1
|
Luo B, Huo J, Zhao L, Guo X, Zhang L, Yang Y, Li S, Zhong J, Lv L, Li M, Guo Y, Xiao X, Li W. The complex association of VRK2 with major depressive disorder in Han Chinese population. J Affect Disord 2025; 383:260-266. [PMID: 40294824 DOI: 10.1016/j.jad.2025.04.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is a polygenic condition with substantial heritability, with genome-wide association studies (GWAS) identifying several risk loci in European populations, including the VRK2 gene. However, the association between VRK2 and MDD in non-European populations, particularly in Han Chinese, remains underexplored. METHODS We genotyped four VRK2 SNPs (rs2678907, rs11682175, rs1568452, rs1518395) in a cohort of 1878 MDD cases and 1800 controls of Han Chinese descent. Genotyping was performed using SNaPShot, and linkage disequilibrium (LD) was assessed with SHEsis. Associations between the SNPs and MDD were evaluated via logistic regression in PLINK. VRK2 mRNA expression in the amygdala and peripheral blood was quantified by RT-qPCR, with statistical significance determined by ANCOVA and t-tests. A meta-analysis incorporating an independent East Asian GWAS cohort was also conducted. RESULTS In our Han Chinese cohort, rs2678907 was significantly associated with MDD (P = 4.17 × 10-5, OR = 1.217). Meta-analysis with independent East Asian GWAS further confirmed the associations of rs2678907 with MDD. Haplotype analysis of VRK2 SNPs in Han Chinese revealed the haplotypes (T-G for rs11682175-rs2678907 and C-G for rs1568452-rs2678907) associated with an increased MDD risk and elevated VRK2 mRNA expression. Additionally, MDD patients showed significantly higher VRK2 mRNA levels in peripheral blood than controls (P = 1.85 × 10-7). CONCLUSIONS These findings provide strong evidence for the role of VRK2 in MDD risk in Han Chinese individuals. Our results underscore the potential of VRK2 as a genetic and expression-based biomarker for MDD, highlighting the importance of accounting for population-specific genetic variations in psychiatric research. Further research is essential to explore the functional implications of VRK2 in MDD pathogenesis.
Collapse
Affiliation(s)
- Binbin Luo
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, China; Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Jinhua Huo
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Lijuan Zhao
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Xiaoge Guo
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, China; Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Luwen Zhang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, China; Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Yongfeng Yang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, China; Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Shiwu Li
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Jingmei Zhong
- Department of Psychiatry, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650201, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, China; Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Ming Li
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Yongbo Guo
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| | - Xiao Xiao
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, China; Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, Xinxiang Medical University, Xinxiang, Henan 453003, China.
| |
Collapse
|
2
|
Hashimoto N, Okada N, Fukunaga M, Nemoto K, Miura K, Matsumoto J, Ishikawa S, Narita H, Morita K, Yasuda Y, Kamishikiryo T, Harada K, Yamamoto M, Ohi K, Matsubara T, Hirano Y, Okada G, Tha KK, Abe O, Onitsuka T, Kawasaki Y, Ozaki N, Kasai K, Hashimoto R. Lithium and valproate affect subcortical brain volumes in individuals with bipolar disorder: Mega-analysis of 235 individuals. J Affect Disord 2025; 381:115-120. [PMID: 40189070 DOI: 10.1016/j.jad.2025.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/01/2024] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION The mega-analysis conducted by the ENIGMA Bipolar Disorder Working Group revealed significant volume increment effects of lithium on the hippocampus in individual with bipolar disorder. However, the study did not assess other medications and other subcortical regions. METHODS Data of 235 individuals with bipolar disorder were taken from a mega-analysis conducted by the COCORO consortium in Japan. The effects of psychotropic prescriptions (lithium, valproate, antipsychotics, antidepressants, benzodiazepines) were assessed using a linear mixed-effects model with volumes of subcortical structures as dependent variables, and age, sex, intracranial volume, duration of illness, and psychotropic prescriptions as independent variables; the type of protocol was incorporated as a random effect. RESULTS Prescriptions of lithium was associated with larger left amygdala volume (Effect size (ES, Cohen's d) = 0.36, p = 0.001). Prescriptions of valproate was associated with smaller left amygdala volume (ES = -0.45, p = 0.001), and larger bilateral ventricle volumes (ES = 0.68, p < 0.001 (left), ES = 0.70, p < 0.001 (right)). Prescriptions of antipsychotics were associated with larger left globus pallidus volume (ES = 0.33, p = 0.014) and smaller left hippocampus volume (ES = -0.33, p = 0.024). Prescriptions of benzodiazepines were associated with smaller left lateral ventricle (ES = -0.40, p = 0.029). Prescriptions of antidepressants were associated with smaller right accumbens volume (ES = -0.22, p = 0.043), bilateral caudate volumes (ES = -0.38, p = 0.013 (left), ES = -0.25, p = 0.050 (right)) and right putamen volume (ES = -0.23, p = 0.024). CONCLUSION We confirmed the association between prescription of valproate and smaller amygdala and larger lateral ventricle volumes in a large sample for the first time. Large sample size, uniform data collection methodology, and robust statistical analysis are strengths of the current study.
Collapse
Affiliation(s)
- Naoki Hashimoto
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Hokkaido, Japan.
| | - Naohiro Okada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; The International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, Japan
| | - Masaki Fukunaga
- Division of Cerebral Integration, National Institute for Physiological Sciences, Aichi, Japan
| | - Kiyotaka Nemoto
- Department of Psychiatry, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kenichiro Miura
- Division of Cerebral Integration, National Institute for Physiological Sciences, Aichi, Japan; Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Junya Matsumoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Shuhei Ishikawa
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Hisashi Narita
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Kentaro Morita
- Department of Rehabilitation, University of Tokyo Hospital, Tokyo, Japan
| | - Yuka Yasuda
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan; Life Grow Brilliant Mental Clinic, Medical Corporation Foster, Osaka, Japan
| | | | - Kenichiro Harada
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Maeri Yamamoto
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, Aichi, Japan
| | - Kazutaka Ohi
- Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan; Department of General Internal Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Toshio Matsubara
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yoji Hirano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Psychiatry, Division of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Go Okada
- Department of Psychiatry and Neuroscience, Hiroshima University, Hiroshima, Japan
| | - Khin K Tha
- Department of Diagnostic Imaging, Hokkaido University Faculty of Medicine, Hokkaido, Japan; Global Center for Biomedical Science and Engineering, Hokkaido University Faculty of Medicine, Hokkaido, Japan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Yasuhiro Kawasaki
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| | - Norio Ozaki
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, Aichi, Japan; Pathophysiology of Mental Disorders, Graduate School of Medicine, Nagoya University, Aichi, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; The International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, Japan; University of Tokyo Institute for Diversity & Adaptation of Human Mind (UTIDAHM), Tokyo, Japan
| | - Ryota Hashimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
3
|
Tonini E, Crouse JJ, Shin M, Scott J, Carpenter JS, Nichles A, Zmicerevska N, Iorfino F, Capon W, Wood SJ, Purcell R, Yung AR, Pantelis C, Nelson B, McGorry PD, Hickie IB. Activation differentiates illness trajectories among youth seeking mental health care. J Affect Disord 2025; 379:680-689. [PMID: 40090388 DOI: 10.1016/j.jad.2025.03.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND The clinical profiles of youth presenting to early intervention mental health services are heterogeneous, with various sub-groups proposed and little information about the longitudinal stability of profiles, especially those associated with bipolarity. METHODS 802 youth aged 12-25-years (Mean = 18.26; 66 % females) accessing primary-care based mental health clinics were assessed at baseline and 417 were re-assessed after 12-months. An exploratory factor analysis of 62 items from six validated rating scales of the severity of mental and physical ill-health was conducted. Seven factors (anxiety, sleep, depression, restlessness, distress, activation, somatic complaints) were derived and modelled using latent profile analysis. Associations between profile membership, clinical outcomes and functioning were examined. Conditional probabilities of shifting to a different profile longitudinally were computed. RESULTS Three profiles were revealed which were psychometrically invariant across baseline and follow-up: (1) 'High distress with high activation' (32 % baseline, 25 % follow-up); (2) 'High distress without activation' (31 % baseline, 26 % follow-up); and (3) 'Moderate distress' (37 % baseline, 33 % follow-up). A fourth profile, 'Low distress' (16 %), emerged at follow-up. Profiles did not differ by age at baseline or sex. 'High distress with high activation' was more likely to be impaired longitudinally, and to meet criteria for a full-threshold mental disorder at follow-up. About 39 % of youth retained the same profile longitudinally, while 16 % shifted to lower distress, and 13 % shifted to higher distress. CONCLUSION These findings suggest that activation is a marker of poorer clinical and functional outcomes in youth presenting for mental health care.
