1
|
Juan Q, Shiwan T, Yurong S, Jiabo S, Yu C, Shui T, Zhijian Y, Qing L. Brain structural and functional abnormalities in affective network are associated with anxious depression. BMC Psychiatry 2024; 24:533. [PMID: 39054442 PMCID: PMC11270941 DOI: 10.1186/s12888-024-05970-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Anxious depression (AD) is a common subtype of major depressive disorder (MDD). Neuroimaging studies of AD have revealed inconsistent and heterogeneous brain alterations with the use of single-model methods. Therefore, it is necessary to explore the pathogenesis of AD using multi-model imaging analyses to obtain more homogeneous and robust results. METHODS One hundred and eighty-two patients with MDD and 64 matched healthy controls (HCs) were recruited. Voxel-based morphometry (VBM) was used to estimate the gray matter volume (GMV) of all subjects. The GMV differences between the AD and non-anxious depression (NAD) participants were used as regions of interest (ROIs) for subsequent resting state functional connectivity (rs-FC) analyses. Correlation analysis was used to evaluate the associations between clinical symptoms and abnormal function in specific brain areas. RESULTS Decreased GMV in the medial frontal gyrus (MFG) and the superior frontal gyrus (SFG) was observed in the AD group compared to the NAD group. Taking the MFG and SFG as ROIs, the rs-FC analysis revealed decreased FC between the left SFG and left temporal pole and between the left SFG and right MFG in the AD group compared to the NAD group. Finally, the FC between the left SFG and left temporal pole was negatively correlated with HAMD-17 scores in the AD group. CONCLUSION By combining the GMV and rs-FC models, this study revealed that structural and functional disruption of the affective network may be an important pathophysiology underlying AD. The structural impairment may serve as the foundation of the functional impairment.
Collapse
Affiliation(s)
- Qiao Juan
- Department of Psychology, The Affiliated Xuzhou Eastern Hospital of Xuzhou Medical University, Xuzhou, 221004, China
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tao Shiwan
- West China Hospital, Mental Health Center, Sichuan University, Chengdu, 610047, China
| | - Sun Yurong
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China
- Child Development and Learning Science, Key Laboratory of Ministry of Education, Nanjing, China
| | - Shi Jiabo
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
- Nanjing Brain Hospital, Medical School of Nanjing University, Nanjing, 210093, China
| | - Chen Yu
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
- Nanjing Brain Hospital, Medical School of Nanjing University, Nanjing, 210093, China
| | - Tian Shui
- Department of Radiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yao Zhijian
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Nanjing Brain Hospital, Medical School of Nanjing University, Nanjing, 210093, China.
| | - Lu Qing
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China.
- Child Development and Learning Science, Key Laboratory of Ministry of Education, Nanjing, China.
| |
Collapse
|
2
|
Xia J, Lin X, Yu T, Yu H, Zou Y, Luo Q, Peng H. Aberrant functional connectivity of the globus pallidus in the modulation of the relationship between childhood trauma and major depressive disorder. J Psychiatry Neurosci 2024; 49:E218-E232. [PMID: 38960625 PMCID: PMC11230669 DOI: 10.1503/jpn.240019] [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: 02/19/2024] [Revised: 04/18/2024] [Accepted: 05/03/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Childhood trauma plays a crucial role in the dysfunctional reward circuitry in major depressive disorder (MDD). We sought to explore the effect of abnormalities in the globus pallidus (GP)-centric reward circuitry on the relationship between childhood trauma and MDD. METHODS We conducted seed-based dynamic functional connectivity (dFC) analysis among people with or without MDD and with or without childhood trauma. We explored the relationship between abnormal reward circuitry, childhood trauma, and MDD. RESULTS We included 48 people with MDD and childhood trauma, 30 people with MDD without childhood trauma, 57 controls with childhood trauma, and 46 controls without childhood trauma. We found that GP subregions exhibited abnormal dFC with several regions, including the inferior parietal lobe, thalamus, superior frontal gyrus (SFG), and precuneus. Abnormal dFC in these GP subregions showed a significant correlation with childhood trauma. Moderation analysis revealed that the dFC between the anterior GP and SFG, as well as between the anterior GP and the precentral gyrus, modulated the relationship between childhood abuse and MDD severity. We observed a negative correlation between childhood trauma and MDD severity among patients with lower dFC between the anterior GP and SFG, as well as higher dFC between the anterior GP and precentral gyrus. This suggests that reduced dFC between the anterior GP and SFG, along with increased dFC between the anterior GP and precentral gyrus, may attenuate the effect of childhood trauma on MDD severity. LIMITATIONS Cross-sectional designs cannot be used to infer causality. CONCLUSION Our findings underscore the pivotal role of reward circuitry abnormalities in MDD with childhood trauma. These abnormalities involve various brain regions, including the postcentral gyrus, precentral gyrus, inferior parietal lobe, precuneus, superior frontal gyrus, thalamus, and middle frontal gyrus. CLINICAL TRIAL REGISTRATION ChiCTR2300078193.
Collapse
Affiliation(s)
- Jinrou Xia
- From the Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China (Xia, Lin, Yu T, Yu H, Zou, Luo, Peng); the Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China (Luo, Peng)
| | - Xiaohui Lin
- From the Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China (Xia, Lin, Yu T, Yu H, Zou, Luo, Peng); the Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China (Luo, Peng)
| | - Tong Yu
- From the Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China (Xia, Lin, Yu T, Yu H, Zou, Luo, Peng); the Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China (Luo, Peng)
| | - Huiwen Yu
- From the Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China (Xia, Lin, Yu T, Yu H, Zou, Luo, Peng); the Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China (Luo, Peng)
| | - Yurong Zou
- From the Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China (Xia, Lin, Yu T, Yu H, Zou, Luo, Peng); the Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China (Luo, Peng)
| | - Qianyi Luo
- From the Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China (Xia, Lin, Yu T, Yu H, Zou, Luo, Peng); the Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China (Luo, Peng)
| | - Hongjun Peng
- From the Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China (Xia, Lin, Yu T, Yu H, Zou, Luo, Peng); the Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China (Luo, Peng)
| |
Collapse
|
3
|
Rozovsky R, Bertocci M, Iyengar S, Stiffler RS, Bebko G, Skeba AS, Brady T, Aslam H, Phillips ML. Identifying tripartite relationship among cortical thickness, neuroticism, and mood and anxiety disorders. Sci Rep 2024; 14:8449. [PMID: 38600283 PMCID: PMC11006921 DOI: 10.1038/s41598-024-59108-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 04/08/2024] [Indexed: 04/12/2024] Open
Abstract
The number of young adults seeking help for emotional distress, subsyndromal-syndromal mood/anxiety symptoms, including those associated with neuroticism, is rising and can be an early manifestation of mood/anxiety disorders. Identification of gray matter (GM) thickness alterations and their relationship with neuroticism and mood/anxiety symptoms can aid in earlier diagnosis and prevention of risk for future mood and anxiety disorders. In a transdiagnostic sample of young adults (n = 252;177 females; age 21.7 ± 2), Hypothesis (H) 1:regularized regression followed by multiple regression examined relationships among GM cortical thickness and clinician-rated depression, anxiety, and mania/hypomania; H2:the neuroticism factor and its subfactors as measured by NEO Personality Inventory (NEO-PI-R) were tested as mediators. Analyses revealed positive relationships between left parsopercularis thickness and depression (B = 4.87, p = 0.002), anxiety (B = 4.68, p = 0.002), mania/hypomania (B = 6.08, p ≤ 0.001); negative relationships between left inferior temporal gyrus (ITG) thickness and depression (B = - 5.64, p ≤ 0.001), anxiety (B = - 6.77, p ≤ 0.001), mania/hypomania (B = - 6.47, p ≤ 0.001); and positive relationships between left isthmus cingulate thickness (B = 2.84, p = 0.011), and anxiety. NEO anger/hostility mediated the relationship between left ITG thickness and mania/hypomania; NEO vulnerability mediated the relationship between left ITG thickness and depression. Examining the interrelationships among cortical thickness, neuroticism and mood and anxiety symptoms enriches the potential for identifying markers conferring risk for mood and anxiety disorders and can provide targets for personalized intervention strategies for these disorders.
Collapse
Affiliation(s)
- Renata Rozovsky
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, 302 Loeffler Building, 121 Meyran Ave., Pittsburgh, PA, USA.
| | - Michele Bertocci
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, 302 Loeffler Building, 121 Meyran Ave., Pittsburgh, PA, USA
| | - Satish Iyengar
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Richelle S Stiffler
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, 302 Loeffler Building, 121 Meyran Ave., Pittsburgh, PA, USA
| | - Genna Bebko
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, 302 Loeffler Building, 121 Meyran Ave., Pittsburgh, PA, USA
| | - Alexander S Skeba
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, 302 Loeffler Building, 121 Meyran Ave., Pittsburgh, PA, USA
| | - Tyler Brady
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, 302 Loeffler Building, 121 Meyran Ave., Pittsburgh, PA, USA
| | - Haris Aslam
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, 302 Loeffler Building, 121 Meyran Ave., Pittsburgh, PA, USA
| | - Mary L Phillips
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, 302 Loeffler Building, 121 Meyran Ave., Pittsburgh, PA, USA
| |
Collapse
|
4
|
Liu H, Hao Z, Qiu S, Wang Q, Zhan L, Huang L, Shao Y, Wang Q, Su C, Cao Y, Sun J, Wang C, Lv Y, Li M, Shen W, Li H, Jia X. Grey matter structural alterations in anxiety disorders: a voxel-based meta-analysis. Brain Imaging Behav 2024; 18:456-474. [PMID: 38150133 DOI: 10.1007/s11682-023-00842-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 12/28/2023]
Abstract
Anxiety disorders (ADs) are a group of prevalent and destructive mental illnesses, but the current understanding of their underlying neuropathology is still unclear. Employing voxel-based morphometry (VBM), previous studies have demonstrated several common brain regions showing grey matter volume (GMV) abnormalities. However, contradictory results have been reported among these studies. Considering that different subtypes of ADs exhibit common core symptoms despite different diagnostic criteria, and previous meta-analyses have found common core GMV-altered brain regions in ADs, the present research aimed to combine the results of individual studies to identify common GMV abnormalities in ADs. Therefore, we first performed a systematic search in PubMed, Embase, and Web of Science on studies investigating GMV differences between patients with ADs and healthy controls (HCs). Then, the anisotropic effect-size signed differential mapping (AES-SDM) was applied in this meta-analysis. A total of 24 studies (including 25 data sets) were included in the current study, and 906 patients with ADs and 1003 HCs were included. Compared with the HCs, the patients with ADs showed increased GMV in the left superior parietal gyrus, right angular gyrus, left precentral gyrus, and right lingual gyrus, and decreased GMV in the bilateral insula, bilateral thalamus, left caudate, and right putamen. In conclusion, the current study has identified some abnormal GMV brain regions that are related to the pathological mechanisms of anxiety disorders. These findings could contribute to a better understanding of the underlying neuropathology of ADs.
Collapse
Affiliation(s)
- Han Liu
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Zeqi Hao
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Shasha Qiu
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Qianqian Wang
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Linlin Zhan
- School of Western Languages, Heilongjiang University, Heilongjiang, China
| | - Lina Huang
- Department of Radiology, Changshu No.2 People's Hospital, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, Jiangsu, China
| | - Youbin Shao
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Qing Wang
- Research Center of Brain and Cognitive Neuroscience, Liaoning Normal University, Dalian, China
| | - Chang Su
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Yikang Cao
- School of Information and Electronics Technology, Jiamusi University, Jiamusi, China
| | - Jiawei Sun
- School of Information and Electronics Technology, Jiamusi University, Jiamusi, China
| | - Chunjie Wang
- Institute of Brain Science, Department of Psychology, School of Education, Hangzhou Normal University, Hangzhou, China
- Center for Cognition and Brain Disorders, the Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Yating Lv
- Center for Cognition and Brain Disorders, the Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Mengting Li
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Wenbin Shen
- Department of Radiology, Changshu No.2 People's Hospital, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, Jiangsu, China
| | - Huayun Li
- School of Psychology, Zhejiang Normal University, Jinhua, China.
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China.
| | - Xize Jia
- School of Psychology, Zhejiang Normal University, Jinhua, China.
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China.
| |
Collapse
|
5
|
He P, Gao Y, Shi L, Li Y, Qiu Y, Feng S, Tie Z, Gong L, Ma G, Zhang Y, Nie K, Wang L. The association of CSF biomarkers and cognitive decline with choroid plexus volume in early Parkinson's disease. Parkinsonism Relat Disord 2024; 120:105987. [PMID: 38183890 DOI: 10.1016/j.parkreldis.2023.105987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024]
Abstract
OBJECTIVE This study aims to determine the link between choroid plexus (CP) volume and cognitive decline in patients with early-stage Parkinson's disease (PD) and to test whether pathological proteins in the cerebrospinal fluid (CSF) are involved in the modulation of any detrimental effects from CP volume. METHODS Data on 95 early-stage PD patients with 5 years of follow-up were collected from the Parkinson's Progression Marker Initiative cohort. The patients were separated into three groups based on tertiles of baseline CP volume. We then used a linear mixed model for longitudinal analysis and conducted path analysis to investigate mediating effects. RESULTS At baseline, the patients in both the upper and middle tertile group were older and had lower concentrations of CSF Aβ1-42 than those in the lowest tertile group. Longitudinal analysis showed that the upper tertile group suffered from a more rapid cognitive decline in the Symbol Digit Modalities test, Hopkins Verbal Learning Test (HVLT)-retention, and HVLT delayed recalled score. Furthermore, path analysis showed that the pathological effects of CP volume on the 5-year decline in memory might be partly mediated by the CSF Aβ1-42/αsyn ratio. CONCLUSION CP enlargement could be an independent risk factor for decreased cognition in patients with early-stage PD, and this risk may be mediated by CSF pathological proteins.
Collapse
Affiliation(s)
- Peikun He
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China; Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuyuan Gao
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China; BrainNow Research Institute, Shenzhen, Guangdong Province, China
| | - Yanyi Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yihui Qiu
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shujun Feng
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zihui Tie
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China; Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Liangxu Gong
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China; Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Guixian Ma
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Kun Nie
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Lijuan Wang
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China; Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
6
|
Bae EB, Han KM. A structural equation modeling approach using behavioral and neuroimaging markers in major depressive disorder. J Psychiatr Res 2024; 171:246-255. [PMID: 38325105 DOI: 10.1016/j.jpsychires.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/16/2023] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Major depressive disorder (MDD) has consistently proven to be a multifactorial and highly comorbid disease. Despite recent depression-related research demonstrating causalities between MDD-related factors and a small number of variables, including brain structural changes, a high-statistical power analysis of the various factors is yet to be conducted. We retrospectively analyzed data from 155 participants (84 healthy controls and 71 patients with MDD). We used magnetic resonance imaging and diffusion tensor imaging data, scales assessing childhood trauma, depression severity, cognitive dysfunction, impulsivity, and suicidal ideation. To simultaneously evaluate the causalities between multivariable, we implemented two types of MDD-specified structural equation models (SEM), the behavioral and neurobehavioral models. Behavioral SEM showed significant results in the MDD group: Comparative Fit Index [CFI] = 1.000, Root Mean Square Error of Approximation [RMSEA]) = 0.000), with a strong correlation in the scales for childhood trauma, depression severity, suicidal ideation, impulsivity, and cognitive dysfunction. Based on behavioral SEM, we established neurobehavioral models showing the best-fit in MDD, especially including the right cingulate cortex, central to the posterior corpus callosum, right putamen, pallidum, whole brainstem, and ventral diencephalon, including the thalamus (CFI >0.96, RMSEA <0.05). Our MDD-specific model revealed that the limbic-associated regions are strongly connected with childhood trauma rather than depression severity, and that they independently affect suicidal ideation and cognitive dysfunction. Furthermore, cognitive dysfunction could affect impulsivity.
