1
|
Li X, Zeng J, Liu N, Yang C, Tao B, Sun H, Gong Q, Zhang W, Li CSR, Lui S. Progressive alterations of resting-state hypothalamic dysconnectivity in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111127. [PMID: 39181307 DOI: 10.1016/j.pnpbp.2024.111127] [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: 04/15/2024] [Revised: 08/07/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The hypothalamus may be involved in the pathogenesis of schizophrenia. Investigating hypothalamus dysfunction in schizophrenia and probing how it is related to symptoms and responds to antipsychotic medication is crucial for understanding the potential mechanism of hypothalamus dysfunction under the long-term illness. METHODS We recruited 216 patients with schizophrenia, including 140 antipsychotic-naïve first-episode patients (FES, including 44 patients with 1-year follow-up data), 76 chronically treated schizophrenia (CTS), and 210 healthy controls (HC). Hypothalamic seed-based functional connectivity (FC) was calculated and compared among the FES, CTS, and HC groups using analysis of covariance. Exploratory analysis was conducted between the FES patients at baseline and after 1-year follow-up. Significantly altered hypothalamic FCs were then related to clinical symptomology, while age- and illness-related regression analyses were also conducted and compared between diagnostic groups. RESULTS The FES patients showed decreased hypothalamic FCs with the midbrain and right thalamus, whereas the CTS patients showed more severe decreased hypothalamic FCs with the midbrain, right thalamus, left putamen, right caudate, and bilateral anterior cingulate cortex compared to HCs. These abnormalities were not correlated to the symptomology or illness duration, or not reversed by the antipsychotic treatment. Age-related hypothalamic FC decrease was also identified in the abovementioned regions, and a faster age-related decline of the hypothalamic FC was observed with the left putamen and bilateral anterior cingulate cortex. CONCLUSION Age-related hypothalamic FC decrease extends the functional alterations that characterize the neurodegenerative nature of schizophrenia. Future studies are required to further probe the hormonal or endocrinal underpinnings of such alterations and trace the precise progressive trajectories.
Collapse
Affiliation(s)
- Xing Li
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China; Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Jiaxin Zeng
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China; Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Naici Liu
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China; Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Chengmin Yang
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China; Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Bo Tao
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China; Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Hui Sun
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China; Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Qiyong Gong
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China; Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Wenjing Zhang
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China; Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China.
| | - Chiang-Shan R Li
- Departments of Psychiatry and of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
| | - Su Lui
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China; Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China.
| |
Collapse
|
2
|
Gao W, Chen Y, Cui D, Zhu C, Jiao Q, Su L, Lu S, Yang R. Alterations of subcortical structure volume in pediatric bipolar disorder patients with manic or depressive first-episode. BMC Psychiatry 2024; 24:762. [PMID: 39487398 PMCID: PMC11531125 DOI: 10.1186/s12888-024-06208-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Bipolar disorder may begin as depression or mania, which can affect the treatment and prognosis. The physiological and pathological differences among pediatric bipolar disorder (PBD) patients with different onset symptoms are not clear. The aims of the present study were to investigate subcortical structural alterations in PBD patients with first-episode depressive (PBD-FED) and first-episode manic (PBD-FEM). METHODS A total of 59 individuals including 28 PBD-FED, 13 PBD-FEM, and 18 healthy controls (HCs) underwent high-resolution structural magnetic resonance scans. FreeSurfer 7.2 was used to detect changes in subcortical volumes. Simultaneously, thalamic, hippocampal, and amygdala subregion volumes were compared between the three groups. RESULTS Analysis of covariance controlling for age, sex, education, and estimated intracranial volume shows third and fourth ventricle enlargement in patients with PBD. Compared with the PBD-FED and HCs, the PBD-FEM group had reduced gray matter volume in the left thalamus, bilateral hippocampus, and right amygdala. Subsequent subregion analyses showed right cortico-amygdaloid transient, bilateral accessory-basal nucleus, left hippocampal tail, right hippocampal head, and body volume reduction in the PBD-FEM group. CONCLUSIONS The present findings provided evidence of decreased subcortical structure in PBD-FEM patients, which might present its trait feature.
Collapse
Affiliation(s)
- Weijia Gao
- Department of Child Psychology, The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, National Children's Regional Medical Center, No. 3333 Binsheng Road, Hangzhou, 310003, Zhejiang, China
| | - Yue Chen
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Key Laboratory of Precision Psychiatry, Zhejiang Engineering Center for Mathematical Mental Health, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
- Faculty of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dong Cui
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shangdong, China
| | - Ce Zhu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Key Laboratory of Precision Psychiatry, Zhejiang Engineering Center for Mathematical Mental Health, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
- Faculty of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Psychiatry, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Qing Jiao
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shangdong, China
| | - Linyan Su
- Mental Health Institute, Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital of Central South University, National Technology Institute of Psychiatry, Changsha, Hunan, China
| | - Shaojia Lu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Key Laboratory of Precision Psychiatry, Zhejiang Engineering Center for Mathematical Mental Health, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Rongwang Yang
- Department of Child Psychology, The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, National Children's Regional Medical Center, No. 3333 Binsheng Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
3
|
Saglam Y, Ermis C, Takir S, Oz A, Hamid R, Kose H, Bas A, Karacetin G. The Contribution of Explainable Machine Learning Algorithms Using ROI-based Brain Surface Morphology Parameters in Distinguishing Early-onset Schizophrenia From Bipolar Disorder. Acad Radiol 2024; 31:3597-3604. [PMID: 38704285 DOI: 10.1016/j.acra.2024.04.013] [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: 12/10/2023] [Revised: 02/25/2024] [Accepted: 04/11/2024] [Indexed: 05/06/2024]
Abstract
RATIONALE AND OBJECTIVES To differentiate early-onset schizophrenia (EOS) from early-onset bipolar disorder (EBD) using surface-based morphometry measurements and brain volumes using machine learning (ML) algorithms. METHOD High-resolution T1-weighted images were obtained to measure cortical thickness (CT), gyrification, gyrification index (GI), sulcal depth (SD), fractal dimension (FD), and brain volumes. After the feature selection step, ML classifiers were applied for each feature set and the combination of them. The SHapley Additive exPlanations (SHAP) technique was implemented to interpret the contribution of each feature. FINDINGS 144 adolescents (16.2 ± 1.4 years, female=39%) with EOS (n = 81) and EBD (n = 63) were included. The Adaptive Boosting (AdaBoost) algorithm had the highest accuracy (82.75%) in the whole dataset that includes all variables from Destrieux atlas. The best-performing algorithms were K-nearest neighbors (KNN) for FD subset, support vector machine (SVM) for SD subset, and AdaBoost for GI subset. The KNN algorithm had the highest accuracy (accuracy=79.31%) in the whole dataset from the Desikan-Killiany-Tourville atlas. CONCLUSION This study demonstrates the use of ML in the differential diagnosis of EOS and EBD using surface-based morphometry measurements. Future studies could focus on multicenter data for the validation of these results.
Collapse
Affiliation(s)
- Yesim Saglam
- Department of Child and Adolescent Psychiatry, University of Health Sciences, Bakirkoy Prof Dr Mazhar Osman Research and Training Hospital for Psychiatry, Neurology and Neurosurgery, Istanbul, Turkey.
| | - Cagatay Ermis
- Queen Silvia Children's Hospital, Department of Child Psychiatry, Gothenburg, Sweden
| | - Seyma Takir
- Department of Artificial Intelligence and Data Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Ahmet Oz
- Department of Radiology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Rauf Hamid
- Department of Radiology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Hatice Kose
- Department of Artificial Intelligence and Data Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Ahmet Bas
- Department of Radiology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gul Karacetin
- Department of Child and Adolescent Psychiatry, University of Health Sciences, Bakirkoy Prof Dr Mazhar Osman Research and Training Hospital for Psychiatry, Neurology and Neurosurgery, Istanbul, Turkey
| |
Collapse
|
4
|
Cheng Y, Cai H, Liu S, Yang Y, Pan S, Zhang Y, Mo F, Yu Y, Zhu J. Brain Network Localization of Gray Matter Atrophy and Neurocognitive and Social Cognitive Dysfunction in Schizophrenia. Biol Psychiatry 2024:S0006-3223(24)01489-6. [PMID: 39103010 DOI: 10.1016/j.biopsych.2024.07.021] [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: 05/13/2024] [Revised: 07/13/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Numerous studies have established the presence of gray matter atrophy and brain activation abnormalities during neurocognitive and social cognitive tasks in schizophrenia. Despite a growing consensus that diseases localize better to distributed brain networks than individual anatomical regions, relatively few studies have examined brain network localization of gray matter atrophy and neurocognitive and social cognitive dysfunction in schizophrenia. METHODS To address this gap, we initially identified brain locations of structural and functional abnormalities in schizophrenia from 301 published neuroimaging studies with 8712 individuals with schizophrenia and 9275 healthy control participants. By applying novel functional connectivity network mapping to large-scale resting-state functional magnetic resonance imaging datasets, we mapped these affected brain locations to 3 brain abnormality networks of schizophrenia. RESULTS The gray matter atrophy network of schizophrenia comprised a broadly distributed set of brain areas predominantly implicating the ventral attention, somatomotor, and default networks. The neurocognitive dysfunction network was also composed of widespread brain areas primarily involving the frontoparietal and default networks. By contrast, the social cognitive dysfunction network consisted of circumscribed brain regions mainly implicating the default, subcortical, and visual networks. CONCLUSIONS Our findings suggest shared and unique brain network substrates of gray matter atrophy and neurocognitive and social cognitive dysfunction in schizophrenia, which may not only refine the understanding of disease neuropathology from a network perspective but may also contribute to more targeted and effective treatments for impairments in different cognitive domains in schizophrenia.
Collapse
Affiliation(s)
- Yan Cheng
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China
| | - Huanhuan Cai
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China
| | - Siyu Liu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China
| | - Yang Yang
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China
| | - Shan Pan
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China
| | - Yongqi Zhang
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China
| | - Fan Mo
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China
| | - Yongqiang Yu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China.
| | - Jiajia Zhu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China.
| |
Collapse
|
5
|
Li X, Wei W, Wang Q, Deng W, Li M, Ma X, Zeng J, Zhao L, Guo W, Hall MH, Li T. Identify Potential Causal Relationships Between Cortical Thickness, Mismatch Negativity, Neurocognition, and Psychosocial Functioning in Drug-Naïve First-Episode Psychosis Patients. Schizophr Bull 2024; 50:827-838. [PMID: 38635296 PMCID: PMC11283193 DOI: 10.1093/schbul/sbae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
BACKGROUND Cortical thickness (CT) alterations, mismatch negativity (MMN) reductions, and cognitive deficits are robust findings in first-episode psychosis (FEP). However, most studies focused on medicated patients, leaving gaps in our understanding of the interrelationships between CT, MMN, neurocognition, and psychosocial functioning in unmedicated FEP. This study aimed to employ multiple mediation analysis to investigate potential pathways among these variables in unmedicated drug-naïve FEP. METHODS We enrolled 28 drug-naïve FEP and 34 age and sex-matched healthy controls. Clinical symptoms, neurocognition, psychosocial functioning, auditory duration MMN, and T1 structural magnetic resonance imaging data were collected. We measured CT in the superior temporal gyrus (STG), a primary MMN-generating region. RESULTS We found a significant negative correlation between MMN amplitude and bilateral CT of STG (CT_STG) in FEP (left: r = -.709, P < .001; right: r = -.612, P = .008). Multiple mediation models revealed that a thinner left STG cortex affected functioning through both direct (24.66%) and indirect effects (75.34%). In contrast, the effects of the right CT_STG on functioning were mainly mediated through MMN and neurocognitive pathways. CONCLUSIONS Bilateral CT_STG showed significant association with MMN, and MMN plays a mediating role between CT and cognition. Both MMN alone and its interaction with cognition mediated the effects of structural alterations on psychosocial function. The decline in overall function in FEP may stem from decreased CT_STG, leading to subsequent MMN deficits and neurocognitive dysfunction. These findings underline the crucial role of MMN in elucidating how subtle structural alterations can impact neurocognition and psychosocial function in FEP.
Collapse
Affiliation(s)
- Xiaojing Li
- Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Wei Wei
- Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Qiang Wang
- Mental Health Center and Psychiatric Laboratory, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Wei Deng
- Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Mingli Li
- Mental Health Center and Psychiatric Laboratory, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiaohong Ma
- Mental Health Center and Psychiatric Laboratory, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jinkun Zeng
- Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liansheng Zhao
- Mental Health Center and Psychiatric Laboratory, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Wanjun Guo
- Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Mei-Hua Hall
- Psychosis Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Tao Li
- Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
6
|
Rodrigue AL, Hayes RA, Waite E, Corcoran M, Glahn DC, Jalbrzikowski M. Multimodal Neuroimaging Summary Scores as Neurobiological Markers of Psychosis. Schizophr Bull 2024; 50:792-803. [PMID: 37844289 PMCID: PMC11283202 DOI: 10.1093/schbul/sbad149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
BACKGROUND AND HYPOTHESIS Structural brain alterations are well-established features of schizophrenia but they do not effectively predict disease/disease risk. Similar to polygenic risk scores in genetics, we integrated multifactorial aspects of brain structure into a summary "Neuroscore" and examined its potential as a marker of disease. STUDY DESIGN We extracted measures from T1-weighted scans and diffusion tensor imaging (DTI) models from three studies with schizophrenia and healthy individuals. We calculated individual-level summary scores (Neuroscores) for T1-weighted and DTI measures and a combined score (Multimodal Neuroscore-MM). We assessed each score's ability to differentiate schizophrenia cases from controls and its relationship to clinical symptomatology, intelligence quotient (IQ), and medication dosage. We assessed Neuroscore specificity by performing all analyses in a more inclusive psychosis sample and by using scores generated from MDD effect sizes. STUDY RESULTS All Neuroscores significantly differentiated schizophrenia cases from controls (T1 d = 0.56, DTI d = 0.29, MM d = 0.64) to a greater degree than individual brain regions. Higher Neuroscores (ie, increased liability) were associated with lower IQ (T1 β = -0.26, DTI β = -0.15, MM β = -0.30). Higher T1-weighted Neuroscores were associated with higher positive and negative symptom severity (Positive β = 0.21, Negative β = 0.16); Higher Multimodal Neuroscores were associated with higher positive symptom severity (β = 0.30). SZ Neuroscores outperformed MDD Neuroscores in predicting IQ (T1: z = 3.5, q = 0.0007; MM: z = 1.8, q = 0.05). CONCLUSIONS Neuroscores are a step toward leveraging widespread structural brain alterations in psychosis to identify robust neurobiological markers of disease. Future studies will assess ways to improve neuroscore calculation, including developing the optimal methods to calculate neuroscores and considering disorder overlap.
Collapse
Affiliation(s)
- Amanda L Rodrigue
- Department of Psychiatry, Boston Children’s Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Rebecca A Hayes
- Department of Psychiatry, Boston Children’s Hospital, Boston, MA, USA
| | - Emma Waite
- Department of Psychiatry, Boston Children’s Hospital, Boston, MA, USA
| | - Mary Corcoran
- Department of Psychiatry, Boston Children’s Hospital, Boston, MA, USA
| | - David C Glahn
- Department of Psychiatry, Boston Children’s Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, USA
| | - Maria Jalbrzikowski
- Department of Psychiatry, Boston Children’s Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Dusi N, Esposito CM, Delvecchio G, Prunas C, Brambilla P. Case report and systematic review of cerebellar vermis alterations in psychosis. Int Clin Psychopharmacol 2024; 39:223-231. [PMID: 38266159 PMCID: PMC11136271 DOI: 10.1097/yic.0000000000000535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
INTRODUCTION Cerebellar alterations, including both volumetric changes in the cerebellar vermis and dysfunctions of the corticocerebellar connections, have been documented in psychotic disorders. Starting from the clinical observation of a bipolar patient with cerebellar hypoplasia, the purpose of this review is to summarize the data in the literature about the association between hypoplasia of the cerebellar vermis and psychotic disorders [schizophrenia (SCZ) and bipolar disorder (BD)]. METHODS A bibliographic search on PubMed has been conducted, and 18 articles were finally included in the review: five used patients with BD, 12 patients with SCZ and one subject at psychotic risk. RESULTS For SCZ patients and subjects at psychotic risk, the results of most of the reviewed studies seem to suggest a gray matter volume reduction coupled with an increase in white matter volumes in the cerebellar vermis, compared to healthy controls. Instead, the results of the studies on BD patients are more heterogeneous with evidence showing a reduction, no difference or even an increase in cerebellar vermis volume compared to healthy controls. CONCLUSIONS From the results of the reviewed studies, a possible correlation emerged between cerebellar vermis hypoplasia and psychotic disorders, especially SCZ, ultimately supporting the hypothesis of psychotic disorders as neurodevelopmental disorders.