Collapse
Affiliation(s)
- Emiliana Tonini
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
| | - Jacob J Crouse
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Mirim Shin
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Jan Scott
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia; Institute of Neuroscience, University of Newcastle, Newcastle, UK
| | | | - Alissa Nichles
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | | | - Frank Iorfino
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - William Capon
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Stephen J Wood
- Orygen, Parkville, VIC, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia; School of Psychology, University of Birmingham, Edgbaston, UK
| | - Rosemary Purcell
- Orygen, Parkville, VIC, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Alison R Yung
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia; School of Health Sciences, University of Manchester, UK
| | - Christos Pantelis
- Department of Psychiatry, University of Melbourne, Western Hospital, Sunshine, St Albans, Vic, Australia; Monash Institute of Pharmaceutical Sciences (MIPS), Monash University, Parkville, Vic, Australia; Florey Institute of Neurosciences and Mental Health, Parkville, Vic, Australia
| | - Barnaby Nelson
- Orygen, Parkville, VIC, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Patrick D McGorry
- Orygen, Parkville, VIC, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Ian B Hickie
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
4
|
Gao Y, Wang S, Li T, Guo X, Lu Z, Luo R, Yang B, Cao J. Modulation of cerebellar homotopic connectivity by modified electroconvulsive therapy at rest: Study of first-episode, drug-naive adolescent major depressive disorder. J Affect Disord 2025; 379:615-623. [PMID: 40044090 DOI: 10.1016/j.jad.2025.03.005] [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: 08/27/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/21/2025]
Abstract
Previous studies of brain function alterations that are associated with modified electroconvulsive therapy (MECT) for major depressive disorder (MDD) have yielded conflicting results because of variations in treatment durations, types of antidepressants, and disease course. Consequently, predicting the efficacy of MECT remains challenging. Thirty patients with first-episode, drug-naive adolescent MDD and 34 healthy controls (HCs) underwent resting-state functional magnetic resonance imaging and neuropsychological tests. The patients received MECT and underwent scanning at two time points (baseline and posttreatment). Voxel-mirrored homotopic connectivity (VMHC), support vector machine (SVM), and support vector regression (SVR) analyses were employed to analyze the imaging data. Compared with HCs, patients at baseline exhibited greater VMHC in the cerebellum_2 and cerebellum_8. Following treatment, patients exhibited the restoration of normal VMHC values. Additionally, SVM and receiver operating characteristic curve analyses revealed that VMHC values in the cerebellum_2 could differentiate MDD patients from HCs with 73.85 % accuracy, 70.00% sensitivity, 79.41 % specificity, and an area under the curve of 0.7486. Furthermore, the SVR results indicated a significant association between predicted and actual symptomatic improvement based on the reduction ratio of Hamilton Depression Rating Scale-17 total scores (R2 = 0.5269, P < 0.0001). This study provides evidence that MECT modulates homotopic connectivity of the cerebellum_2 in first-episode, drug-naive adolescent MDD. Moreover, VMHC values in the cerebellum_2 may serve as a valuable neuroimaging biomarker for distinguishing MDD from HCs and potentially predicting early treatment response in MDD.
Collapse
Affiliation(s)
- Yujun Gao
- Department of Psychiatry, Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, China.
| | - Sanwang Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430000, China; Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Tingting Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Xin Guo
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430000, China
| | - Zhaoyuan Lu
- Department of Psychiatry, Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Ruqin Luo
- Department of Psychiatry, Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Bin Yang
- Department of Psychiatry, Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Jingchen Cao
- Department of Psychiatry, Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Laroy M, Emsell L, Vandenbulcke M, Bouckaert F. Mapping electroconvulsive therapy induced neuroplasticity: Towards a multilevel understanding of the available clinical literature - A scoping review. Neurosci Biobehav Rev 2025; 173:106143. [PMID: 40222573 DOI: 10.1016/j.neubiorev.2025.106143] [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: 01/06/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
Since its introduction in 1938, the precise mechanism underlying the efficacy of electroconvulsive therapy (ECT) in treating severe psychiatric disorders remains elusive. This paper presents a comprehensive scoping review aimed to collate and summarize findings from clinical studies on neuroplastic changes induced by ECT. The review categorizes neuroplasticity into molecular, structural, and functional domains, offering a multilevel view of current research and its limitations. Molecular findings detail the varied responses of neurotrophic factors and neurotransmitters post-ECT, highlighting inconsistent evidence on their clinical relevance. Structural neuroplasticity is explored through changes in brain volume, cortical thickness, and white matter properties, presenting ECT as a potent stimulator of brain architecture alterations. Functional plasticity examines ECT's impact on brain function through diverse neuroimaging techniques, suggesting significant yet complex modifications in brain network connectivity and activity. The review emphasizes the multilevel nature of these neuroplasticity levels and their collective role in ECT's therapeutic outcomes. Methodological considerations-including sample size, patient heterogeneity, and variability in assessment timing-emerge as recurring themes in the literature, underscoring the need for more consistent and rigorous research designs. By outlining a cohesive framework of changes in neuroplasticity due to ECT, this review provides initial steps towards a deeper comprehension of ECT's mechanisms.
Collapse
Affiliation(s)
- Maarten Laroy
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven B-3000, Belgium; Psychiatric Neuromodulation Centre, University Psychiatric Center KU Leuven, Leuven B-3000, Belgium.
| | - Louise Emsell
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven B-3000, Belgium; Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven B-3000, Belgium; KU Leuven, Leuven Brain Institute, Department of Imaging and Pathology, Translational MRI, Leuven B-3000, Belgium
| | - Mathieu Vandenbulcke
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven B-3000, Belgium; Psychiatric Neuromodulation Centre, University Psychiatric Center KU Leuven, Leuven B-3000, Belgium; Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven B-3000, Belgium
| | - Filip Bouckaert
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven B-3000, Belgium; Psychiatric Neuromodulation Centre, University Psychiatric Center KU Leuven, Leuven B-3000, Belgium; Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven B-3000, Belgium
| |
Collapse
|
6
|
Kraus A, Dohm K, Borgers T, Goltermann J, Grotegerd D, Winter A, Thiel K, Flinkenflügel K, Schürmeyer N, Hahn T, Langer S, Kircher T, Nenadić I, Straube B, Jamalabadi H, Alexander N, Jansen A, Stein F, Brosch K, Usemann P, Teutenberg L, Thomas-Odenthal F, Meinert S, Dannlowski U. Brain structural correlates of an impending initial major depressive episode. Neuropsychopharmacology 2025; 50:1176-1185. [PMID: 40074869 PMCID: PMC12089404 DOI: 10.1038/s41386-025-02075-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/20/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Neuroimaging research has yet to elucidate whether reported gray matter volume (GMV) alterations in major depressive disorder (MDD) exist already before the onset of the first episode. Recruitment of presently healthy individuals with a subsequent transition to MDD (converters) is extremely challenging but crucial to gain insights into neurobiological vulnerability. Hence, we compared converters to patients with MDD and sustained healthy controls (HC) to distinguish pre-existing neurobiological markers from those emerging later in the course of depression. Combining two clinical cohorts (n = 1709), voxel-based morphometry was utilized to analyze GMV of n = 45 converters, n = 748 patients with MDD, and n = 916 HC in a region-of-interest approach and exploratory whole-brain. By contrasting the subgroups and considering both remission state and reported recurrence at a 2-year clinical follow-up, we stepwise disentangled effects of (1) vulnerability, (2) the acute depressive state, and (3) an initial vs. a recurrent episode. Analyses revealed higher amygdala GMV in converters relative to HC (ptfce-FWE = 0.037, d = 0.447) and patients (ptfce-FWE = 0.005, d = 0.508), remaining significant when compared to remitted patients with imminent recurrence. Lower GMV in the dorsolateral prefrontal cortex (ptfce-FWE < 0.001, d = 0.188) and insula (ptfce-FWE = 0.010, d = 0.186) emerged in patients relative to HC but not to converters, driven by patients with acute MDD. By examining one of the largest available converter samples in psychiatric neuroimaging, this study allowed a first determination of neural markers for an impending initial depressive episode. Our findings suggest a temporary vulnerability, which in combination with other common risk factors might facilitate prediction and in turn improve prevention of depression.