Collapse
Affiliation(s)
- Eun Bit Bae
- Research Institute for Medical Bigdata Science, Korea University, Seoul, Republic of Korea; 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.
| |
Collapse
|
7
|
Zhou E, Wang W, Ma S, Xie X, Kang L, Xu S, Deng Z, Gong Q, Nie Z, Yao L, Bu L, Wang F, Liu Z. Prediction of anxious depression using multimodal neuroimaging and machine learning. Neuroimage 2024; 285:120499. [PMID: 38097055 DOI: 10.1016/j.neuroimage.2023.120499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/15/2023] [Accepted: 12/11/2023] [Indexed: 12/18/2023] Open
Abstract
Anxious depression is a common subtype of major depressive disorder (MDD) associated with adverse outcomes and severely impaired social function. It is important to clarify the underlying neurobiology of anxious depression to refine the diagnosis and stratify patients for therapy. Here we explored associations between anxiety and brain structure/function in MDD patients. A total of 260 MDD patients and 127 healthy controls underwent three-dimensional T1-weighted structural scanning and resting-state functional magnetic resonance imaging. Demographic data were collected from all participants. Differences in gray matter volume (GMV), (fractional) amplitude of low-frequency fluctuation ((f)ALFF), regional homogeneity (ReHo), and seed point-based functional connectivity were compared between anxious MDD patients, non-anxious MDD patients, and healthy controls. A random forest model was used to predict anxiety in MDD patients using neuroimaging features. Anxious MDD patients showed significant differences in GMV in the left middle temporal gyrus and ReHo in the right superior parietal gyrus and the left precuneus than HCs. Compared with non-anxious MDD patients, patients with anxious MDD showed significantly different GMV in the left inferior temporal gyrus, left superior temporal gyrus, left superior frontal gyrus (orbital part), and left dorsolateral superior frontal gyrus; fALFF in the left middle temporal gyrus; ReHo in the inferior temporal gyrus and the superior frontal gyrus (orbital part); and functional connectivity between the left superior temporal gyrus(temporal pole) and left medial superior frontal gyrus. A diagnostic predictive random forest model built using imaging features and validated by 10-fold cross-validation distinguished anxious from non-anxious MDD with an AUC of 0.802. Patients with anxious depression exhibit dysregulation of brain regions associated with emotion regulation, cognition, and decision-making, and our diagnostic model paves the way for more accurate, objective clinical diagnosis of anxious depression.
Collapse
Affiliation(s)
- Enqi Zhou
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Simeng Ma
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinhui Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijun Kang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuxian Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zipeng Deng
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Gong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaowen Nie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lihua Yao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lihong Bu
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China; Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China.
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
8
|
Jain PR, Yates M, de Celis CR, Drineas P, Jahanshad N, Thompson P, Paschou P. Multiomic approach and Mendelian randomization analysis identify causal associations between blood biomarkers and subcortical brain structure volumes. Neuroimage 2023; 284:120466. [PMID: 37995919 DOI: 10.1016/j.neuroimage.2023.120466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/17/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023] Open
Abstract
Alterations in subcortical brain structure volumes have been found to be associated with several neurodegenerative and psychiatric disorders. At the same time, genome-wide association studies (GWAS) have identified numerous common variants associated with brain structure. In this study, we integrate these findings, aiming to identify proteins, metabolites, or microbes that have a putative causal association with subcortical brain structure volumes via a two-sample Mendelian randomization approach. This method uses genetic variants as instrument variables to identify potentially causal associations between an exposure and an outcome. The exposure data that we analyzed comprised genetic associations for 2994 plasma proteins, 237 metabolites, and 103 microbial genera. The outcome data included GWAS data for seven subcortical brain structure volumes including accumbens, amygdala, caudate, hippocampus, pallidum, putamen, and thalamus. Eleven proteins and six metabolites were found to have a significant association with subcortical structure volumes, with nine proteins and five metabolites replicated using independent exposure data. We found causal associations between accumbens volume and plasma protease c1 inhibitor as well as strong association between putamen volume and Agouti signaling protein. Among metabolites, urate had the strongest association with thalamic volume. No significant associations were detected between the microbial genera and subcortical brain structure volumes. We also observed significant enrichment for biological processes such as proteolysis, regulation of the endoplasmic reticulum apoptotic signaling pathway, and negative regulation of DNA binding. Our findings provide insights to the mechanisms through which brain volumes may be affected in the pathogenesis of neurodevelopmental and psychiatric disorders and point to potential treatment targets for disorders that are associated with subcortical brain structure volumes.
Collapse
Affiliation(s)
- Pritesh R Jain
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Madison Yates
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Carlos Rubin de Celis
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Petros Drineas
- Department of Computer Science, Purdue University, United States
| | - Neda Jahanshad
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California, United States
| | - Paul Thompson
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California, United States
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
9
|
He P, Gao Y, Shi L, Li Y, Jiang S, Tie Z, Qiu Y, Ma G, Zhang Y, Nie K, Wang L. Motor progression phenotypes in early-stage Parkinson's Disease: A clinical prediction model and the role of glymphatic system imaging biomarkers. Neurosci Lett 2023; 814:137435. [PMID: 37562710 DOI: 10.1016/j.neulet.2023.137435] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Substantial heterogeneity of motor symptoms in Parkinson's disease (PD) poses a challenge to disease prediction. OBJECTIVES The aim of this study was to construct a nomogram model that can distinguish different longitudinal trajectories of motor symptom changes in early-stage PD patients. METHODS Data on 90 patients with 5-years of follow-up were collected from the Parkinson's Progression Marker Initiative (PPMI) cohort. We used a latent class mixed modeling (LCMM) to identify distinct progression patterns of motor symptoms, and backward stepwise logistic regression with baseline information was conducted to identify the potential predictors for motor trajectory and to develop a nomogram. The performance of the nomogram model was then evaluated using the optimism-corrected C-index for internal validation, the area under the curve (AUC) of the receiver operating characteristic (ROC) curve for discrimination, the calibration curve for predictive accuracy, and decision curve analysis (DCA) for its clinical value. RESULTS We identified two trajectories for motor progression patterns. The first, Class 1 (Motor deteriorated group), was characterized by sustained, continuously worsening motor symptoms, and the second, Class 2 (Motor stable group), had stable motor symptoms throughout the follow-up period. The best combination of 7 baseline variables was identified and assembled into the nomogram: Scopa-AUT [odds ratio (OR), 1.11; p = 0.091], Letter number sequencing (LNS) (OR, 0.76; p = 0.068), the asymmetry index of putamen (OR, 0.95; p = 0.034), mean caudate uptake (OR, 0.14; p = 0.086), CSF pTau/α-synuclein (OR, 0.00; p = 0.011), CSF tTau/Aβ (OR, 25434806; p = 0.025), and the index for diffusion tensor image analysis along the perivascular space (ALPS-index) (OR, 0.02; p = 0.030). The nomogram achieved good discrimination, with an original AUC of 0.901 (95% CI, 0.813-0.989), and the bias-corrected concordance index (C-index) with 1,000 bootstraps was 0.834. The calibration curve and DCA also suggested both the high accuracy and clinical usefulness of the nomogram, respectively. CONCLUSIONS This study proposes an effective nomogram to predict different motor progression patterns in early-stage PD. Furthermore, the imaging biomarker indicating glymphatic function could be an independent predictive factor for PD motor progression.
Collapse
Affiliation(s)
- Peikun He
- School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuyuan Gao
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China; BrainNow Research Institute, Shenzhen, Guangdong Province, China
| | - Yanyi Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shuolin Jiang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zihui Tie
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yihui Qiu
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guixian Ma
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Kun Nie
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Lijuan Wang
- School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Bashford-Largo J, R Blair RJ, Blair KS, Dobbertin M, Dominguez A, Hatch M, Bajaj S. Identification of structural brain alterations in adolescents with depressive symptomatology. Brain Res Bull 2023; 201:110723. [PMID: 37536609 PMCID: PMC10451038 DOI: 10.1016/j.brainresbull.2023.110723] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/10/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023]
Abstract
INTRODUCTION Depressive symptoms can emerge as early as childhood and may lead to adverse situations in adulthood. Studies have examined structural brain alternations in individuals with depressive symptoms, but findings remain inconclusive. Furthermore, previous studies have focused on adults or used a categorical approach to assess depression. The current study looks to identify grey matter volumes (GMV) that predict depressive symptomatology across a clinically concerning sample of adolescents. METHODS Structural MRI data were collected from 338 clinically concerning adolescents (mean age = 15.30 SD=2.07; mean IQ = 101.01 SD=12.43; 132 F). Depression symptoms were indexed via the Mood and Feelings Questionnaire (MFQ). Freesurfer was used to parcellate the brain into 68 cortical regions and 14 subcortical regions. GMV was extracted from all 82 brain areas. Multiple linear regression was used to look at the relationship between MFQ scores and region-specific GMV parameter. Follow up regressions were conducted to look at potential effects of psychiatric diagnoses and medication intake. RESULTS Our regression analysis produced a significant model (R2 = 0.446, F(86, 251) = 2.348, p < 0.001). Specifically, there was a negative association between GMV of the left parahippocampal (B = -0.203, p = 0.005), right rostral anterior cingulate (B = -0.162, p = 0.049), and right frontal pole (B = -0.147, p = 0.039) and a positive association between GMV of the left bank of the superior temporal sulcus (B = 0.173, p = 0.029). Follow up analyses produced results proximal to the main analysis. CONCLUSIONS Altered regional brain volumes may serve as biomarkers for the development of depressive symptoms during adolescence. These findings suggest a homogeneity of altered cortical structures in adolescents with depressive symptoms.
Collapse
Affiliation(s)
- Johannah Bashford-Largo
- Multimodal Clinical Neuroimaging Laboratory (MCNL), Center for Neurobehavioral Research, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Brain, Biology, and Behavior, University of Nebraska-Lincoln, Lincoln, NE, USA.
| | - R James R Blair
- Child and Adolescent Mental Health Centre, Mental Health Services, Capital Region of Denmark, Copenhagen, Denmark
| | - Karina S Blair
- Multimodal Clinical Neuroimaging Laboratory (MCNL), Center for Neurobehavioral Research, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Matthew Dobbertin
- Multimodal Clinical Neuroimaging Laboratory (MCNL), Center for Neurobehavioral Research, Boys Town National Research Hospital, Boys Town, NE, USA; Child and Adolescent Inpatient Psychiatric Unit, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Ahria Dominguez
- Multimodal Clinical Neuroimaging Laboratory (MCNL), Center for Neurobehavioral Research, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Melissa Hatch
- Multimodal Clinical Neuroimaging Laboratory (MCNL), Center for Neurobehavioral Research, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Sahil Bajaj
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
11
|
Francis AM, Bissonnette JN, Hull KM, Leckey J, Pimer L, Lawrence MA, Berrigan LI, Fisher DJ. Measuring the attention networks and quantitative-electroencephalography correlates of attention in depression. Psychiatry Res Neuroimaging 2023; 333:111661. [PMID: 37331318 DOI: 10.1016/j.pscychresns.2023.111661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023]
Abstract
Given the association between major depressive disorder (MDD) and cortical inefficiency related to executive control, specifically in the sense that individuals with MDD may recruit more cognitive resources to complete tasks at the same capacity as those without MDD, the current study was interested in examining the attention networks and executive functioning of those with MDD. Past research has used the Attention Network Test (ANT) to measure changes of attention in clinical vs. healthy populations; however, theoretical concerns have been raised regarding the task. The Combined Attention Systems Task (CAST) was developed to address these concerns and was used in our study in combination with quantitative-electroencephalography (QEEG) to assess both behavioural and neurophysiological changes in participants with MDD (n = 18) compared to healthy controls (HCs; n = 22). We found no behavioural differences between MDD and HC groups suggesting individuals with MDD in our sample were not experiencing the executive functioning deficits previously reported in the literature. Neurophysiological measures of attention revealed that MDD participants had greater theta and alpha1 activity relative to HCs, suggesting that although individuals with MDD do not show deficits in behavioural attention, they exhibit altered neural processing which underlies cognitive function.
Collapse
Affiliation(s)
- Ashley M Francis
- Department of Psychology, Saint Mary's University, Halifax, NS, Canada; Department of Psychology, St. Francis Xavier University, Antigonish, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Nova Scotia, Canada
| | - Jenna N Bissonnette
- Department of Psychology, Mount Saint Vincent University, Halifax, NS, Canada; Department of Psychology & Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Krista M Hull
- Department of Psychology, Mount Saint Vincent University, Halifax, NS, Canada
| | - Jennifer Leckey
- Department of Psychology, Mount Saint Vincent University, Halifax, NS, Canada
| | - Laura Pimer
- Department of Psychiatry, Dalhousie University, Nova Scotia, Canada; Department of Psychology, Mount Saint Vincent University, Halifax, NS, Canada
| | - Michael A Lawrence
- Department of Psychology & Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Lindsay I Berrigan
- Department of Psychology, St. Francis Xavier University, Antigonish, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Nova Scotia, Canada
| | - Derek J Fisher
- Department of Psychology, Saint Mary's University, Halifax, NS, Canada; Department of Psychiatry, Dalhousie University, Nova Scotia, Canada; Department of Psychology, Mount Saint Vincent University, Halifax, NS, Canada; Department of Psychology & Neuroscience, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
12
|
Hu X, Yuan X, Yang M, Han M, Ommati MM, Ma Y. Arsenic exposure induced anxiety-like behaviors in male mice via influencing the GABAergic Signaling in the prefrontal cortex. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:86352-86364. [PMID: 37402917 DOI: 10.1007/s11356-023-28426-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 06/21/2023] [Indexed: 07/06/2023]
Abstract
Arsenic contamination in drinking water causes a global public health problem. Emerging evidence suggests that arsenic may act as an environmental risk factor for anxiety disorders. However, the exact mechanism underlying the adverse effects has not been fully elucidated. This study aimed to evaluate the anxiety-like behaviors of mice exposed to arsenic trioxide (As2O3), to observe the neuropathological changes, and to explore the link between the GABAergic system and behavioral manifestations. For this purpose, male C57BL/6 mice were exposed to various doses of As2O3 (0, 0.15, 1.5, and 15 mg/L) through drinking water for 12 weeks. Anxiety-like behaviors were assessed using the open field test (OFT), light/dark choice test, and elevated zero maze (EZM). Neuronal injuries in the cerebral cortex and hippocampus were assessed by light microscopy with H&E and Nissl staining. Ultrastructural alteration in the cerebral cortex was assessed by transmission electron microscope (TEM). The expression levels of GABAergic system-related molecules (i.e., glutamate decarboxylase, GABA transporter, and GABAB receptor subunits) in the prefrontal cortex (PFC) were determined by qRT-PCR and western blotting. Arsenic exposure showed a striking anxiogenic effect on mice, especially in the group exposed to 15 mg/L As2O3. Light microscopy showed neuron necrosis and reduced cell counts. TEM revealed marked ultrastructural changes, including the vacuolated mitochondria, disrupted Nissl bodies, an indentation in the nucleus membrane, and delamination of myelin sheath in the cortex. In addition, As2O3 influenced the GABAergic system in the PFC by decreasing the expression of the glutamate decarboxylase 1 (GAD1) and the GABAB2 receptor subunit, but not the GABAB1 receptor subunit. To sum up, sub-chronic exposure to As2O3 is associated with increased anxiety-like behaviors, which may be mediated by altered GABAergic signaling in the PFC. These findings shed light on the mechanisms responsible for the neurotoxic effects of arsenic and therefore more cautions should be taken.