Collapse
Affiliation(s)
- Nicola Dusi
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan
| | | | - Giuseppe Delvecchio
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan
| | - Cecilia Prunas
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan
- Department of Pathophisiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
8
|
Krug A, Stein F, David FS, Schmitt S, Brosch K, Pfarr JK, Ringwald KG, Meller T, Thomas-Odenthal F, Meinert S, Thiel K, Winter A, Waltemate L, Lemke H, Grotegerd D, Opel N, Repple J, Hahn T, Streit F, Witt SH, Rietschel M, Andlauer TFM, Nöthen MM, Philipsen A, Nenadić I, Dannlowski U, Kircher T, Forstner AJ. Factor analysis of lifetime psychopathology and its brain morphometric and genetic correlates in a transdiagnostic sample. Transl Psychiatry 2024; 14:235. [PMID: 38830892 PMCID: PMC11148082 DOI: 10.1038/s41398-024-02936-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
There is a lack of knowledge regarding the relationship between proneness to dimensional psychopathological syndromes and the underlying pathogenesis across major psychiatric disorders, i.e., Major Depressive Disorder (MDD), Bipolar Disorder (BD), Schizoaffective Disorder (SZA), and Schizophrenia (SZ). Lifetime psychopathology was assessed using the OPerational CRITeria (OPCRIT) system in 1,038 patients meeting DSM-IV-TR criteria for MDD, BD, SZ, or SZA. The cohort was split into two samples for exploratory and confirmatory factor analyses. All patients were scanned with 3-T MRI, and data was analyzed with the CAT-12 toolbox in SPM12. Psychopathological factor scores were correlated with gray matter volume (GMV) and cortical thickness (CT). Finally, factor scores were used for exploratory genetic analyses including genome-wide association studies (GWAS) and polygenic risk score (PRS) association analyses. Three factors (paranoid-hallucinatory syndrome, PHS; mania, MA; depression, DEP) were identified and cross-validated. PHS was negatively correlated with four GMV clusters comprising parts of the hippocampus, amygdala, angular, middle occipital, and middle frontal gyri. PHS was also negatively associated with the bilateral superior temporal, left parietal operculum, and right angular gyrus CT. No significant brain correlates were observed for the two other psychopathological factors. We identified genome-wide significant associations for MA and DEP. PRS for MDD and SZ showed a positive effect on PHS, while PRS for BD showed a positive effect on all three factors. This study investigated the relationship of lifetime psychopathological factors and brain morphometric and genetic markers. Results highlight the need for dimensional approaches, overcoming the limitations of the current psychiatric nosology.
Collapse
Affiliation(s)
- Axel Krug
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany.
| | - Friederike S David
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Simon Schmitt
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Julia-Katharina Pfarr
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Kai G Ringwald
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Tina Meller
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Florian Thomas-Odenthal
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Katharina Thiel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Alexandra Winter
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Lena Waltemate
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Hannah Lemke
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Nils Opel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- German Centre for Mental Health (DZPG), Site Jena-Magdeburg-Halle, Jena, Germany
- Department of Psychiatry and Psychotherapy, University Hospital Jena, Jena, Germany
| | - Jonathan Repple
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Fabian Streit
- Department of Genetic Epidemiology 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
- Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159, Mannheim, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159, Mannheim, Germany
| | - Till F M Andlauer
- Department of Neurology, Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Alexandra Philipsen
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
- Centre for Human Genetics, University of Marburg, Marburg, Germany
| |
Collapse
|
9
|
Li M, Liu Y, Sun M, Yang Y, Zhang L, Liu Y, Li F, Liu H. SEP-363856 exerts neuroprotection through the PI3K/AKT/GSK-3β signaling pathway in a dual-hit neurodevelopmental model of schizophrenia-like mice. Drug Dev Res 2024; 85:e22225. [PMID: 38879781 DOI: 10.1002/ddr.22225] [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: 12/18/2023] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 10/11/2024]
Abstract
Schizophrenia (SZ) is a serious, destructive neurodevelopmental disorder. Antipsychotic medications are the primary therapy approach for this illness, but it's important to pay attention to the adverse effects as well. Clinical studies for SZ are currently in phase ΙΙΙ for SEP-363856 (SEP-856)-a new antipsychotic that doesn't work on dopamine D2 receptors. However, the underlying action mechanism of SEP-856 remains unknown. This study aimed to evaluate the impact and underlying mechanisms of SEP-856 on SZ-like behavior in a perinatal MK-801 treatment combined with social isolation from the weaning to adulthood model (MK-SI). First, we created an animal model that resembles SZ that combines the perinatal MK-801 with social isolation from weaning to adulthood. Then, different classical behavioral tests were used to evaluate the antipsychotic properties of SEP-856. The levels of proinflammatory cytokines (tumor necrosis factor-α, interleukin-6, and interleukin-1β), apoptosis-related genes (Bax and Bcl-2), and synaptic plasticity-related genes (brain-derived neurotrophic factor [BDNF] and PSD-95) in the hippocampus were analyzed by quantitative real-time PCR. Hematoxylin and eosin staining were used to observe the morphology of neurons in the hippocampal DG subregions. Western blot was performed to detect the protein expression levels of BDNF, PSD-95, Bax, Bcl-2, PI3K, p-PI3K, AKT, p-AKT, GSK-3β, p-GSK-3β in the hippocampus. MK-SI neurodevelopmental disease model studies have shown that compared with sham group, MK-SI group exhibit higher levels of autonomic activity, stereotyped behaviors, withdrawal from social interactions, dysregulated sensorimotor gating, and impaired recognition and spatial memory. These findings imply that the MK-SI model can mimic symptoms similar to those of SZ. Compared with the MK-SI model, high doses of SEP-856 all significantly reduced increased activity, improved social interaction, reduced stereotyping behavior, reversed sensorimotor gating dysregulation, and improved recognition memory and spatial memory impairment in MK-SI mice. In addition, SEP-856 can reduce the release of proinflammatory factors in the MK-SI model, promote the expression of BDNF and PSD-95 in the hippocampus, correct the Bax/Bcl-2 imbalance, turn on the PI3K/AKT/GSK-3β signaling pathway, and ultimately help the MK-SI mice's behavioral abnormalities. SEP-856 may play an antipsychotic role in MK-SI "dual-hit" model-induced SZ-like behavior mice by promoting synaptic plasticity recovery, decreasing death of hippocampal neurons, lowering the production of pro-inflammatory substances in the hippocampal region, and subsequently initiating the PI3K/AKT/GSK-3β signaling cascade.
Collapse
Affiliation(s)
- Mengdie Li
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yunxiao Liu
- Department of Psychiatry, School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Meng Sun
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yating Yang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ling Zhang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuexia Liu
- The Second People's Hospital of Huizhou, Huizhou, Guangdong, China
| | - Fujin Li
- The Second People's Hospital of Huizhou, Huizhou, Guangdong, China
| | - Huanzhong Liu
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
10
|
Bolat E, Polat S, Tunç M, Çoban M, Göker P. Investigation of Skull Cortical Thickness Changes in Healthy Population and Patients With Schizophrenia on Computed Tomography Images. J Craniofac Surg 2024; 35:1284-1288. [PMID: 38727232 DOI: 10.1097/scs.0000000000010261] [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: 02/09/2024] [Accepted: 04/03/2024] [Indexed: 06/04/2024] Open
Abstract
Cortical bone thickness is essential for the mechanical function of bone. Some factors including aging, sex, body size, hormone levels, behavior, and genetics lead to changes in cranial cortical robusticity. Moreover, the skull is one of the hardest and most durable structures in the human body. Schizophrenia is defined as a psychiatric disease characterized by delusions and hallucinations, and these patients have reduced brain volume; however, there is no study including cortical bone structure. For this reason, the aim of this study was to determine whether there is a difference in the skull cortical thickness of patients with schizophrenia and, compare it with healthy subjects. The cranial length, cranial width, anterior cortical thickness, right and left anterior cortical thickness, right and left lateral cortical thickness, right and left posterior lateral thickness, and posterior cortical thickness were measured with axial computed tomography images of 30 patients with schizophrenia and 132 healthy individuals aged between 18 and 69years. A statistically significant difference was found between the two groups in the measurements of right and left posterior lateral thickness, and posterior cortical thickness ( P = 0.006, P = 0.001, and P = 0.047, respectively). The sexes were compared, and it was found that the cranial width, anterior thickness, left anterior thickness, and right and left posterior thickness measurements of patients with schizophrenia showed a statistically significant difference compared with the control group ( P < 0.001, P = 0.003, P = 0.001, P < 0.001 and P < 0.001, respectively). The authors observed that skull cortical thickness may be different in schizophrenia. The results obtained from this study may be beneficial for evaluating these structures for clinical and pathological processes. Furthermore, knowledge about the skull cortical thickness in planning surgical procedures will increase the reliability and effectiveness of the surgical method, and this will minimize the risk of complications.
Collapse
Affiliation(s)
- Esra Bolat
- Department of Anatomy, Çukurova University Faculty of Medicine
| | - Sema Polat
- Department of Anatomy, Çukurova University Faculty of Medicine
| | - Mahmut Tunç
- Department of Therapy and Rehabilitation, Vocational School of Health Services, Baskent University
| | - Muhammet Çoban
- Department of Radiology, Kozan State Hospital, Adana, Turkey
| | - Pinar Göker
- Department of Anatomy, Çukurova University Faculty of Medicine
| |
Collapse
|
11
|
Viejo-Romero M, Whalley HC, Shen X, Stolicyn A, Smith DJ, Howard DM. An epidemiological study of season of birth, mental health, and neuroimaging in the UK Biobank. PLoS One 2024; 19:e0300449. [PMID: 38776272 PMCID: PMC11111058 DOI: 10.1371/journal.pone.0300449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 02/27/2024] [Indexed: 05/24/2024] Open
Abstract
Environmental exposures during the perinatal period are known to have a long-term effect on adult physical and mental health. One such influential environmental exposure is the time of year of birth which affects the amount of daylight, nutrients, and viral load that an individual is exposed to within this key developmental period. Here, we investigate associations between season of birth (seasonality), four mental health traits (n = 137,588) and multi-modal neuroimaging measures (n = 33,212) within the UK Biobank. Summer births were associated with probable recurrent Major Depressive Disorder (β = 0.026, pcorr = 0.028) and greater mean cortical thickness in temporal and occipital lobes (β = 0.013 to 0.014, pcorr<0.05). Winter births were associated with greater white matter integrity globally, in the association fibers, thalamic radiations, and six individual tracts (β = -0.013 to -0.022, pcorr<0.05). Results of sensitivity analyses adjusting for birth weight were similar, with an additional association between winter birth and white matter microstructure in the forceps minor and between summer births, greater cingulate thickness and amygdala volume. Further analyses revealed associations between probable depressive phenotypes and a range of neuroimaging measures but a paucity of interactions with seasonality. Our results suggest that seasonality of birth may affect later-life brain structure and play a role in lifetime recurrent Major Depressive Disorder. Due to the small effect sizes observed, and the lack of associations with other mental health traits, further research is required to validate birth season effects in the context of different latitudes, and by co-examining genetic and epigenetic measures to reveal informative biological pathways.
Collapse
Affiliation(s)
- Maria Viejo-Romero
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, United Kingdom
| | - Heather C. Whalley
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, United Kingdom
| | - Xueyi Shen
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, United Kingdom
| | - Aleks Stolicyn
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, United Kingdom
| | - Daniel J. Smith
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, United Kingdom
| | - David M. Howard
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, United Kingdom
- Institute of Psychiatry, Social, Genetic and Developmental Psychiatry Centre, Psychology & Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|
12
|
Danyeli LV, Sen ZD, Colic L, Opel N, Refisch A, Blekic N, Macharadze T, Kretzschmar M, Munk MJ, Gaser C, Speck O, Walter M, Li M. Cortical thickness of the posterior cingulate cortex is associated with the ketamine-induced altered sense of self: An ultra-high field MRI study. J Psychiatr Res 2024; 172:136-143. [PMID: 38382237 DOI: 10.1016/j.jpsychires.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
Subanesthetic doses of ketamine induce an antidepressant effect within hours in individuals with treatment-resistant depression while it furthermore induces immediate but transient psychotomimetic effects. Among these psychotomimetic effects, an altered sense of self has specifically been associated with the antidepressant response to ketamine as well as psychedelics. However, there is plenty of variation in the extent of the drug-induced altered sense of self experience that might be explained by differences in basal morphological characteristics, such as cortical thickness. Regions that have been previously associated with a psychedelics-induced sense of self and with ketamine's mechanism of action, are the posterior cingulate cortex (PCC) and the pregenual anterior cingulate cortex (pgACC). In this randomized, placebo-controlled, double-blind cross-over magnetic resonance imaging study, thirty-five healthy male participants (mean age ± standard deviation (SD) = 25.1 ± 4.2 years) were scanned at 7 T. We investigated whether the cortical thickness of two DMN regions, the PCC and the pgACC, are associated with disembodiment and experience of unity scores, which were used to index the ketamine-induced altered sense of self. We observed a negative correlation between the PCC cortical thickness and the disembodiment scores (R = -0.54, p < 0.001). In contrast, no significant association was found between the pgACC cortical thickness and the ketamine-induced altered sense of self. In the context of the existing literature, our findings highlight the importance of the PCC as a structure involved in the mechanism of ketamine-induced altered sense of self that seems to be shared with different antidepressant agents with psychotomimetic effects operating on different classes of transmitter systems.
Collapse
Affiliation(s)
- Lena Vera Danyeli
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany; Department of Psychiatry and Psychotherapy, University Tübingen, Tübingen, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Zümrüt Duygu Sen
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Lejla Colic
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany; German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany
| | - Nils Opel
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany; German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany
| | - Alexander Refisch
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Nikolai Blekic
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
| | - Tamar Macharadze
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Department Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany; Department of Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Moritz Kretzschmar
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - MatthiasH J Munk
- Department of Psychiatry and Psychotherapy, University Tübingen, Tübingen, Germany; Systems Neurophysiology, Department of Biology, Darmstadt University of Technology, Darmstadt, Germany
| | - Christian Gaser
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany; German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany; Department of Neurology, Jena University Hospital, Jena, Germany
| | - Oliver Speck
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany; German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany; Department of Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany; Department of Biomedical Magnetic Resonance, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany; Department of Psychiatry and Psychotherapy, University Tübingen, Tübingen, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany; German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany; Department of Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany.
| | - Meng Li
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany.
| |
Collapse
|
13
|
Onwordi EC, Whitehurst T, Shatalina E, Mansur A, Arumuham A, Osugo M, Marques TR, Jauhar S, Gupta S, Mehrotra R, Rabiner EA, Gunn RN, Natesan S, Howes OD. Synaptic Terminal Density Early in the Course of Schizophrenia: An In Vivo UCB-J Positron Emission Tomographic Imaging Study of SV2A. Biol Psychiatry 2024; 95:639-646. [PMID: 37330164 PMCID: PMC10923626 DOI: 10.1016/j.biopsych.2023.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND The synaptic hypothesis is an influential theory of the pathoetiology of schizophrenia (SCZ), which is supported by the finding that there is lower uptake of the synaptic terminal density marker [11C]UCB-J in patients with chronic SCZ than in control participants. However, it is unclear whether these differences are present early in the illness. To address this, we investigated [11C]UCB-J volume of distribution (VT) in antipsychotic-naïve/free patients with SCZ who were recruited from first-episode services compared with healthy volunteers. METHODS Forty-two volunteers (SCZ n = 21, healthy volunteers n = 21) underwent [11C]UCB-J positron emission tomography to index [11C]UCB-J VT and distribution volume ratio in the anterior cingulate, frontal, and dorsolateral prefrontal cortices; the temporal, parietal and occipital lobes; and the hippocampus, thalamus, and amygdala. Symptom severity was assessed in the SCZ group using the Positive and Negative Syndrome Scale. RESULTS We found no significant effects of group on [11C]UCB-J VT or distribution volume ratio in most regions of interest (effect sizes from d = 0.0-0.7, p > .05), with two exceptions: we found lower distribution volume ratio in the temporal lobe (d = 0.7, uncorrected p < .05) and lower VT/fp in the anterior cingulate cortex in patients (d = 0.7, uncorrected p < .05). The Positive and Negative Syndrome Scale total score was negatively associated with [11C]UCB-J VT in the hippocampus in the SCZ group (r = -0.48, p = .03). CONCLUSIONS These findings indicate that large differences in synaptic terminal density are not present early in SCZ, although there may be more subtle effects. When taken together with previous evidence of lower [11C]UCB-J VT in patients with chronic illness, this may indicate synaptic density changes during the course of SCZ.