Collapse
Grants
- This work is part of the German multicenter consortium “Neurobiology of Affective Disorders. A translational perspective on brain structure and function“, funded by the consortia grants from the German Research Foundation (Deutsche Forschungsgemeinschaft DFG; Forschungsgruppe/Research Unit FOR2107) FOR 2107 and SFB/TRR 393 (grant FOR2107 KI588/14-1, KI588/14-2, KI588/15-1, KI588/17-1, KI588/20-1, KI588/22-1 to TK, DA1151/5-1, DA1151/5-2, DA1151/6-1, DA1151/9-1, DA1151/10-1, DA1151/11-1 to UD, STR1146/18-1 to BS, NE2254/1-2, NE2254/2-1, NE2254/3-1, NE2254/4-1 to IN, JA1890/7-1, JA1890/7-2 to AJ, HA7070/2-2 to TH; grant SFB-TRR393, Projects A01 and S03 to TH, A02 and Z to TK, A02 and S02 to UD; A04 to SM, A04 and C02 to IN, B01 and INF to AJ, B03 and RTG to BS, B03 and S03 to HJ, B05 and S02 to NA, INF to FS), the Interdisciplinary Center for Clinical Research (IZKF) of the medical faculty of Münster (grant Dan3/022/22 to UD) and the “Innovative Medizinische Forschung“ (IMF) of the medical faculty Münster (grant ME122205 to SM; grant KO-121806 to KD).
Collapse
Affiliation(s)
- Anna Kraus
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Dohm
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tiana Borgers
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Alexandra Winter
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Thiel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Kira Flinkenflügel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Navid Schürmeyer
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Simon Langer
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Benjamin Straube
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Hamidreza Jamalabadi
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Nina Alexander
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
- Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Paula Usemann
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Lea Teutenberg
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Florian Thomas-Odenthal
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany.
- Institute for Translational Neuroscience, University of Münster, Münster, Germany.
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| |
Collapse
|
7
|
Parker AJ, Sorcher LK, Cutshaw OP, Botdorf M, Dunstan J, Riggins T, Dougherty LR. Hippocampal subregion volumes and preadolescent depression risk in the ABCD sample. J Affect Disord 2025; 378:165-174. [PMID: 40023259 DOI: 10.1016/j.jad.2025.02.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/19/2024] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
The hippocampus is central in the pathophysiology of depression. Subregions of the hippocampus (head, body, tail) have been implicated in adult depression, though research examining depression and hippocampal subregions in youth has been limited. This study aimed to examine associations between preadolescent hippocampal subregions and depression risk as well as their interactions with factors associated with depression risk, including biological sex and socioeconomic status (SES). Hippocampal subregions were extracted from the Adolescent Brain and Cognitive Development Study baseline sample (N = 10,469, ages 9-10 years). Depression risk factors included maternal lifetime depression, child depressive symptoms, and child internalizing and externalizing symptoms. Maternal depression was measured through the Family History Questionnaire, and child symptoms were measured through the Child Behavioral Checklist. Results identified associations between hippocampal volumes and future increases in internalizing symptoms (N = 9738). Further, associations between hippocampal subregions and depression risk were moderated by biological sex and SES: males, but not females, with maternal depression exhibited lower hippocampal tail volumes (N = 9826), and for preadolescents with low, but not high, SES, greater hippocampal head volumes predicted increased internalizing symptoms at baseline (N = 10,294) and at the 24-month follow up (N = 7069-7086). Together, this study demonstrates the importance of hippocampal subregions within preadolescent depression risk and identifies subgroups, including preadolescent males and those with low SES, that may be at particular risk.
Collapse
Affiliation(s)
- Alyssa J Parker
- University of Maryland, College Park, Department of Psychology, United States of America.
| | - Leah K Sorcher
- University of Maryland, College Park, Department of Psychology, United States of America
| | - Olivia P Cutshaw
- University of Maryland, College Park, Department of Psychology, United States of America
| | - Morgan Botdorf
- University of Maryland, College Park, Department of Psychology, United States of America; Children's Hospital of Philadelphia, United States of America
| | - Jade Dunstan
- University of Maryland, College Park, Department of Psychology, United States of America
| | - Tracy Riggins
- University of Maryland, College Park, Department of Psychology, United States of America
| | - Lea R Dougherty
- University of Maryland, College Park, Department of Psychology, United States of America
| |
Collapse
|
8
|
Rabl U, Bartova L, Sezen P, Keller J, Schatzberg A, Pezawas L. HPA axis in psychotic and non-psychotic major depression: Cortisol plasma levels and hippocampal volume. J Affect Disord 2025; 377:14-22. [PMID: 39955073 DOI: 10.1016/j.jad.2025.02.014] [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: 04/04/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Psychotic major depression (PMD) differs from non-psychotic MD (NPMD) in psychopathology and is linked to changes in brain volumetry and hypothalamic-pituitary-adrenal (HPA) axis function that can be reflected by its principal output - the glucocorticoid cortisol. NPMD patients exhibit smaller hippocampi than healthy controls (HC), purportedly representing exposure to chronic stress. However, the relationship between the individual clinical phenotype, hippocampal volume and diurnal cortisol signaling remains unclear. METHODS Since understanding the interplay among symptoms, neuroimaging and HPA function is crucial for discerning biological differences between PMD and NPMD, this study explored the link between clinical phenotype, hippocampal structural MRI and circadian plasma cortisol levels in 32 HC, 27 NPMD and 26 PMD patients. RESULTS PMD patients showed significantly elevated evening (6 p.m. - 1 a.m.) cortisol levels compared to NPMD and HC, while NPMD and HC did not differ. No group differences in hippocampal volume were observed, but a significant interaction effect emerged between overnight (1 a.m. - 9 a.m.) cortisol levels, hippocampal volume, and clinical phenotype. NPMD patients displayed a negative correlation between overnight cortisol levels and hippocampal volume, which was specific to the ascending cortisol curve (2 a.m. - 5 a.m.) and absent in PMD and HC. The hippocampus-cortisol interaction was associated with depressive symptom severity in NPMD but not PMD, where cortisol alone predicted greater severity. CONCLUSIONS These findings imply a time-dependent relationship between hippocampal volume and overnight cortisol in NPMD, which is absent in PMD and HC. In contrast, PMD patients exhibited increased evening cortisol levels. In an exploratory analysis, these effects were also related to symptom severity at similar timepoints. While correlational, these results point to distinct neurobiological mechanisms underlying NPMD and PMD, which are potentially related to the heterogeneous clinical manifestations.
Collapse
Affiliation(s)
- U Rabl
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - L Bartova
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - P Sezen
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - J Keller
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - A Schatzberg
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - L Pezawas
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Gibson K, Walsh M, Hynd M, Eisenlohr-Moul T, Walsh E, Bondy E, Gray R, Brierley J, Bizzell J, Styner M, Dichter GS, Schiller CE. The effects of estradiol on subcortical brain volumes in perimenopausal-onset depression. J Affect Disord 2025; 377:45-52. [PMID: 39983774 PMCID: PMC11997973 DOI: 10.1016/j.jad.2025.02.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/05/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Perimenopause is associated with increases in depressive and vasomotor symptoms (VMS), which can be alleviated with transdermal estradiol (TE2) administration. Subcortical brain regions are commonly implicated in depression, are dense with E2 receptors and are susceptible to volumetric changes resulting from E2 regulation of synaptic density. No studies have examined linkages among TE2 administration, perimenopausal-onset major depression (PO-MDD) and subcortical brain volumes. METHODS This is an exploratory data analysis of change in subcortical brain volumes measured via 3 T MRI before and after three-weeks of TE2 administration in 14 women with PO-MDD and 17 euthymic controls. Regions of interest were the hippocampus, amygdala, putamen, thalamus, and caudate nucleus. Multilevel models examined relations between baseline volumes and volumetric changes with symptom trajectories in the PO-MDD group. RESULTS In the PO-MDD group, anhedonia (p < 0.004) and VMS (p < 0.001) significantly reduced following TE2 administration. There was a significant Group X Time interaction in the right hippocampus (p < 0.01), driven by volume increases in the control group (p < 0.001). In the PO-MDD group, change in right hippocampal volumes significantly predicted decreases in anhedonia trajectories from baseline to week 2 and week 3 (p's < 0.001) and decreases in VMS across all timepoints (p's < 0.001). DISCUSSION Women with PO-MDD, who presented with more severe baseline anhedonia and VMS, experienced greater reductions in anhedonia, VMS, and hippocampal volumes, demonstrating a greater response to E2. Hippocampal volume change may be a candidate for predicting treatment response to E2 for anhedonia and vasomotor symptoms in women with PO-MDD. These findings should be validated with a placebo-controlled trial.