Collapse
Affiliation(s)
- Xin Hu
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Xiaohong Yuan
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Mingyu Yang
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Mingsheng Han
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Mohammad Mehdi Ommati
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Yanqin Ma
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China.
| |
Collapse
|
13
|
Zhou W, He J, Zhang C, Pan Y, Sang T, Qiu X. Fiber-specific white matter alterations in Parkinson's disease patients with freezing of gait. Brain Res 2023:148440. [PMID: 37271491 DOI: 10.1016/j.brainres.2023.148440] [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: 11/20/2022] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
Freezing of gait (FOG) is a gait disorder that usually occurs in advanced stages of Parkinson's disease (PD). Understanding the underlying mechanism of FOG is important for treatment and prevention. Previous studies have investigated white matter (WM) structure to explore the pathology of FOG. However, the pathology is still unclear, possibly due to the methodological limitation in identifying specific fiber tracts. This study aimed to investigate tract-specific WM structural changes in FOG patients and their relationships with clinical characteristics. We enrolled 19 PD patients with FOG (PD-FOG), 19 without FOG (PD-woFOG) and 21 controls. Fixel-based analysis is a novel framework to avoid the effect of crossing fibers, which provides the metrics to assess WM morphology. By combining a method for segmenting fibers, we identified abnormalities in the specific fiber tracts. Compared to PD-woFOG, PD-FOG showed significant increased fiber-bundle cross-section (FC) in the corpus callosum (CC), fornix (FX), inferior longitudinal fasciculus (ILF), striato-premotor (ST_PREM), superior thalamic radiation (STR), thalamo-premotor (T_PREM), increased fiber density and cross-section (FDC) in the STR, and decreased fiber density (FD) in the CC and ILF. Additionally, the ILF was correlated with motor, cognition and memory, the CC was correlated with anxiety, and the T_PREM was also correlated with cognition. In conclusion, in addition to impairments of WM found in PD-FOG, we found enhancements in WM, which may imply compensatory mechanisms. Furthermore, multiple fiber tracts were correlated with clinical characteristics, especially the ILF, validating the involvement of transmission circuits of multiple distinct information in mechanisms of FOG.
Collapse
Affiliation(s)
- Wenyang Zhou
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Jianzhong He
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Chengzhe Zhang
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Yiang Pan
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Tian Sang
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Xiang Qiu
- Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Department of Automation, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China.
| |
Collapse
|
14
|
Shokouh Alaei H, Ghoshuni M, Vosough I. Directed brain network analysis in anxious and non-anxious depression based on EEG source reconstruction and graph theory. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2023.104666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
15
|
Jain P, Yates M, de Celis CR, Drineas P, Jahanshad N, Thompson P, Paschou P. Multiomic approach and Mendelian randomization analysis identify causal associations between blood biomarkers and subcortical brain structure volumes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.30.23287968. [PMID: 37066330 PMCID: PMC10104218 DOI: 10.1101/2023.03.30.23287968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Alterations in subcortical brain structure volumes have been found to be associated with several neurodegenerative and psychiatric disorders. At the same time, genome-wide association studies (GWAS) have identified numerous common variants associated with brain structure. In this study, we integrate these findings, aiming to identify proteins, metabolites, or microbes that have a putative causal association with subcortical brain structure volumes via a two-sample Mendelian randomization approach. This method uses genetic variants as instrument variables to identify potentially causal associations between an exposure and an outcome. The exposure data that we analyzed comprised genetic associations for 2,994 plasma proteins, 237 metabolites, and 103 microbial genera. The outcome data included GWAS data for seven subcortical brain structure volumes including accumbens, amygdala, caudate, hippocampus, pallidum, putamen, and thalamus. Eleven proteins and six metabolites were found to have a significant association with subcortical structure volumes. We found causal associations between amygdala volume and granzyme A as well as association between accumbens volume and plasma protease c1 inhibitor. Among metabolites, urate had the strongest association with thalamic volume. No significant associations were detected between the microbial genera and subcortical brain structure volumes. We also observed significant enrichment for biological processes such as proteolysis, regulation of the endoplasmic reticulum apoptotic signaling pathway, and negative regulation of DNA binding. Our findings provide insights to the mechanisms through which brain volumes may be affected in the pathogenesis of neurodevelopmental and psychiatric disorders and point to potential treatment targets for disorders that are associated with subcortical brain structure volumes.
Collapse
Affiliation(s)
- Pritesh Jain
- Department of Biological Sciences, Purdue University
| | - Madison Yates
- Department of Biological Sciences, Purdue University
| | | | | | - Neda Jahanshad
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California
| | - Paul Thompson
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California
| | | |
Collapse
|
16
|
Plausible Role of Stem Cell Types for Treating and Understanding the Pathophysiology of Depression. Pharmaceutics 2023; 15:pharmaceutics15030814. [PMID: 36986674 PMCID: PMC10058940 DOI: 10.3390/pharmaceutics15030814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Major Depressive Disorder (MDD), colloquially known as depression, is a debilitating condition affecting an estimated 3.8% of the population globally, of which 5.0% are adults and 5.7% are above the age of 60. MDD is differentiated from common mood changes and short-lived emotional responses due to subtle alterations in gray and white matter, including the frontal lobe, hippocampus, temporal lobe, thalamus, striatum, and amygdala. It can be detrimental to a person’s overall health if it occurs with moderate or severe intensity. It can render a person suffering terribly to perform inadequately in their personal, professional, and social lives. Depression, at its peak, can lead to suicidal thoughts and ideation. Antidepressants manage clinical depression and function by modulating the serotonin, norepinephrine, and dopamine neurotransmitter levels in the brain. Patients with MDD positively respond to antidepressants, but 10–30% do not recuperate or have a partial response accompanied by poor life quality, suicidal ideation, self-injurious behavior, and an increased relapse rate. Recent research shows that mesenchymal stem cells and iPSCs may be responsible for lowering depression by producing more neurons with increased cortical connections. This narrative review discusses the plausible functions of various stem cell types in treating and understanding depression pathophysiology.
Collapse
|
17
|
Sachdeva B, Sachdeva P, Ghosh S, Ahmad F, Sinha JK. Ketamine as a therapeutic agent in major depressive disorder and posttraumatic stress disorder: Potential medicinal and deleterious effects. IBRAIN 2023; 9:90-101. [PMID: 37786516 PMCID: PMC10528797 DOI: 10.1002/ibra.12094] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 10/04/2023]
Abstract
Major depressive disorder (MDD) and posttraumatic stress disorder (PTSD) are the most common causes of emotional distress that impair an individual's quality of life. MDD is a chronic mental illness that affects 300 million people across the world. Clinical manifestations of MDD include fatigue, loss of interest in routine tasks, psychomotor agitation, impaired ability to focus, suicidal ideation, hypersomnolence, altered psychosocial functioning, and appetite loss. Individuals with depression also demonstrate a reduced behavioral response while experiencing pleasure, a symptom known as anhedonia. Like MDD, PTSD is a prevalent and debilitating psychiatric disorder resulting from a traumatic incident such as sexual assault, war, severe accident, or natural disaster. Symptoms such as recalling event phases, hypervigilance, irritability, and anhedonia are common in PTSD. Both MDD and PTSD pose enormous socioeconomic burdens across the globe. The search for effective treatment with minimal side effects is still ongoing. Ketamine is known for its anesthetic and analgesic properties. Psychedelic and psychotropic effects of ketamine have been found on the nervous system, which highlights its toxicity. In this article, the effectiveness of ketamine as a potential therapeutic for PTSD and MDD along with its mechanisms of action, clinical trials, and possible side effects have been discussed.
Collapse
Affiliation(s)
- Bhuvi Sachdeva
- Department of Physics and Astrophysics, Bhagini Nivedita CollegeUniversity of DelhiDelhiIndia
| | | | - Shampa Ghosh
- GloNeuro AcademyNoidaUttar PradeshIndia
- ICMR—National Institute of NutritionTarnakaHyderabadIndia
| | - Faizan Ahmad
- Department of Medical Elementology and ToxicologyJamia HamdardDelhiIndia
| | | |
Collapse
|
18
|
Fowler CH, Gaffrey MS. Reduced cortical surface area globally and in reward-related cortex is associated with elevated depressive symptoms in preschoolers. J Affect Disord 2022; 319:286-293. [PMID: 36162658 DOI: 10.1016/j.jad.2022.09.075] [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: 11/24/2021] [Revised: 08/29/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Elevated depressive symptoms in early childhood strongly predict depression onset in youth. Nevertheless, little is known about the neural correlates of these symptoms, information that is key for understanding the early development of depression. As a result, the present study conducted a novel investigation of the association between cortical structure and depressive symptoms in preschoolers. METHODS Forty-six preschool age children (Mage = 5.90, SD = 0.75), some (N = 15) at high risk for depression, participated in the study. Data included parent-report of child depressive symptoms and measures of child whole brain and regional cortical structure acquired via 3T MRI. RESULTS After adjustment for maternal depression, socio-economic status, child age, child sex, and intracranial volume, reduced total cortical surface area and reduced surface area of the lateral orbitofrontal cortex were associated with elevated depressive symptoms. Cortical thickness was not associated with depressive symptoms. LIMITATIONS The present data are cross-sectional, limiting any causal interpretations. CONCLUSIONS Results suggest that reduced cortical surface area, rather than thickness, is a neural correlate of depressive symptoms in preschoolers. Findings highlight the importance of surface area in reward processing regions (i.e., lateral orbitofrontal cortex) in particular. The present results provide novel insight into early emerging associations between brain structure and features of depression in young children and underscore early childhood as an important developmental period for understanding depression.
Collapse
Affiliation(s)
- Carina H Fowler
- Duke University, Department of Psychology & Neuroscience, Durham, NC 27708, United States of America.
| | - Michael S Gaffrey
- Duke University, Department of Psychology & Neuroscience, Durham, NC 27708, United States of America
| |
Collapse
|
19
|
Fouche JP, Groenewold NA, Sevenoaks T, Heany S, Lochner C, Alonso P, Batistuzzo MC, Cardoner N, Ching CRK, de Wit SJ, Gutman B, Hoexter MQ, Jahanshad N, Kim M, Kwon JS, Mataix-Cols D, Menchon JM, Miguel EC, Nakamae T, Phillips ML, Pujol J, Sakai Y, Yun JY, Soriano-Mas C, Thompson PM, Yamada K, Veltman DJ, van den Heuvel OA, Stein DJ. Shape analysis of subcortical structures in obsessive-compulsive disorder and the relationship with comorbid anxiety, depression, and medication use: A meta-analysis by the OCD Brain Imaging Consortium. Brain Behav 2022; 12:e2755. [PMID: 36106505 PMCID: PMC9575597 DOI: 10.1002/brb3.2755] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Neuroimaging studies of obsessive-compulsive disorder (OCD) patients have highlighted the important role of deep gray matter structures. Less work has however focused on subcortical shape in OCD patients. METHODS Here we pooled brain MRI scans from 412 OCD patients and 368 controls to perform a meta-analysis utilizing the ENIGMA-Shape protocol. In addition, we investigated modulating effects of medication status, comorbid anxiety or depression, and disease duration on subcortical shape. RESULTS There was no significant difference in shape thickness or surface area between OCD patients and healthy controls. For the subgroup analyses, OCD patients with comorbid depression or anxiety had lower thickness of the hippocampus and caudate nucleus and higher thickness of the putamen and pallidum compared to controls. OCD patients with comorbid depression had lower shape surface area in the thalamus, caudate nucleus, putamen, hippocampus, and nucleus accumbens and higher shape surface area in the pallidum. OCD patients with comorbid anxiety had lower shape surface area in the putamen and the left caudate nucleus and higher shape surface area in the pallidum and the right caudate nucleus. Further, OCD patients on medication had lower shape thickness of the putamen, thalamus, and hippocampus and higher thickness of the pallidum and caudate nucleus, as well as lower shape surface area in the hippocampus and amygdala and higher surface area in the putamen, pallidum, and caudate nucleus compared to controls. There were no significant differences between OCD patients without co-morbid anxiety and/or depression and healthy controls on shape measures. In addition, there were also no significant differences between OCD patients not using medication and healthy controls. CONCLUSIONS The findings here are partly consistent with prior work on brain volumes in OCD, insofar as they emphasize that alterations in subcortical brain morphology are associated with comorbidity and medication status. Further work is needed to understand the biological processes contributing to subcortical shape.
Collapse
Affiliation(s)
- Jean-Paul Fouche
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Nynke A Groenewold
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Tatum Sevenoaks
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Sarah Heany
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Christine Lochner
- SAMRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| | - Pino Alonso
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain.,Carlos III Health Institute, Centro de Investigacion Biomedica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Marcelo C Batistuzzo
- Department & Institute of Psychiatry, University of Sao Paulo Medical School, Sao Paulo, Brazil.,Department of Methods and Techniques in Psychology, Pontifical Catholic University, Sao Paulo, SP, Brazil
| | - Narcis Cardoner
- Carlos III Health Institute, Centro de Investigacion Biomedica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Sant Pau Mental Health Group, Institut d'Investigacio Biomedica Sant Pau (IBB-Sant Pau), Hospital de la Sant Creu i Sant Pau, Barcelona, Spain.,Department of Psychiatry and Forensic Medicine, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Christopher R K Ching
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Marina del Rey, California, USA
| | - Stella J de Wit
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Department of Anatomy & Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Boris Gutman
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Marcelo Q Hoexter
- Department & Institute of Psychiatry, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Neda Jahanshad
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Marina del Rey, California, USA
| | - Minah Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jun Soo Kwon
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.,Institute of Human Behavioral Medicine, SNU MRC, Seoul, Republic of Korea.,Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - David Mataix-Cols
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jose M Menchon
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain.,Carlos III Health Institute, Centro de Investigacion Biomedica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Euripedes C Miguel
- Department & Institute of Psychiatry, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Takashi Nakamae
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mary L Phillips
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Jesus Pujol
- MRI Research Unit, Radiology Department, Hospital del Mar, Barcelona, Spain
| | - Yuki Sakai
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.,ATR Brain Information Communication Research Laboratory Group, Kyoto, Japan
| | - Je-Yeon Yun
- Seoul National University Hospital, Seoul, Republic of Korea.,Yeongeon Student Support Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Carles Soriano-Mas
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain.,Carlos III Health Institute, Centro de Investigacion Biomedica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Social Psychology and Quantitative Psychology, Universitat de Barcelona-UB, Barcelona, Spain
| | - Paul M Thompson
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Marina del Rey, California, USA
| | - Kei Yamada
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Department of Anatomy & Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Odile A van den Heuvel
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Department of Anatomy & Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Dan J Stein
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa.,SAMRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
20
|
Li GZ, Liu PH, Zhang AX, Andari E, Zhang KR. A resting state fMRI study of major depressive disorder with and without anxiety. Psychiatry Res 2022; 315:114697. [PMID: 35839636 DOI: 10.1016/j.psychres.2022.114697] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 06/17/2022] [Accepted: 06/25/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND The neurobiology of the Major depressive disorder (MDD) with anxiety is still unclear. The present study aimed to explore the brain correlates of MDD with and without anxiety in men and women during resting-state fMRI. METHODS Two hundred and fifty-four patients with MDD (MDD with anxiety, N = 152) and MDD without anxiety, N = 102) and 228 healthy controls (HCs) participated in this study. We compared the fALFF(fractional amplitude of low-frequency fluctuations) and ReHo(regional homogeneity) of ACC(anterior cingulate cortex) and insula among these three groups. We also compared gender difference between MDD with anxiety and MDD without anxiety. RESULTS We found that the fALFF values within the ACC and insula were significantly lower in MDD with anxiety compared to without anxiety and HCs. However, we did not find differences in ReHo values among the three groups. In women, we found significant differences in fALFF values between MDD with and without anxiety. These differences were not observed in men. CONCLUSIONS It is possible that MDD with anxiety show less spontaneous BOLD-fMRI signal intensity within the ACC and insula compared to MDD without anxiety, especially in women. The fALFF within the ACC and insula can be a potential biomarker for severe MDD phenotype.