Collapse
Affiliation(s)
- Ellis Chika Onwordi
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Centre for Psychiatry and Mental Health, Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom.
| | - Thomas Whitehurst
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Ekaterina Shatalina
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Ayla Mansur
- Department of Brain Sciences, Imperial College London, The Commonwealth Building, Hammersmith Hospital, London, United Kingdom; Invicro, Burlington Danes Building, London, United Kingdom
| | - Atheeshaan Arumuham
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Martin Osugo
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Tiago Reis Marques
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Sameer Jauhar
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Susham Gupta
- Early Detection and Early Intervention, East London National Health Service Foundation Trust, London, United Kingdom
| | - Ravi Mehrotra
- Early Intervention in Psychosis Team, West Middlesex University Hospital, West London National Health Service Trust, Isleworth, London, United Kingdom
| | - Eugenii A Rabiner
- Invicro, Burlington Danes Building, London, United Kingdom; Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Roger N Gunn
- Department of Brain Sciences, Imperial College London, The Commonwealth Building, Hammersmith Hospital, London, United Kingdom; Invicro, Burlington Danes Building, London, United Kingdom
| | - Sridhar Natesan
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Oliver D Howes
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
14
|
Niu L, Fang K, Han S, Xu C, Sun X. Resolving heterogeneity in schizophrenia, bipolar I disorder, and attention-deficit/hyperactivity disorder through individualized structural covariance network analysis. Cereb Cortex 2024; 34:bhad391. [PMID: 38142281 DOI: 10.1093/cercor/bhad391] [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: 09/10/2023] [Revised: 09/30/2023] [Accepted: 10/01/2023] [Indexed: 12/25/2023] Open
Abstract
Disruptions in large-scale brain connectivity are hypothesized to contribute to psychiatric disorders, including schizophrenia, bipolar I disorder, and attention-deficit/hyperactivity disorder. However, high inter-individual variation among patients with psychiatric disorders hinders achievement of unified findings. To this end, we adopted a newly proposed method to resolve heterogeneity of differential structural covariance network in schizophrenia, bipolar I disorder, and attention-deficit/hyperactivity disorder. This method could infer individualized structural covariance aberrance by assessing the deviation from healthy controls. T1-weighted anatomical images of 114 patients with psychiatric disorders (schizophrenia: n = 37; bipolar I disorder: n = 37; attention-deficit/hyperactivity disorder: n = 37) and 110 healthy controls were analyzed to obtain individualized differential structural covariance network. Patients exhibited tremendous heterogeneity in profiles of individualized differential structural covariance network. Despite notable heterogeneity, patients with the same disorder shared altered edges at network level. Moreover, individualized differential structural covariance network uncovered two distinct psychiatric subtypes with opposite differences in structural covariance edges, that were otherwise obscured when patients were merged, compared with healthy controls. These results provide new insights into heterogeneity and have implications for the nosology in psychiatric disorders.
Collapse
Affiliation(s)
- Lianjie Niu
- Department of Breast Disease, Henan Breast Cancer Center. The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Keke Fang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Chunmiao Xu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Xianfu Sun
- Department of Breast Disease, Henan Breast Cancer Center. The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| |
Collapse
|
15
|
Pan TY, Pan YJ, Tsai SJ, Tsai CW, Yang FY. Focused Ultrasound Stimulates the Prefrontal Cortex and Prevents MK-801-Induced Psychiatric Symptoms of Schizophrenia in Rats. Schizophr Bull 2024; 50:120-131. [PMID: 37301986 PMCID: PMC10754174 DOI: 10.1093/schbul/sbad078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
BACKGROUND AND HYPOTHESIS Treatment of schizophrenia remains a major challenge. Recent studies have focused on glutamatergic signaling hypoactivity through N-methyl-D-aspartate (NMDA) receptors. Low-intensity pulsed ultrasound (LIPUS) improves behavioral deficits and ameliorates neuropathology in dizocilpine (MK-801)-treated rats. The aim of this study was to investigate the efficacy of LIPUS against psychiatric symptoms and anxiety-like behaviors. STUDY DESIGN Rats assigned to 4 groups were pretreated with or without LIPUS for 5 days. The open field and prepulse inhibition tests were performed after saline or MK-801 (0.3 mg/kg) administration. Then, the neuroprotective effects of LIPUS on the MK-801-treated rats were evaluated using western blotting and immunohistochemical staining. STUDY RESULTS LIPUS stimulation of the prefrontal cortex (PFC) prevented deficits in locomotor activity and sensorimotor gating and improved anxiety-like behavior. MK-801 downregulated the expression of NR1, the NMDA receptor, in rat medial PFC (mPFC). NR1 expression was significantly higher in animals receiving LIPUS pretreatment compared to those receiving only MK-801. In contrast, a significant increase in c-Fos-positive cells in the mPFC and ventral tegmental area was observed in the MK-801-treated rats compared to those receiving only saline; this change was suppressed by pretreatment with LIPUS. CONCLUSIONS This study provides new evidence for the role of LIPUS stimulation in regulating the NMDA receptor and modulating c-Fos activity, which makes it a potentially valuable antipsychotic treatment for schizophrenia.
Collapse
Affiliation(s)
- Tsung-Yu Pan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ju Pan
- Department of Psychiatry, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Institute of Public Health, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Che-Wen Tsai
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
16
|
Barth C, Nerland S, Jørgensen KN, Haatveit B, Wortinger LA, Melle I, Haukvik UK, Ueland T, Andreassen OA, Agartz I. Altered Sex Differences in Hippocampal Subfield Volumes in Schizophrenia. Schizophr Bull 2024; 50:107-119. [PMID: 37354490 PMCID: PMC10754184 DOI: 10.1093/schbul/sbad091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/26/2023]
Abstract
BACKGROUND AND HYPOTHESIS The hippocampus is a heterogenous brain structure that differs between the sexes and has been implicated in the pathophysiology of psychiatric illnesses. Here, we explored sex and diagnostic group differences in hippocampal subfield volumes, in individuals with schizophrenia spectrum disorder (SZ), bipolar disorders (BD), and healthy controls (CTL). STUDY DESIGN One thousand and five hundred and twenty-one participants underwent T1-weighted magnetic resonance imaging (SZ, n = 452, mean age 30.7 ± 9.2 [SD] years, males 59.1%; BD, n = 316, 33.7 ± 11.4, 41.5%; CTL, n = 753, 34.1 ± 9.1, 55.6%). Total hippocampal, subfield, and intracranial volumes were estimated with Freesurfer (v6.0.0). Analysis of covariance and multiple regression models were fitted to examine sex-by-diagnostic (sub)group interactions in volume. In SZ and BD, separately, associations between volumes and clinical as well as cognitive measures were examined between the sexes using regression models. STUDY RESULTS Significant sex-by-group interactions were found for the total hippocampus, dentate gyrus, molecular layer, presubiculum, fimbria, hippocampal-amygdaloid transition area, and CA4, indicating a larger volumetric deficit in male patients relative to female patients when compared with same-sex CTL. Subgroup analyses revealed that this interaction was driven by males with schizophrenia. Effect sizes were overall small (partial η < 0.02). We found no significant sex differences in the associations between hippocampal volumes and clinical or cognitive measures in SZ and BD. CONCLUSIONS Using a well-powered sample, our findings indicate that the pattern of morphological sex differences in hippocampal subfields is altered in individuals with schizophrenia relative to CTL, due to higher volumetric deficits in males.
Collapse
Affiliation(s)
- Claudia Barth
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
| | - Stener Nerland
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
| | - Kjetil N Jørgensen
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Department of Psychiatry, Telemark Hospital, Skien, Norway
| | - Beathe Haatveit
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
| | - Laura A Wortinger
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
| | - Ingrid Melle
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
| | - Unn K Haukvik
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
- Department of Adult Mental Health, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre of Research and Education in Forensic Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Torill Ueland
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
17
|
Chen G, Li L, Sun T, Jiang C, Xu W, Chen S, Hu C, Yue Y, Wang T, Jiang W, Yuan Y. The Interaction of LAMA2 and Duration of Illness Affects the Thickness of the Right Transverse Temporal Gyrus in Major Depressive Disorder. Neuropsychiatr Dis Treat 2023; 19:2807-2816. [PMID: 38144699 PMCID: PMC10749177 DOI: 10.2147/ndt.s435025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/14/2023] [Indexed: 12/26/2023] Open
Abstract
Background Depression is a heritable brain disorder. Laminin genes were recently identified to affect the brain's overall thickness through neurogenesis, differentiation, and migration in depression. This study aims to explore the effects of the LAMA2's single nucleotide polymorphisms (SNP), a subunit gene of laminin, on the detected brain regions of patients with major depressive disorder (MDD). Methods The study included 89 patients with MDD and 60 healthy controls with T1-weighted structural magnetic resonance imaging and blood samples for genotyping. The interactions between LAMA2 gene SNPs and diagnosis as well as duration of illness (DOI) were explored on brain measures controlled for age, gender, and site. Results The right transverse temporal gyrus and right parahippocampal gyrus showed reduced thickness in MDD. Almost all seven LAMA2 SNPs showed significant interactions with diagnosis on both gyrus (corrected p < 0.05 or trending). In MDD, rs6569604, rs2229848, rs2229849, rs2229850, and rs2784895 interacted with DOI on the right transverse temporal gyrus (corrected p < 0.05), but not the right parahippocampal gyrus. Conclusion The thickness of the right transverse temporal gyrus in patients with MDD may be affected by LAMA2 gene and DOI.
Collapse
Affiliation(s)
- Gang Chen
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Department of Medical Psychology, Huai’an NO 3 People’s Hospital, Huaian, People’s Republic of China
| | - Lei Li
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Department of Sleep Medicine, The Fourth People’s Hospital of Lianyungang, Lianyungang, People’s Republic of China
| | - Taipeng Sun
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Department of Medical Psychology, Huai’an NO 3 People’s Hospital, Huaian, People’s Republic of China
| | - Chenguang Jiang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Wei Xu
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Suzhen Chen
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Changchun Hu
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Yingying Yue
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Tianyu Wang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Wenhao Jiang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
18
|
Harmata GIS, Barsotti EJ, Casten LG, Fiedorowicz JG, Williams A, Shaffer JJ, Richards JG, Sathyaputri L, Schmitz SL, Christensen GE, Long JD, Gaine ME, Xu J, Michaelson JJ, Wemmie JA, Magnotta VA. Cerebellar morphological differences and associations with extrinsic factors in bipolar disorder type I. J Affect Disord 2023; 340:269-279. [PMID: 37562560 PMCID: PMC10529949 DOI: 10.1016/j.jad.2023.08.018] [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: 01/04/2023] [Revised: 07/18/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND The neural underpinnings of bipolar disorder (BD) remain poorly understood. The cerebellum is ideally positioned to modulate emotional regulation circuitry yet has been understudied in BD. Literature suggests differences in cerebellar activity and metabolism in BD, however findings on structural differences remain contradictory. Potential reasons include combining BD subtypes, small sample sizes, and potential moderators such as genetics, adverse childhood experiences (ACEs), and pharmacotherapy. METHODS We collected 3 T MRI scans from participants with (N = 131) and without (N = 81) BD type I, as well as blood and questionnaires. We assessed differences in cerebellar volumes and explored potentially influential factors. RESULTS The cerebellar cortex was smaller bilaterally in participants with BD. Polygenic propensity score did not predict any cerebellar volumes, suggesting that non-genetic factors may have greater influence on the cerebellar volume difference we observed in BD. Proportionate cerebellar white matter volumes appeared larger with more ACEs, but this may result from reduced ICV. Time from onset and symptom burden were not associated with cerebellar volumes. Finally, taking sedatives was associated with larger cerebellar white matter and non-significantly larger cortical volume. LIMITATIONS This study was cross-sectional, limiting interpretation of possible mechanisms. Most of our participants were White, which could limit the generalizability. Additionally, we did not account for potential polypharmacy interactions. CONCLUSIONS These findings suggest that external factors, such as sedatives and childhood experiences, may influence cerebellum structure in BD and may mask underlying differences. Accounting for such variables may be critical for consistent findings in future studies.
Collapse
Affiliation(s)
- Gail I S Harmata
- Department of Psychiatry, The University of Iowa, United States; Iowa Neuroscience Institute, The University of Iowa, United States; Department of Radiology, The University of Iowa, United States
| | - Ercole John Barsotti
- Department of Psychiatry, The University of Iowa, United States; Department of Epidemiology, The University of Iowa, United States
| | - Lucas G Casten
- Department of Psychiatry, The University of Iowa, United States; Interdisciplinary Graduate Program in Genetics, The University of Iowa, United States
| | - Jess G Fiedorowicz
- Department of Psychiatry, The University of Iowa, United States; Iowa Neuroscience Institute, The University of Iowa, United States; Department of Psychiatry, University of Ottawa, Canada
| | - Aislinn Williams
- Department of Psychiatry, The University of Iowa, United States; Iowa Neuroscience Institute, The University of Iowa, United States
| | - Joseph J Shaffer
- Department of Psychiatry, The University of Iowa, United States; Iowa Neuroscience Institute, The University of Iowa, United States; Department of Radiology, The University of Iowa, United States; Department of Biosciences, Kansas City University, United States
| | | | | | | | - Gary E Christensen
- Department of Electrical and Computer Engineering, The University of Iowa, United States; Department of Radiation Oncology, The University of Iowa, United States
| | - Jeffrey D Long
- Department of Psychiatry, The University of Iowa, United States; Department of Biostatistics, The University of Iowa, United States
| | - Marie E Gaine
- Department of Psychiatry, The University of Iowa, United States; Iowa Neuroscience Institute, The University of Iowa, United States; Department of Pharmaceutical Sciences and Experimental Therapeutics (PSET), College of Pharmacy, The University of Iowa, United States
| | - Jia Xu
- Department of Radiology, The University of Iowa, United States
| | - Jake J Michaelson
- Department of Psychiatry, The University of Iowa, United States; Iowa Neuroscience Institute, The University of Iowa, United States; Interdisciplinary Graduate Program in Genetics, The University of Iowa, United States
| | - John A Wemmie
- Department of Psychiatry, The University of Iowa, United States; Iowa Neuroscience Institute, The University of Iowa, United States; Department of Molecular Physiology and Biophysics, The University of Iowa, United States; Department of Neurosurgery, The University of Iowa, United States; Veterans Affairs Medical Center, Iowa City, United States
| | - Vincent A Magnotta
- Department of Psychiatry, The University of Iowa, United States; Iowa Neuroscience Institute, The University of Iowa, United States; Department of Radiology, The University of Iowa, United States; Department of Biomedical Engineering, The University of Iowa, United States.
| |
Collapse
|
19
|
Wang Z, Lin X, Luo X, Xiao J, Zhang Y, Xu J, Wang S, Zhao F, Wang H, Zheng H, Zhang W, Lin C, Tan Z, Cao L, Wang Z, Tan Y, Chen W, Cao Y, Guo X, Pittenger C, Luo X. Pleiotropic Association of CACNA1C Variants With Neuropsychiatric Disorders. Schizophr Bull 2023; 49:1174-1184. [PMID: 37306960 PMCID: PMC10483336 DOI: 10.1093/schbul/sbad073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
BACKGROUND Neuropsychiatric disorders are highly heritable and have overlapping genetic underpinnings. Single nucleotide polymorphisms (SNPs) in the gene CACNA1C have been associated with several neuropsychiatric disorders, across multiple genome-wide association studies. METHOD A total of 70,711 subjects from 37 independent cohorts with 13 different neuropsychiatric disorders were meta-analyzed to identify overlap of disorder-associated SNPs within CACNA1C. The differential expression of CACNA1C mRNA in five independent postmortem brain cohorts was examined. Finally, the associations of disease-sharing risk alleles with total intracranial volume (ICV), gray matter volumes (GMVs) of subcortical structures, cortical surface area (SA), and average cortical thickness (TH) were tested. RESULTS Eighteen SNPs within CACNA1C were nominally associated with more than one neuropsychiatric disorder (P < .05); the associations shared among schizophrenia, bipolar disorder, and alcohol use disorder survived false discovery rate correction (five SNPs with P < 7.3 × 10-4 and q < 0.05). CACNA1C mRNA was differentially expressed in brains from individuals with schizophrenia, bipolar disorder, and Parkinson's disease, relative to controls (three SNPs with P < .01). Risk alleles shared by schizophrenia, bipolar disorder, substance dependence, and Parkinson's disease were significantly associated with ICV, GMVs, SA, or TH (one SNP with P ≤ 7.1 × 10-3 and q < 0.05). CONCLUSION Integrating multiple levels of analyses, we identified CACNA1C variants associated with multiple psychiatric disorders, and schizophrenia and bipolar disorder were most strongly implicated. CACNA1C variants may contribute to shared risk and pathophysiology in these conditions.