Collapse
Affiliation(s)
- Kathryn Gibson
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA.
| | - Melissa Walsh
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| | - Megan Hynd
- Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514, USA
| | - Tory Eisenlohr-Moul
- Department of Psychiatry, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Erin Walsh
- Department of Psychiatry, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Erin Bondy
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| | - Reese Gray
- Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514, USA
| | - James Brierley
- Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514, USA
| | - Joshua Bizzell
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| | - Martin Styner
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| | - Gabriel S Dichter
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA; Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, Chapel Hill, NC 27510, USA
| | - Crystal E Schiller
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| |
Collapse
|
10
|
Owen MJ, Bray NJ, Walters JTR, O'Donovan MC. Genomics of schizophrenia, bipolar disorder and major depressive disorder. Nat Rev Genet 2025:10.1038/s41576-025-00843-0. [PMID: 40355602 DOI: 10.1038/s41576-025-00843-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Schizophrenia, bipolar disorder and major depressive disorder - which are the most common adult disorders requiring psychiatric care - contribute substantially to premature mortality and morbidity globally. Treatments for these disorders are suboptimal, there are no diagnostic pathologies or biomarkers and their pathophysiologies are poorly understood. Novel therapeutic and diagnostic approaches are thus badly needed. Given the high heritability of psychiatric disorders, psychiatry has potentially much to gain from the application of genomics to identify molecular risk mechanisms and to improve diagnosis. Recent large-scale, genome-wide association studies and sequencing studies, together with advances in functional genomics, have begun to illuminate the genetic architectures of schizophrenia, bipolar disorder and major depressive disorder and to identify potential biological mechanisms. Genomic findings also point to the aetiological relationships between different diagnoses and to the relationships between adult psychiatric disorders and childhood neurodevelopmental conditions.
Collapse
Affiliation(s)
- Michael J Owen
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.
| | - Nicholas J Bray
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - James T R Walters
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
11
|
Whittle S, Rakesh D, Simmons JG, Schwartz O, Vijayakumar N, Allen NB. Prospective Associations Between Structural Brain Development and Onset of Depressive Disorder During Adolescence and Emerging Adulthood. Am J Psychiatry 2025:appiajp20240588. [PMID: 40329643 DOI: 10.1176/appi.ajp.20240588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
OBJECTIVE Brain structural alterations are consistently reported in depressive disorders, yet it remains unclear whether these alterations exist prior to disorder onset and thus may reflect a preexisting vulnerability. The authors investigated prospective adolescent neurodevelopmental risk markers for depressive disorder onset, using data from a 15-year longitudinal study. METHODS A community sample of 161 adolescents participated in neuroimaging assessments conducted during early (age 12), mid (age 16), and late (age 19) adolescence. Onsets of depressive disorders were assessed for the period spanning early adolescence through emerging adulthood (ages 12-27). Forty-six participants (28 female) experienced a first episode of a depressive disorder during the follow-up period; 83 participants (36 female) received no mental disorder diagnosis. Joint modeling was used to investigate whether brain structure (subcortical volume, cortical thickness, and surface area) or age-related changes in brain structure were associated with the risk of depressive disorder onset. RESULTS Age-related increases in amygdala volume (hazard ratio=3.01), and more positive age-related changes (i.e., greater thickening or attenuated thinning) of temporal (parahippocampal gyrus, hazard ratio=3.73; fusiform gyrus, hazard ratio=4.14), insula (hazard ratio=4.49), and occipital (lingual gyrus, hazard ratio=4.19) regions were statistically significantly associated with the onset of depressive disorder. CONCLUSIONS Relative increases in amygdala volume and temporal, insula, and occipital cortical thickness across adolescence may reflect disturbances in brain development, contributing to depression onset. This raises the possibility that prior findings of reduced gray matter in clinically depressed individuals instead reflect alterations that are caused by disorder-related factors after onset.
Collapse
Affiliation(s)
- Sarah Whittle
- Centre for Youth Mental Health, University of Melbourne, Parkville, Victoria, Australia (Whittle); Orygen, Parkville, Victoria, Australia (Whittle); Neuroimaging Department, Institute of Psychology, Psychiatry, and Neuroscience, King's College London (Whittle); Melbourne School of Psychological Sciences (Simmons) and Department of Psychiatry (Schwartz), University of Melbourne, Parkville, Victoria, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia (Vijayakumar); Deakin University, Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Geelong, Victoria, Australia (Vijayakumar); Department of Psychology, University of Oregon, Eugene (Allen)
| | - Divyangana Rakesh
- Centre for Youth Mental Health, University of Melbourne, Parkville, Victoria, Australia (Whittle); Orygen, Parkville, Victoria, Australia (Whittle); Neuroimaging Department, Institute of Psychology, Psychiatry, and Neuroscience, King's College London (Whittle); Melbourne School of Psychological Sciences (Simmons) and Department of Psychiatry (Schwartz), University of Melbourne, Parkville, Victoria, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia (Vijayakumar); Deakin University, Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Geelong, Victoria, Australia (Vijayakumar); Department of Psychology, University of Oregon, Eugene (Allen)
| | - Julian G Simmons
- Centre for Youth Mental Health, University of Melbourne, Parkville, Victoria, Australia (Whittle); Orygen, Parkville, Victoria, Australia (Whittle); Neuroimaging Department, Institute of Psychology, Psychiatry, and Neuroscience, King's College London (Whittle); Melbourne School of Psychological Sciences (Simmons) and Department of Psychiatry (Schwartz), University of Melbourne, Parkville, Victoria, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia (Vijayakumar); Deakin University, Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Geelong, Victoria, Australia (Vijayakumar); Department of Psychology, University of Oregon, Eugene (Allen)
| | - Orli Schwartz
- Centre for Youth Mental Health, University of Melbourne, Parkville, Victoria, Australia (Whittle); Orygen, Parkville, Victoria, Australia (Whittle); Neuroimaging Department, Institute of Psychology, Psychiatry, and Neuroscience, King's College London (Whittle); Melbourne School of Psychological Sciences (Simmons) and Department of Psychiatry (Schwartz), University of Melbourne, Parkville, Victoria, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia (Vijayakumar); Deakin University, Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Geelong, Victoria, Australia (Vijayakumar); Department of Psychology, University of Oregon, Eugene (Allen)
| | - Nandita Vijayakumar
- Centre for Youth Mental Health, University of Melbourne, Parkville, Victoria, Australia (Whittle); Orygen, Parkville, Victoria, Australia (Whittle); Neuroimaging Department, Institute of Psychology, Psychiatry, and Neuroscience, King's College London (Whittle); Melbourne School of Psychological Sciences (Simmons) and Department of Psychiatry (Schwartz), University of Melbourne, Parkville, Victoria, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia (Vijayakumar); Deakin University, Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Geelong, Victoria, Australia (Vijayakumar); Department of Psychology, University of Oregon, Eugene (Allen)
| | - Nicholas B Allen
- Centre for Youth Mental Health, University of Melbourne, Parkville, Victoria, Australia (Whittle); Orygen, Parkville, Victoria, Australia (Whittle); Neuroimaging Department, Institute of Psychology, Psychiatry, and Neuroscience, King's College London (Whittle); Melbourne School of Psychological Sciences (Simmons) and Department of Psychiatry (Schwartz), University of Melbourne, Parkville, Victoria, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia (Vijayakumar); Deakin University, Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Geelong, Victoria, Australia (Vijayakumar); Department of Psychology, University of Oregon, Eugene (Allen)
| |
Collapse
|
12
|
Ping L, Chu Z, Zhou B, Sun D, Chu J, Xu J, Li Z, Zhang D, Cheng Y. Structural alterations after repetitive transcranial magnetic stimulation in depression and the link to neurotransmitter profiles. Asian J Psychiatr 2025; 107:104445. [PMID: 40117801 DOI: 10.1016/j.ajp.2025.104445] [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: 11/24/2024] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Repetitive Transcranial Magnetic Stimulation (rTMS) is widely used to treat depression, showing good efficacy and tolerability. However, the neurobiological mechanisms of its antidepressant effects remain unclear. This study explores the potential impact of rTMS on brain structure in depressed patients and its link to neurotransmitter systems. METHODS Thirty-six MDD patients were randomized to receive 5 times per week for 3 weeks of active or sham rTMS targeting the dorsolateral prefrontal cortex (DLPFC) within a double-blind, sham-controlled trial. The Hamilton Depression Rating Scale-17 items (HAMD-17) was used to assess depressive symptoms at baseline and the end of 1 W, 2 W and 3 W after treatment. We analyzed the differences in efficacy between the two groups of patients at different time points, and the grey matter changes of the brain before and after treatment in both groups. In addition, we analyzed the spatial correlations between abnormal grey matter and the neurotransmitter receptors and transporters map. RESULTS Both the active and sham groups showed significant improvement in depression and anxiety symptoms following rTMS treatment, with the Active group demonstrating greater improvement. Additionally, the Active group exhibited increased grey matter volume in regions associated with the frontal-limbic network, and these changes were significantly correlated with the spatial distribution of D1 receptors. CONCLUSION This study suggests that rTMS targeting the left DLPFC produces antidepressant effects by enhancing structural plasticity in the frontal-limbic network, and that dopamine system modulation may underlie rTMS therapeutic effects. These findings provide insight into the neurobiological basis of rTMS for depression treatment.