Collapse
Affiliation(s)
- Gai-Zhi Li
- Department of psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China; Shanxi Medical University, Taiyuan, Shanxi, China
| | - Peng-Hong Liu
- Department of psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China; Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ai-Xia Zhang
- Department of psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China; Shanxi Medical University, Taiyuan, Shanxi, China
| | - Elissar Andari
- Department of Psychiatry, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.
| | - Ke-Rang Zhang
- Department of psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China; Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
21
|
Sheehan AE, Heilner E, Bounoua N, Miglin R, Spielberg JM, Sadeh N. Cortical thickness in parietal regions link perseverative thinking with suicidal ideation. J Affect Disord 2022; 306:131-137. [PMID: 35304233 PMCID: PMC9100854 DOI: 10.1016/j.jad.2022.03.019] [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: 07/19/2021] [Revised: 12/17/2021] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Suicide represents a major public health concern, as the tenth leading cause of death in the United States. Links between perseverative thinking (PT) and suicidal ideation have previously been examined, while their biological underpinnings remain understudied. The present study had two aims: 1) investigate whether cortical thickness varied as a function of PT, and 2) examine whether variation in thickness partially explained associations between PT and lifetime history of ideation. We hypothesized that cortical thickness would vary as a function of PT and PT would be positively associated with lifetime history of ideation. METHODS A community sample of 73 adults (ages 18-55; 42.5% female) completed self-report measures examining PT and ideation, as well as a neuroimaging protocol. Mean scores on the Perseverative Thinking Questionnaire were entered as the explanatory variable in the analysis of cortical thickness clusters related to PT. The indirect effect of PT on ideation through thickness was tested cross-sectionally. RESULTS PT was positively associated with i) thickness in three clusters bilaterally in the parietal cortex and ii) suicidal ideation. Follow-up analyses revealed a significant indirect effect of PT on suicidal ideation through left superior parietal thickness. LIMITATIONS Limitations of the study include the use of cross-sectional data and a modest sample size. CONCLUSIONS PT is associated with variations in cortical thickness, and increased thickness in the left parietal region may partially explain the link between PT and suicidal ideation, identifying a novel neurobiological mechanism of ideation.
Collapse
Affiliation(s)
- Ana E Sheehan
- Department of Psychological and Brain Sciences, University of Delaware, United States of America.
| | - Emily Heilner
- Department of Psychological and Brain Sciences, University of Delaware, United States of America
| | - Nadia Bounoua
- Department of Psychological and Brain Sciences, University of Delaware, United States of America
| | - Rickie Miglin
- Department of Psychological and Brain Sciences, University of Delaware, United States of America
| | - Jeffrey M Spielberg
- Department of Psychological and Brain Sciences, University of Delaware, United States of America
| | - Naomi Sadeh
- Department of Psychological and Brain Sciences, University of Delaware, United States of America
| |
Collapse
|
22
|
Mertse N, Denier N, Walther S, Breit S, Grosskurth E, Federspiel A, Wiest R, Bracht T. Associations between anterior cingulate thickness, cingulum bundle microstructure, melancholia and depression severity in unipolar depression. J Affect Disord 2022; 301:437-444. [PMID: 35026360 DOI: 10.1016/j.jad.2022.01.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Structural and functional alterations of the anterior cingulate cortex (ACC) have been related to emotional, cognitive and behavioral domains of major depressive disorder. In this study, we investigate cortical thickness of rostral and caudal ACC. In addition, we explore white matter microstructure of the cingulum bundle (CB), a white matter pathway connecting multiple segments of the ACC. We hypothesized reduced cortical thickness and reduced white matter microstructure of the CB in MDD, in particular in the melancholic subtype. In addition, we expect an association between depression severity and CB microstructure. METHODS Fifty-four patients with a current depressive episode and 22 healthy controls matched for age, gender and handedness underwent structural and diffusion-weighted MRI-scans. Cortical thickness of rostral and caudal ACC were computed. The CB was reconstructed bilaterally using manual tractography. Cortical thickness and fractional anisotropy (FA) of bilateral CB were compared first between all patients and healthy controls and second between healthy controls, melancholic and non-melancholic patients. Correlations between FA and depression severity were calculated. RESULTS We found no group differences in rostral and caudal ACC cortical thickness or in FA of the CB comparing all patients with healthy controls. Melancholic patients had reduced cortical thickness of bilateral caudal ACC compared to non-melancholic patients and compared to healthy controls. Across all patients, depression severity was associated with reduced FA in bilateral CB. LIMITATIONS Impact of medication CONCLUSIONS: Cortical thickness of the caudal ACC is associated with the melancholic syndrome. CB microstructure may represent a marker of depression severity.
Collapse
Affiliation(s)
- Nicolas Mertse
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, 3000 Bern 60, Bern, Switzerland
| | - Niklaus Denier
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, 3000 Bern 60, Bern, Switzerland
| | - Sebastian Walther
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, 3000 Bern 60, Bern, Switzerland
| | - Sigrid Breit
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, 3000 Bern 60, Bern, Switzerland
| | - Elmar Grosskurth
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, 3000 Bern 60, Bern, Switzerland
| | - Andrea Federspiel
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, 3000 Bern 60, Bern, Switzerland
| | - Roland Wiest
- Institute of Diagnostic and Interventional Neuroradiology, University of Bern, Bern, Switzerland
| | - Tobias Bracht
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, 3000 Bern 60, Bern, Switzerland.
| |
Collapse
|
23
|
Auerbach RP, Pagliaccio D, Hubbard NA, Frosch I, Kremens R, Cosby E, Jones R, Siless V, Lo N, Henin A, Hofmann SG, Gabrieli JDE, Yendiki A, Whitfield-Gabrieli S, Pizzagalli DA. Reward-Related Neural Circuitry in Depressed and Anxious Adolescents: A Human Connectome Project. J Am Acad Child Adolesc Psychiatry 2022; 61:308-320. [PMID: 33965516 PMCID: PMC8643367 DOI: 10.1016/j.jaac.2021.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/17/2021] [Accepted: 04/26/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Although depression and anxiety often have distinct etiologies, they frequently co-occur in adolescence. Recent initiatives have underscored the importance of developing new ways of classifying mental illness based on underlying neural dimensions that cut across traditional diagnostic boundaries. Accordingly, the aim of the study was to clarify reward-related neural circuitry that may characterize depressed-anxious youth. METHOD The Boston Adolescent Neuroimaging of Depression and Anxiety Human Connectome Project tested group differences regarding subcortical volume and nucleus accumbens activation during an incentive processing task among 14- to 17-year-old adolescents presenting with a primary depressive and/or anxiety disorder (n = 129) or no lifetime history of mental disorders (n = 64). In addition, multimodal modeling examined predictors of depression and anxiety symptom change over a 6-month follow-up period. RESULTS Our findings highlighted considerable convergence. Relative to healthy youth, depressed-anxious adolescents exhibited reduced nucleus accumbens volume and activation following reward receipt. These findings remained when removing all medicated participants (∼59% of depressed-anxious youth). Subgroup analyses comparing anxious-only, depressed-anxious, and healthy youth also were largely consistent. Multimodal modeling showed that only structural alterations predicted depressive symptoms over time. CONCLUSION Multimodal findings highlight alterations within nucleus accumbens structure and function that characterize depressed-anxious adolescents. In the current hypothesis-driven analyses, however, only reduced nucleus accumbens volume predicted depressive symptoms over time. An important next step will be to clarify why structural alterations have an impact on reward-related processes and associated symptoms.
Collapse
|
24
|
Liu PH, Li Y, Zhang AX, Sun N, Li GZ, Chen X, Bai TJ, Bo QJ, Chen GM, Chen NX, Chen TL, Chen W, Cheng C, Cheng YQ, Cui XL, Duan J, Fang YR, Gong QY, Guo WB, Hou ZH, Hu L, Kuang L, Li F, Li KM, Li T, Liu YS, Liu ZN, Long YC, Luo QH, Meng HQ, Peng DH, Qiu HT, Qiu J, Shen YD, Shi YS, Wang F, Wang K, Wang L, Wang X, Wang Y, Wu XP, Wu XR, Xie CM, Xie GR, Xie HY, Xie P, Xu XF, Yang H, Yang J, Yao JS, Yao SQ, Yin YY, Yuan YG, Zhang H, Zhang L, Zhang ZJ, Zhou RB, Zhou YT, Zhu JJ, Zou CJ, Si TM, Zuo XN, Yan CG, Zhang KR. Brain structural alterations in MDD patients with gastrointestinal symptoms: Evidence from the REST-meta-MDD project. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110386. [PMID: 34119573 DOI: 10.1016/j.pnpbp.2021.110386] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE While gastrointestinal (GI) symptoms are very common in patients with major depressive disorder (MDD), few studies have investigated the neural basis behind these symptoms. In this study, we sought to elucidate the neural basis of GI symptoms in MDD patients by analyzing the changes in regional gray matter volume (GMV) and gray matter density (GMD) in brain structure. METHOD Subjects were recruited from 13 clinical centers and categorized into three groups, each of which is based on the presence or absence of GI symptoms: the GI symptoms group (MDD patients with at least one GI symptom), the non-GI symptoms group (MDD patients without any GI symptoms), and the healthy control group (HCs). Structural magnetic resonance images (MRI) were collected of 335 patients in the GI symptoms group, 149 patients in the non-GI symptoms group, and 446 patients in the healthy control group. The 17-item Hamilton Depression Rating Scale (HAMD-17) was administered to all patients. Correlation analysis and logistic regression analysis were used to determine if there was a correlation between the altered brain regions and the clinical symptoms. RESULTS There were significantly higher HAMD-17 scores in the GI symptoms group than that of the non-GI symptoms group (P < 0.001). Both GMV and GMD were significant different among the three groups for the bilateral superior temporal gyrus, bilateral middle temporal gyrus, left lingual gyrus, bilateral caudate nucleus, right Fusiform gyrus and bilateral Thalamus (GRF correction, cluster-P < 0.01, voxel-P < 0.001). Compared to the HC group, the GI symptoms group demonstrated increased GMV and GMD in the bilateral superior temporal gyrus, and the non-GI symptoms group demonstrated an increased GMV and GMD in the right superior temporal gyrus, right fusiform gyrus and decreased GMV in the right Caudate nucleus (GRF correction, cluster-P < 0.01, voxel-P < 0.001). Compared to the non-GI symptoms group, the GI symptoms group demonstrated significantly increased GMV and GMD in the bilateral thalamus, as well as decreased GMV in the bilateral superior temporal gyrus and bilateral insula lobe (GRF correction, cluster-P < 0.01, voxel-P < 0.001). While these changed brain areas had significantly association with GI symptoms (P < 0.001), they were not correlated with depressive symptoms (P > 0.05). Risk factors for gastrointestinal symptoms in MDD patients (p < 0.05) included age, increased GMD in the right thalamus, and decreased GMV in the bilateral superior temporal gyrus and left Insula lobe. CONCLUSION MDD patients with GI symptoms have more severe depressive symptoms. MDD patients with GI symptoms exhibited larger GMV and GMD in the bilateral thalamus, and smaller GMV in the bilateral superior temporal gyrus and bilateral insula lobe that were correlated with GI symptoms, and some of them and age may contribute to the presence of GI symptoms in MDD patients.