Collapse
Affiliation(s)
- Zuxing Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of medicine, Shanghai 200030, China
- Sichuan Provincial Center for Mental Health, The Center of Psychosomatic Medicine of Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Fujian Provincial Cancer Hospital, the Teaching Hospital of Fujian Medical University, Fuzhou, Fujian 350014, China
| | - Xinqun Luo
- Department of Neurosurgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350001, China
| | - Jun Xiao
- Sichuan Provincial Center for Mental Health, The Center of Psychosomatic Medicine of Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin 300180, China
| | - Jianying Xu
- Zhuhai Center for Maternal and Child Health Care, Zhuhai, Guangdong 519000, China
| | - Shibin Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of medicine, Shanghai 200030, China
| | - Fen Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of medicine, Shanghai 200030, China
| | - Huifen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of medicine, Shanghai 200030, China
| | - Hangxiao Zheng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of medicine, Shanghai 200030, China
| | - Wei Zhang
- Department of Pharmacology, Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Chen Lin
- Beijing Huilongguan Hospital, Peking University Huilongguan School of Clinical Medicine, Beijing 100096, China
| | - Zewen Tan
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510370, China
| | - Liping Cao
- Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510370, China
| | - Zhiren Wang
- Beijing Huilongguan Hospital, Peking University Huilongguan School of Clinical Medicine, Beijing 100096, China
| | - Yunlong Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan School of Clinical Medicine, Beijing 100096, China
| | - Wenzhong Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of medicine, Shanghai 200030, China
| | - Yuping Cao
- Department of Psychiatry, Second Xiangya Hospital, Central South University; China National Clinical Research Center on Mental Disorders, China National Technology Institute on Mental Disorders, Changsha, Hunan 410011, China
| | - Xiaoyun Guo
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of medicine, Shanghai 200030, China
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, US
| | - Christopher Pittenger
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, US
| | - Xingguang Luo
- Beijing Huilongguan Hospital, Peking University Huilongguan School of Clinical Medicine, Beijing 100096, China
| |
Collapse
|
20
|
Luckett PH, Olufawo M, Lamichhane B, Park KY, Dierker D, Verastegui GT, Yang P, Kim AH, Chheda MG, Snyder AZ, Shimony JS, Leuthardt EC. Predicting survival in glioblastoma with multimodal neuroimaging and machine learning. J Neurooncol 2023; 164:309-320. [PMID: 37668941 PMCID: PMC10522528 DOI: 10.1007/s11060-023-04439-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/26/2023] [Indexed: 09/06/2023]
Abstract
PURPOSE Glioblastoma (GBM) is the most common and aggressive malignant glioma, with an overall median survival of less than two years. The ability to predict survival before treatment in GBM patients would lead to improved disease management, clinical trial enrollment, and patient care. METHODS GBM patients (N = 133, mean age 60.8 years, median survival 14.1 months, 57.9% male) were retrospectively recruited from the neurosurgery brain tumor service at Washington University Medical Center. All patients completed structural neuroimaging and resting state functional MRI (RS-fMRI) before surgery. Demographics, measures of cortical thickness (CT), and resting state functional network connectivity (FC) were used to train a deep neural network to classify patients based on survival (< 1y, 1-2y, >2y). Permutation feature importance identified the strongest predictors of survival based on the trained models. RESULTS The models achieved a combined cross-validation and hold out accuracy of 90.6% in classifying survival (< 1y, 1-2y, >2y). The strongest demographic predictors were age at diagnosis and sex. The strongest CT predictors of survival included the superior temporal sulcus, parahippocampal gyrus, pericalcarine, pars triangularis, and middle temporal regions. The strongest FC features primarily involved dorsal and inferior somatomotor, visual, and cingulo-opercular networks. CONCLUSION We demonstrate that machine learning can accurately classify survival in GBM patients based on multimodal neuroimaging before any surgical or medical intervention. These results were achieved without information regarding presentation symptoms, treatments, postsurgical outcomes, or tumor genomic information. Our results suggest GBMs have a global effect on the brain's structural and functional organization, which is predictive of survival.
Collapse
Affiliation(s)
- Patrick H Luckett
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Michael Olufawo
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bidhan Lamichhane
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center for Health Sciences, Oklahoma State University, Tulsa, OK, 74136, USA
| | - Ki Yun Park
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Donna Dierker
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Peter Yang
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Brain Tumor Center at Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Milan G Chheda
- Brain Tumor Center at Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Abraham Z Snyder
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Brain Tumor Center at Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric C Leuthardt
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Brain Tumor Center at Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, 63130, USA
- Department of Mechanical Engineering and Materials Science, Washington University in Saint Louis, St. Louis, MO, 63130, USA
- Center for Innovation in Neuroscience and Technology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Brain Laser Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- National Center for Adaptive Neurotechnologies, Albany, USA
| |
Collapse
|
21
|
Zak N, Moberget T, Bøen E, Boye B, Rygvold TW, Malt UF, Andreassen OA, Andersson S, Westlye LT, Elvsåshagen T. Baseline long-term potentiation-like cortical plasticity is associated with longitudinal cortical thinning in healthy adults and in adults with bipolar disorder type II. Eur J Neurosci 2023; 58:2824-2837. [PMID: 37163975 DOI: 10.1111/ejn.16038] [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: 08/06/2021] [Revised: 03/20/2023] [Accepted: 05/06/2023] [Indexed: 05/12/2023]
Abstract
The precise neurobiological processes underlying cerebral cortical thinning in aging and psychiatric illnesses remain undetermined, yet aging- and synaptic dysfunction-related loss of synapses are potentially important mechanisms. We used long-term potentiation-like plasticity of the visual evoked potential as an index of synaptic function in the cortex and hypothesized that plasticity at baseline would be negatively associated with future cortical thinning in healthy adults and in adults with bipolar disorder type II. Thirty-two healthy adults and 15 adults with bipolar disorder type II underwent electroencephalography-based measurement of visual evoked potential plasticity and 3T magnetic resonance imaging of the brain at baseline and a follow-up brain scan on average 2.3 years later. The relationships between visual evoked potential plasticity at baseline and longitudinal cortical thickness changes were examined using Freesurfer and the Permutation Analysis of Linear Models tool. The analyses showed a negative association between the plasticity of the N1 visual evoked potential amplitude at baseline and thinning rate in the medial and lateral parietal and medial occipital cortices in healthy adults and in the right medial occipital cortex in the total sample of healthy adults and adults with bipolar disorder type II, indicating greater thinning over time in subjects with less N1 plasticity (pFWER < .05). Although preliminary, the results indicate an association between visual evoked potential plasticity and the future rate of cortical thinning in healthy adults and in bipolar disorder type II, supporting the hypothesis that cortical thinning might be related to synaptic dysfunction.
Collapse
Affiliation(s)
- Nathalia Zak
- Norwegian Centre for Mental Disorders Research (NORMENT), KG Jebsen Centre for Psychosis Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torgeir Moberget
- Norwegian Centre for Mental Disorders Research (NORMENT), KG Jebsen Centre for Psychosis Research, Oslo University Hospital, Oslo, Norway
| | - Erlend Bøen
- Unit for Psychosomatics and C-L psychiatry for adults, Oslo University Hospital, Oslo, Norway
| | - Birgitte Boye
- Unit for Psychosomatics and C-L psychiatry for adults, Oslo University Hospital, Oslo, Norway
- Department of Behavioral Medicine, University of Oslo, Oslo, Norway
| | | | - Ulrik F Malt
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Research and Education, Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), KG Jebsen Centre for Psychosis Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research (NORMENT), KG Jebsen Centre for Psychosis Research, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Torbjørn Elvsåshagen
- Norwegian Centre for Mental Disorders Research (NORMENT), KG Jebsen Centre for Psychosis Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
22
|
Newbury JB, Arseneault L, Moffitt TE, Odgers CL, Howe LD, Bakolis I, Reuben A, Danese A, Sugden K, Williams B, Rasmussen LJH, Trotta A, Ambler AP, Fisher HL. Socioenvironmental Adversity and Adolescent Psychotic Experiences: Exploring Potential Mechanisms in a UK Longitudinal Cohort. Schizophr Bull 2023; 49:1042-1054. [PMID: 36934309 PMCID: PMC10318878 DOI: 10.1093/schbul/sbad017] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2023]
Abstract
BACKGROUND AND HYPOTHESIS Children exposed to socioenvironmental adversities (eg, urbanicity, pollution, neighborhood deprivation, crime, and family disadvantage) are more likely to subsequently develop subclinical psychotic experiences during adolescence (eg, hearing voices, paranoia). However, the pathways through which this occurs have not been previously investigated. We hypothesized that cognitive ability and inflammation would partly explain this association. STUDY DESIGN Data were utilized from the Environmental-Risk Longitudinal Twin Study, a cohort of 2232 children born in 1994-1995 in England and Wales and followed to age 18. Socioenvironmental adversities were measured from birth to age 10 and classified into physical risk (defined by high urbanicity and air pollution) and socioeconomic risk (defined by high neighborhood deprivation, neighborhood disorder, and family disadvantage). Cognitive abilities (overall, crystallized, fluid, and working memory) were assessed at age 12; and inflammatory markers (C-reactive protein, interleukin-6, soluble urokinase plasminogen activator receptor) were measured at age 18 from blood samples. Participants were interviewed at age 18 regarding psychotic experiences. STUDY RESULTS Higher physical risk and socioeconomic risk were associated with increased odds of psychotic experiences in adolescence. The largest mediation pathways were from socioeconomic risk via overall cognitive ability and crystallized ability, which accounted for ~11% and ~19% of the association with psychotic experiences, respectively. No statistically significant pathways were found via inflammatory markers in exploratory (partially cross-sectional) analyses. CONCLUSIONS Cognitive ability, especially crystallized ability, may partly explain the association between childhood socioenvironmental adversity and adolescent psychotic experiences. Interventions to support cognitive development among children living in disadvantaged settings could buffer them against developing subclinical psychotic phenomena.
Collapse
Affiliation(s)
- Joanne B Newbury
- King’s College London, Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Louise Arseneault
- King’s College London, Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Terrie E Moffitt
- King’s College London, Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Centre for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Candice L Odgers
- Social Science Research Institute, Duke University, Durham, NC, USA
- Department of Psychological Science, School of Social Ecology, University of California, Irvine, Irvine, CA, USA
| | - Laura D Howe
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ioannis Bakolis
- King’s College London, Centre for Implementation Science, Department of Health Service and Population Research, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- King’s College London, Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Aaron Reuben
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Andrea Danese
- King’s College London, Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- King’s College London, Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- National and Specialist CAMHS Clinic for Trauma, Anxiety, and Depression, South London and Maudsley NHS Foundation Trust, London, UK
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Benjamin Williams
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Line J H Rasmussen
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Antonella Trotta
- King’s College London, Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- School of Health and Social Care, University of Essex, Colchester, UK
| | - Antony P Ambler
- King’s College London, Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Helen L Fisher
- King’s College London, Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- ESRC Centre for Society and Mental Health, King’s College London, London, UK
| |
Collapse
|
23
|
Seitz-Holland J, Nägele FL, Kubicki M, Pasternak O, Cho KIK, Hough M, Mulert C, Shenton ME, Crow TJ, James ACD, Lyall AE. Shared and distinct white matter abnormalities in adolescent-onset schizophrenia and adolescent-onset psychotic bipolar disorder. Psychol Med 2023; 53:4707-4719. [PMID: 35796024 PMCID: PMC11119277 DOI: 10.1017/s003329172200160x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND While adolescent-onset schizophrenia (ADO-SCZ) and adolescent-onset bipolar disorder with psychosis (psychotic ADO-BPD) present a more severe clinical course than their adult forms, their pathophysiology is poorly understood. Here, we study potentially state- and trait-related white matter diffusion-weighted magnetic resonance imaging (dMRI) abnormalities along the adolescent-onset psychosis continuum to address this need. METHODS Forty-eight individuals with ADO-SCZ (20 female/28 male), 15 individuals with psychotic ADO-BPD (7 female/8 male), and 35 healthy controls (HCs, 18 female/17 male) underwent dMRI and clinical assessments. Maps of extracellular free-water (FW) and fractional anisotropy of cellular tissue (FAT) were compared between individuals with psychosis and HCs using tract-based spatial statistics and FSL's Randomise. FAT and FW values were extracted, averaged across all voxels that demonstrated group differences, and then utilized to test for the influence of age, medication, age of onset, duration of illness, symptom severity, and intelligence. RESULTS Individuals with adolescent-onset psychosis exhibited pronounced FW and FAT abnormalities compared to HCs. FAT reductions were spatially more widespread in ADO-SCZ. FW increases, however, were only present in psychotic ADO-BPD. In HCs, but not in individuals with adolescent-onset psychosis, FAT was positively related to age. CONCLUSIONS We observe evidence for cellular (FAT) and extracellular (FW) white matter abnormalities in adolescent-onset psychosis. Although cellular white matter abnormalities were more prominent in ADO-SCZ, such alterations may reflect a shared trait, i.e. neurodevelopmental pathology, present across the psychosis spectrum. Extracellular abnormalities were evident in psychotic ADO-BPD, potentially indicating a more dynamic, state-dependent brain reaction to psychosis.
Collapse
Affiliation(s)
- Johanna Seitz-Holland
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Felix L. Nägele
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Psychiatry Neuroimaging Branch, Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
| | - Marek Kubicki
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ofer Pasternak
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kang Ik K. Cho
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Morgan Hough
- SANE POWIC, University Department of Psychiatry, Warneford Hospital, Oxford, UK
- Highfield Unit, University Department of Psychiatry, Warneford Hospital, Oxford, UK
| | - Christoph Mulert
- Psychiatry Neuroimaging Branch, Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
- Centre for Psychiatry and Psychotherapy, Justus-Liebig-University, Giessen, Germany
| | - Martha E. Shenton
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy J. Crow
- SANE POWIC, University Department of Psychiatry, Warneford Hospital, Oxford, UK
| | - Anthony C. D. James
- SANE POWIC, University Department of Psychiatry, Warneford Hospital, Oxford, UK
- Highfield Unit, University Department of Psychiatry, Warneford Hospital, Oxford, UK
| | - Amanda E. Lyall
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Vecchio D, Piras F, Ciullo V, Piras F, Natalizi F, Ducci G, Ambrogi S, Spalletta G, Banaj N. Brain Network Topology in Deficit and Non-Deficit Schizophrenia: Application of Graph Theory to Local and Global Indices. J Pers Med 2023; 13:jpm13050799. [PMID: 37240969 DOI: 10.3390/jpm13050799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Patients with deficit schizophrenia (SZD) suffer from primary and enduring negative symptoms. Limited pieces of evidence and neuroimaging studies indicate they differ from patients with non-deficit schizophrenia (SZND) in neurobiological aspects, but the results are far from conclusive. We applied for the first time, graph theory analyses to discriminate local and global indices of brain network topology in SZD and SZND patients compared with healthy controls (HC). High-resolution T1-weighted images were acquired for 21 SZD patients, 21 SZND patients, and 21 HC to measure cortical thickness from 68 brain regions. Graph-based metrics (i.e., centrality, segregation, and integration) were computed and compared among groups, at both global and regional networks. When compared to HC, at the regional level, SZND were characterized by temporoparietal segregation and integration differences, while SZD showed widespread alterations in all network measures. SZD also showed less segregated network topology at the global level in comparison to HC. SZD and SZND differed in terms of centrality and integration measures in nodes belonging to the left temporoparietal cortex and to the limbic system. SZD is characterized by topological features in the network architecture of brain regions involved in negative symptomatology. Such results help to better define the neurobiology of SZD (SZD: Deficit Schizophrenia; SZND: Non-Deficit Schizophrenia; SZ: Schizophrenia; HC: healthy controls; CC: clustering coefficient; L: characteristic path length; E: efficiency; D: degree; CCnode: CC of a node; CCglob: the global CC of the network; Eloc: efficiency of the information transfer flow either within segregated subgraphs or neighborhoods nodes; Eglob: efficiency of the information transfer flow among the global network; FDA: Functional Data Analysis; and Dmin: estimated minimum densities).
Collapse
Affiliation(s)
- Daniela Vecchio
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Valentina Ciullo
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Federica Piras
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Federica Natalizi
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
- Department of Psychology, "Sapienza" University of Rome, Via dei Marsi 78, 00185 Rome, Italy
- PhD Program in Behavioral Neuroscience, Sapienza University of Rome, 00161 Rome, Italy
| | - Giuseppe Ducci
- Department of Mental Health, ASL Roma 1, 00135 Rome, Italy
| | - Sonia Ambrogi
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Gianfranco Spalletta
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nerisa Banaj
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| |
Collapse
|
25
|
Long X, Li L, Wang X, Cao Y, Wu B, Roberts N, Gong Q, Kemp GJ, Jia Z. Gray matter alterations in adolescent major depressive disorder and adolescent bipolar disorder. J Affect Disord 2023; 325:550-563. [PMID: 36669567 DOI: 10.1016/j.jad.2023.01.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 12/24/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023]
Abstract
BACKGROUND Gray matter volume (GMV) alterations in several emotion-related brain areas are implicated in mood disorders, but findings have been inconsistent in adolescents with major depressive disorder (MDD) or bipolar disorder (BD). METHODS We conducted a comprehensive meta-analysis of 35 region-of-interest (ROI) and 18 whole-brain voxel-based morphometry (VBM) MRI studies in adolescent MDD and adolescent BD, and indirectly compared the results in the two groups. The effects of age, sex, and other demographic and clinical scale scores were explored using meta-regression analysis. RESULTS In the ROI meta-analysis, right putamen volume was decreased in adolescents with MDD, while bilateral amygdala volume was decreased in adolescents with BD compared to healthy controls (HC). In the whole-brain VBM meta-analysis, GMV was increased in right middle frontal gyrus and decreased in left caudate in adolescents with MDD compared to HC, while in adolescents with BD, GMV was increased in left superior frontal gyrus and decreased in limbic regions compared with HC. MDD vs BD comparison revealed volume alteration in the prefrontal-limbic system. LIMITATION Different clinical features limit the comparability of the samples, and small sample size and insufficient clinical details precluded subgroup analysis or meta-regression analyses of these variables. CONCLUSIONS Distinct patterns of GMV alterations in adolescent MDD and adolescent BD could help to differentiate these two populations and provide potential diagnostic biomarkers.