Collapse
Affiliation(s)
- Liangliang Ping
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; Department of Psychiatry, Xiamen Xianyue Hospital, Xiamen, Fujian 361000, China; Xianyue Hospital Affiliated with Xiamen Medical College, Xiamen, Fujian 361000, China; Fujian Psychiatric Center, Xiamen, Fujian 361000, China; Fujian Clinical Research Center for Mental Disorders, Xiamen, Fujian 361000, China
| | - Zhaosong Chu
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Biao Zhou
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Duo Sun
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Jiangmin Chu
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Jian Xu
- Department of Rheumatology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Zhenhui Li
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, Yunnan 650118, China
| | - Dafu Zhang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, Yunnan 650118, China.
| | - Yuqi Cheng
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310063, China.
| |
Collapse
|
13
|
Vater M, Rost N, Eckstein G, Sauer S, Tontsch A, Erhardt A, Lucae S, Brückl T, Klopstock T, Sämann PG, Binder EB. Huntingtin CAG repeat size variations below the Huntington's disease threshold: associations with depression, anxiety and basal ganglia structure. Eur J Hum Genet 2025; 33:624-632. [PMID: 39572770 PMCID: PMC12048612 DOI: 10.1038/s41431-024-01737-1] [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: 06/01/2024] [Revised: 09/19/2024] [Accepted: 11/05/2024] [Indexed: 05/04/2025] Open
Abstract
Huntington's disease (HD) is strongly associated with psychiatric symptoms, yet, associations between huntingtin gene (HTT) CAG repeat size variations and psychiatric phenotypes outside the HD complex are still under-investigated. In this genetic case-control study we compared the distribution of HTT CAG repeat sizes in predefined ranges between patients with major depressive disorder (MDD) (n = 2136) and anxiety disorders (ANX) (n = 493), and healthy controls (CON) (n = 1566). We used regression models to study interactions between the alleles and associations with fine-granular clinical phenotypes and basal ganglia structure. HD mutations in the range of incomplete penetrance (36-39 repeats) were not overrepresented in patients. In participants older than 48 years, 13-20 repeats on both HTT alleles were associated with a reduced ANX risk whereas a 13-20 | 21-26 combination was associated with an increased ANX risk. Post-hoc analyses confirmed a turning point around 21 repeats and trends in the same direction were detected for MDD. The joint patient | CON analysis of the full spectrum of allele combinations confirmed interaction effects and age-dependent allele | risk profiles. A short-by-long interaction effect and an age-dependent negative correlation of the short allele on the nucleus accumbens volume was detected, independently of the diagnostic group. In conclusion, we revealed that HTT CAG repeat sizes of both alleles in the non-HD range are associated with a risk modulation for common psychiatric disorders as well as basal ganglia structure differences in an age-dependent way, possibly implying that normal variation of the functionally diverse wildtype huntingtin protein may impact brain function.
Collapse
Affiliation(s)
- Magdalena Vater
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
| | - Nicolas Rost
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Gertrud Eckstein
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Susann Sauer
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Alina Tontsch
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Angelika Erhardt
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | | | - Tanja Brückl
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Thomas Klopstock
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Friedrich-Baur-Institute, Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | | | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
14
|
Linli Z, Liang X, Zhang Z, Hu K, Guo S. Enhancing brain age estimation under uncertainty: A spectral-normalized neural gaussian process approach utilizing 2.5D slicing. Neuroimage 2025; 311:121184. [PMID: 40180003 DOI: 10.1016/j.neuroimage.2025.121184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/19/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025] Open
Abstract
Brain age gap, the difference between estimated brain age and chronological age via magnetic resonance imaging, has emerged as a pivotal biomarker in the detection of brain abnormalities. While deep learning is accurate in estimating brain age, the absence of uncertainty estimation may pose risks in clinical use. Moreover, current 3D brain age models are intricate, and using 2D slices hinders comprehensive dimensional data integration. Here, we introduced Spectral-normalized Neural Gaussian Process (SNGP) accompanied by 2.5D slice approach for seamless uncertainty integration in a single network with low computational expenses, and extra dimensional data integration without added model complexity. Subsequently, we compared different deep learning methods for estimating brain age uncertainty via the Pearson correlation coefficient, a metric that helps circumvent systematic underestimation of uncertainty during training. SNGP shows excellent uncertainty estimation and generalization on a dataset of 11 public datasets (N = 6327), with competitive predictive performance (MAE=2.95). Besides, SNGP demonstrates superior generalization performance (MAE=3.47) on an independent validation set (N = 301). Additionally, we conducted five controlled experiments to validate our method. Firstly, uncertainty adjustment in brain age estimation improved the detection of accelerated brain aging in adolescents with ADHD, with a 38% increase in effect size after adjustment. Secondly, the SNGP model exhibited OOD detection capabilities, showing significant differences in uncertainty across Asian and non-Asian datasets. Thirdly, the performance of DenseNet as a backbone for SNGP was slightly better than ResNeXt, attributed to DenseNet's feature reuse capability, with robust generalization on an independent validation set. Fourthly, site effect harmonization led to a decline in model performance, consistent with previous studies. Finally, the 2.5D slice approach significantly outperformed 2D methods, improving model performance without increasing network complexity. In conclusion, we present a cost-effective method for estimating brain age with uncertainty, utilizing 2.5D slicing for enhanced performance, showcasing promise for clinical applications.
Collapse
Affiliation(s)
- Zeqiang Linli
- School of Mathematics and Statistics, Guangdong University of Foreign Studies, Guangzhou, 510420, PR China; Laboratory of Language Engineering and Computing, Guangdong University of Foreign Studies, 510420, Guangzhou, PR China; MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha, 410006, PR China.
| | - Xingcheng Liang
- School of Mathematics and Statistics, Guangdong University of Foreign Studies, Guangzhou, 510420, PR China; Laboratory of Language Engineering and Computing, Guangdong University of Foreign Studies, 510420, Guangzhou, PR China.
| | - Zhenhua Zhang
- School of Mathematics and Statistics, Guangdong University of Foreign Studies, Guangzhou, 510420, PR China; Laboratory of Language Engineering and Computing, Guangdong University of Foreign Studies, 510420, Guangzhou, PR China.
| | - Kang Hu
- School of Information Engineering, Wuhan Business University, Wuhan, 430056, PR China.
| | - Shuixia Guo
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha, 410006, PR China; Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha, 410006, PR China.
| |
Collapse
|
15
|
Yang M, Qin X, Liu X. The effect of mitochondrial-associated endoplasmic reticulum membranes (MAMs) modulation: New insights into therapeutic targets for depression. Neurosci Biobehav Rev 2025; 172:106087. [PMID: 40031998 DOI: 10.1016/j.neubiorev.2025.106087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/07/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
Depression is a prevalent mental disorder with high morbidity and mortality and its pathogenesis remains exactly unclarified. However, mitochondria and endoplasmic reticulum (ER) are two highly dynamic organelles that perform an indispensable role in the development of depression. Mitochondrial dysfunction and ER stress are recognized as vital pathological hallmarks in depression. The changes of intracellular activities such as mitochondrial dynamics, mitophagy, energy metabolism and ER stress are closely correlated with the progression of depression. Moreover, organelles interactions are conducive to homeostasis and cellular functions, and mitochondrial-associated endoplasmic reticulum membranes (MAMs) serve as signaling hubs of the two organelles and the coupling of the pathological progression. The main roles of MAMs are involved in metabolism, signal transduction, lipid transport, and maintenance of its structure and function. At present, accumulating studies elucidated that MAMs have gradually become a novel therapeutic target in treatment of depression. In the review, we focus on influence of mitochondria dysfunction and ER stress on depression. Furthermore, we discuss the underlying role of MAMs in depression and highlight natural products targeting MAMs as potential antidepressants to treat depression.
Collapse
Affiliation(s)
- Maohui Yang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China.
| | - Xiaojie Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
16
|
Li ZY, Fei CJ, Yin RY, Kang JJ, Ma Q, He XY, Wu XR, Zhao YJ, Zhang W, Liu WS, Wu BS, Yang L, Zhu Y, Feng JF, Yu JT, Cheng W. Whole exome sequencing identified six novel genes for depressive symptoms. Mol Psychiatry 2025; 30:1925-1936. [PMID: 39472661 DOI: 10.1038/s41380-024-02804-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 04/24/2025]
Abstract
Previous genome-wide association studies of depression have primarily focused on common variants, limiting our comprehensive understanding of the genetic architecture. In contrast, whole-exome sequencing can capture rare coding variants, helping to explore the phenotypic consequences of altering protein-coding genes. Here, we conducted a large-scale exome-wide association study on 296,199 participants from the UK Biobank, assessing their depressive symptom scores through the Patient Health Questionnaire-4. We identified 22 genes associated with depressive symptoms, including 6 newly discovered genes (TRIM27, UBD, SVOP, ADGRB2, IRF2BPL, and ANKRD12). Both ontology enrichment analysis and plasma proteomics association analysis consistently revealed that the identified genes were associated with immune responses. Furthermore, we identified associations between these genes and brain regions related to depression, such as anterior cingulate cortex and orbitofrontal cortex. Additionally, phenome-wide association analysis demonstrated that TRIM27 and UBD were associated with neuropsychiatric, cognitive, biochemistry, and inflammatory traits. Our findings offer new insights into the potential mechanisms and genetic architecture of depressive symptoms.