Collapse
Affiliation(s)
- Peng-Hong Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, China; Department of First Clinical Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Yan Li
- Department of Clinical Medicine, Fenyang College of Shanxi Medical University, 032200, China
| | - Ai-Xia Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Ning Sun
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, China; Department of First Clinical Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Gai-Zhi Li
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, China; Department of First Clinical Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Xiao Chen
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100054, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100054, China
| | - Tong-Jian Bai
- Anhui Medical University, Hefei, Anhui 230022, China
| | - Qi-Jing Bo
- Beijing Anding Hospital, Capital Medical University, Beijing 100054, China
| | - Guan-Mao Chen
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Ning-Xuan Chen
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100054, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100054, China
| | - Tao-Lin Chen
- Mental Health Center, West China Hospital, Sichuan University Chengdu, Sichuan 610041, China
| | - Wei Chen
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310012, China
| | - Chang Cheng
- The Institute of Mental Health, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yu-Qi Cheng
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650221, China
| | - Xi-Long Cui
- The Institute of Mental Health, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Jia Duan
- Department of Psychiatry, First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Yi-Ru Fang
- Department of Psychiatry, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Qi-Yong Gong
- Department of Radiology, Huaxi MR Research Center,West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Psychoradiology Research Unit of Chinese Academy of Medical Sciences, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Wen-Bin Guo
- The Institute of Mental Health, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Zheng-Hua Hou
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lan Hu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Li Kuang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Feng Li
- Anhui Medical University, Hefei, Anhui 230022, China
| | - Kai-Ming Li
- Department of Radiology, Huaxi MR Research Center,West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Tao Li
- Mental Health Center, West China Hospital, Sichuan University Chengdu, Sichuan 610041, China
| | - Yan-Song Liu
- Department of Clinical Psychology, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu 215137, China
| | - Zhe-Ning Liu
- The Institute of Mental Health, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yi-Cheng Long
- The Institute of Mental Health, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Qing-Hua Luo
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hua-Qing Meng
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dai-Hui Peng
- Department of Psychiatry, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Hai-Tang Qiu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiang Qiu
- Faculty of Psychology, Southwest University, Chongqing 400716, China
| | - Yue-Di Shen
- Department of Diagnostics, Affiliated Hospital, Hangzhou Normal University Medical School, Hangzhou, Zhejiang 311121, China
| | - Yu-Shu Shi
- Mental Health Center, West China Hospital, Sichuan University Chengdu, Sichuan 610041, China
| | - Fei Wang
- Department of Psychiatry, First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Kai Wang
- Anhui Medical University, Hefei, Anhui 230022, China
| | - Li Wang
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing 100191, China; Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing 100191, China
| | - Xiang Wang
- The Institute of Mental Health, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ying Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Xiao-Ping Wu
- Xi'an Central Hospital, Xi'an, Shannxi 710003, China
| | - Xin-Ran Wu
- Faculty of Psychology, Southwest University, Chongqing 400716, China
| | - Chun-Ming Xie
- Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing, Jiangsu 210009, China
| | - Guang-Rong Xie
- The Institute of Mental Health, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Hai-Yan Xie
- Department of Psychiatry, The Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Peng Xie
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiu-Feng Xu
- Beijing Anding Hospital, Capital Medical University, Beijing 100054, China
| | - Hong Yang
- Department of Radiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jian Yang
- The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710061, China
| | - Jia-Shu Yao
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310012, China
| | - Shu-Qiao Yao
- The Institute of Mental Health, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ying-Ying Yin
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yong-Gui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hong Zhang
- Xi'an Central Hospital, Xi'an, Shannxi 710003, China
| | - Lei Zhang
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100054, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100054, China
| | - Zhi-Jun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing, Jiangsu 210009, China
| | - Ru-Bai Zhou
- Department of Psychiatry, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Yi-Ting Zhou
- Mental Health Center, West China Hospital, Sichuan University Chengdu, Sichuan 610041, China
| | - Jun-Juan Zhu
- Department of Psychiatry, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Chao-Jie Zou
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650221, China
| | - Tian-Mei Si
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing 100191, China; Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing 100191, China
| | - Xi-Nian Zuo
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100054, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100054, China; Magnetic Resonance Imaging Research Center and Research Center for Lifespan Development of Mind and Brain (CLIMB), Institute of Psychology, Chinese Academy of Sciences, Beijing 100054, China
| | - Chao-Gan Yan
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100054, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100054, China; Magnetic Resonance Imaging Research Center and Research Center for Lifespan Development of Mind and Brain (CLIMB), Institute of Psychology, Chinese Academy of Sciences, Beijing 100054, China; Department of Child and Adolescent Psychiatry, NYU Langone Medical Center School of Medicine, New York, NY 10016, USA
| | - Ke-Rang Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
25
|
Tozzi L, Anene ET, Gotlib IH, Wintermark M, Kerr AB, Wu H, Seok D, Narr KL, Sheline YI, Whitfield-Gabrieli S, Williams LM. Convergence, preliminary findings and future directions across the four human connectome projects investigating mood and anxiety disorders. Neuroimage 2021; 245:118694. [PMID: 34732328 PMCID: PMC8727513 DOI: 10.1016/j.neuroimage.2021.118694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/11/2021] [Accepted: 10/29/2021] [Indexed: 12/31/2022] Open
Abstract
In this paper we provide an overview of the rationale, methods, and preliminary results of the four Connectome Studies Related to Human Disease investigating mood and anxiety disorders. The first study, "Dimensional connectomics of anxious misery" (HCP-DAM), characterizes brain-symptom relations of a transdiagnostic sample of anxious misery disorders. The second study, "Human connectome Project for disordered emotional states" (HCP-DES), tests a hypothesis-driven model of brain circuit dysfunction in a sample of untreated young adults with symptoms of depression and anxiety. The third study, "Perturbation of the treatment resistant depression connectome by fast-acting therapies" (HCP-MDD), quantifies alterations of the structural and functional connectome as a result of three fast-acting interventions: electroconvulsive therapy, serial ketamine therapy, and total sleep deprivation. Finally, the fourth study, "Connectomes related to anxiety and depression in adolescents" (HCP-ADA), investigates developmental trajectories of subtypes of anxiety and depression in adolescence. The four projects use comparable and standardized Human Connectome Project magnetic resonance imaging (MRI) protocols, including structural MRI, diffusion-weighted MRI, and both task and resting state functional MRI. All four projects also conducted comprehensive and convergent clinical and neuropsychological assessments, including (but not limited to) demographic information, clinical diagnoses, symptoms of mood and anxiety disorders, negative and positive affect, cognitive function, and exposure to early life stress. The first round of analyses conducted in the four projects offered novel methods to investigate relations between functional connectomes and self-reports in large datasets, identified new functional correlates of symptoms of mood and anxiety disorders, characterized the trajectory of connectome-symptom profiles over time, and quantified the impact of novel treatments on aberrant connectivity. Taken together, the data obtained and reported by the four Connectome Studies Related to Human Disease investigating mood and anxiety disorders describe a rich constellation of convergent biological, clinical, and behavioral phenotypes that span the peak ages for the onset of emotional disorders. These data are being prepared for open sharing with the scientific community following screens for quality by the Connectome Coordinating Facility (CCF). The CCF also plans to release data from all projects that have been pre-processed using identical state-of-the-art pipelines. The resultant dataset will give researchers the opportunity to pool complementary data across the four projects to study circuit dysfunctions that may underlie mood and anxiety disorders, to map cohesive relations among circuits and symptoms, and to probe how these relations change as a function of age and acute interventions. This large and combined dataset may also be ideal for using data-driven analytic approaches to inform neurobiological targets for future clinical trials and interventions focused on clinical or behavioral outcomes.
Collapse
Affiliation(s)
- Leonardo Tozzi
- Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Esther T Anene
- Psychiatry, Neurology, Radiology, University of Pennsylvania, Philadelphia PA, USA
| | | | | | - Adam B Kerr
- Center for Cognitive and Neurobiological Imaging, Stanford University, CA, USA; Electrical Engineering, Stanford University, CA, USA
| | - Hua Wu
- Electrical Engineering, Stanford University, CA, USA
| | - Darsol Seok
- Department of Psychiatry, University of Pennsylvania, Philadelphia PA, USA
| | - Katherine L Narr
- Neurology, Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA
| | - Yvette I Sheline
- Neurology, Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA.
| | | | - Leanne M Williams
- Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Sierra-Pacific Mental Illness Research, Education, and Clinical Center (MIRECC) Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
26
|
Chien YL, Chen YC, Chiu YN, Tsai WC, Gau SSF. A translational exploration of the effects of WNT2 variants on altered cortical structures in autism spectrum disorder. J Psychiatry Neurosci 2021; 46:E647-E658. [PMID: 34862305 PMCID: PMC8648347 DOI: 10.1503/jpn.210022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/19/2021] [Accepted: 07/28/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Evidence suggests that cortical anatomy may be aytpical in autism spectrum disorder. The wingless-type MMTV integration site family, member 2 (WNT2), a candidate gene for autism spectrum disorder, may regulate cortical development. However, it is unclear whether WNT2 variants are associated with altered cortical thickness in autism spectrum disorder. METHODS In a sample of 118 people with autism spectrum disorder and 122 typically developing controls, we investigated cortical thickness using FreeSurfer software. We then examined the main effects of the WNT2 variants and the interactions of group × SNP and age × SNP for each hemisphere and brain region that was altered in people with autism spectrum disorder. RESULTS Compared to neurotypical controls, people with autism spectrum disorder showed reduced mean cortical thickness in both hemispheres and 9 cortical regions after false discovery rate correction, including the right cingulate gyrus, the orbital gyrus, the insula, the inferior frontal gyrus (orbital part and triangular part), the lateral occipitotemporal gyrus, the posterior transverse collateral sulcus, the lateral sulcus and the superior temporal sulcus. In the full sample, 2 SNPs of WNT2 (rs6950765 and rs2896218) showed age × SNP interactions for the mean cortical thickness of both hemispheres, the middle-posterior cingulate cortex and the superior temporal cortex. LIMITATIONS We examined the genetic effect for each hemisphere and the 9 regions that were altered in autism spectrum disorder. The age effect we found in this cross-sectional study needs to be examined in longitudinal studies. CONCLUSION Based on neuroimaging and genetic data, our findings suggest that WNT2 variants might be associated with altered cortical thickness in autism spectrum disorder. Whether and how these WNT2 variants might involve cortical thinning requires further investigation. TRIAL REGISTRATION ClinicalTrials.gov no. NCT01582256. PROTOCOL REGISTRATION National Institutes of Health no. NCT00494754.
Collapse
Affiliation(s)
| | | | | | | | - Susan Shur-Fen Gau
- From the Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan (Chien, Chen, Chiu, Tsai, Gau); and the Graduate Institute of Clinical Medicine, and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (Chen, Gau)
| |
Collapse
|
27
|
Liu P, Tu H, Zhang A, Yang C, Liu Z, Lei L, Wu P, Sun N, Zhang K. Brain functional alterations in MDD patients with somatic symptoms: A resting-state fMRI study. J Affect Disord 2021; 295:788-796. [PMID: 34517253 DOI: 10.1016/j.jad.2021.08.143] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/22/2021] [Accepted: 08/27/2021] [Indexed: 01/22/2023]
Abstract
OBJECTIVE It has been established that major depressive disorder (MDD) is accompanied by various somatic symptoms that are related to the clinical course and severity of depression. However, the mechanisms of somatic symptoms in MDD have rarely been studied. In this study, we sought to investigate the functional neurological changes in MDD patients with somatic symptoms based off the regional homogeneity (ReHo) and the amplitude of low-frequency fluctuation (ALFF). METHOD Study participants included 74 first-episode, drug naïve MDD patients as well as 70 healthy subjects (HCs). Patients diagnosed with MDD were separated into two groups based on the presence (n=50) or absence (n=24) of somatic symptoms. Functional images were obtained and analyzed. Alterations in ReHo/ALFF and the severity of clinical symptoms were investigated using correlation analysis. RESULTS More severe depressive symptoms were observed in the somatic depression group than that of the pure depression group (P< 0.001). Furthermore, there was a significant reduction in ReHo and ALFF in the bilateral precentral gyrus, bilateral postcentral gyrus, and left paracentral gyrus in the somatic MDD group as compared to the pure depression group (GRF correction, voxel-P< 0.001, cluster-P < 0.01). Pearson correlation analysis revealed a negative correlation between ReHo and ALFF values in these abnomal regions with the severity of somatic and depressive symptoms (P< 0.01). CONCLUSION Somatic depression is more severe than pure depression. The ReHo and ALFF changes in the precentral gyrus, postcentral gyrus, and paracentral gyrus may serve a significant role in the pathophysiology of somatic symptoms in MDD.
Collapse
Affiliation(s)
- Penghong Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China; Shanxi Medical University, Taiyuan 030001, PR China
| | - Hongwei Tu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China; Shanxi Medical University, Taiyuan 030001, PR China
| | - Aixia Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Chunxia Yang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Zhifen Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Lei Lei
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China; Department of Psychiatry, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Peiyi Wu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China; Shanxi Medical University, Taiyuan 030001, PR China
| | - Ning Sun
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Kerang Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| |
Collapse
|
28
|
Yeung HW, Shen X, Stolicyn A, de Nooij L, Harris MA, Romaniuk L, Buchanan CR, Waiter GD, Sandu AL, McNeil CJ, Murray A, Steele JD, Campbell A, Porteous D, Lawrie SM, McIntosh AM, Cox SR, Smith KM, Whalley HC. Spectral clustering based on structural magnetic resonance imaging and its relationship with major depressive disorder and cognitive ability. Eur J Neurosci 2021; 54:6281-6303. [PMID: 34390586 DOI: 10.1111/ejn.15423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/09/2021] [Indexed: 11/29/2022]
Abstract
There is increasing interest in using data-driven unsupervised methods to identify structural underpinnings of common mental illnesses, including major depressive disorder (MDD) and associated traits such as cognition. However, studies are often limited to severe clinical cases with small sample sizes and most do not include replication. Here, we examine two relatively large samples with structural magnetic resonance imaging (MRI), measures of lifetime MDD and cognitive variables: Generation Scotland (GS subsample, N = 980) and UK Biobank (UKB, N = 8,900), for discovery and replication, using an exploratory approach. Regional measures of FreeSurfer derived cortical thickness (CT), cortical surface area (CSA), cortical volume (CV) and subcortical volume (subCV) were input into a clustering process, controlling for common covariates. The main analysis steps involved constructing participant K-nearest neighbour graphs and graph partitioning with Markov stability to determine optimal clustering of participants. Resultant clusters were (1) checked whether they were replicated in an independent cohort and (2) tested for associations with depression status and cognitive measures. Participants separated into two clusters based on structural brain measurements in GS subsample, with large Cohen's d effect sizes between clusters in higher order cortical regions, commonly associated with executive function and decision making. Clustering was replicated in the UKB sample, with high correlations of cluster effect sizes for CT, CSA, CV and subCV between cohorts across regions. The identified clusters were not significantly different with respect to MDD case-control status in either cohort (GS subsample: pFDR = .2239-.6585; UKB: pFDR = .2003-.7690). Significant differences in general cognitive ability were, however, found between the clusters for both datasets, for CSA, CV and subCV (GS subsample: d = 0.2529-.3490, pFDR < .005; UKB: d = 0.0868-0.1070, pFDR < .005). Our results suggest that there are replicable natural groupings of participants based on cortical and subcortical brain measures, which may be related to differences in cognitive performance, but not to the MDD case-control status.
Collapse
Affiliation(s)
- Hon Wah Yeung
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Xueyi Shen
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Aleks Stolicyn
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Laura de Nooij
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Mathew A Harris
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Liana Romaniuk
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Colin R Buchanan
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Gordon D Waiter
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Anca-Larisa Sandu
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Christopher J McNeil
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Alison Murray
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - J Douglas Steele
- School of Medicine, University of Dundee, Dundee, UK.,Department of Neurology, NHS Tayside, Ninewells Hospital and Medical School, Dundee, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - David Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.,Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | | | - Andrew M McIntosh
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK.,Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Simon R Cox
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Keith M Smith
- Usher Institute, University of Edinburgh, Edinburgh, UK.,Health Data Research UK, London, UK
| | | |
Collapse
|
29
|
Quantitative evaluation of brain volumes in drug-free major depressive disorder using MRI-Cloud method. Neuroreport 2021; 32:1027-1034. [PMID: 34075004 DOI: 10.1097/wnr.0000000000001682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Quantitative analysis of the high-resolution T1-weighted images provides useful markers to measure anatomical changes during brain degeneration related to major depressive disorder (MDD). However, there are controversial findings regarding these volume alterations in MDD indicating even to increased volumes in some specific regions in MDD patients. METHODS This study is a case-controlled study including 23 depression patients and 15 healthy subject person and 20-38 years of age, who have been treated at the Neurology and Psychiatry Department here. We compared specific anatomic regions between drug-free MDD patients and control group through MRI-Cloud, which is a novel brain imaging method that enables to analyze multiple brain regions simultaneously. RESULTS We have found that frontal, temporal, and parietal hemispheric volumes and middle frontal gyrus, inferior frontal gyrus, superior parietal gyrus, cingulum-hippocampus, lateral fronto-orbital gyrus, superior temporal gyrus, superior temporal white matter, middle temporal gyrus subanatomic regions were significantly reduced bilaterally in MDD patients compared to the control group, while striatum, amygdala, putamen, and nucleus accumbens bilaterally increased in MDD group compared to the control group suggesting that besides the heterogeneity among studies, also comorbid factors such as anxiety and different personal traits could be responsible for these discrepant results. CONCLUSION Our study gives a strong message that depression is associated with altered structural brain volumes, especially, in drug-free and first-episode MDD patients who present with similar duration and severity of depression while the role of demographic and comorbid risk factors should not be neglected.
Collapse
|
30
|
Im S, Lee J, Kim S. Preliminary Comparison of Subcortical Structures in Elderly Subclinical Depression: Structural Analysis with 3T MRI. Exp Neurobiol 2021; 30:183-202. [PMID: 33972469 PMCID: PMC8118753 DOI: 10.5607/en20056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/19/2021] [Accepted: 02/17/2021] [Indexed: 01/23/2023] Open
Abstract
Depression in the elderly population has shown increased likelihood of neurological disorders due to structural changes in the subcortical area. However, further investigation into depression related subcortical changes is needed due to mismatches in structural analysis results between studies as well as scarcities in research regarding subcortical connectivity patterns of subclinical depression populations. This study aims to investigate structural differences in subcortical regions of aged participants with subclinical depression using 3Tesla MRI. In structural analysis, volumes of each subcortical region were measured to observe the volumetric difference and asymmetry between groups, but no significant difference was found. In addition, fractional anisotropy (FA) and apparent diffusion coefficient (ADC) did not show any significant differences between groups. Structural analysis using probabilistic tractography indicated that the connection strength between left nucleus accumbens-right hippocampus, and right thalamus-right caudate was higher in the control group than the subclinical depression group. The differences in subcortical connection strength of subclinical depression groups, have shown to correlate with emotional and cognitive disorders, such as anxiety and memory impairment. We believe that the analysis of structural differences and cross-regional network measures in subcortical structures can help identify neurophysiological changes occurring in subclinical depression.
