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease characterized by preferential neuronal loss in the striatum. The mechanism underlying striatal selective neurodegeneration remains unclear, making it difficult to develop effective treatments for HD. In the brains of nonhuman primates, we examined the expression of Huntingtin (HTT), the gene responsible for HD. We found that HTT protein is highly expressed in striatal neurons due to its slow degradation in the striatum. We also identified tripartite motif-containing 37 (TRIM37) as a primate-specific protein that interacts with HTT and is selectively reduced in the primate striatum. TRIM37 promotes the ubiquitination and degradation of mutant HTT (mHTT) in vitro and modulates mHTT aggregation in mouse and monkey brains. Our findings suggest that nonhuman primates are crucial for understanding the mechanisms of human diseases such as HD and support TRIM37 as a potential therapeutic target for treating HD.
Collapse
Affiliation(s)
- Yiyang Qin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Laiqiang Chen
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Wenzhen Zhu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Jiahong Song
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Jingpan Lin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yuwei Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Jiawei Zhang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Xichen Song
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Tingting Xing
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Tingting Guo
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Xuezhi Duan
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yiran Zhang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Eshu Ruan
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Qi Wang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Bang Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Weili Yang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Peng Yin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Su Yang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| |
Collapse
|
43
|
Zhang L, Ding Y, Li T, Li H, Liu F, Li P, Zhao J, Lv D, Lang B, Guo W. Similar imaging changes and their relations to genetic profiles in bipolar disorder across different clinical stages. Psychiatry Res 2024; 335:115868. [PMID: 38554494 DOI: 10.1016/j.psychres.2024.115868] [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: 01/04/2024] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 04/01/2024]
Abstract
Bipolar disorder (BD) across different clinical stages may present shared and distinct changes in brain activity. We aimed to reveal the neuroimaging homogeneity and heterogeneity of BD and its relationship with clinical variables and genetic variations. In present study, we conducted fractional amplitude of low-frequency fluctuations (fALFF), functional connectivity (FC) and genetic neuroimaging association analyses with 32 depressed, 26 manic, 35 euthymic BD patients and 87 healthy controls (HCs). Significant differences were found in the bilateral pre/subgenual anterior cingulate cortex (ACC) across the four groups, and all bipolar patients exhibited decreased fALFF values in the ACC when compared to HCs. Furthermore, positive associations were significantly observed between fALFF values in the pre/subgenual ACC and participants' cognitive functioning. No significant changes were found in ACC-based FC. We identified fALFF-alteration-related genes in BD, with enrichment in biological progress including synaptic and ion transmission. Taken together, abnormal activity in ACC is a characteristic change associated with BD, regardless of specific mood stages, serving as a potential neuroimaging feature in BD patients. Our genetic neuroimaging association analysis highlights possible heterogeneity in biological processes that could be responsible for different clinical stages in BD.
Collapse
Affiliation(s)
- Leyi Zhang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yudan Ding
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Tingting Li
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Huabing Li
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Feng Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Li
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Jingping Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Dongsheng Lv
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Center of Mental Health, Inner Mongolia Autonomous Region, Hohhot 010010, China.
| | - Bing Lang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - 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 410011, Hunan, China.