Collapse
Affiliation(s)
- Ze-Yu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Chen-Jie Fei
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Rui-Ying Yin
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Qing Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Xiao-Yu He
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xin-Rui Wu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yu-Jie Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei-Shi Liu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Liu Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ying Zhu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, UK
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, China
| | - Jin-Tai Yu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, China.
| |
Collapse
|
17
|
Zhou X, Zhang L, Gao W, Li H, Guo Q, Dai J, Gao F, Wang L. Esketamine alleviates cognitive impairment signs induced by modified electroconvulsive therapy in a depression rat model via the KLF4/p38 MAPK pathway. J Affect Disord 2025; 376:302-312. [PMID: 39938694 DOI: 10.1016/j.jad.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/05/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Depression is a common and serious psychiatric disorder with significant impacts on individuals. Modified electroconvulsive therapy (MECT) is an established treatment for severe and treatment-resistant depression, but its cognitive side effects, particularly memory impairments, limit its use. Esketamine (ESK), an FDA-approved antidepressant, has shown neuroprotective effects. However, its role in mitigating MECT-induced cognitive deficits remains unexplored. This study investigates whether ESK could alleviate MECT-induced signs of cognitive impairments in a rat model of depression and explores the underlying mechanisms. METHODS Male Sprague-Dawley rats were exposed to chronic unpredictable mild stress (CUMS) model to induce depressive-like behaviors. Rats were then subjected to MECT, ESK treatment, or both. Depression-like behaviors and cognitive functions were evaluated using various tests. Molecular and cellular assays were performed to assess hippocampal neuronal apoptosis, inflammation, and synaptic plasticity, with a focus on the Krüppel-like factor 4 (KLF4) and p38 MAPK signaling pathways. RESULTS MECT treatment significantly alleviated depressive-like symptoms but exacerbated cognitive impairments, hippocampal neuronal apoptosis, and neuroinflammation. ESK co-treatment improved depressive behaviors while reversing MECT-induced cognitive deficits, reducing hippocampal apoptosis, and decreasing inflammatory cytokine levels. Furthermore, ESK enhanced synaptic plasticity and upregulated KLF4 expression, which in turn inhibited the activation of the p38 MAPK pathway. Functionally, knockdown of KLF4 diminished the neuroprotective effects of ESK, confirming its critical role in mediating cognitive protection. CONCLUSIONS Esketamine mitigates METC-induced cognitive impairment in the animal model, by upregulating KLF4, which inhibits the p38 MAPK pathway, offering a potential therapeutic strategy for improving cognitive outcomes in patients undergoing ECT.
Collapse
Affiliation(s)
- Xiaohui Zhou
- Department of Anesthesiology, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, Hebei 050000, China
| | - Li Zhang
- Department of Anesthesiology, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, Hebei 050000, China
| | - Weiwei Gao
- Department of Anesthesiology, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, Hebei 050000, China
| | - Huili Li
- Department of Anesthesiology, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, Hebei 050000, China
| | - Qiongmei Guo
- Department of Anesthesiology, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, Hebei 050000, China
| | - Jiajia Dai
- Department of Anesthesiology, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, Hebei 050000, China
| | - Fei Gao
- Department of Anesthesiology, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, Hebei 050000, China
| | - Li Wang
- Department of Anesthesiology, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, Hebei 050000, China.
| |
Collapse
|
18
|
Chen C, Liu Y, Sun Y, Jiang W, Yuan Y, Qing Z. Abnormal structural covariance network in major depressive disorder: Evidence from the REST-meta-MDD project. Neuroimage Clin 2025; 46:103794. [PMID: 40328096 DOI: 10.1016/j.nicl.2025.103794] [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: 03/07/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is a common mental illness associated with brain morphological abnormalities. Although extensive studies have examined gray matter volume (GMV) changes in MDD, inconsistencies persist in reported findings. In the current study, we employed source-based morphometry (SBM) and structural covariance network (SCN) analyses to a large multi-center sample from the REST-meta-MDD database, aiming to characterize robust results of structural abnormalities in MDD. METHODS We analyzed 798 MDD patients and 974 healthy controls (HCs) from the REST-meta-MDD consortium. Voxel-based morphometry was applied to generate GMV maps. SBM was used to adaptively parcellate brain into different components, and SCN was constructed based on SBM components. Volume scores in each component and SCNs between the components were both compared between MDD and HC groups, as well as between first-episode drug-naive (FEDN) and recurrent MDD subgroups. RESULTS SBM identified 20 stable components. Three components encompassing the middle temporal gyrus, middle orbitofrontal gyrus and superior frontal gyrus exhibited volumetric differences between the MDD and HC groups. Volume differences were observed in the cingulate cortex and medial frontal gyrus between the FEDN and recurrent groups. SCN analysis revealed 9 aberrant pairs in MDD vs. HCs, and 7 pairs in FEDN vs. recurrent groups. All aberrant component pairs in the SCN implicated the prefrontal cortex. CONCLUSIONS These findings demonstrated brain structural deficits in MDD, and highlighted the prefrontal cortex as a central hub of SCN alterations. Our findings advance the understanding of MDD's neural mechanisms and suggest directions for diagnostic research.
Collapse
Affiliation(s)
- Changmin Chen
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Yuhan Liu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Yu Sun
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China; Joint Research Center for Biomedical Engineering, Southeast University-University of Birmingham, Nanjing 210096, China
| | - Wenhao Jiang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China
| | - Zhao Qing
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China; Shing-Tung Yau Center, Southeast University, Nanjing 210096, China; Joint Research Center for Biomedical Engineering, Southeast University-University of Birmingham, Nanjing 210096, China.
| |
Collapse
|
19
|
Goes FS, Collado-Torres L, Zandi PP, Huuki-Myers L, Tao R, Jaffe AE, Pertea G, Shin JH, Weinberger DR, Kleinman JE, Hyde TM. Large-scale transcriptomic analyses of major depressive disorder reveal convergent dysregulation of synaptic pathways in excitatory neurons. Nat Commun 2025; 16:3981. [PMID: 40295477 PMCID: PMC12037741 DOI: 10.1038/s41467-025-59115-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Major Depressive Disorder (MDD) is a common, complex disorder that is a leading cause of disability worldwide and a significant risk factor for suicide. In this study, we have performed the largest molecular analysis of MDD in postmortem human brains (846 samples across 458 individuals) in the subgenual Anterior Cingulate Cortex (sACC) and the Amygdala, two regions central to mood regulation and the pathophysiology of MDD. We found extensive expression differences, particularly at the level of specific transcripts, with prominent enrichment for genes associated with the vesicular functioning, the postsynaptic density, GTPase signaling, and gene splicing. We find associated transcriptional features in 107 of 243 genome-wide significant loci for MDD and, through integrative analyses, highlight convergence of genetic risk, gene expression, and network-based analyses on dysregulated glutamatergic signaling and synaptic vesicular functioning. Together, these results provide an initial mechanistic understanding of MDD and highlight potential targets for novel drug discovery.
Collapse
Affiliation(s)
- Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Stanley and Elizabeth Star Precision Medicine Center of Excellence in Mood Disorders, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Leonardo Collado-Torres
- The Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Peter P Zandi
- Department of Psychiatry and Behavioral Sciences, Stanley and Elizabeth Star Precision Medicine Center of Excellence in Mood Disorders, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Ran Tao
- The Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Andrew E Jaffe
- The Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Geo Pertea
- The Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Joo Heon Shin
- The Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Daniel R Weinberger
- The Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joel E Kleinman
- Department of Psychiatry and Behavioral Sciences, Stanley and Elizabeth Star Precision Medicine Center of Excellence in Mood Disorders, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Thomas M Hyde
- The Lieber Institute for Brain Development, Baltimore, MD, USA.