Collapse
Affiliation(s)
- SangJin Im
- Lee Gil Ya Cancer & Diabetes Institute, Gachon University, Incheon 21999, Korea
| | - Jeonghwan Lee
- Department of Psychiatry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Siekyeong Kim
- Department of Psychiatry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| |
Collapse
|
31
|
Ndlovu NA, Morgan N, Malapile S, Subramaney U, Daniels W, Naidoo J, van den Heuvel MP, Calvey T. Fronto-temporal cortical atrophy in 'nyaope' combination heroin and cannabis use disorder. Drug Alcohol Depend 2021; 221:108630. [PMID: 33667779 DOI: 10.1016/j.drugalcdep.2021.108630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 12/21/2022]
Abstract
Sub-Saharan Africa is one of the top three regions with the highest rates of opioid-related premature mortality. Nyaope is the street name for what is believed to be a drug cocktail in South Africa although recent research suggests that it is predominantly heroin. Nyaope powder is most commonly smoked together with cannabis, a drug-use pattern unique to the region. Due to the increasing burden of this drug in low-income communities and the absence of human structural neuroimaging data of combination heroin and cannabis use disorder, we initiated an important cohort study in order to identify neuroanatomical sequelae. Twenty-eight male nyaope users and thirty healthy, matched controls were recruited from drug rehabilitation centers and the community, respectively. T1-weighted MRI images were obtained using a 3 T General Electric Discovery and cortical thickness was examined and compared. Nyaope users displayed extensive grey matter atrophy in the right hemispheric medial orbitofrontal, rostral middle frontal, superior temporal, superior frontal, and supramarginal gyri (two-sided t-test, p < 0.05, corrected for multiple comparisons). Our findings indicate cortical abnormality in nyaope users in regions involved in impulse control, decision making, social- and self-perception, and working memory. Importantly, affected brain regions show large overlap with the pattern of cortical abnormalities shown in heroin use disorder.
Collapse
Affiliation(s)
- Nhanisi A Ndlovu
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nirvana Morgan
- Department of Psychiatry, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stella Malapile
- The Nelson Mandela Children's Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Ugasvaree Subramaney
- Department of Psychiatry, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - William Daniels
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jaishree Naidoo
- Department of Radiology, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - Martijn P van den Heuvel
- Department of Complex Trait Genetics, CNCR, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Child Psychiatry, Amsterdam UMC, Amsterdam, the Netherlands
| | - Tanya Calvey
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
32
|
MORC1 methylation and BDI are associated with microstructural features of the hippocampus and medial prefrontal cortex. J Affect Disord 2021; 282:91-97. [PMID: 33401128 DOI: 10.1016/j.jad.2020.12.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND Alterations in the hippocampus and prefrontal cortex (PFC) have frequently been reported in depressed patients. These parameters might prove to be a consistent finding in depression. In addition, peripheral DNA methylation of the MORC1 gene promoter showed stable associations with depression across independent samples. However, the question arises whether MORC1, supposedly acting as transcription factor, might also be involved in neurobiological alterations accompanying depression. This study further analyses the role of MORC1 in depression by investigating a potential correlation between peripheral MORC1 DNA methylation and neuronal structural properties previously associated with depression in humans. METHODS Beck Depression Inventory (BDI) was assessed in 52 healthy participants. DNA was extracted from buccal cells and MORC1 methylation correlated with micro- and macrostructural properties derived from magnetic resonance imaging (MRI) and neurite orientation dispersion and density imaging (NODDI) in the hippocampus and medial prefrontal cortex (mPFC). RESULTS MORC1 methylation was associated with volume reduction and neurite orientation dispersion and density markers in the hippocampus and mPFC. BDI was positively associated with neurite orientation dispersion and density markers in the hippocampus. LIMITATIONS The study was conducted in a small sample of healthy participants with subclinical depressive symptoms. Peripheral tissue was analyzed. CONCLUSION We found significant negative associations between peripheral MORC1 methylation and macro- and microstructural markers in the hippocampus and mPFC. Thus, MORC1 might be involved in neurobiological properties. Studies investigating neuronal methylation patterns of MORC1 are needed to support this hypothesis.
Collapse
|
33
|
Sampedro F, Farrés CCI, Soler J, Elices M, Schmidt C, Corripio I, Domínguez-Clavé E, Pomarol-Clotet E, Salvador R, Pascual JC. Structural brain abnormalities in borderline personality disorder correlate with clinical severity and predict psychotherapy response. Brain Imaging Behav 2021; 15:2502-2512. [PMID: 33634348 DOI: 10.1007/s11682-021-00451-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2021] [Indexed: 11/24/2022]
Abstract
Although previous imaging studies in borderline personality disorder (BPD) have found brain abnormalities, the results have been inconsistent. This study aimed to investigate structural brain abnormalities using voxel-based morphometry (VBM) and cortical thickness (Cth) analyses in a large sample of patients with BPD. Additionally, we aimed to determine the correlation between structural abnormalities and clinical severity and to assess its potential value at predicting psychotherapeutic response. Sixty-one individuals with BPD and 19 healthy controls underwent magnetic resonance imaging. Participants with BPD completed several self-report clinical scales, received dialectical-behavioral therapy skills training and post-therapy changes in clinical scores were also recorded. Gray matter volume (GMV) and Cth differences between groups were compared. Within the BPD group, we further characterized the structural brain correlates of clinical severity and investigated the relationship between pre-therapy structural abnormalities and therapeutic response. As potential confounders we included age, sex, educational level, and total intracranial volume (the latter only in VBM analyses). Compared to controls, the BPD group showed a reduced GMV/Cth in prefrontal areas but increased GMV in the limbic structures (amygdala and parahippocampal regions). Prefrontal abnormalities correlated with higher baseline scores on impulsivity and general BPD severity. Increased GMV in the parahippocampal area correlated with a greater emotion dysregulation. Importantly, several baseline structural abnormalities correlated with worse response to psychotherapy. Patients with BPD showed a reduced GMV in the prefrontal areas but a greater GMV in the limbic structures. Several structural abnormalities (i.e. middle and inferior prefrontal areas, anterior insula, or parahippocampal area) correlated with clinical severity and could potentially be used as imaging biological correlates biomarkers to predict psychotherapy response.
Collapse
Affiliation(s)
- Frederic Sampedro
- Movement Disorders Unit Neurology Department Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Cristina Carmona I Farrés
- Department of Psychiatry Hospital de la Santa Creu i Sant Pau, C/ Sant Antoni Mª Claret, 167.08025, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM, Madrid, Spain
| | - Joaquim Soler
- Department of Psychiatry Hospital de la Santa Creu i Sant Pau, C/ Sant Antoni Mª Claret, 167.08025, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM, Madrid, Spain. .,Department of Psychiatry and Legal Medicine, Autonomous University of Barcelona, UAB, Barcelona, Spain.
| | - Matilde Elices
- Department of Psychiatry Hospital de la Santa Creu i Sant Pau, C/ Sant Antoni Mª Claret, 167.08025, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM, Madrid, Spain
| | - Carlos Schmidt
- Department of Psychiatry Hospital de la Santa Creu i Sant Pau, C/ Sant Antoni Mª Claret, 167.08025, Barcelona, Spain
| | - Iluminada Corripio
- Department of Psychiatry Hospital de la Santa Creu i Sant Pau, C/ Sant Antoni Mª Claret, 167.08025, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM, Madrid, Spain.,Department of Psychiatry and Legal Medicine, Autonomous University of Barcelona, UAB, Barcelona, Spain
| | - Elisabet Domínguez-Clavé
- Department of Psychiatry Hospital de la Santa Creu i Sant Pau, C/ Sant Antoni Mª Claret, 167.08025, Barcelona, Spain
| | - Edith Pomarol-Clotet
- Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM, Madrid, Spain.,FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
| | - Raymond Salvador
- Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM, Madrid, Spain.,FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
| | - Juan C Pascual
- Department of Psychiatry Hospital de la Santa Creu i Sant Pau, C/ Sant Antoni Mª Claret, 167.08025, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM, Madrid, Spain.,Department of Psychiatry and Legal Medicine, Autonomous University of Barcelona, UAB, Barcelona, Spain
| |
Collapse
|
34
|
Wang X, Wang T, Sun L, Zhang H, Liu C, Zhang C, Yu L. B-vitamin supplementation ameliorates anxiety- and depression-like behavior induced by gestational urban PM 2.5 exposure through suppressing neuroinflammation in mice offspring. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 266:115146. [PMID: 32663728 DOI: 10.1016/j.envpol.2020.115146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/03/2020] [Accepted: 06/28/2020] [Indexed: 06/11/2023]
Abstract
PM2.5 exposure is an emerging environmental concern and severe health insult closely related to psychological conditions such as anxiety and depression in adolescence. Adolescence is a critical period for neural system development characterized by continuous brain maturation, especially in the prefrontal cortex. The etiology of these adolescent conditions may derive from fetal origin, probably attributed to the adverse effects induced by intrauterine environmental exposure. Anxiety- and depression-like behavior can be induced by gestational exposure to PM2.5 in mice offspring which act as a useful model system. Recent studies show that B-vitamin may alleviate PM2.5-induced hippocampal neuroinflammation- and function-related spatial memory impairment in adolescent mice offspring. However, cortical damage and related neurobehavioral defects induced by gestational PM2.5 exposure, as well as the potential reversibility by interventions in mice offspring require to be elucidated. Here, we aimed to investigate whether B-vitamin would protect mice offspring from the adverse effects derived from gestational exposure to urban PM2.5 on cortical areas to which anxiety and depression are closely related. Pregnant mice were divided into three groups: control group (treated with PBS alone), model group (treated with both PM2.5 and PBS), and intervention group (treated with both PM2.5 and B-vitamin), respectively. The mice offspring were then applied to comprehensive neurobehavioral, ultrastructural, biochemical, and molecular biological analyses. Interestingly, we observed that gestational PM2.5 exposure led to neurobehavioral defects including anxiety- and depression-like behavior. In addition, neuroinflammation, oxidative damage, increased apoptosis, and caspase-1-mediated inflammasome activation in the prefrontal cortex were observed. Notably, both behavioral and molecular changes could be significantly alleviated by B-vitamin treatment. In summary, our results suggest that the anxiety- and depression-like behavior induced by gestational PM2.5 exposure in mice offspring can be ameliorated by B-vitamin supplementation, probably through the suppression of apoptosis, oxidative damage, neuroinflammation, and caspase-1-mediated inflammasome activation.
Collapse
Affiliation(s)
- Xia Wang
- School of Public Health, Weifang Medical University, Weifang, China
| | - Tingting Wang
- Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Department of Histology and Embryology, Weifang Medical University, Weifang, China
| | - Lijuan Sun
- Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Department of Histology and Embryology, Weifang Medical University, Weifang, China
| | - Haoyun Zhang
- Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Department of Histology and Embryology, Weifang Medical University, Weifang, China
| | - Chong Liu
- Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Department of Histology and Embryology, Weifang Medical University, Weifang, China
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Li Yu
- Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Department of Histology and Embryology, Weifang Medical University, Weifang, China.
| |
Collapse
|
35
|
Espinoza Oyarce DA, Shaw ME, Alateeq K, Cherbuin N. Volumetric brain differences in clinical depression in association with anxiety: a systematic review with meta-analysis. J Psychiatry Neurosci 2020; 45:406-429. [PMID: 32726102 PMCID: PMC7595741 DOI: 10.1503/jpn.190156] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Structural differences associated with depression have not been confirmed in brain regions apart from the hippocampus. Comorbid anxiety has been inconsistently assessed, and may explain discrepancies in previous findings. We investigated the link between depression, comorbid anxiety and brain structure. METHODS We followed Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines (PROSPERO CRD42018089286). We searched the Cochrane Library, MEDLINE, PsycInfo, PubMed and Scopus, from database inception to Sept. 13, 2018, for MRI case-control studies that reported brain volumes in healthy adults and adults with clinical depression. We summarized mean volumetric differences using meta-analyses, and we assessed demographics, depression factors and segmentation procedure as moderators using meta-regressions. RESULTS We included 112 studies in the meta-analyses, assessing 4911 healthy participants and 5934 participants with depression (mean age 49.8 yr, 68.2% female). Volume effects were greater in late-onset depression and in multiple episodes of depression. Adults with depression and no comorbidity showed significantly lower volumes in the putamen, pallidum and thalamus, as well as significantly lower grey matter volume and intracranial volume; the largest effects were in the hippocampus (6.8%, p < 0.001). Adults with depression and comorbid anxiety showed significantly higher volumes in the amygdala (3.6%, p < 0.001). Comorbid anxiety lowered depression effects by 3% on average. Sex moderated reductions in intracranial volume. LIMITATIONS High heterogeneity in hippocampus effects could not be accounted for by any moderator. Data on symptom severity and medication were sparse, but other factors likely made significant contributions. CONCLUSION Depression-related differences in brain structure were modulated by comorbid anxiety, chronicity of symptoms and onset of illness. Early diagnosis of anxiety symptomatology will prove crucial to ensuring effective, tailored treatments for improving long-term mental health and mitigating cognitive problems, given the effects in the hippocampus.
Collapse
Affiliation(s)
- Daniela A Espinoza Oyarce
- From the Centre for Research on Ageing, Health and Wellbeing, The Australian National University, Canberra, ACT, Australia (Espinoza Oyarce, Alateeq, Cherbuin); and the College of Engineering and Computer Science, The Australian National University, Canberra, ACT, Australia (Shaw)
| | - Marnie E Shaw
- From the Centre for Research on Ageing, Health and Wellbeing, The Australian National University, Canberra, ACT, Australia (Espinoza Oyarce, Alateeq, Cherbuin); and the College of Engineering and Computer Science, The Australian National University, Canberra, ACT, Australia (Shaw)
| | - Khawlah Alateeq
- From the Centre for Research on Ageing, Health and Wellbeing, The Australian National University, Canberra, ACT, Australia (Espinoza Oyarce, Alateeq, Cherbuin); and the College of Engineering and Computer Science, The Australian National University, Canberra, ACT, Australia (Shaw)
| | - Nicolas Cherbuin
- From the Centre for Research on Ageing, Health and Wellbeing, The Australian National University, Canberra, ACT, Australia (Espinoza Oyarce, Alateeq, Cherbuin); and the College of Engineering and Computer Science, The Australian National University, Canberra, ACT, Australia (Shaw)
| |
Collapse
|
36
|
Marijuana use and major depressive disorder are additively associated with reduced verbal learning and altered cortical thickness. COGNITIVE AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2020; 19:1047-1058. [PMID: 30809764 DOI: 10.3758/s13415-019-00704-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Marijuana (MJ) use and major depressive disorder (MDD) have both been associated with deficits in verbal learning and memory as well as structural brain abnormalities. It is not known if MJ use by those with MDD confers additional impairment. The goal of this study was to examine unique and combined effects of MDD and MJ use on verbal memory and brain structure. Young adults (n=141) aged 18-25 years with MJ use and no lifetime MDD (MJ, n=46), MDD and no MJ use (MDD, n=23), MJ use and lifetime MDD (MDD+MJ, n=24), and healthy controls without MDD or MJ use (CON, n=48) were enrolled. Participants completed the California Verbal Learning Test, Second Edition (CVLT-II), a measure of verbal learning and memory. A sub-sample of 82 participants also underwent a structural magnetic resonance imaging (MRI) scan. Group differences in CVLT-II performance, cortical thickness, and hippocampal volume were assessed. We found an additive effect of MDD and MJ on memory recall. Only MDD, but not MJ, was associated with poorer initial learning, fewer words recalled, more intrusion errors, and lower percent retention. There was also an additive effect of MDD and MJ use on reduced cortical thickness in the middle temporal gyrus. Findings indicate that MJ use and MDD have additive adverse associations with verbal recall and cortical thickness in the middle temporal gyrus, suggesting that MJ use among those with MDD may be contraindicated. Prospective studies are warranted to determine whether this association may be causal.