Collapse
Affiliation(s)
- Xipeng Long
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu 610041, Sichuan, PR China; Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Lei Li
- Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, PR China; Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xiuli Wang
- Department of Clinical Psychiatry, the Fourth People's Hospital of Chengdu, Chengdu 610041, Sichuan, PR China
| | - Yuan Cao
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu 610041, Sichuan, PR China
| | - Baolin Wu
- Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, PR China; Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Neil Roberts
- The Queens Medical Research Institute (QMRI), School of Clinical Sciences, University of Edinburgh, Edinburgh, UK
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, PR China; Department of Radiology, West China Xiamen Hospital of Sichuan University, 699Jinyuan Xi Road, Jimei District, 361021 Xiamen, Fujian, PR China
| | - Graham J Kemp
- Liverpool Magnetic Resonance Imaging Center (LiMRIC) and Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu 610041, Sichuan, PR China; Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, PR China.
| |
Collapse
|
26
|
Curtis MT, Sklar AL, Coffman BA, Salisbury DF. Functional connectivity and gray matter deficits within the auditory attention circuit in first-episode psychosis. Front Psychiatry 2023; 14:1114703. [PMID: 36860499 PMCID: PMC9968732 DOI: 10.3389/fpsyt.2023.1114703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/24/2023] [Indexed: 02/16/2023] Open
Abstract
Background Selective attention deficits in first episode of psychosis (FEP) can be indexed by impaired attentional modulation of auditory M100. It is unknown if the pathophysiology underlying this deficit is restricted to auditory cortex or involves a distributed attention network. We examined the auditory attention network in FEP. Methods MEG was recorded from 27 FEP and 31 matched healthy controls (HC) while alternately ignoring or attending tones. A whole-brain analysis of MEG source activity during auditory M100 identified non-auditory areas with increased activity. Time-frequency activity and phase-amplitude coupling were examined in auditory cortex to identify the attentional executive carrier frequency. Attention networks were defined by phase-locking at the carrier frequency. Spectral and gray matter deficits in the identified circuits were examined in FEP. Results Attention-related activity was identified in prefrontal and parietal regions, markedly in precuneus. Theta power and phase coupling to gamma amplitude increased with attention in left primary auditory cortex. Two unilateral attention networks were identified with precuneus seeds in HC. Network synchrony was impaired in FEP. Gray matter thickness was reduced within the left hemisphere network in FEP but did not correlate with synchrony. Conclusion Several extra-auditory attention areas with attention-related activity were identified. Theta was the carrier frequency for attentional modulation in auditory cortex. Left and right hemisphere attention networks were identified, with bilateral functional deficits and left hemisphere structural deficits, though FEP showed intact auditory cortex theta phase-gamma amplitude coupling. These novel findings indicate attention-related circuitopathy early in psychosis potentially amenable to future non-invasive interventions.
Collapse
Affiliation(s)
| | | | | | - Dean F. Salisbury
- Clinical Neurophysiology Research Laboratory, Department of Psychiatry, Western Psychiatric Hospital, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
27
|
Du X, Hare S, Summerfelt A, Adhikari BM, Garcia L, Marshall W, Zan P, Kvarta M, Goldwaser E, Bruce H, Gao S, Sampath H, Kochunov P, Simon JZ, Hong LE. Cortical connectomic mediations on gamma band synchronization in schizophrenia. Transl Psychiatry 2023; 13:13. [PMID: 36653335 PMCID: PMC9849210 DOI: 10.1038/s41398-022-02300-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/07/2022] [Accepted: 12/22/2022] [Indexed: 01/20/2023] Open
Abstract
Aberrant gamma frequency neural oscillations in schizophrenia have been well demonstrated using auditory steady-state responses (ASSR). However, the neural circuits underlying 40 Hz ASSR deficits in schizophrenia remain poorly understood. Sixty-six patients with schizophrenia spectrum disorders and 85 age- and gender-matched healthy controls completed one electroencephalography session measuring 40 Hz ASSR and one imaging session for resting-state functional connectivity (rsFC) assessments. The associations between the normalized power of 40 Hz ASSR and rsFC were assessed via linear regression and mediation models. We found that rsFC among auditory, precentral, postcentral, and prefrontal cortices were positively associated with 40 Hz ASSR in patients and controls separately and in the combined sample. The mediation analysis further confirmed that the deficit of gamma band ASSR in schizophrenia was nearly fully mediated by three of the rsFC circuits between right superior temporal gyrus-left medial prefrontal cortex (MPFC), left MPFC-left postcentral gyrus (PoG), and left precentral gyrus-right PoG. Gamma-band ASSR deficits in schizophrenia may be associated with deficient circuitry level connectivity to support gamma frequency synchronization. Correcting gamma band deficits in schizophrenia may require corrective interventions to normalize these aberrant networks.
Collapse
Affiliation(s)
- Xiaoming Du
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Stephanie Hare
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ann Summerfelt
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bhim M Adhikari
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Laura Garcia
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wyatt Marshall
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peng Zan
- Department of Electrical & Computer Engineering, University of Maryland, College Park, MD, USA
| | - Mark Kvarta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eric Goldwaser
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Heather Bruce
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Si Gao
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hemalatha Sampath
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jonathan Z Simon
- Department of Electrical & Computer Engineering, University of Maryland, College Park, MD, USA
- Department of Biology, University of Maryland, College Park, MD, USA
- Institute for Systems Research, University of Maryland, College Park, MD, USA
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Kia SM, Huijsdens H, Rutherford S, de Boer A, Dinga R, Wolfers T, Berthet P, Mennes M, Andreassen OA, Westlye LT, Beckmann CF, Marquand AF. Closing the life-cycle of normative modeling using federated hierarchical Bayesian regression. PLoS One 2022; 17:e0278776. [PMID: 36480551 PMCID: PMC9731431 DOI: 10.1371/journal.pone.0278776] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Clinical neuroimaging data availability has grown substantially in the last decade, providing the potential for studying heterogeneity in clinical cohorts on a previously unprecedented scale. Normative modeling is an emerging statistical tool for dissecting heterogeneity in complex brain disorders. However, its application remains technically challenging due to medical data privacy issues and difficulties in dealing with nuisance variation, such as the variability in the image acquisition process. Here, we approach the problem of estimating a reference normative model across a massive population using a massive multi-center neuroimaging dataset. To this end, we introduce a federated probabilistic framework using hierarchical Bayesian regression (HBR) to complete the life-cycle of normative modeling. The proposed model provides the possibilities to learn, update, and adapt the model parameters on decentralized neuroimaging data. Our experimental results confirm the superiority of HBR in deriving more accurate normative ranges on large multi-site neuroimaging datasets compared to the current standard methods. In addition, our approach provides the possibility to recalibrate and reuse the learned model on local datasets and even on datasets with very small sample sizes. The proposed method will facilitate applications of normative modeling as a medical tool for screening the biological deviations in individuals affected by complex illnesses such as mental disorders.
Collapse
Affiliation(s)
- Seyed Mostafa Kia
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department for Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hester Huijsdens
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Saige Rutherford
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department for Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Augustijn de Boer
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Richard Dinga
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department for Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas Wolfers
- Department of Psychology, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Pierre Berthet
- Department of Psychology, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Maarten Mennes
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Lars T. Westlye
- Department of Psychology, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Christian F. Beckmann
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department for Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
- Centre for Functional MRI of the Brain, University of Oxford, Oxford, United Kingdom
| | - Andre F. Marquand
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department for Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Neuroimaging, Institute of Psychiatry, King’s College London, London, United Kingdom
| |
Collapse
|
29
|
Mendelian randomization analyses support causal relationships between brain imaging-derived phenotypes and risk of psychiatric disorders. Nat Neurosci 2022; 25:1519-1527. [PMID: 36216997 DOI: 10.1038/s41593-022-01174-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/29/2022] [Indexed: 01/13/2023]
Abstract
Observational studies have reported the correlations between brain imaging-derived phenotypes (IDPs) and psychiatric disorders; however, whether the relationships are causal is uncertain. We conducted bidirectional two-sample Mendelian randomization (MR) analyses to explore the causalities between 587 reliable IDPs (N = 33,224 individuals) and 10 psychiatric disorders (N = 9,725 to 161,405). We identified nine IDPs for which there was evidence of a causal influence on risk of schizophrenia, anorexia nervosa and bipolar disorder. For example, 1 s.d. increase in the orientation dispersion index of the forceps major was associated with 32% lower odds of schizophrenia risk. Reverse MR indicated that only genetically predicted schizophrenia was positively associated with two IDPs, the cortical surface area and the volume of the right pars orbitalis. We established the BrainMR database ( http://www.bigc.online/BrainMR/ ) to share our results. Our findings provide potential strategies for the prediction and intervention for psychiatric disorder risk at the brain-imaging level.
Collapse
|
30
|
Ghosh A, Kaur S, Shah R, Oomer F, Avasthi A, Ahuja CK, Basu D, Nehra R, Khandelwal N. Surface-based brain morphometry in schizophrenia vs. cannabis-induced psychosis: A controlled comparison. J Psychiatr Res 2022; 155:286-294. [PMID: 36170756 DOI: 10.1016/j.jpsychires.2022.09.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/19/2022] [Accepted: 09/16/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND & AIM We examined group differences in cortical thickness and surface-parameters among age and handedness--matched persons with cannabis-induced psychosis (CIP), schizophrenia with heavy cannabis use (SZC), and healthy controls (HC). METHODS We recruited 31 men with SZC, 28 with CIP, and 30 with HC. We used the Psychiatric Research Interview for Substance and Mental Disorders to differentiate between CIP and SZC. We processed and analyzed T1 MR images using the Surface-based Brain Morphometry (SBM) pipeline of the CAT-12 toolbox within the statistical parametric mapping. After pre-processing, volumes were segmented using surface and thickness estimation for the analysis of the region of interest. We used the projection-based thickness method to assess the cortical thickness and Desikan-Killiany atlas for cortical parcellation. RESULTS We observed the lowest cortical thickness, depth, and gyrification in the SZC, followed by CIP and the control groups. The differences were predominantly seen in frontal cortices, with limited parietal and temporal regions involvement. After False Discovery Rate (FDR) corrections and post-hoc analysis, SZC had reduced cortical thickness than HC in the middle and inferior frontal, right entorhinal, and left postcentral regions. Cortical thickness of SZC was also significantly lower than CIP in bilateral postcentral and right middle frontal regions. We found negative correlations (after FDR corrections) between the duration of cannabis use and cortical thickness in loci of parietal and occipital cortices. CONCLUSION Our study suggested cortical structural abnormalities in schizophrenia, in reference to healthy controls and cannabis-induced psychosis, indicating different pathophysiology of SZC and CIP.
Collapse
Affiliation(s)
- Abhishek Ghosh
- Drug De-addiction and Treatment Centre, Department of Psychiatry, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Simranjit Kaur
- Thapar Institute of Engineering and Technology, Punjab, India
| | - Raghav Shah
- Drug De-addiction and Treatment Centre, Department of Psychiatry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Fareed Oomer
- Chasefarm Hospital, Barnet, Enfield & Haringey Mental Health Trust, Enfield, UK
| | - Ajit Avasthi
- Department of Psychiatry, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Chirag K Ahuja
- Department of Radio-diagnosis and Imaging, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Debasish Basu
- Chasefarm Hospital, Barnet, Enfield & Haringey Mental Health Trust, Enfield, UK
| | - Ritu Nehra
- Department of Psychiatry, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Niranjan Khandelwal
- Department of Radio-diagnosis and Imaging, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
31
|
Kochunov P, Ma Y, Hatch KS, Gao S, Jahanshad N, Thompson PM, Adhikari BM, Bruce H, Van der vaart A, Goldwaser EL, Sotiras A, Kvarta MD, Ma T, Chen S, Nichols TE, Hong LE. Brain-wide versus genome-wide vulnerability biomarkers for severe mental illnesses. Hum Brain Mapp 2022; 43:4970-4983. [PMID: 36040723 PMCID: PMC9582367 DOI: 10.1002/hbm.26056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 01/06/2023] Open
Abstract
Severe mental illnesses (SMI), including major depressive (MDD), bipolar (BD), and schizophrenia spectrum (SSD) disorders have multifactorial risk factors and capturing their complex etiopathophysiology in an individual remains challenging. Regional vulnerability index (RVI) was used to measure individual's brain-wide similarity to the expected SMI patterns derived from meta-analytical studies. It is analogous to polygenic risk scores (PRS) that measure individual's similarity to genome-wide patterns in SMI. We hypothesized that RVI is an intermediary phenotype between genome and symptoms and is sensitive to both genetic and environmental risks for SMI. UK Biobank sample of N = 17,053/19,265 M/F (age = 64.8 ± 7.4 years) and an independent sample of SSD patients and controls (N = 115/111 M/F, age = 35.2 ± 13.4) were used to test this hypothesis. UKBB participants with MDD had significantly higher RVI-MDD (Cohen's d = 0.20, p = 1 × 10-23 ) and PRS-MDD (d = 0.17, p = 1 × 10-15 ) than nonpsychiatric controls. UKBB participants with BD and SSD showed significant elevation in the respective RVIs (d = 0.65 and 0.60; p = 3 × 10-5 and .009, respectively) and PRS (d = 0.57 and 1.34; p = .002 and .002, respectively). Elevated RVI-SSD were replicated in an independent sample (d = 0.53, p = 5 × 10-5 ). RVI-MDD and RVI-SSD but not RVI-BD were associated with childhood adversity (p < .01). In nonpsychiatric controls, elevation in RVI and PRS were associated with lower cognitive performance (p < 10-5 ) in six out of seven domains and showed specificity with disorder-associated deficits. In summary, the RVI is a novel brain index for SMI and shows similar or better specificity for SMI than PRS, and together they may complement each other in the efforts to characterize the genomic to brain level risks for SMI.
Collapse
Affiliation(s)
- Peter Kochunov
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Yizhou Ma
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Kathryn S. Hatch
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Si Gao
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Neda Jahanshad
- Imaging Genetics Center, Stevens Neuroimaging & Informatics InstituteKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Paul M. Thompson
- Imaging Genetics Center, Stevens Neuroimaging & Informatics InstituteKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Bhim M. Adhikari
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Heather Bruce
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Andrew Van der vaart
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Eric L. Goldwaser
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Aris Sotiras
- Institute of Informatics, University of WashingtonSchool of MedicineSt. LouisMissouriUSA
| | - Mark D. Kvarta
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Tianzhou Ma
- Department of Epidemiology and BiostatisticsUniversity of MarylandCollege ParkMarylandUSA
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Thomas E. Nichols
- Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - L. Elliot Hong
- Maryland Psychiatric Research Center, Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
32
|
Nian R, Gao M, Zhang S, Yu J, Gholipour A, Kong S, Wang R, Sui Y, Velasco-Annis C, Tomas-Fernandez X, Li Q, Lv H, Qian Y, Warfield SK. Toward evaluation of multiresolution cortical thickness estimation with FreeSurfer, MaCRUISE, and BrainSuite. Cereb Cortex 2022; 33:5082-5096. [PMID: 36288912 DOI: 10.1093/cercor/bhac401] [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: 06/06/2022] [Revised: 09/09/2022] [Accepted: 09/11/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
Advances in Magnetic Resonance Imaging hardware and methodologies allow for promoting the cortical morphometry with submillimeter spatial resolution. In this paper, we generated 3D self-enhanced high-resolution (HR) MRI imaging, by adapting 1 deep learning architecture, and 3 standard pipelines, FreeSurfer, MaCRUISE, and BrainSuite, have been collectively employed to evaluate the cortical thickness. We systematically investigated the differences in cortical thickness estimation for MRI sequences at multiresolution homologously originated from the native image. It has been revealed that there systematically exhibited the preferences in determining both inner and outer cortical surfaces at higher resolution, yielding most deeper cortical surface placements toward GM/WM or GM/CSF boundaries, which directs a consistent reduction tendency of mean cortical thickness estimation; on the contrary, the lower resolution data will most probably provide a more coarse and rough evaluation in cortical surface reconstruction, resulting in a relatively thicker estimation. Although the differences of cortical thickness estimation at the diverse spatial resolution varied with one another, almost all led to roughly one-sixth to one-fifth significant reduction across the entire brain at the HR, independent to the pipelines we applied, which emphasizes on generally coherent improved accuracy in a data-independent manner and endeavors to cost-efficiency with quantitative opportunities.