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
20
|
Le Berre A, Attali D, Uszynski I, Debacker C, Lui M, Charron S, Moyal M, Ramon F, Henensal A, Benzakoun J, Mekaoui L, Gorwood P, Poupon C, Cachia A, Oppenheim C, Plaze M. Hippocampal microstructural changes following electroconvulsive therapy in severe depression. Mol Psychiatry 2025:10.1038/s41380-025-03016-x. [PMID: 40199994 DOI: 10.1038/s41380-025-03016-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 03/20/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
Electroconvulsive therapy (ECT) induces hippocampal volume increases in depressed patients, potentially reflecting neuroplasticity. We hypothesized that Neurite Orientation Dispersion and Density Imaging (NODDI) could provide in vivo evidence of hippocampal neuroplasticity following ECT. This longitudinal study evaluated 43 depressed patients undergoing ECT and 24 controls. MRI and clinical assessments were performed at baseline (V1), after 5 sessions (V2), and post-treatment (V3). Evaluations included a 3 T MR-scan with 3DT1-weighted and multi-shell diffusion (b = 200/1500/2500 s/mm², 30/45/60directions) sequences. Q-ball, Diffusion Tensor, and NODDI models provided: axial diffusivity (AD), radial diffusivity (RD), mean diffusivity (MD), fractional anisotropy (FA), generalized FA (GFA), neurite density index (NDI), isotropic fraction (Fiso), and orientation dispersion index (ODI). FreeSurfer extracted whole hippocampal and subfield volumes from T1-weighted images. Longitudinal changes were assessed with linear mixed-effect models. 107 MRIs from patients and 24 MRIs from controls were analyzed. ECT induced significant bilateral hippocampal volume increases (p < 0.001). Group comparisons showed consistently higher FA, lower GFA and ODI in patients compared to controls at all time-points. Following ECT, significant diffusion changes included decreased hippocampal GFA, FA, AD, MD and Fiso, along with increased ODI and NDI. NDI and Fiso changes were localized to the dentate gyrus but not the hippocampal tail. ECT responders showed a significant right hippocampal volume increase at V2 compared to non-responders. After ECT, hippocampal volume increases are accompanied by bilateral changes in NODDI parameters, particularly in the dentate gyrus, consistent with hippocampal neuroplasticity.
Collapse
Affiliation(s)
- A Le Berre
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France.
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France.
| | - D Attali
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
- Physics for Medicine Paris, Inserm U1273, CNRS UMR 8063, ESPCI Paris, PSL University, Paris, France
| | - I Uszynski
- BAOBAB, UMR 9027, NeuroSpin, Université Paris-Saclay, CNRS, CEA, Gif-sur-Yvette, France
| | - C Debacker
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - M Lui
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - S Charron
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - M Moyal
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - F Ramon
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
| | - A Henensal
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - J Benzakoun
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - L Mekaoui
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - P Gorwood
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - C Poupon
- BAOBAB, UMR 9027, NeuroSpin, Université Paris-Saclay, CNRS, CEA, Gif-sur-Yvette, France
| | - A Cachia
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- Université Paris Cité, Laboratory for the Psychology of Child Development and Education, CNRS UMR 8240, F-75005, Paris, France
| | - C Oppenheim
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - M Plaze
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Ima-Brain team, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| |
Collapse
|
21
|
Kochunov P, Adhikari BM, Keator D, Amen D, Gao S, Karcher NR, Labate D, Azencott R, Huang Y, Syed H, Ke H, Thompson PM, Wang DJJ, Mitchell BD, Turner JA, van Erp TG, Jahanshad N, Ma Y, Du X, Burroughs W, Chen S, Ma T, Soares JC, Hong LE. Functional vs Structural Cortical Deficit Pattern Biomarkers for Major Depressive Disorder. JAMA Psychiatry 2025:2832270. [PMID: 40172866 PMCID: PMC11966481 DOI: 10.1001/jamapsychiatry.2025.0192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/01/2025] [Indexed: 04/04/2025]
Abstract
Importance Major depressive disorder (MDD) is a severe mental illness characterized more by functional rather than structural brain abnormalities. The pattern of regional homogeneity (ReHo) deficits in MDD may relate to underlying regional hypoperfusion. Capturing this functional deficit pattern provides a brain pattern-based biomarker for MDD that is linked to the underlying pathophysiology. Objective To examine whether cortical ReHo patterns provide a replicable biomarker for MDD that is more sensitive than reduced cortical thickness and evaluate whether the ReHo MDD deficit pattern reflects regional cerebral blood flow (RCBF) deficit patterns in MDD and whether a regional vulnerability index (RVI) thus constructed may provide a concise brain pattern-based biomarker for MDD. Design, Settings, and Participants The UK Biobank (UKBB) participants had ReHo and structural measurements. Participants from the Enhancing Neuroimaging Genetics Through Meta-Analysis (ENIGMA) Consortium were included for measuring the MDD structural cortical deficit pattern. The UKBB ReHo and ENIGMA cortical thickness effect sizes for MDD were used to test the deficit patterns in the Amish Connectome Project (ACP) with ReHo, structural, and RCBF data. Finally, the Ament Clinic Inc (ACI) sample had RCBF data measured using single-photon emission computed tomography. Data were analyzed from August 2021 to September 2024. Exposures ReHo and structural measurements. Results Included in this analysis were 4 datasets: (1) UKBB (N = 4810 participants; 2220 with recurrent MDD and 2590 controls; mean [SD] age, 63.0 [7.5] years; 1121 female [50%]), (2) ENIGMA (N = 10 115 participants; 2148 with MDD and 7957 healthy controls; mean [SD] age, 39.9 [10.0] years; 5927 female [59%]), (3) ACP (N = 204 participants; 68 with a lifetime diagnosis of MDD and 136 controls; mean [SD] age, 41.0 [14.5] years; 104 female [51%]), and (4) ACI (N = 372 participants; 296 with recurrent MDD and 76 controls; mean [SD] age, 45.3 [17.2] years; 189 female [51%]). MDD participants had lower cortical ReHo in the cingulum, superior temporal lobe, frontal lobe, and several other areas, with no significant differences in cortical thickness. The regional pattern of ReHo MDD effect sizes was significantly correlated with that of RCBF obtained from 2 independent datasets (Pearson r = 0.52 and Pearson r = 0.46; P < 10-4). ReHo and RCBF functional RVIs showed numerically stronger effect sizes (Cohen d = 0.33-0.90) compared with structural RVIs (Cohen d = 0.09-0.20). Elevated ReHo-based RVI-MDD values in individuals with MDD were associated with higher depression symptom severity across cohorts. Conclusions and Relevance Results of this case-control study suggest that the ReHo MDD deficit pattern reflected cortical hypoperfusion and was regionally specific in MDD. ReHo-based RVI may serve as a sensitive functional biomarker for MDD.
Collapse
Affiliation(s)
- Peter Kochunov
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston School of Behavioral Health Sciences, University of Texas Health Science Center at Houston, Houston
| | - Bhim M. Adhikari
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston School of Behavioral Health Sciences, University of Texas Health Science Center at Houston, Houston
| | - David Keator
- Amen Clinics Inc, Costa Mesa, California
- Department of Psychiatry and Human Behavior, University of California, Irvine
- Change Your Brain Change Your Life Foundation, Costa Mesa, California
| | - Daniel Amen
- Amen Clinics Inc, Costa Mesa, California
- Change Your Brain Change Your Life Foundation, Costa Mesa, California
| | - Si Gao
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston School of Behavioral Health Sciences, University of Texas Health Science Center at Houston, Houston
| | - Nicole R. Karcher
- Department of Psychiatry, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Demetrio Labate
- Departments of Mathematics, University of Houston, Houston, Texas
| | - Robert Azencott
- Departments of Mathematics, University of Houston, Houston, Texas
| | - Yewen Huang
- Departments of Mathematics, University of Houston, Houston, Texas
| | - Hussain Syed
- Departments of Mathematics, University of Houston, Houston, Texas
| | - Hongjie Ke
- Department of Biostatistics, University of Maryland College Park, College Park
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey
| | - Danny J. J. Wang
- Laboratory of Functional MRI Technology, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles
| | - Braxton D. Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, Maryland
| | - Jessica A. Turner
- Department of Psychiatry and Behavioral Science, The Ohio State University College of Medicine, Columbus
| | - Theo G.M. van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine
- Center for the Neurobiology of Learning and Memory, University of California Irvine
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey
| | - Yizhou Ma
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston School of Behavioral Health Sciences, University of Texas Health Science Center at Houston, Houston
| | - Xiaoming Du
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston School of Behavioral Health Sciences, University of Texas Health Science Center at Houston, Houston
| | - William Burroughs
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston School of Behavioral Health Sciences, University of Texas Health Science Center at Houston, Houston
| | - Shuo Chen
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore
| | - Tianzhou Ma
- Department of Biostatistics, University of Maryland College Park, College Park
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston School of Behavioral Health Sciences, University of Texas Health Science Center at Houston, Houston
| | - L. Elliot Hong
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston School of Behavioral Health Sciences, University of Texas Health Science Center at Houston, Houston
| |
Collapse
|
22
|
Wang J, Chen W, Zhu Q, Liu Y, Kang Z, Liu D, Zeng G. Effects of Qingyangshen glycosides on neuroplasticity in a mouse model of social defeat. Animal Model Exp Med 2025; 8:581-594. [PMID: 39921215 PMCID: PMC12008452 DOI: 10.1002/ame2.12499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 08/16/2024] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Qingyangshen (Cynanchum otophyllum C.K. Schneid) is a folk drug for treating depression and other mental disorders induced by social defeat stress. Neuroplasticity in the hippocampus is essential for the modulation of cognition and emotion, and its impairment may contribute to the development and progression of depression. Our previous studies have found that Qingyangshen glycosides (QYS) can improve depression-like behavior in social failure mouse models, mainly through PGC-1α/FNDC5/BDNF signaling pathways activation, but its effects and mechanisms on hippocampal neuroplasticity remain unknown. METHODS Chronic social defeat stress (CSDS) was used to induce social defeat in mice. Morphological changes in the hippocampus were observed by H&E staining and Golgi staining. Immunofluorescence double staining was used to detect the expression of synaptophysin (SYN) and postsynaptic density protein-95 (PSD-95), while western blot was employed to evaluate PSD-95, SYN, and doublecortin (DCX) proteins. The pathological processing of social defeat and the therapeutic effects of QYS on it was confirmed through behavioral assessment associated with morphologic observation. RESULTS During the whole study, the sucrose preference indices and OFT activity time of CSDS mice were significantly decreased (p ≤ 0.05), and the tail suspension immobility time was significantly increased (p ≤ 0.05), suggesting that the mice had significant depressive symptoms. Treatment with QYS (25, 50, and 100 mg/kg) significantly alleviated depressive symptoms in CSDS mice, which was demonstrated by significantly (p ≤ 0.05 or p ≤ 0.01) reducing the duration of tail-hanging immobility and increasing the tendency of sucrose preference indices and OFT activity time. QYS treatment also significantly increased the expression of DCX, PSD-95, and SYN proteins, which play a crucial role in depression. CONCLUSIONS QYS alleviated these symptoms by enhancing hippocampal neuroplasticity through upregulating the expression of synapse-associated proteins (SAPs). The therapeutic mechanism of QYS may involve modulating the neuroplasticity of hippocampus neurons by altering the expression of SAPs.