Collapse
|
37
|
Kang W, Shin JH, Han KM, Kim A, Kang Y, Kang J, Tae WS, Paik JW, Lee HW, Seong JK, Ham BJ. Local shape volume alterations in subcortical structures of suicide attempters with major depressive disorder. Hum Brain Mapp 2020; 41:4925-4934. [PMID: 32804434 PMCID: PMC7643352 DOI: 10.1002/hbm.25168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/14/2020] [Accepted: 08/01/2020] [Indexed: 01/18/2023] Open
Abstract
Suicide is among the most important global health concerns; accordingly, an increasing number of studies have shown the risks for suicide attempt(s) in terms of brain morphometric features and their clinical correlates. However, brain studies addressing suicidal vulnerability have been more focused on demonstrating impairments in cortical structures than in the subcortical structures. Using local shape volumes (LSV) analysis, we investigated subcortical structures with their clinical correlates in depressed patients who attempted suicide. Then we compared them with depressed patients without a suicidal history and age- and sex-matched healthy controls (HCs; i.e., 47 suicide attempters with depression, 47 non-suicide attempters with depression, and 109 HCs). Significant volumetric differences were found between suicidal and nonsuicidal depressed patients in several vertices: 16 in the left amygdala; 201 in the left hippocampus; 1,057 in the left putamen; and 140 in the left pallidum; 1 in the right pallidum; and 6 in the bilateral thalamus. These findings indicated subcortical alterations in LSV in components of the limbic-cortical-striatal-pallidal-thalamic circuits. Moreover, our results demonstrated that the basal ganglia was correlated with perceived stress levels, and the thalamus was correlated with suicidal ideation. We suggest that suicidality in major depressive disorder may involve subcortical volume alterations.
Collapse
Affiliation(s)
- Wooyoung Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jeong-Hyeon Shin
- Medical & Health Device Division, Korea Testing Laboratory, 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
| | - Youbin Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - June Kang
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Woo-Suk Tae
- Brain Convergence Research Center, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Jong-Woo Paik
- Department of Neuropsychiatry, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hae-Woo Lee
- Department of Psychiatry, Seoul Medical Center, Seoul, Republic of Korea
| | - Joon-Kyung Seong
- School of Biomedical Engineering, Department of Artificial Intelligence, Korea University, Seoul, Republic of Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
38
|
Yang C, Chang J, Liang X, Bao X, Wang R. Gray Matter Alterations in Parkinson's Disease With Rapid Eye Movement Sleep Behavior Disorder: A Meta-Analysis of Voxel-Based Morphometry Studies. Front Aging Neurosci 2020; 12:213. [PMID: 32903450 PMCID: PMC7434963 DOI: 10.3389/fnagi.2020.00213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Gray matter (GM) alterations in Parkinson's disease (PD) patients with rapid eye movement sleep behavior disorder (RBD) have been demonstrated in many neuroimaging studies using voxel-based morphometry (VBM). However, the inconsistent findings between studies cannot be applied to clinical practice as a neuroimaging biomarker. We performed a meta-analysis of VBM studies at a whole-brain level to investigate GM differences between PD patients with and without RBD. Methods: A systematic search was conducted in PubMed, Embase, and Web of Science from inception to November 2019 to identify eligible VBM studies. We adopted the latest Seed-based d Mapping with Permutation of Subject Images technique to quantitatively estimate the difference of regional GM volume between PD patients with and without RBD. Results: We included five studies comprising 105 PD patients with RBD and 140 PD patients without RBD. The pooled meta-analysis revealed that PD patients with RBD showed a significant reduction of GM volume in the right superior temporal gyrus (STG) compared with those without RBD. This result was confirmed to be robust by the jackknife sensitivity analysis. Conclusion: Our finding shows significantly and robustly reduced GM volume in the right STG in PD patients with RBD, preliminarily suggesting the association of GM atrophy in this brain region with the occurrence of RBD in PD patients.
Collapse
Affiliation(s)
- Chengxian Yang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianbo Chang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohang Liang
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
39
|
Wu H, Wu C, Wu F, Zhan Q, Peng H, Wang J, Zhao J, Ning Y, Zheng Y, She S. Covariation between Childhood-Trauma Related Resting-State Functional Connectivity and Affective Temperaments is Impaired in Individuals with Major Depressive Disorder. Neuroscience 2020; 453:102-112. [PMID: 32795554 DOI: 10.1016/j.neuroscience.2020.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 06/25/2020] [Accepted: 08/03/2020] [Indexed: 01/23/2023]
Abstract
Affective temperaments and childhood-trauma experiences are associated with major depressive disorder (MDD). So far, how the covariation between distinct affective temperaments and childhood-trauma insulted brain functional connectivities (FCs) contribute to MDD remains unclear. Here, we aimed to investigate whether certain brain FC patterns are related to certain affective temperaments and whether the FCs contribute to depressive symptom dimensions of MDD patients. Twenty-nine medication-free MDD patients and 58 healthy controls underwent magnetic resonance imaging scanning and completed the Hamilton Depression Rating Scale (HDRS), the Hamilton Anxiety Rating Scale (HARS), the Childhood Trauma Questionnaire-Short Form (CTQ-SF), and the Temperament Evaluation of Memphis, Pisa, Paris and San Diego (TEMPS). Two multivariate analyses of partial least squares (PLS) regression were used to explore the associations among childhood-trauma related resting-state FCs, affective temperaments and depressive symptom dimensions. In all participants, a linear combination of 81 FCs (involving parahippocampus, amygdala, cingulate cortex, insula, frontal-temporal-parietal-occipital cortex, pallidum, and cerebellum) were associated with a linear combination of increased depressive, irritable, anxious, and cyclothymic temperaments. Moreover, the covariation between the PLS FC profile and the PLS affective-temperament profile were enhanced in the MDD patients compared to healthy controls. In MDD participants alone, the affective-temperament modulated FC profile (mainly of the lingual and temporal cortex) was associated with the somatization symptom dimension when age, sex, ill-duration, age-of-onset, and HARS scores were adjusted. The findings imply possible neural correlates of affective temperaments and may find applications in intervention of the somatization-depression symptoms by stimulation of the related neural correlates.
Collapse
Affiliation(s)
- Huawang Wu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China
| | - Chao Wu
- School of Nursing, Peking University Health Science Center, Beijing 100091, China
| | - Fengchun Wu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China
| | - Qianqian Zhan
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou 510370, China
| | - Hongjun Peng
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China
| | - Jiaojian Wang
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 625014, China
| | - Jingping Zhao
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou 510370, China; The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yuping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China
| | - Yingjun Zheng
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China.
| | - Shenglin She
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou 510370, China.
| |
Collapse
|
40
|
Liu P, Li G, Zhang A, Yang C, Liu Z, Sun N, Kerang Z. Brain structural and functional alterations in MDD patient with gastrointestinal symptoms: A resting-state MRI study. J Affect Disord 2020; 273:95-105. [PMID: 32421626 DOI: 10.1016/j.jad.2020.03.107] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/30/2020] [Accepted: 03/28/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE It is common for major depressive disorder (MDD) to be accompanied by gastrointestinal (GI) symptoms, which are known to negatively impact the course and severity of the disease. Although previous studies have attempted to explore the neuropathology of MDD, few studies have focused on the pathogenesis of GI symptoms in MDD. In this study, we investigated the changes in regional gray matter volume (GMV) and regional homogeneity (ReHo) present in MDD accompanied by GI symptoms. METHOD The following images were obtained and analyzed: Structural and functional magnetic resonance images (MRI) of 36 patients with MDD accompanied by GI symptoms (GI symptoms group), 22 patients without GI symptoms (Non-GI symptoms group), and 27 healthy controls (HC. The 24-item Hamilton Depression Rating Scale (HAMD) was administered. A correlation analysis was used to identify the possible associations between altered regional GMV, ReHo symptoms, GI symptoms, and depressive symptoms. RESULTS The total scores from the HAMD-24 in the GI symptoms group were significantly higher than in the Non-GI symptoms group (P<0.05). Significant differences in both GMV and ReHo were observed among the three groups for the right parahippocampal gyrus, left precentral gyrus, left middle frontal gyrus, right superior frontal gyrus, right middle frontal gyrus, and left inferior orbitofrontal gyrus (AlphaSim correction, P <0.001). The GI symptoms group exhibited significantly decreased GMV and ReHo in the left middle frontal gyrus, precentral gyrus, right superior frontal gyrus, and middle frontal gyrus. Additionally, the GI symptoms group exhibited increased ReHo in the left superior temporal gyrus at a higher level than the non-GI symptoms group. (AlphaSim correction, P <0.001). These altered brain areas were correlated with GI symptoms (P<0.001) but not depressive symptoms (P>0.05). CONCLUSION Patients with MDD accompanied by GI symptoms have more severe depressive symptoms. The structural and functional changes of the brain may be the pathogenesis for the GI symptoms in patients with MDD.
Collapse
Affiliation(s)
- Penghong Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, PR China, 030001; Shanxi Medical University, Taiyuan, PR China, 030001.
| | - Gaizhi Li
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, PR China, 030001.
| | - Aixia Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, PR China, 030001.
| | - Chunxia Yang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, PR China, 030001.
| | - Zhifen Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, PR China, 030001
| | - Ning Sun
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, PR China, 030001.
| | - Zhang Kerang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, PR China, 030001.
| |
Collapse
|
41
|
Nan D, Yuqi C, Zonglin S, Chenglong D, Na L, Fang L, Cong Z, Xiufeng X. Association of a SIRT1 polymorphism with changes of gray matter volume in patients with first-episode medication-naïve major depression. Psychiatry Res Neuroimaging 2020; 301:111101. [PMID: 32447184 DOI: 10.1016/j.pscychresns.2020.111101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/18/2020] [Accepted: 04/23/2020] [Indexed: 01/08/2023]
Abstract
A single nucleotide polymorphism (SNP) rs12415800 of the silent mating type information regulation 2 homolog 1 gene (SIRT1) has shown a genome-wide significant association with major depression disorder (MDD) in a recent GWAS using a large sample. Subsequent studies of SIRT1's biological functions were supportive of a possible role in the pathophysiology of MDD. However, SIRT1-mediated physiopathology of MDD may be brain region specific. In the present study, we investigated the impact of SIRT1 rs12415800 genotypes on gray matter volumes (GMV) among different brain regions in both MDD patients and healthy controls. The rs12415800 was genotyped in 170 patients with first-episode medication-naïve MDD (cases) and 170 healthy controls. Magnetic resonance imaging was conducted and the voxel-based morphometry (VBM) approach was employed to analyze obtained images. When compared with the cases carrying GG genotype, the cases carrying GA or AA genotypes (A for risk allele) showed decreased GMV in right precuneus, left cuneus/precuneus, and right frontal superior. In contrast, the rs12415800-associated GMV abnormalities were not observed in controls. The SIRT1-rs12415800 polymorphism may be associated with the changes of GMV in MDD patients.
Collapse
Affiliation(s)
- Dai Nan
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang RD, Kunming 650032, Kunming, Yunnan, China
| | - Cheng Yuqi
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang RD, Kunming 650032, Kunming, Yunnan, China
| | - Shen Zonglin
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang RD, Kunming 650032, Kunming, Yunnan, China
| | - Dong Chenglong
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang RD, Kunming 650032, Kunming, Yunnan, China
| | - Li Na
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang RD, Kunming 650032, Kunming, Yunnan, China
| | - Liu Fang
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang RD, Kunming 650032, Kunming, Yunnan, China
| | - Zhou Cong
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang RD, Kunming 650032, Kunming, Yunnan, China
| | - Xu Xiufeng
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang RD, Kunming 650032, Kunming, Yunnan, China.
| |
Collapse
|
42
|
Kim BH, Choi YH, Yang JJ, Kim S, Nho K, Lee JM. Identification of Novel Genes Associated with Cortical Thickness in Alzheimer’s Disease: Systems Biology Approach to Neuroimaging Endophenotype. J Alzheimers Dis 2020; 75:531-545. [DOI: 10.3233/jad-191175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Bo-Hyun Kim
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Yong-Ho Choi
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Jin-Ju Yang
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine and Clinical Neuroscience Center of Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jong-Min Lee
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | | |
Collapse
|
43
|
Setroikromo SN, Bauduin SE, Reesen JE, van der Werff SJ, Smit AS, Vermetten E, van der Wee NJ. Cortical Thickness in Dutch Police Officers: An Examination of Factors Associated with Resilience. J Trauma Stress 2020; 33:181-189. [PMID: 32162369 PMCID: PMC7216895 DOI: 10.1002/jts.22494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 08/18/2019] [Accepted: 08/25/2019] [Indexed: 12/31/2022]
Abstract
Previous neuroimaging studies on resilience have generally compared resilience and psychopathology after stress exposure, which does not allow for conclusions regarding correlates specific to resilience. The aim of the present study was to investigate resilience-specific correlates in cortical thickness and/or cortical surface area and their correlations with psychometric measurements, using a three-group design that included a non-trauma-exposed control group in order to disentangle effects related to resilience from those related to psychopathology. Structural magnetic resonance imaging scans were acquired from 82 Dutch police officers. Participants were categorized into resilient (n = 31; trauma exposure, no psychopathology), vulnerable (n = 32; trauma exposure, psychopathology), and control groups (n = 19; no trauma exposure, no psychopathology). Specific regions of interest (ROIs) were identified based on previous studies that found the rostral and caudal anterior cingulate cortex (ACC) to be implicated in trauma-related psychopathology. Cortical thickness and surface area of the ROIs-the rostral and caudal ACC-and of the whole brain were examined. No significant differences in cortical thickness or surface area were found between the resilient group and other groups in the ROI and whole-brain analyses. Thus, the results of the present study provide no evidence of an association between resilience to traumatic stress and measures of thickness and surface area in cortical regions of the brain in a sample of Dutch police officers.