| |
Collapse
|
44
|
Sartorius AM, Rokicki J, Birkeland S, Bettella F, Barth C, de Lange AMG, Haram M, Shadrin A, Winterton A, Steen NE, Schwarz E, Stein DJ, Andreassen OA, van der Meer D, Westlye LT, Theofanopoulou C, Quintana DS. An evolutionary timeline of the oxytocin signaling pathway. Commun Biol 2024; 7:471. [PMID: 38632466 PMCID: PMC11024182 DOI: 10.1038/s42003-024-06094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
Oxytocin is a neuropeptide associated with both psychological and somatic processes like parturition and social bonding. Although oxytocin homologs have been identified in many species, the evolutionary timeline of the entire oxytocin signaling gene pathway has yet to be described. Using protein sequence similarity searches, microsynteny, and phylostratigraphy, we assigned the genes supporting the oxytocin pathway to different phylostrata based on when we found they likely arose in evolution. We show that the majority (64%) of genes in the pathway are 'modern'. Most of the modern genes evolved around the emergence of vertebrates or jawed vertebrates (540 - 530 million years ago, 'mya'), including OXTR, OXT and CD38. Of those, 45% were under positive selection at some point during vertebrate evolution. We also found that 18% of the genes in the oxytocin pathway are 'ancient', meaning their emergence dates back to cellular organisms and opisthokonta (3500-1100 mya). The remaining genes (18%) that evolved after ancient and before modern genes were classified as 'medium-aged'. Functional analyses revealed that, in humans, medium-aged oxytocin pathway genes are highly expressed in contractile organs, while modern genes in the oxytocin pathway are primarily expressed in the brain and muscle tissue.
Collapse
Affiliation(s)
- Alina M Sartorius
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Jaroslav Rokicki
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Centre of Research and Education in Forensic Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Siri Birkeland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
- Natural History Museum, University of Oslo, Oslo, Norway
| | - Francesco Bettella
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| | - Claudia Barth
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Ann-Marie G de Lange
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Marit Haram
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Mental Health and Suicide, Norwegian Institute of Public Health, Oslo, Norway
| | - Alexey Shadrin
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Adriano Winterton
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Nils Eiel Steen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Emanuel Schwarz
- Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dan J Stein
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Dennis van der Meer
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Daniel S Quintana
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway.
- Department of Psychology, University of Oslo, Oslo, Norway.
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway.
- NevSom, Department of Rare Disorders, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
45
|
Shen Y, Shao M, Hao ZZ, Huang M, Xu N, Liu S. Multimodal Nature of the Single-cell Primate Brain Atlas: Morphology, Transcriptome, Electrophysiology, and Connectivity. Neurosci Bull 2024; 40:517-532. [PMID: 38194157 PMCID: PMC11003949 DOI: 10.1007/s12264-023-01160-4] [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: 03/22/2023] [Accepted: 09/23/2023] [Indexed: 01/10/2024] Open
Abstract
Primates exhibit complex brain structures that augment cognitive function. The neocortex fulfills high-cognitive functions through billions of connected neurons. These neurons have distinct transcriptomic, morphological, and electrophysiological properties, and their connectivity principles vary. These features endow the primate brain atlas with a multimodal nature. The recent integration of next-generation sequencing with modified patch-clamp techniques is revolutionizing the way to census the primate neocortex, enabling a multimodal neuronal atlas to be established in great detail: (1) single-cell/single-nucleus RNA-seq technology establishes high-throughput transcriptomic references, covering all major transcriptomic cell types; (2) patch-seq links the morphological and electrophysiological features to the transcriptomic reference; (3) multicell patch-clamp delineates the principles of local connectivity. Here, we review the applications of these technologies in the primate neocortex and discuss the current advances and tentative gaps for a comprehensive understanding of the primate neocortex.
Collapse
Affiliation(s)
- Yuhui Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Mingting Shao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Mengyao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Nana Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, 510080, China.