Collapse
Affiliation(s)
- Rui Nian
- School of Electronic Engineering, Ocean University of China, 238 Songling Road, Qingdao, China
- Harvard Medical School, 25 Shattuck Street, Boston, MA, United States
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, United States
| | - Mingshan Gao
- Citigroup Services and Technology Limited, 1000 Chenhi Road, Shanghai, China
| | | | - Junjie Yu
- School of Electronic Engineering, Ocean University of China, 238 Songling Road, Qingdao, China
| | - Ali Gholipour
- Harvard Medical School, 25 Shattuck Street, Boston, MA, United States
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, United States
| | - Shuang Kong
- School of Electronic Engineering, Ocean University of China, 238 Songling Road, Qingdao, China
| | - Ruirui Wang
- School of Electronic Engineering, Ocean University of China, 238 Songling Road, Qingdao, China
| | - Yao Sui
- Harvard Medical School, 25 Shattuck Street, Boston, MA, United States
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, United States
| | - Clemente Velasco-Annis
- Harvard Medical School, 25 Shattuck Street, Boston, MA, United States
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, United States
| | - Xavier Tomas-Fernandez
- Harvard Medical School, 25 Shattuck Street, Boston, MA, United States
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, United States
| | - Qiuying Li
- School of Electronic Engineering, Ocean University of China, 238 Songling Road, Qingdao, China
| | - Hangyu Lv
- School of Electronic Engineering, Ocean University of China, 238 Songling Road, Qingdao, China
| | - Yuqi Qian
- School of Electronic Engineering, Ocean University of China, 238 Songling Road, Qingdao, China
| | - Simon K Warfield
- Harvard Medical School, 25 Shattuck Street, Boston, MA, United States
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, United States
| |
Collapse
|
33
|
Zhou H, Wang D, Cao B, Zhang X. Association of reduced cortical thickness and psychopathological symptoms in patients with first-episode drug-naïve schizophrenia. Int J Psychiatry Clin Pract 2022; 27:42-50. [PMID: 36193901 DOI: 10.1080/13651501.2022.2129067] [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] [Indexed: 10/10/2022]
Abstract
OBJECTIVE There is growing evidence that reduced cortical thickness has been considered to be a central abnormality in schizophrenia. Brain imaging studies have demonstrated that the cerebral cortex becomes thinner in patients with first-episode schizophrenia. This study aimed to examine whether cortical thickness is altered in drug-naïve schizophrenia in a Chinese Han population and the relationship between cortical thickness and clinical symptoms. METHODS We compared cortical thickness in 41 schizophrenia patients and 30 healthy controls. Psychopathology of patients with schizophrenia was assessed using the Positive and Negative Syndrome Scale (PANSS). RESULTS The cortical thickness of left banks of superior temporal sulcus, left lateral occipital gyrus, left rostral middle frontal gyrus, right inferior parietal lobule and right lateral occipital gyrus in schizophrenia patients was generally thinner compared with healthy controls. Correlation analysis revealed a negative correlation between cortical thickness of the left banks of superior temporal sulcus and general psychopathology of PANSS. CONCLUSIONS Our results suggest that cortical thickness abnormalities are already present early in the onset of schizophrenia and are associated with psychopathological symptoms, suggesting that it plays an important role in the pathogenesis and symptomatology of schizophrenia.Key points(1) The first-episode drug-naïve schizophrenia had reduced cortical thickness than the controls.(2) Cortical thickness was associated with psychopathological symptoms in patients with schizophrenia.
Collapse
Affiliation(s)
- Huixia Zhou
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, PR China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, PR China
| | - Dongmei Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, PR China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, PR China
| | - Bo Cao
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.,Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiangyang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, PR China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
34
|
Ringin E, Cropley V, Zalesky A, Bruggemann J, Sundram S, Weickert CS, Weickert TW, Bousman CA, Pantelis C, Van Rheenen TE. The impact of smoking status on cognition and brain morphology in schizophrenia spectrum disorders. Psychol Med 2022; 52:3097-3115. [PMID: 33443010 DOI: 10.1017/s0033291720005152] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Cigarette smoking is associated with worse cognition and decreased cortical volume and thickness in healthy cohorts. Chronic cigarette smoking is prevalent in schizophrenia spectrum disorders (SSD), but the effects of smoking status on the brain and cognition in SSD are not clear. This study aimed to understand whether cognitive performance and brain morphology differed between smoking and non-smoking individuals with SSD compared to healthy controls. METHODS Data were obtained from the Australian Schizophrenia Research Bank. Cognitive functioning was measured in 299 controls and 455 SSD patients. Cortical volume, thickness and surface area data were analysed from T1-weighted structural scans obtained in a subset of the sample (n = 82 controls, n = 201 SSD). Associations between smoking status (cigarette smoker/non-smoker), cognition and brain morphology were tested using analyses of covariance, including diagnosis as a moderator. RESULTS No smoking by diagnosis interactions were evident, and no significant differences were revealed between smokers and non-smokers across any of the variables measured, with the exception of a significantly thinner left posterior cingulate in smokers compared to non-smokers. Several main effects of smoking in the cognitive, volume and thickness analyses were initially significant but did not survive false discovery rate (FDR) correction. CONCLUSIONS Despite the general absence of significant FDR-corrected findings, trend-level effects suggest the possibility that subtle smoking-related effects exist but were not uncovered due to low statistical power. An investigation of this topic is encouraged to confirm and expand on our findings.
Collapse
Affiliation(s)
- Elysha Ringin
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
| | - Vanessa Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- Centre for Mental Health, Faculty of Health, Arts and Design, School of Health Sciences, Swinburne University, Melbourne, Australia
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- Department of Electrical and Electronic Engineering, University of Melbourne, Melbourne, VIC, Australia
| | - Jason Bruggemann
- School of Psychiatry, University of New South Wales, New South Wales, Australia
- Neuroscience Research Australia, New South Wales, Australia
| | - Suresh Sundram
- Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Australia
- Mental Health Program, Monash Health, Clayton, Victoria, Australia
| | - Cynthia Shannon Weickert
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- School of Psychiatry, University of New South Wales, New South Wales, Australia
- Neuroscience Research Australia, New South Wales, Australia
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, New York 13210, USA
| | - Thomas W Weickert
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- School of Psychiatry, University of New South Wales, New South Wales, Australia
- Neuroscience Research Australia, New South Wales, Australia
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, New York 13210, USA
| | - Chad A Bousman
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- Departments of Medical Genetics, Psychiatry, and Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Tamsyn E Van Rheenen
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- Centre for Mental Health, Faculty of Health, Arts and Design, School of Health Sciences, Swinburne University, Melbourne, Australia
| |
Collapse
|
35
|
Lei D, Li W, Tallman MJ, Strakowski SM, DelBello MP, Rodrigo Patino L, Fleck DE, Lui S, Gong Q, Sweeney JA, Strawn JR, Nery FG, Welge JA, Rummelhoff E, Adler CM. Changes in the structural brain connectome over the course of a nonrandomized clinical trial for acute mania. Neuropsychopharmacology 2022; 47:1961-1968. [PMID: 35585125 PMCID: PMC9485114 DOI: 10.1038/s41386-022-01328-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/17/2022] [Accepted: 04/11/2022] [Indexed: 02/05/2023]
Abstract
Disrupted topological organization of brain functional networks has been widely reported in bipolar disorder. However, the potential clinical implications of structural connectome abnormalities have not been systematically investigated. The present study included 109 unmedicated subjects with acute mania who were assigned to 8 weeks of treatment with quetiapine or lithium and 60 healthy controls. High resolution 3D-T1 weighted magnetic resonance images (MRI) were collected from both groups at baseline, week 1 and week 8. Brain networks were constructed based on the similarity of morphological features across brain regions and analyzed using graph theory approaches. At baseline, individuals with bipolar disorder illness showed significantly lower clustering coefficient (Cp) (p = 0.012) and normalized characteristic path length (λ) (p = 0.004) compared to healthy individuals, as well as differences in nodal centralities across multiple brain regions. No baseline or post-treatment differences were identified between drug treatment conditions, so change after treatment were considered in the combined treatment groups. Relative to healthy individuals, differences in Cp, λ and cingulate gyrus nodal centrality were significantly reduced with treatment; changes in these parameters correlated with changes in Young Mania Rating Scale scores. Baseline structural connectome matrices significantly differentiated responder and non-responder groups at 8 weeks with 74% accuracy. Global and nodal network alterations evident at baseline were normalized with treatment and these changes associated with symptomatic improvement. Further, baseline structural connectome matrices predicted treatment response. These findings suggest that structural connectome abnormalities are clinically significant and may be useful for predicting clinical outcome of treatment and tracking drug effects on brain anatomy in bipolar disorder. CLINICAL TRIALS REGISTRATION Name: Functional and Neurochemical Brain Changes in First-episode Bipolar Mania Following Successful Treatment with Lithium or Quetiapine. URL: https://clinicaltrials.gov/ . REGISTRATION NUMBER NCT00609193. Name: Neurofunctional and Neurochemical Markers of Treatment Response in Bipolar Disorder. URL: https://clinicaltrials.gov/ . REGISTRATION NUMBER NCT00608075.
Collapse
Affiliation(s)
- Du Lei
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA.
| | - Wenbin Li
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
- Huaxi MR Research Center (HMRRC), Department of Radiology, The Center for Medical Imaging, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China
- Department of the Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Maxwell J Tallman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
| | - Stephen M Strakowski
- Department of Psychiatry & Behavioral Sciences, Dell Medical School of The University of Texas at Austin, Austin, 78712, TX, USA
| | - Melissa P DelBello
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
| | - L Rodrigo Patino
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
| | - David E Fleck
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
| | - Su Lui
- Huaxi MR Research Center (HMRRC), Department of Radiology, The Center for Medical Imaging, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, The Center for Medical Imaging, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China
| | - John A Sweeney
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
- Huaxi MR Research Center (HMRRC), Department of Radiology, The Center for Medical Imaging, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China
| | - Jeffrey R Strawn
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
| | - Fabiano G Nery
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
| | - Jeffrey A Welge
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
| | - Emily Rummelhoff
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
| | - Caleb M Adler
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, 45219, OH, USA
| |
Collapse
|
36
|
Uenishi S, Tamaki A, Yamada S, Yasuda K, Ikeda N, Mizutani-Tiebel Y, Keeser D, Padberg F, Tsuji T, Kimoto S, Takahashi S. Computational modeling of electric fields for prefrontal tDCS across patients with schizophrenia and mood disorders. Psychiatry Res Neuroimaging 2022; 326:111547. [PMID: 36240572 DOI: 10.1016/j.pscychresns.2022.111547] [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: 10/25/2021] [Revised: 07/30/2022] [Accepted: 10/01/2022] [Indexed: 02/25/2023]
Abstract
This cross-diagnostic study aims to computationally model electric field (efield) for prefrontal transcranial direct current stimulation in mood disorders and schizophrenia. Enrolled were patients with major depressive disorder (n = 23), bipolar disorder (n = 24), schizophrenia (n = 23), and healthy controls (n = 23). The efield was simulated using SimNIBS software (ver.2.1.1). Electrodes were placed at the left and right prefrontal areas and the current intensity was set to 2 mA intensity. Schizophrenia and major depressive disorder groups showed significantly lower 99.5th percentile efield strength than healthy controls. In voxel-wise analysis, patients with schizophrenia showed a significant reduction of simulated efield strength in the bilateral frontal lobe, cerebellum and brain stem compared with healthy controls. Among the patients with schizophrenia, reduction of simulated efield strength was not significantly correlated with psychiatric symptoms or global functioning. The patients with bipolar disorder showed no significant difference in simulated efield strength compared with healthy controls, and there was no significant difference between the clinical groups. Our results suggest attenuated electrophysiological response to transcranial direct current stimulation to the prefrontal cortex in patients with schizophrenia, and to some extent in patients with major depressive disorder.
Collapse
Affiliation(s)
- Shinya Uenishi
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan; Department of Psychiatry, Hidaka Hospital, Gobo, Japan.
| | - Atsushi Tamaki
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan; Department of Psychiatry, Hidaka Hospital, Gobo, Japan
| | - Shinichi Yamada
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan
| | - Kasumi Yasuda
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan
| | - Natsuko Ikeda
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan; Department of Psychiatry, Wakayama Prefectural Mental Health Care Center, Aridagawa, Japan
| | - Yuki Mizutani-Tiebel
- Department of Psychiatry and Psychotherapy, University Hospital LMU Munich, Munich, Germany
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, University Hospital LMU Munich, Munich, Germany; Department of Radiology, University Hospital LMU Munich, Munich, Germany
| | - Frank Padberg
- Department of Psychiatry and Psychotherapy, University Hospital LMU Munich, Munich, Germany
| | - Tomikimi Tsuji
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan
| | - Sohei Kimoto
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan
| | - Shun Takahashi
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan; Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan; Graduate School of Rehabilitation Science, Osaka Metropolitan University, Habikino, Japan; Clinical Research and Education Center, Asakayama General Hospital, Sakai, Japan
| |
Collapse
|
37
|
Allebone J, Wilson SJ, Bradlow RCJ, Maller J, O'Brien T, Mullen SA, Cook M, Adams SJ, Vogrin S, Vaughan DN, Connelly A, Kwan P, Berkovic SF, D'Souza WJ, Jackson G, Velakoulis D, Kanaan RA. Increased cortical thickness in nodes of the cognitive control and default mode networks in psychosis of epilepsy. Seizure 2022; 101:244-252. [PMID: 36116283 DOI: 10.1016/j.seizure.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/07/2022] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE To explore the cortical morphological associations of the psychoses of epilepsy. METHODS Psychosis of epilepsy (POE) has two main subtypes - postictal psychosis and interictal psychosis. We used automated surface-based analysis of magnetic resonance images to compare cortical thickness, area, and volume across the whole brain between: (i) all patients with POE (n = 23) relative to epilepsy-without psychosis controls (EC; n = 23), (ii) patients with interictal psychosis (n = 10) or postictal psychosis (n = 13) relative to EC, and (iii) patients with postictal psychosis (n = 13) relative to patients with interictal psychosis (n = 10). RESULTS POE is characterised by cortical thickening relative to EC, occurring primarily in nodes of the cognitive control network; (rostral anterior cingulate, caudal anterior cingulate, middle frontal gyrus), and the default mode network (posterior cingulate, medial paracentral gyrus, and precuneus). Patients with interictal psychosis displayed cortical thickening in the left hemisphere in occipital and temporal regions relative to EC (lateral occipital cortex, lingual, fusiform, and inferior temporal gyri), which was evident to a lesser extent in postictal psychosis patients. There were no significant differences in cortical thickness, area, or volume between the postictal psychosis and EC groups, or between the postictal psychosis and interictal psychosis groups. However, prior to correction for multiple comparisons, both the interictal psychosis and postictal psychosis groups displayed cortical thickening relative to EC in highly similar regions to those identified in the POE group overall. SIGNIFICANCE The results show cortical thickening in POE overall, primarily in nodes of the cognitive control and default mode networks, compared to patients with epilepsy without psychosis. Additional thickening in temporal and occipital neocortex implicated in the dorsal and ventral visual pathways may differentiate interictal psychosis from postictal psychosis. A novel mechanism for cortical thickening in POE is proposed whereby normal synaptic pruning processes are interrupted by seizure onset.
Collapse
Affiliation(s)
- James Allebone
- Melbourne School of Psychological Sciences, University of Melbourne, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Sarah J Wilson
- Melbourne School of Psychological Sciences, University of Melbourne, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Comprehensive Epilepsy Program, Austin Health, University of Melbourne, Victoria, Australia
| | | | - Jerome Maller
- ANU College of Health and Medicine, Australian National University, Canberra, Victoria, Australia; Monash Alfred Psychiatry Research Centre, The Alfred and Monash University, Melbourne, Australia
| | - Terry O'Brien
- Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Neuroscience, Alfred Hospital, Monash University, Melbourne, Australia
| | - Saul A Mullen
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Mark Cook
- Graeme Clark Institute, University of Melbourne, Melbourne, Australia
| | - Sophia J Adams
- Department of Psychiatry, Austin Health, University of Melbourne, Melbourne, Australia
| | - Simon Vogrin
- St Vincent's Hospital, Melbourne, Victoria, Australia
| | - David N Vaughan
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Comprehensive Epilepsy Program, Austin Health, University of Melbourne, Victoria, Australia
| | - Alan Connelly
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Comprehensive Epilepsy Program, Austin Health, University of Melbourne, Victoria, Australia
| | - Patrick Kwan
- Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Neuroscience, Alfred Hospital, Monash University, Melbourne, Australia
| | - Samuel F Berkovic
- Comprehensive Epilepsy Program, Austin Health, University of Melbourne, Victoria, Australia
| | - Wendyl J D'Souza
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Australia
| | - Graeme Jackson
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Comprehensive Epilepsy Program, Austin Health, University of Melbourne, Victoria, Australia
| | - Dennis Velakoulis
- Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Melbourne Neuropsychiatry Centre, University of Melbourne and Melbourne Health, Melbourne, Australia
| | - Richard A Kanaan
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Department of Psychiatry, Austin Health, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
38
|
Choi KW, Han KM, Kim A, Kang W, Kang Y, Tae WS, Ham BJ. Decreased cortical gyrification in patients with bipolar disorder. Psychol Med 2022; 52:2232-2244. [PMID: 33190651 DOI: 10.1017/s0033291720004079] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND An aberrant neural connectivity has been known to be associated with bipolar disorder (BD). Local gyrification may reflect the early neural development of cortical connectivity and has been studied as a possible endophenotype of psychiatric disorders. This study aimed to investigate differences in the local gyrification index (LGI) in each cortical region between patients with BD and healthy controls (HCs). METHODS LGI values, as measured using FreeSurfer software, were compared between 61 patients with BD and 183 HCs. The values were also compared between patients with BD type I and type II as a sub-group analysis. Furthermore, we evaluated whether there was a correlation between LGI values and illness duration or depressive symptom severity in patients with BD. RESULTS Patients with BD showed significant hypogyria in various cortical regions, including the left inferior frontal gyrus (pars opercularis), precentral gyrus, postcentral gyrus, superior temporal cortex, insula, right entorhinal cortex, and both transverse temporal cortices, compared to HCs after the Bonferroni correction (p < 0.05/66, 0.000758). LGI was not associated with clinical factors such as illness duration, depressive symptom severity, and lithium treatment. No significant differences in cortical gyrification according to the BD subtype were found. CONCLUSIONS BD appears to be characterized by a significant regionally localized hypogyria, in various cortical areas. This abnormality may be a structural and developmental endophenotype marking the risk for BD, and it might help to clarify the etiology of BD.