Collapse
Affiliation(s)
- Santoucha N.W. Setroikromo
- Department of PsychiatryLeiden University Medical CenterLeidenThe Netherlands,Leiden Institute for Brain and CognitionLeidenthe Netherlands
| | - Stephanie E.E.C. Bauduin
- Department of PsychiatryLeiden University Medical CenterLeidenThe Netherlands,Leiden Institute for Brain and CognitionLeidenthe Netherlands
| | - Joyce E. Reesen
- Department of PsychiatryLeiden University Medical CenterLeidenThe Netherlands
| | - Steven J.A. van der Werff
- Department of PsychiatryLeiden University Medical CenterLeidenThe Netherlands,Leiden Institute for Brain and CognitionLeidenthe Netherlands
| | | | - Eric Vermetten
- Department of PsychiatryLeiden University Medical CenterLeidenThe Netherlands
| | - Nic. J.A. van der Wee
- Department of PsychiatryLeiden University Medical CenterLeidenThe Netherlands,Leiden Institute for Brain and CognitionLeidenthe Netherlands
| |
Collapse
|
44
|
Liu S, Zhao W, Li Y, Li X, Li J, Cao H, Yang Z, Xu Y. Improve cognition of depressive patients through the regulation of basal ganglia connectivity: Combined medication using Shuganjieyu capsule. J Psychiatr Res 2020; 123:39-47. [PMID: 32035307 DOI: 10.1016/j.jpsychires.2020.01.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/24/2020] [Accepted: 01/25/2020] [Indexed: 01/25/2023]
Abstract
Shuganjieyu capsule (Shugan) is a combined extract of Hypericum perforatum (HP) and Eleutherococcus senticosus (ES). Both HP and ES have been proven effective in the treatment of depression and impaired cognition. However, for mild to moderate depression (MMD), the treatment effect and underlying mechanism by combining both HP and ES are largely unknown. Here, we aim to evaluate the therapeutic effects on impaired cognition using Shugan, a combined medication of HP and ES. Resting-state magnetic resonance imaging (MRI) data and cognitive assessment have been collected from 54 healthy controls and 55 MMD patients that have been undergoing 8-week Shugan-treatment. The functional connectivity (FC) and brain region volume changes of the basal ganglia seeded circuit have been measured, and their relation with the cognitive assessment score was calculated. After that, a literature-based pathway analysis has been conducted to explore the biological relations among Shugan, brain regions, and depression. Compared to healthy controls, MMD patients demonstrated a significantly higher FC (P= 0.0025) between right ventral caudate (vCa) and left orbitofrontal cortex (OFC), which was decreased after the treatment (P < 0.001). A volume of the right caudate, which is increased in MMD, has also been reduced by Shugan treatment (P= 0.017). Importantly, the cognitive scores were strongly correlated with both Shugan treatment and the FC between vCa and OFC (r= 0.321, P= 0.02). Besides, we identified multiple signaling pathways, through which Shugan might improve the cognition of MMD patients. Our results support the therapeutic effects of Shugan on cognition in MMD, which may be realized partly through the regulation within two brain regions, vCa and OFC.
Collapse
Affiliation(s)
- Sha Liu
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China; Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Wentao Zhao
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China; Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Yaojun Li
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China; Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Xinrong Li
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China; Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China; Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Hongbao Cao
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China; Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China; School of Systems Biology, George Mason University (GMU), Fairfax, VA, USA
| | - Zhi Yang
- Laboratory of Psychological Health and Imaging, Shanghai Mental Health Center, Shanghai Jiao Tong University Medical School, Shanghai, China
| | - Yong Xu
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China; Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
45
|
Su J, Pan YW, Wang SQ, Li XZ, Huang F, Ma SP. Saikosaponin-d attenuated lipopolysaccharide-induced depressive-like behaviors via inhibiting microglia activation and neuroinflammation. Int Immunopharmacol 2020; 80:106181. [PMID: 31926446 DOI: 10.1016/j.intimp.2019.106181] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/20/2019] [Accepted: 12/31/2019] [Indexed: 12/15/2022]
Abstract
Saikosaponin-d (SSd), a triterpenoid saponins compound extracted from Radix Bupleuri, has been demonstrated to effectively alleviate chronic mild stress-induced depressive behaviors in rats, but the underlying molecular mechanisms are still uncertain. Increasing evidence indicated that microglia activation and inflammatory responses were involved in the pathogenesis of depression. Thus, we desired to induce inflammation-related depressive-like behaviors in mice by injecting lipopolysaccharide (LPS) to investigate whether the antidepressant effect of SSd is related to inhibiting inflammation. The results of behavioral tests showed that SSd administration ameliorated LPS-induced depressive-like behaviors, as shown by increased sucrose consumption in the sucrose preference test and decreased immobility time in the tail suspension test and forced swimming test. Furthermore, immunostaining results showed that SSd pretreatment inhibited LPS-induced microglia activation in the hippocampus of mice and primary microglia cells. Enzyme-linked immunosorbent assay (ELISA) results showed that SSd pretreatment suppressed LPS-induced overexpression of inflammatory factors such as interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α both in vivo and in vitro. Immunostaining and western blot analysis results demonstrated that SSd pretreatment also inhibited LPS-induced HMGB1 translocation from nuclear to extracellular and decreased the protein levels of TLR4, p-IκB-α, NF-κBp65. These results suggested that SSd effectively improved LPS-induced inflammation-related depressive-like behaviors by inhibiting LPS-induced microglia activation and neuroinflammation, and the possible mechanism might associate with the regulation of the HMGB1/TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jing Su
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yi-Wei Pan
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - Si-Qi Wang
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xue-Zhen Li
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China
| | - Fang Huang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Shi-Ping Ma
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, PR China; Qinba Traditional Chinese Medicine Resources Research and Development Center, AnKang University, AnKang 725000, PR China.
| |
Collapse
|
46
|
Shen J, Li DL, Tan XX, Tao WW, Xie CJ, Shi XG, Wang Y. A transcranial sonography study of brainstem and its association with depression in idiopathic generalized epilepsy with tonic-clonic seizures. Epilepsy Behav 2020; 102:106589. [PMID: 31726317 DOI: 10.1016/j.yebeh.2019.106589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/19/2019] [Accepted: 09/22/2019] [Indexed: 01/18/2023]
Abstract
Brainstem raphe (BR) hypoechogenicity in transcranial sonography (TCS) has been depicted in patients with depression. But, up to date, the association of BR alterations in TCS with depression in patients with epilepsy has never been reported. This study was to investigate the possible role of BR examination via TCS in patients with idiopathic generalized epilepsy with tonic-clonic seizures (IGE-TCS) and depression. Forty-six patients with IGE-TCS and 45 healthy controls were recruited. Echogenicity of the caudate nuclei (CN), lentiform nuclei (LN), substantia nigra (SN), and BR and widths of the lateral ventricle (LV) frontal horns and the third ventricle (TV) were assessed via TCS. The determination of depression was based on the criteria of the Diagnostic and Statistical Manual of Mental Disorders IV (DSM-IV), and depression severity measured by Chinese version Neurological Disorders Depression Inventory for Epilepsy (C-NDDI-E) and Beck Depression Inventory-II (BDI-II). The width of TV in patients with epilepsy was found significantly larger than that in healthy controls (p = 0.001), but there was no significant difference in TV width between patients with IGE-TCS with and without depression. There were no significant differences between patients with IGE-TCS and healthy controls in LV frontal horn width, as well as in SN, CN, LN, and BR echogenicity. Here, it seems that patients with IGE-TCS were detected with smaller SN echogenic area compared with controls though they had no statistical significance. Patients with IGE-TCS with hypoechogenic BR had significantly higher C-NDDI-E and BDI-II scores than those with normal BR signal, and most patients with IGE-TCS with depression exhibited hypoechogenic BR, but few patients with IGE-TCS without depression exhibited hypoechogenic BR. In conclusion, BR echogenic signal alterations in TCS can be a biomarker for depression in epilepsy, but it might not be associated with epilepsy itself. The alterations of SN echogenic area and TV width in TCS may reflect a potential role of SN and diencephalon structure in the pathogenesis of epilepsy, which needs to be further elucidated.
Collapse
Affiliation(s)
- Jie Shen
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Dong-Lin Li
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Xiu-Xiu Tan
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Wei-Wei Tao
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Cheng-Juan Xie
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Xue-Gong Shi
- Department of Echocardiography, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Yu Wang
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China; Department of Neurology, the Fourth Affiliated Hospital of Anhui Medical University, Huaihai Avenue 100, Hefei 230000, China.
| |
Collapse
|
47
|
Hung CI, Liu CY, Yang CH, Gan ST. Comorbidity with more anxiety disorders associated with a poorer prognosis persisting at the 10-year follow-up among patients with major depressive disorder. J Affect Disord 2020; 260:97-104. [PMID: 31493646 DOI: 10.1016/j.jad.2019.08.085] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 06/30/2019] [Accepted: 08/28/2019] [Indexed: 11/29/2022]
Abstract
OBJECTIVE No study has investigated the association between number of anxiety disorders (NADs) and long-term outcome over 10 years among patients with major depressive disorder (MDD). This study investigated this issue. METHODS At baseline, 290 outpatients with MDD were enrolled, 149 with at least one anxiety disorder (AD). Subjects were followed-up at six-month, two-year, and 10-year points. The Structured Clinical Interview for DSM-IV-TR was used to confirm psychiatric diagnoses. NADs at baseline was recorded. The Hamilton Depression Rating Scale (HAMD), the anxiety subscale of the Hospital Anxiety and Depression Scale (HADS-A), and the somatic subscale (SS) of the Depression and Somatic Symptoms Scale were employed. Generalized Estimating Equation models were used for statistical analysis. RESULTS MDD patients with ADs had greater depression, anxiety, and somatic severities at the three follow-up points than those without. NADs was significantly and positively correlated with the three dimensions and total duration of pharmacotherapy at follow-up. NADs was independently associated with symptom severity after controlling for depression and anxiety at baseline and pharmacotherapy. When the DSM-5 criteria for ADs were applied, the results were unchanged. Specific phobia, panic disorder and social phobia, and panic disorder and specific phobia were independently associated with depression, anxiety, and somatic symptoms, respectively. LIMITATION Pharmacotherapy at follow-up was not controlled. The three follow-up intervals were unequal. CONCLUSIONS Comorbidity with more ADs was associated with a poorer prognosis. The negative impacts of ADs on MDD persisted at the ten-year follow-up point. NADs was associated with the long-term prognosis of MDD.
Collapse
Affiliation(s)
- Ching-I Hung
- Department of Psychiatry, Chang-Gung Memorial Hospital at Linkou and Chang-Gung University College of Medicine, 5 Fu-Shing St, Kweishan,Tao-Yuan 333 Taiwan.
| | - Chia-Yih Liu
- Department of Psychiatry, Chang-Gung Memorial Hospital at Linkou and Chang-Gung University College of Medicine, 5 Fu-Shing St, Kweishan,Tao-Yuan 333 Taiwan
| | - Ching-Hui Yang
- Department of Nursing, Mackay Medical College, New Taipei City, Taiwan
| | - Shu-Ting Gan
- Center for Big Data Analytics and Statistics, Chang-Gung Memorial Hospital at Linkou, Tao-Yuan, Taiwan
| |
Collapse
|
48
|
Heij GJ, Penninx BWHJ, van Velzen LS, van Tol MJ, van der Wee NJA, Veltman DJ, Aghajani M. White matter architecture in major depression with anxious distress symptoms. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109664. [PMID: 31158389 DOI: 10.1016/j.pnpbp.2019.109664] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/29/2019] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Comorbid anxious distress is common in Major Depressive Disorder (MDD), and associated with significantly worse clinical course and treatment response. While DSM-5 recently introduced the Anxious Distress (AD) specifier as a potentially useful symptom-based subtyping scheme for MDD, its neurobiological underpinnings remain unclear. The current study hence uniquely probed whether MDD with co-occurring AD (MDD/AD+) relates to distinct perturbations in frontolimbic white matter (WM) pathways tentatively theorized in MDD/AD+ pathophysiology. METHODS Tract-based spatial statistics (TBSS) was therefore used to analyze diffusion tensor imaging data on WM microstructure, in MDD/AD+ patients (N = 20) relative to MDD patients without AD (MDD/AD-; N = 29) and healthy controls (HC; N = 39). Using TBSS, we probed fractional anisotropy and axial/radial/mean diffusivity as proxies for WM integrity. Categorical (between-groups) and dimensional (within-patients) analyses subsequently assessed how Anxious Distress in MDD impacts frontolimbic WM connectivity. Receiver-Operating Characteristics additionally assessed classification capabilities of between-groups WM effects. RESULTS Compared to MDD/AD- and HC participants, MDD/AD+ patients exhibited diminished integrity within the anterior thalamic radiation (ATR). Higher AD specifier scores within MDD patients additionally related to diminished integrity of the uncinate fasciculus and cingulum pathways. These effects were not confounded by key clinical (e.g., comorbid anxiety disorder) and sociodemographic (e.g., age/sex) factors, with altered ATR integrity moreover successfully classifying MDD/AD+ patients from MDD/AD- and HC participants (90% sensitivity | 73% specificity | 77% accuracy). CONCLUSIONS These findings collectively link MDD/AD+ to distinct WM anomalies in frontolimbic tracts important to adaptive emotional functioning, and may as such provide relevant, yet preliminary, clues on MDD/AD+ pathophysiology.
Collapse
Affiliation(s)
- Gijs J Heij
- VU University, Faculty of Earth and Life Sciences, the Netherlands; Amsterdam UMC, Location VUMC, Dept. of Psychiatry & Amsterdam Neuroscience, the Netherlands
| | - Brenda W H J Penninx
- Amsterdam UMC, Location VUMC, Dept. of Psychiatry & Amsterdam Neuroscience, the Netherlands; GGZ InGeest Specialized Mental Health Care, the Netherlands
| | - Laura S van Velzen
- Amsterdam UMC, Location VUMC, Dept. of Psychiatry & Amsterdam Neuroscience, the Netherlands; GGZ InGeest Specialized Mental Health Care, the Netherlands
| | - Marie-José van Tol
- University Medical Center Groningen, Dept. of Psychiatry, the Netherlands
| | | | - Dick J Veltman
- Amsterdam UMC, Location VUMC, Dept. of Psychiatry & Amsterdam Neuroscience, the Netherlands; GGZ InGeest Specialized Mental Health Care, the Netherlands
| | - Moji Aghajani
- Amsterdam UMC, Location VUMC, Dept. of Psychiatry & Amsterdam Neuroscience, the Netherlands; GGZ InGeest Specialized Mental Health Care, the Netherlands.
| |
Collapse
|
49
|
Duman JG, Mulherkar S, Tu YK, Erikson KC, Tzeng CP, Mavratsas VC, Ho TSY, Tolias KF. The adhesion-GPCR BAI1 shapes dendritic arbors via Bcr-mediated RhoA activation causing late growth arrest. eLife 2019; 8:47566. [PMID: 31461398 PMCID: PMC6713510 DOI: 10.7554/elife.47566] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/03/2019] [Indexed: 12/17/2022] Open
Abstract
Dendritic arbor architecture profoundly impacts neuronal connectivity and function, and aberrant dendritic morphology characterizes neuropsychiatric disorders. Here, we identify the adhesion-GPCR BAI1 as an important regulator of dendritic arborization. BAI1 loss from mouse or rat hippocampal neurons causes dendritic hypertrophy, whereas BAI1 overexpression precipitates dendrite retraction. These defects specifically manifest as dendrites transition from growth to stability. BAI1-mediated growth arrest is independent of its Rac1-dependent synaptogenic function. Instead, BAI1 couples to the small GTPase RhoA, driving late RhoA activation in dendrites coincident with growth arrest. BAI1 loss lowers RhoA activation and uncouples it from dendrite dynamics, causing overgrowth. None of BAI1's known downstream effectors mediates BAI1-dependent growth arrest. Rather, BAI1 associates with the Rho-GTPase regulatory protein Bcr late in development and stimulates its cryptic RhoA-GEF activity, which functions together with its Rac1-GAP activity to terminate arborization. Our results reveal a late-acting signaling pathway mediating a key transition in dendrite development.
Collapse
Affiliation(s)
- Joseph G Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Yen-Kuei Tu
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, United States
| | - Kelly C Erikson
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Christopher P Tzeng
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Vasilis C Mavratsas
- Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Rice University, Houston, United States
| | - Tammy Szu-Yu Ho
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, United States.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
| |
Collapse
|
50
|
Häberling I, Baumgartner N, Emery S, Keller P, Strumberger M, Nalani K, Schmeck K, Erb S, Bachmann S, Wöckel L, Müller-Knapp U, Contin-Waldvogel B, Rhiner B, Walitza S, Berger G. Anxious depression as a clinically relevant subtype of pediatric major depressive disorder. J Neural Transm (Vienna) 2019; 126:1217-1230. [DOI: 10.1007/s00702-019-02069-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022]
|