| |
Collapse
|
46
|
Cai M, Ji Y, Zhao Q, Xue H, Sun Z, Wang H, Zhang Y, Chen Y, Zhao Y, Zhang Y, Lei M, Wang C, Zhuo C, Liu N, Liu H, Liu F. Homotopic functional connectivity disruptions in schizophrenia and their associated gene expression. Neuroimage 2024; 289:120551. [PMID: 38382862 DOI: 10.1016/j.neuroimage.2024.120551] [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: 12/11/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024] Open
Abstract
It has been revealed that abnormal voxel-mirrored homotopic connectivity (VMHC) is present in patients with schizophrenia, yet there are inconsistencies in the relevant findings. Moreover, little is known about their association with brain gene expression profiles. In this study, transcription-neuroimaging association analyses using gene expression data from Allen Human Brain Atlas and case-control VMHC differences from both the discovery (meta-analysis, including 9 studies with a total of 386 patients and 357 controls) and replication (separate group-level comparisons within two datasets, including a total of 258 patients and 287 controls) phases were performed to identify genes associated with VMHC alterations. Enrichment analyses were conducted to characterize the biological functions and specific expression of identified genes, and Neurosynth decoding analysis was performed to examine the correlation between cognitive-related processes and VMHC alterations in schizophrenia. In the discovery and replication phases, patients with schizophrenia exhibited consistent VMHC changes compared to controls, which were correlated with a series of cognitive-related processes; meta-regression analysis revealed that illness duration was negatively correlated with VMHC abnormalities in the cerebellum and postcentral/precentral gyrus. The abnormal VMHC patterns were stably correlated with 1287 genes enriched for fundamental biological processes like regulation of cell communication, nervous system development, and cell communication. In addition, these genes were overexpressed in astrocytes and immune cells, enriched in extensive cortical regions and wide developmental time windows. The present findings may contribute to a more comprehensive understanding of the molecular mechanisms underlying VMHC alterations in patients with schizophrenia.
Collapse
Affiliation(s)
- Mengjing Cai
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yuan Ji
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qiyu Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hui Xue
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - He Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yijing Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yayuan Chen
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yao Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yujie Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Minghuan Lei
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chunyang Wang
- Department of Scientific Research, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chuanjun Zhuo
- Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PGNP_Lab), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Nana Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Huaigui Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
47
|
Zheng C, Xiao X, Zhao W, Yang Z, Guo S. Functional brain network controllability dysfunction in Alzheimer's disease and its relationship with cognition and gene expression profiling. J Neural Eng 2024; 21:026018. [PMID: 38502960 DOI: 10.1088/1741-2552/ad357e] [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: 09/11/2023] [Accepted: 03/19/2024] [Indexed: 03/21/2024]
Abstract
Objective. In recent studies, network control theory has been applied to clarify transitions between brain states, emphasizing the significance of assessing the controllability of brain networks in facilitating transitions from one state to another. Despite these advancements, the potential alterations in functional network controllability associated with Alzheimer's disease (AD), along with the underlying genetic mechanisms responsible for these alterations, remain unclear.Approach. We conducted a comparative analysis of functional network controllability measures between patients with AD (n= 64) and matched normal controls (NCs,n= 64). We investigated the association between altered controllability measures and cognitive function in AD. Additionally, we conducted correlation analyses in conjunction with the Allen Human Brain Atlas to identify genes whose expression was correlated with changes in functional network controllability in AD, followed by a set of analyses on the functional features of the identified genes.Main results. In comparison to NCs, patients with AD exhibited a reduction in average controllability, predominantly within the default mode network (DMN) (63% of parcellations), and an increase in average controllability within the limbic (LIM) network (33% of parcellations). Conversely, AD patients displayed a decrease in modal controllability within the LIM network (27% of parcellations) and an increase in modal controllability within the DMN (80% of parcellations). In AD patients, a significant positive correlation was found between the average controllability of the salience network and the mini-mental state examination scores. The changes in controllability measures exhibited spatial correlation with transcriptome profiles. The significant genes identified exhibited enrichment in neurobiologically relevant pathways and demonstrated preferential expression in various tissues, cell types, and developmental periods.Significance. Our findings have the potential to offer new insights into the genetic mechanisms underlying alterations in the controllability of functional networks in AD. Additionally, these results offered perspectives for a deeper understanding of the pathogenesis and the development of therapeutic strategies for AD.