Collapse
Affiliation(s)
- Kwan Woo Choi
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyu-Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Aram Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Wooyoung Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Youbin Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo-Suk Tae
- Brain Convergence Research Center, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- Brain Convergence Research Center, Korea University Anam Hospital, Seoul, Republic of Korea
| |
Collapse
|
39
|
Xie Y, Ding H, Du X, Chai C, Wei X, Sun J, Zhuo C, Wang L, Li J, Tian H, Liang M, Zhang S, Yu C, Qin W. Morphometric Integrated Classification Index: A Multisite Model-Based, Interpretable, Shareable and Evolvable Biomarker for Schizophrenia. Schizophr Bull 2022; 48:1217-1227. [PMID: 35925032 PMCID: PMC9673259 DOI: 10.1093/schbul/sbac096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND HYPOTHESIS Multisite massive schizophrenia neuroimaging data sharing is becoming critical in understanding the pathophysiological mechanism and making an objective diagnosis of schizophrenia; it remains challenging to obtain a generalizable and interpretable, shareable, and evolvable neuroimaging biomarker for schizophrenia diagnosis. STUDY DESIGN A Morphometric Integrated Classification Index (MICI) was proposed as a potential biomarker for schizophrenia diagnosis based on structural magnetic resonance imaging data of 1270 subjects from 10 sites (588 schizophrenia patients and 682 normal controls). An optimal XGBoost classifier plus sample-weighted SHapley Additive explanation algorithms were used to construct the MICI measure. STUDY RESULTS The MICI measure achieved comparable performance with the sample-weighted ensembling model and merged model based on raw data (Delong test, P > 0.82) while outperformed the single-site models (Delong test, P < 0.05) in either the independent-sample testing datasets from the 9 sites or the independent-site dataset (generalizable). Besides, when new sites were embedded in, the performance of this measure was gradually increasing (evolvable). Finally, MICI was strongly associated with the severity of schizophrenia brain structural abnormality, with the patients' positive and negative symptoms, and with the brain expression profiles of schizophrenia risk genes (interpretable). CONCLUSIONS In summary, the proposed MICI biomarker may provide a simple and explainable way to support clinicians for objectively diagnosing schizophrenia. Finally, we developed an online model share platform to promote biomarker generalization and provide free individual prediction services (http://micc.tmu.edu.cn/mici/index.html).
Collapse
Affiliation(s)
- Yingying Xie
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Ding
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China,School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Xiaotong Du
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Chai
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaotong Wei
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Jie Sun
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Chuanjun Zhuo
- Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin, China
| | - Lina Wang
- Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin, China
| | - Jie Li
- Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin, China
| | | | - Meng Liang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China,School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | | | | | - Wen Qin
- To whom correspondence should be addressed; Department of Radiology, and Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital. Anshan Road No 154, Heping District, Tianjin 300052, China.
| |
Collapse
|
40
|
Luckhoff HK, Asmal L, Scheffler F, Phahladira L, Smit R, van den Heuvel L, Fouche JP, Seedat S, Emsley R, du Plessis S. Associations between BMI and brain structures involved in food intake regulation in first-episode schizophrenia spectrum disorders and healthy controls. J Psychiatr Res 2022; 152:250-259. [PMID: 35753245 DOI: 10.1016/j.jpsychires.2022.06.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/04/2022] [Accepted: 06/10/2022] [Indexed: 11/28/2022]
Abstract
Structural brain differences have been described in first-episode schizophrenia spectrum disorders (FES), and often overlap with those evident in the metabolic syndrome (MetS). We examined the associations between body mass index (BMI) and brain structures involved in food intake regulation in minimally treated FES patients (n = 117) compared to healthy controls (n = 117). The effects of FES diagnosis, BMI and their interactions on our selected prefrontal cortical thickness and subcortical gray matter volume regions of interest (ROIs) were investigated with hierarchical multivariate regressions, followed by post-hoc regressions for the individual ROIs. In a secondary analysis, we examined the relationships of other MetS risk factors and psychopathology with the brain ROIs. Both illness and BMI significantly predicted the grouped prefrontal cortical thickness ROIs, whereas only BMI predicted the grouped subcortical volume ROIs. For the individual ROIs, schizophrenia diagnosis predicted thinner left and right frontal pole and right lateral OFC thickness, and increased BMI predicted thinner left and right caudal ACC thickness. There were no significant main or interaction effects for diagnosis and BMI on any of the individual subcortical volume ROIs. Secondary analyses suggest associations between several brain ROIs and individual MetS risk factors, but not with psychopathology. Our findings indicate differential, independent effects for FES diagnosis and BMI on brain structures. Limited evidence suggests that the BMI effects are more prominent in FES. Exploratory analyses suggest associations between other MetS risk factors and some brain ROIs.
Collapse
Affiliation(s)
- H K Luckhoff
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa.
| | - L Asmal
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa
| | - F Scheffler
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa
| | - L Phahladira
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa
| | - R Smit
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa
| | - L van den Heuvel
- South African Medical Research Council, Stellenbosch University Genomics of Brain Disorders Research Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa
| | - J P Fouche
- South African Medical Research Council, Stellenbosch University Genomics of Brain Disorders Research Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa
| | - S Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa
| | - R Emsley
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa
| | - S du Plessis
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, 7550, South Africa
| |
Collapse
|
41
|
Rani T, Behl T, Sharma N, Makeen HA, Albratty M, Alhazmi HA, Meraya AM, Bhatia S, Bungau SG. Exploring the role of biologics in depression. Cell Signal 2022; 98:110409. [PMID: 35843573 DOI: 10.1016/j.cellsig.2022.110409] [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: 06/21/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 11/03/2022]
Abstract
Depression is a chronic and prevalent neuropsychiatric disorder; clinical symptoms include excessive sad mood, anhedonia, increased anxiety, disturbed sleep, and cognitive deficits. The exact etiopathogenesis of depression is not well understood. Studies have suggested that tumor necrosis factor-alpha (TNF-α) and interleukins (ILs) perform vital roles in the pathogenesis and treatment of depression. Increasing evidence suggests the upregulation of TNF-α and ILs expression in patients with depression. Therefore, biologics like TNF inhibitors (etanercept, infliximab, adalimumab) and IL inhibitors (ustekinumab) have become key compounds in the treatment of depression. Interestingly, treatment with an antidepressant has been found to decrease the TNF-α level and improve depression-like behaviors in several preclinical and clinical studies. In the current article, we have reviewed the recent findings linking TNF-α and the pathogenesis of depression proving TNF-α inhibitors as potential new therapeutic agents. Animal models and clinical studies further support that TNF-α inhibitors are effective in ameliorating depression-like behaviors. Moreover, studies showed that peripheral injection of TNF-α exhibits depressive symptoms. These symptoms have been improved by treatment with TNF-α inhibitors. Hence suggesting TNF-α inhibitors as potential new antidepressants for the management of depressive disorder.
Collapse
Affiliation(s)
- Tarapati Rani
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A Alhazmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia; Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Abdulkarim M Meraya
- Pharmacy Parctice Research Unit, Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman; School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania; Doctoral School of Biomedical Sciences, University of Oradea, Oradea, Romania
| |
Collapse
|
42
|
Cortical changes in patients with schizophrenia across two ethnic backgrounds. Sci Rep 2022; 12:10810. [PMID: 35752706 PMCID: PMC9233668 DOI: 10.1038/s41598-022-14914-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 06/15/2022] [Indexed: 11/24/2022] Open
Abstract
While it is known that cultural background influences the healthy brain, less is known about how it affects cortical changes in schizophrenia. Here, we tested whether schizophrenia differentially affected the brain in Japanese and German patients. In a sample of 155 patients with a diagnosis of schizophrenia and 191 healthy controls from Japan and Germany, we acquired 3 T-MRI of the brain. We subsequently compared cortical thickness and cortical surface area to identify whether differences between healthy controls and patients might be influenced by ethnicity. Additional analyses were performed to account for effects of duration of illness and medication. We found pronounced interactions between schizophrenia and cultural background in the cortical thickness of several areas, including the left inferior and middle temporal gyrus, as well as the right lateral occipital cortex. Regarding cortical surface area, interaction effects appeared in the insula and the occipital cortex, among others. Some of these brain areas are related to the expression of psychotic symptoms, which are known to differ across cultures. Our results indicate that cultural background impacts cortical structures in different ways, probably resulting in varying clinical manifestations, and call for the inclusion of more diverse samples in schizophrenia research.
Collapse
|
43
|
Schmidt SD, Zinn CG, Cavalcante LE, Ferreira FF, Furini CRG, Izquierdo I, de Carvalho Myskiw J. Participation of Hippocampal 5-HT 5A, 5-HT 6 and 5-HT 7 Serotonin Receptors on the Consolidation of Social Recognition Memory. Neuroscience 2022; 497:171-183. [PMID: 35718219 DOI: 10.1016/j.neuroscience.2022.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/31/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022]
Abstract
Social recognition is the ability of animals to identify and recognize a conspecific. The consolidation of social stimuli in long-term memory is crucial for the establishment and maintenance of social groups, reproduction and species survival. Despite its importance, little is known about the circuitry and molecular mechanisms involved in the social recognition memory (SRM). Serotonin (5-hydroxytryptamine, 5-HT) is acknowledged as a major neuromodulator, which plays a key role in learning and memory. Focusing on the more recently described 5-HT receptors, we investigated in the CA1 region of the dorsal hippocampus the participation of 5-HT5A, 5-HT6 and 5-HT7 receptors in the consolidation of SRM. Male Wistar rats cannulated in CA1 were subjected to a social discrimination task. In the sample phase the animals were exposed to a juvenile conspecific for 1 h. Immediately after, they received different pharmacological treatments. Twenty-four hours later, they were submitted to a 5 min retention test in the presence of the previously presented juvenile (familiar) and a novel juvenile. The animals that received infusions of 5-HT5A receptor antagonist SB-699551 (10 µg/µL), 5-HT6 receptor agonist WAY-208466 (0.63 µg/µL) or 5-HT7 receptor agonist AS-19 (5 µg/µL) intra-CA1 were unable to recognize the familiar juvenile. This effect was blocked by the coinfusion of WAY-208466 plus 5-HT6 receptor antagonist SB-271046 (10 µg/µL) or AS-19 plus 5-HT7 receptor antagonist SB-269970 (5 µg/µL). The present study helps to clarify the neurobiological functions of the 5-HT receptors more recently described and extends our knowledge about mechanisms underlying the SRM.
Collapse
Affiliation(s)
- Scheila Daiane Schmidt
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil.
| | - Carolina Garrido Zinn
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil
| | - Lorena Evelyn Cavalcante
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil
| | - Flávia Fagundes Ferreira
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil
| | - Cristiane Regina Guerino Furini
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil; National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Ivan Izquierdo
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil; National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Jociane de Carvalho Myskiw
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil; National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil; Psychobiology and Neurocomputation Laboratory (LPBNC), Department of Biophysics, Institute of Biosciences, Federal University of Rio Grande do Sul (UFRGS), Av. Bento Gonçalves, 9500, Building 43422, Room 208A, 91501-970 Porto Alegre, RS, Brazil.
| |
Collapse
|
44
|
Bellon A, Feuillet V, Cortez-Resendiz A, Mouaffak F, Kong L, Hong LE, De Godoy L, Jay TM, Hosmalin A, Krebs MO. Dopamine-induced pruning in monocyte-derived-neuronal-like cells (MDNCs) from patients with schizophrenia. Mol Psychiatry 2022; 27:2787-2802. [PMID: 35365810 PMCID: PMC9156413 DOI: 10.1038/s41380-022-01514-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 02/05/2022] [Accepted: 02/25/2022] [Indexed: 01/10/2023]
Abstract
The long lapse between the presumptive origin of schizophrenia (SCZ) during early development and its diagnosis in late adolescence has hindered the study of crucial neurodevelopmental processes directly in living patients. Dopamine, a neurotransmitter consistently associated with the pathophysiology of SCZ, participates in several aspects of brain development including pruning of neuronal extensions. Excessive pruning is considered the cause of the most consistent finding in SCZ, namely decreased brain volume. It is therefore possible that patients with SCZ carry an increased susceptibility to dopamine's pruning effects and that this susceptibility would be more obvious in the early stages of neuronal development when dopamine pruning effects appear to be more prominent. Obtaining developing neurons from living patients is not feasible. Instead, we used Monocyte-Derived-Neuronal-like Cells (MDNCs) as these cells can be generated in only 20 days and deliver reproducible results. In this study, we expanded the number of individuals in whom we tested the reproducibility of MDNCs. We also deepened the characterization of MDNCs by comparing its neurostructure to that of human developing neurons. Moreover, we studied MDNCs from 12 controls and 13 patients with SCZ. Patients' cells differentiate more efficiently, extend longer secondary neurites and grow more primary neurites. In addition, MDNCs from medicated patients expresses less D1R and prune more primary neurites when exposed to dopamine. Haloperidol did not influence our results but the role of other antipsychotics was not examined and thus, needs to be considered as a confounder.
Collapse
Affiliation(s)
- Alfredo Bellon
- Department of Psychiatry and Behavioral Health, Penn State Hershey Medical Center, Hershey, PA, USA.
- Department of Pharmacology, Penn State Hershey Medical Center, Hershey, PA, USA.
| | - Vincent Feuillet
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014, Paris, France
| | - Alonso Cortez-Resendiz
- Department of Psychiatry and Behavioral Health, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Faycal Mouaffak
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
- Pôle de Psychiatrie d'Adultes 93G04, EPS Ville Evrard, Saint Denis, France
| | - Lan Kong
- Department of Public Health Sciences, Penn State Hershey Medical Center, Hershey, PA, USA
| | - L Elliot Hong
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Therese M Jay
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
| | - Anne Hosmalin
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014, Paris, France
| | - Marie-Odile Krebs
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
- Groupe-Hospitalo-Universitaire de Paris, Psychiatrie et Neuroscience, Pôle PEPIT, University of Paris, Paris, France
| |
Collapse
|
45
|
Perez-Rando M, Elvira UKA, García-Martí G, Gadea M, Aguilar EJ, Escarti MJ, Ahulló-Fuster MA, Grasa E, Corripio I, Sanjuan J, Nacher J. Alterations in the volume of thalamic nuclei in patients with schizophrenia and persistent auditory hallucinations. Neuroimage Clin 2022; 35:103070. [PMID: 35667173 PMCID: PMC9168692 DOI: 10.1016/j.nicl.2022.103070] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/02/2022] [Accepted: 05/30/2022] [Indexed: 11/29/2022]
Abstract
Analysis of structural MRI images using a probabilistic atlas for segmentation of several nuclei of the thalamus. Comparison of chronic patients with schizophrenia, with and without auditory hallucinations and matched healthy controls. Volumetric reductions in patients with AH vs controls: Medial geniculate nucleus, anterior pulvinar nucleus and lateral and medial mediodorsal nuclei. In patients without AH we found reductions in the volume of the pulvinar and mediodorsal nuclei, but not in the medial geniculate nucleus. Found also some significant correlations between the volume of these nuclei and the total score of the PSYRATS scale.
The thalamus is a subcortical structure formed by different nuclei that relay information to the neocortex. Several reports have already described alterations of this structure in patients of schizophrenia that experience auditory hallucinations. However, to date no study has addressed whether the volumes of specific thalamic nuclei are altered in chronic patients experiencing persistent auditory hallucinations. We have processed structural MRI images using Freesurfer, and have segmented them into 25 nuclei using the probabilistic atlas developed by Iglesias and collaborators (Iglesias et al., 2018). To homogenize the sample, we have matched patients of schizophrenia, with and without persistent auditory hallucinations, with control subjects, considering sex, age and their estimated intracranial volume. This rendered a group number of 41 patients experiencing persistent auditory hallucinations, 35 patients without auditory hallucinations, and 55 healthy controls. In addition, we have also correlated the volume of the altered thalamic nuclei with the total score of the PSYRATS, a clinical scale used to evaluate the positive symptoms of this disorder. We have found alterations in the volume of 8 thalamic nuclei in both cohorts of patients with schizophrenia: The medial and lateral geniculate nuclei, the anterior, inferior, and lateral pulvinar nuclei, the lateral complex and the lateral and medial mediodorsal nuclei. We have also found some significant correlations between the volume of these nuclei in patients experiencing auditory hallucinations, and the total score of the PSYRATS scale. Altogether our results indicate that volumetric alterations of thalamic nuclei involved in audition may be related to persistent auditory hallucinations in chronic schizophrenia patients, whereas alterations in nuclei related to association cortices are evident in all patients. Future studies should explore whether the structural alterations are cause or consequence of these positive symptoms and whether they are already present in first episodes of psychosis.
Collapse
Affiliation(s)
- Marta Perez-Rando
- Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; Spanish National Network for Research in Mental Health, (CIBERSAM), Madrid, Spain; Institute of Research of the Clinic Hospital from Valencia (INCLIVA), Valencia, Spain.
| | - Uriel K A Elvira
- Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; Institutes of Biomedical Technologies and Neuroscience, University of La Laguna, San Cristóbal de La Laguna, Spain
| | - Gracian García-Martí
- Spanish National Network for Research in Mental Health, (CIBERSAM), Madrid, Spain; Quironsalud Hospital, Valencia, Spain
| | - Marien Gadea
- Spanish National Network for Research in Mental Health, (CIBERSAM), Madrid, Spain; Institute of Research of the Clinic Hospital from Valencia (INCLIVA), Valencia, Spain; Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - Eduardo J Aguilar
- Spanish National Network for Research in Mental Health, (CIBERSAM), Madrid, Spain; Psychiatry Unit, Faculty of Medicine, Universitat de València, Valencia, Spain
| | - Maria J Escarti
- Spanish National Network for Research in Mental Health, (CIBERSAM), Madrid, Spain
| | - Mónica Alba Ahulló-Fuster
- Department of Radiology, Rehabilitation and Physiotherapy. Faculty of Nursing, Physiotherapy and Podiatry. Universidad Complutense de Madrid, Spain
| | - Eva Grasa
- Spanish National Network for Research in Mental Health, (CIBERSAM), Madrid, Spain; Servicio de Psiquiatría. Instituto de Investigación Biomédica Sant Pau (IIB-SANT PAU), Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
| | - Iluminada Corripio
- Spanish National Network for Research in Mental Health, (CIBERSAM), Madrid, Spain; Servicio de Psiquiatría. Instituto de Investigación Biomédica Sant Pau (IIB-SANT PAU), Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
| | - Julio Sanjuan
- Spanish National Network for Research in Mental Health, (CIBERSAM), Madrid, Spain; Quironsalud Hospital, Valencia, Spain
| | - Juan Nacher
- Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; Spanish National Network for Research in Mental Health, (CIBERSAM), Madrid, Spain; Institute of Research of the Clinic Hospital from Valencia (INCLIVA), Valencia, Spain.
| |
Collapse
|
46
|
Hørlyck LD, Jespersen AE, King JA, Ullum H, Miskowiak KW. Impaired allocentric spatial memory in patients with affective disorders. J Psychiatr Res 2022; 150:153-159. [PMID: 35378488 DOI: 10.1016/j.jpsychires.2022.01.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Memory disturbances are frequent in unipolar depression (UD) and bipolar disorder (BD) and may comprise important predisposing and maintaining factors. Previous studies have demonstrated hippocampal abnormalities in UD and BD but there is a lack of studies specifically assessing hippocampus-dependent memory. METHODS We used a virtual task to assess hippocampus-dependent (allocentric) vs non-hipppocampal (egocentric) spatial memory in remitted and partially remitted patients with UD or BD (N = 22) and a healthy control group (N = 32). Participants also completed a range of standard neuropsychological and functional assessments. RESULTS Participants in the UD/BD group showed selective impairments on high-load hippocampal (allocentric) memory compared to egocentric memory and this effect was independent of residual mood symptoms. Across both samples, both allocentric and egocentric spatial memory correlated with more general measures of memory and other aspects of cognition measured on standard neuropsychological tests but only high-load allocentric memory showed a significant relationship with functional capacity. CONCLUSION Results show a selective impairment in high-load allocentric spatial memory compared to egocentric memory in the patient group, suggesting impaired hippocampal functioning in patients with remitted UD/BD.
Collapse
Affiliation(s)
- Lone D Hørlyck
- Neurocognition and Emotion in Affective Disorder (NEAD) Group, Copenhagen Affective Disorder Research Centre (CADIC), Copenhagen Psychiatric Centre, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark; Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353, Copenhagen, Denmark
| | - Andreas E Jespersen
- Neurocognition and Emotion in Affective Disorder (NEAD) Group, Copenhagen Affective Disorder Research Centre (CADIC), Copenhagen Psychiatric Centre, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark; Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353, Copenhagen, Denmark
| | - John A King
- Department of Clinical and Health Psychology, University College London, 1-19 Torrington Place, London, WC1E 7HB, United Kingdom
| | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Kamilla W Miskowiak
- Neurocognition and Emotion in Affective Disorder (NEAD) Group, Copenhagen Affective Disorder Research Centre (CADIC), Copenhagen Psychiatric Centre, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark; Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353, Copenhagen, Denmark.
| |
Collapse
|
47
|
Mizutani-Tiebel Y, Takahashi S, Karali T, Mezger E, Bulubas L, Papazova I, Dechantsreiter E, Stoecklein S, Papazov B, Thielscher A, Padberg F, Keeser D. Differences in electric field strength between clinical and non-clinical populations induced by prefrontal tDCS: A cross-diagnostic, individual MRI-based modeling study. Neuroimage Clin 2022; 34:103011. [PMID: 35487132 PMCID: PMC9125784 DOI: 10.1016/j.nicl.2022.103011] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/17/2022] [Accepted: 04/13/2022] [Indexed: 01/25/2023]
Abstract
MDD and SCZ showed lower prefrontal tDCS-induced e-field strengths compared to HC. Average e-field strengths did not significantly differ between MDD and SCZ patients. Inter-individual variability of e-field intensities and distribution was prominent. Inter-rater variability emphasizes the importance of standardized positioning.
Introduction Prefrontal cortex (PFC) regions are promising targets for therapeutic applications of non-invasive brain stimulation, e.g. transcranial direct current stimulation (tDCS), which has been proposed as a novel intervention for major depressive disorder (MDD) and negative symptoms of schizophrenia (SCZ). However, the effects of tDCS vary inter-individually, and dose–response relationships have not been established. Stimulation parameters are often tested in healthy subjects and transferred to clinical populations. The current study investigates the variability of individual MRI-based electric fields (e-fields) of standard bifrontal tDCS across individual subjects and diagnoses. Method The study included 74 subjects, i.e. 25 patients with MDD, 24 patients with SCZ, and 25 healthy controls (HC). Individual e-fields of a common tDCS protocol (i.e. 2 mA stimulation intensity, bifrontal anode-F3/cathode-F4 montage) were modeled by two investigators using SimNIBS (2.0.1) based on structural MRI scans. Result On a whole-brain level, the average e-field strength was significantly reduced in MDD and SCZ compared to HC, but MDD and SCZ did not differ significantly. Regions of interest (ROI) analysis for PFC subregions showed reduced e-fields in Sallet areas 8B and 9 for MDD and SCZ compared to HC, whereas there was again no difference between MDD and SCZ. Within groups, we generally observed high inter-individual variability of e-field intensities at a higher percentile of voxels. Conclusion MRI-based e-field modeling revealed significant differences in e-field strengths between clinical and non-clinical populations in addition to a general inter-individual variability. These findings support the notion that dose–response relationships for tDCS cannot be simply transferred from healthy to clinical cohorts and need to be individually established for clinical groups. In this respect, MRI-based e-field modeling may serve as a proxy for individualized dosing.
Collapse
Affiliation(s)
- Yuki Mizutani-Tiebel
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany; NeuroImaging Core Unit Munich (NICUM), Munich, Germany.
| | - Shun Takahashi
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany; Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan; Clinical Research and Education Center, Asakayama General Hospital, Sakai, Japan; Graduate School of Rehabilitation Science, Osaka Metropolitan University, Habikino, Japan; Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Temmuz Karali
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany; Department of Radiology, University Hospital LMU, Munich, Germany
| | - Eva Mezger
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany
| | - Lucia Bulubas
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany; International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Irina Papazova
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany; Department of Psychiatry and Psychotherapy, University of Augsburg, Germany
| | - Esther Dechantsreiter
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany
| | | | - Boris Papazov
- NeuroImaging Core Unit Munich (NICUM), Munich, Germany; Department of Radiology, University Hospital LMU, Munich, Germany
| | - Axel Thielscher
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark; Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Frank Padberg
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany; NeuroImaging Core Unit Munich (NICUM), Munich, Germany; Department of Radiology, University Hospital LMU, Munich, Germany; Munich Center for Neurosciences (MCN) - Brain & Mind, 82152 Planegg-Martinsried, Germany.
| |
Collapse
|
48
|
The Limits between Schizophrenia and Bipolar Disorder: What Do Magnetic Resonance Findings Tell Us? Behav Sci (Basel) 2022; 12:bs12030078. [PMID: 35323397 PMCID: PMC8944966 DOI: 10.3390/bs12030078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/10/2022] [Accepted: 03/10/2022] [Indexed: 02/01/2023] Open
Abstract
Schizophrenia and bipolar disorder, two of the most severe psychiatric illnesses, have historically been regarded as dichotomous entities but share many features of the premorbid course, clinical profile, genetic factors and treatment approaches. Studies focusing on neuroimaging findings have received considerable attention, as they plead for an improved understanding of the brain regions involved in the pathophysiology of schizophrenia and bipolar disorder. In this review, we summarize the main magnetic resonance imaging findings in both disorders, aiming at exploring the neuroanatomical and functional similarities and differences between the two. The findings show that gray and white matter structural changes and functional dysconnectivity predominate in the frontal and limbic areas and the frontotemporal circuitry of the brain areas involved in the integration of executive, cognitive and affective functions, commonly affected in both disorders. Available evidence points to a considerable overlap in the affected regions between the two conditions, therefore possibly placing them at opposite ends of a psychosis continuum.
Collapse
|
49
|
Fan L, Yu M, Pinkham A, Zhu Y, Tang X, Wang X, Zhang X, Ma J, Zhang J, Zhang X, Dai Z. Aberrant large-scale brain modules in deficit and non-deficit schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2022; 113:110461. [PMID: 34688810 DOI: 10.1016/j.pnpbp.2021.110461] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Schizophrenia is a heterogenous psychiatric disease, and deficit schizophrenia (DS) is a clinical subgroup with primary and enduring negative symptoms. Although previous neuroimaging studies have identified functional connectome alterations in schizophrenia, the modular organizations in DS and nondeficit schizophrenia (NDS) remain poorly understood. Therefore, this study aimed to investigate the modular-level alterations in DS patients compared with the NDS and healthy control (HC) groups. METHODS A previously collected dataset was re-analyzed, in which 74 chronic male schizophrenia patients (33 DS and 41 NDS) and 40 HC underwent resting-state functional magnetic resonance imaging with eyes closed in a Siemens 3 T scanner (scanning duration = 8 min). Modular- (intramodule and intermodule connectivity) and nodal- [normalized within-module degree (Zi) and participation coefficient (PCi)] level graph theory properties were computed and compared among the three groups. Receiver operating characteristic curve (ROC) analyses were performed to examine the classification ability of these measures, and partial correlations were conducted between network measures and symptom severity. Validation analyses on head motion, network sparsity, and parcellation scheme were also performed. RESULTS Both schizophrenia subgroups showed decreased intramodule connectivity in salience network (SN), somatosensory-motor network (SMN), and visual network (VN), and increased intermodule connectivity in SMN-default mode network (DMN) and SMN-frontoparietal network (FPN). Compared with NDS patients, DS patients showed weaker intramodule connectivity in SN and stronger intermodule connectivity in SMN-FPN and SMN-VN. At the nodal level, the schizophrenia-related alterations were distributed in SN, SMN, VN, and DMN, and 7 DS-specific nodal alterations were identified. Intramodule connectivity of SN, intermodule connectivity of SMN-VN, and Zi of left precuneus successfully distinguished the three groups. Partial correlational analyses revealed that these measures were related to negative symptoms, general psychiatric symptoms, and neurocognitive function. CONCLUSION Our findings suggest that functional connectomes, especially SN, SMN, and VN, may capture the distinct and common disruptions of DS and NDS. These findings may help to understand the neuropathology of negative symptoms of schizophrenia and inform targets for treating different schizophrenia subtypes.
Collapse
Affiliation(s)
- Linlin Fan
- Department of Psychology, Sun Yat-sen University, Guangzhou, Guangdong, China; School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
| | - Miao Yu
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, No. 264 Guangzhou Road, Nanjing, Jiangsu, China
| | - Amy Pinkham
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
| | - Yiyi Zhu
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
| | - Xiaowei Tang
- Department of Geriatric Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiang Wang
- Medical Psychological Institute of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaobin Zhang
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Junji Ma
- Department of Psychology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jinbo Zhang
- Department of Psychology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiangrong Zhang
- Department of Geriatric Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Zhengjia Dai
- Department of Psychology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
50
|
Ohi K, Ishibashi M, Torii K, Hashimoto M, Yano Y, Shioiri T. Differences in subcortical brain volumes among patients with schizophrenia and bipolar disorder and healthy controls. J Psychiatry Neurosci 2022; 47:E77-E85. [PMID: 35232800 PMCID: PMC8896343 DOI: 10.1503/jpn.210144] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/09/2021] [Accepted: 10/10/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Patients with schizophrenia and bipolar disorder have an overlapping polygenic architecture and clinical similarities, although the 2 disorders are distinct diagnoses with clinical dissimilarities. It remains unclear whether there are specific differences in subcortical volumes between schizophrenia and bipolar disorder, and whether the subcortical differences are affected by any clinical characteristics. We investigated differences in subcortical volumes bilaterally among patients with schizophrenia, patients with bipolar disorder and healthy controls. We also investigated the influences of clinical characteristics on specific subcortical volumes in these patient groups. METHODS We collected 3 T T 1-weighted MRI brain scans from 413 participants (157 with schizophrenia, 51 with bipolar disorder and 205 controls) with a single scanner at a single institute. We used FreeSurfer version 6.0 for processing the T 1-weighted images to segment the following subcortical brain volumes: thalamus, caudate, putamen, globus pallidus, hippocampus, amygdala and nucleus accumbens. Differences in the 7 subcortical volumes were investigated among the groups. We also evaluated correlations between subcortical volumes and clinical variables in these patient groups. RESULTS Of 7 subcortical regions, patients with schizophrenia had significantly smaller volumes in the left thalamus (Cohen d = -0.29, p = 5.83 × 10-3), bilateral hippocampi (left, d = -0.36, p = 8.85 × 10-4; right, d = -0.41, p = 1.15 × 10-4) and left amygdala (d = -0.31, p = 4.02 × 10-3) than controls. Compared with controls, patients with bipolar disorder had bilateral reductions only in the hippocampal volumes (left, d = -0.52, p = 1.12 × 10-3; right, d = -0.58, p = 0.30 × 10-4). We also found that patients with schizophrenia had significantly smaller volumes in the bilateral amygdalae (left, d = -0.43, p = 4.22 × 10-3; right, d = -0.45, p = 4.56 × 10-3) than patients with bipolar disorder. We did not find any significant volumetric differences in the other 6 subcortical structures between patient groups (p > 0.05). Smaller left amygdalar volumes were significantly correlated with younger onset age only in patients with schizophrenia (r = 0.22, p = 5.78 × 10-3). LIMITATIONS We did not evaluate the differences in subcortical volumes between patients stratified based on clinical bipolar disorder subtype and a history of psychotic episodes because our sample size of patients with bipolar disorder was limited. CONCLUSION Our findings suggest that volumetric differences in the amygdala between patients with schizophrenia and those with bipolar disorder may be a putative biomarker for distinguishing 2 clinically similar diagnoses.
Collapse
Affiliation(s)
- Kazutaka Ohi
- From the Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan (Ohi, Shioiri); the Department of General Internal Medicine, Kanazawa Medical University, Ishikawa, Japan (Ohi); and the School of Medicine, Gifu University, Gifu, Japan (Ishibashi, Torii, Hashimoto, Yano)
| | | | | | | | | | | |
Collapse
|