Collapse
Affiliation(s)
- Chuchu Zheng
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| | - Xiaoxia Xiao
- School of Informatics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, People's Republic of China
| | - Wei Zhao
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| | - Zeyu Yang
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| | - Shuixia Guo
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| |
Collapse
|
48
|
Chu C, Li W, Shi W, Wang H, Wang J, Liu Y, Liu B, Elmenhorst D, Eickhoff SB, Fan L, Jiang T. Co-representation of Functional Brain Networks Is Shaped by Cortical Myeloarchitecture and Reveals Individual Behavioral Ability. J Neurosci 2024; 44:e0856232024. [PMID: 38290847 PMCID: PMC10977027 DOI: 10.1523/jneurosci.0856-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 01/16/2024] [Accepted: 01/20/2024] [Indexed: 02/01/2024] Open
Abstract
Large-scale functional networks are spatially distributed in the human brain. Despite recent progress in differentiating their functional roles, how the brain navigates the spatial coordination among them and the biological relevance of this coordination is still not fully understood. Capitalizing on canonical individualized networks derived from functional MRI data, we proposed a new concept, that is, co-representation of functional brain networks, to delineate the spatial coordination among them. To further quantify the co-representation pattern, we defined two indexes, that is, the co-representation specificity (CoRS) and intensity (CoRI), for separately measuring the extent of specific and average expression of functional networks at each brain location by using the data from both sexes. We found that the identified pattern of co-representation was anchored by cortical regions with three types of cytoarchitectural classes along a sensory-fugal axis, including, at the first end, primary (idiotypic) regions showing high CoRS, at the second end, heteromodal regions showing low CoRS and high CoRI, at the third end, paralimbic regions showing low CoRI. Importantly, we demonstrated the critical role of myeloarchitecture in sculpting the spatial distribution of co-representation by assessing the association with the myelin-related neuroanatomical and transcriptomic profiles. Furthermore, the significance of manifesting the co-representation was revealed in its prediction of individual behavioral ability. Our findings indicated that the spatial coordination among functional networks was built upon an anatomically configured blueprint to facilitate neural information processing, while advancing our understanding of the topographical organization of the brain by emphasizing the assembly of functional networks.
Collapse
Affiliation(s)
- Congying Chu
- Brainnetome Center, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Wen Li
- Brainnetome Center, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiyang Shi
- Brainnetome Center, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Haiyan Wang
- Brainnetome Center, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jiaojian Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing 100876, China
| | - Bing Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - David Elmenhorst
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Forschungszentrum Jülich, Jülich 52428, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University, Düsseldorf 40204, Germany
| | - Lingzhong Fan
- Brainnetome Center, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100049, China
| | - Tianzi Jiang
- Brainnetome Center, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100049, China
- Xiaoxiang Institute for Brain Health and Yongzhou Central Hospital, Yongzhou 425000, Hunan Province, China
| |
Collapse
|
49
|
Tissink EP, Shadrin AA, van der Meer D, Parker N, Hindley G, Roelfs D, Frei O, Fan CC, Nagel M, Nærland T, Budisteanu M, Djurovic S, Westlye LT, van den Heuvel MP, Posthuma D, Kaufmann T, Dale AM, Andreassen OA. Abundant pleiotropy across neuroimaging modalities identified through a multivariate genome-wide association study. Nat Commun 2024; 15:2655. [PMID: 38531894 DOI: 10.1038/s41467-024-46817-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Genetic pleiotropy is abundant across spatially distributed brain characteristics derived from one neuroimaging modality (e.g. structural, functional or diffusion magnetic resonance imaging [MRI]). A better understanding of pleiotropy across modalities could inform us on the integration of brain function, micro- and macrostructure. Here we show extensive genetic overlap across neuroimaging modalities at a locus and gene level in the UK Biobank (N = 34,029) and ABCD Study (N = 8607). When jointly analysing phenotypes derived from structural, functional and diffusion MRI in a genome-wide association study (GWAS) with the Multivariate Omnibus Statistical Test (MOSTest), we boost the discovery of loci and genes beyond previously identified effects for each modality individually. Cross-modality genes are involved in fundamental biological processes and predominantly expressed during prenatal brain development. We additionally boost prediction of psychiatric disorders by conditioning independent GWAS on our multimodal multivariate GWAS. These findings shed light on the shared genetic mechanisms underlying variation in brain morphology, functional connectivity, and tissue composition.
Collapse
Affiliation(s)
- E P Tissink
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 HV, Amsterdam, The Netherlands.
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
| | - A A Shadrin
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway
| | - D van der Meer
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - N Parker
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway
| | - G Hindley
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway
- Psychosis Studies, Institute of Psychiatry, Psychology and Neurosciences, King's College London, 16 De Crespigny Park, London, SE5 8AB, United Kingdom
| | - D Roelfs
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway
| | - O Frei
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway
| | - C C Fan
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, 92037, USA
| | - M Nagel
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 HV, Amsterdam, The Netherlands
| | - T Nærland
- K.G. Jebsen Centre for Neurodevelopmental disorders, Division of Paediatric Medicine, Institute of Clinical Medicine, University of Oslo, Building 31, Oslo, Norway
| | - M Budisteanu
- Prof. Dr. Alex Obregia Clinical Hospital of Psychiatry, Bucharest, Romania
- "Victor Babes" National Institute of Pathology, Bucharest, Romania
| | - S Djurovic
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental disorders, Division of Paediatric Medicine, Institute of Clinical Medicine, University of Oslo, Building 31, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - L T Westlye
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental disorders, Division of Paediatric Medicine, Institute of Clinical Medicine, University of Oslo, Building 31, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - M P van den Heuvel
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 HV, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, Amsterdam Neuroscience, VU University Medical Centre, Amsterdam, The Netherlands
| | - D Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 HV, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, Amsterdam Neuroscience, VU University Medical Centre, Amsterdam, The Netherlands
| | - T Kaufmann
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
| | - A M Dale
- Department of Radiology, University of California San Diego, La Jolla, CA, 92037, USA
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, 92037, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92037, USA
| | - O A Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Building 48, Oslo, Norway.
- K.G. Jebsen Centre for Neurodevelopmental disorders, Division of Paediatric Medicine, Institute of Clinical Medicine, University of Oslo, Building 31, Oslo, Norway.
| |
Collapse
|
50
|
Ji Y, Cai M, Zhou Y, Ma J, Zhang Y, Zhang Z, Zhao J, Wang Y, Jiang Y, Zhai Y, Xu J, Lei M, Xu Q, Liu H, Liu F. Exploring functional dysconnectivity in schizophrenia: alterations in eigenvector centrality mapping and insights into related genes from transcriptional profiles. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:37. [PMID: 38491019 PMCID: PMC10943118 DOI: 10.1038/s41537-024-00457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Schizophrenia is a mental health disorder characterized by functional dysconnectivity. Eigenvector centrality mapping (ECM) has been employed to investigate alterations in functional connectivity in schizophrenia, yet the results lack consistency, and the genetic mechanisms underlying these changes remain unclear. In this study, whole-brain voxel-wise ECM analyses were conducted on resting-state functional magnetic resonance imaging data. A cohort of 91 patients with schizophrenia and 91 matched healthy controls were included during the discovery stage. Additionally, in the replication stage, 153 individuals with schizophrenia and 182 healthy individuals participated. Subsequently, a comprehensive analysis was performed using an independent transcriptional database derived from six postmortem healthy adult brains to explore potential genetic factors influencing the observed functional dysconnectivity, and to investigate the roles of identified genes in neural processes and pathways. The results revealed significant and reliable alterations in the ECM across multiple brain regions in schizophrenia. Specifically, there was a significant decrease in ECM in the bilateral superior and middle temporal gyrus, and an increase in the bilateral thalamus in both the discovery and replication stages. Furthermore, transcriptional analysis revealed 420 genes whose expression patterns were related to changes in ECM, and these genes were enriched mainly in biological processes associated with synaptic signaling and transmission. Together, this study enhances our knowledge of the neural processes and pathways involved in schizophrenia, shedding light on the genetic factors that may be linked to functional dysconnectivity in this disorder.
Collapse
Affiliation(s)
- Yuan Ji
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengjing Cai
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yujing Zhou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Juanwei Ma
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yijing Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhihui Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiaxuan Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yurong Jiang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Zhai
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinglei Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Minghuan Lei
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China.
| | - Huaigui Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China.
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|