Collapse
Affiliation(s)
- Jingru Wang
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Pharmacological research department, Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs and Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| | - Weishi Chen
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qiang Zhu
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yao Liu
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zheng Kang
- Pharmacological research department, Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs and Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| | - Dingding Liu
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Guirong Zeng
- Pharmacological research department, Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs and Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| |
Collapse
|
23
|
Sălcudean A, Bodo CR, Popovici RA, Cozma MM, Păcurar M, Crăciun RE, Crisan AI, Enatescu VR, Marinescu I, Cimpian DM, Nan AG, Sasu AB, Anculia RC, Strete EG. Neuroinflammation-A Crucial Factor in the Pathophysiology of Depression-A Comprehensive Review. Biomolecules 2025; 15:502. [PMID: 40305200 PMCID: PMC12024626 DOI: 10.3390/biom15040502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Depression is a multifactorial psychiatric condition with complex pathophysiology, increasingly linked to neuroinflammatory processes. The present review explores the role of neuroinflammation in depression, focusing on glial cell activation, cytokine signaling, blood-brain barrier dysfunction, and disruptions in neurotransmitter systems. The article highlights how inflammatory mediators influence brain regions implicated in mood regulation, such as the hippocampus, amygdala, and prefrontal cortex. The review further discusses the involvement of the hypothalamic-pituitary-adrenal (HPA) axis, oxidative stress, and the kynurenine pathway, providing mechanistic insights into how chronic inflammation may underlie emotional and cognitive symptoms of depression. The bidirectional relationship between inflammation and depressive symptoms is emphasized, along with the role of peripheral immune responses and systemic stress. By integrating molecular, cellular, and neuroendocrine perspectives, this review supports the growing field of immunopsychiatry and lays the foundation for novel diagnostic biomarkers and anti-inflammatory treatment approaches in depression. Further research in this field holds promise for developing more effective and personalized interventions for individuals suffering from depression.
Collapse
Affiliation(s)
- Andreea Sălcudean
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania; (A.S.); (M.-M.C.); (D.-M.C.)
| | - Cristina-Raluca Bodo
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania; (A.S.); (M.-M.C.); (D.-M.C.)
| | - Ramona-Amina Popovici
- Department of Management and Communication in Dental Medicine, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania
| | - Maria-Melania Cozma
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania; (A.S.); (M.-M.C.); (D.-M.C.)
| | - Mariana Păcurar
- Orthodontic Department, Faculty of Dental Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mures, Romania;
| | | | - Andrada-Ioana Crisan
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| | - Virgil-Radu Enatescu
- Department of Psychiatry, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania;
| | - Ileana Marinescu
- Discipline of Psychiatry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Dora-Mihaela Cimpian
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania; (A.S.); (M.-M.C.); (D.-M.C.)
| | - Andreea-Georgiana Nan
- First Department of Psychiatry, Clinical County Hospital of Targu Mures, 540142 Târgu Mureș, Romania; (A.-G.N.); (A.-B.S.)
| | - Andreea-Bianca Sasu
- First Department of Psychiatry, Clinical County Hospital of Targu Mures, 540142 Târgu Mureș, Romania; (A.-G.N.); (A.-B.S.)
| | - Ramona-Camelia Anculia
- Discipline of Occupational Medicine, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 300041 Timișoara, Romania;
| | - Elena-Gabriela Strete
- Department of Psychiatry, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| |
Collapse
|
24
|
Lei AA, Phang VWX, Lee YZ, Kow ASF, Tham CL, Ho YC, Lee MT. Chronic Stress-Associated Depressive Disorders: The Impact of HPA Axis Dysregulation and Neuroinflammation on the Hippocampus-A Mini Review. Int J Mol Sci 2025; 26:2940. [PMID: 40243556 PMCID: PMC11988747 DOI: 10.3390/ijms26072940] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Chronic stress significantly contributes to the development of depressive disorders, with the hypothalamic-pituitary-adrenal (HPA) axis playing a central role in mediating stress responses. This review examines the neurobiological alterations in the hippocampus linked to HPA axis dysregulation in chronic stress-associated depressive disorders. The prolonged activation of the HPA axis disrupts cortisol regulation, leading to the decline of both physical and mental health. The chronic stress-induced HPA axis dysfunction interacts with inflammatory pathways and generates oxidative stress, contributing to cellular damage and neuroinflammation that further aggravates depressive symptoms. These processes result in structural and functional alterations in the hippocampus, which is essential for emotional regulation and cognitive function. Comprehending the impact of chronic stress on the HPA axis and associated neurobiological pathways is essential for formulating effective interventions for depressive disorders. This review summarises the existing findings and underscores the necessity for future investigations into intervention strategies to improve physical and psychological wellbeing targeting at HPA axis dysregulation for the betterment of psychological wellbeing and human health.
Collapse
Affiliation(s)
- Ai Ai Lei
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | | | - Yu Zhao Lee
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | | | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Yu-Cheng Ho
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Ming Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
- Office of Postgraduate Studies, UCSI University, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
25
|
Sun X, Xia M. Schizophrenia and Neurodevelopment: Insights From Connectome Perspective. Schizophr Bull 2025; 51:309-324. [PMID: 39209793 PMCID: PMC11908871 DOI: 10.1093/schbul/sbae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
BACKGROUND Schizophrenia is conceptualized as a brain connectome disorder that can emerge as early as late childhood and adolescence. However, the underlying neurodevelopmental basis remains unclear. Recent interest has grown in children and adolescent patients who experience symptom onset during critical brain development periods. Inspired by advanced methodological theories and large patient cohorts, Chinese researchers have made significant original contributions to understanding altered brain connectome development in early-onset schizophrenia (EOS). STUDY DESIGN We conducted a search of PubMed and Web of Science for studies on brain connectomes in schizophrenia and neurodevelopment. In this selective review, we first address the latest theories of brain structural and functional development. Subsequently, we synthesize Chinese findings regarding mechanisms of brain structural and functional abnormalities in EOS. Finally, we highlight several pivotal challenges and issues in this field. STUDY RESULTS Typical neurodevelopment follows a trajectory characterized by gray matter volume pruning, enhanced structural and functional connectivity, improved structural connectome efficiency, and differentiated modules in the functional connectome during late childhood and adolescence. Conversely, EOS deviates with excessive gray matter volume decline, cortical thinning, reduced information processing efficiency in the structural brain network, and dysregulated maturation of the functional brain network. Additionally, common functional connectome disruptions of default mode regions were found in early- and adult-onset patients. CONCLUSIONS Chinese research on brain connectomes of EOS provides crucial evidence for understanding pathological mechanisms. Further studies, utilizing standardized analyses based on large-sample multicenter datasets, have the potential to offer objective markers for early intervention and disease treatment.
Collapse
Affiliation(s)
- Xiaoyi Sun
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- School of Systems Science, Beijing Normal University, Beijing, China
| | - Mingrui Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
26
|
Li J, Long Z, Ji GJ, Han S, Chen Y, Yao G, Xu Y, Zhang K, Zhang Y, Cheng J, Wang K, Chen H, Liao W. Major depressive disorder on a neuromorphic continuum. Nat Commun 2025; 16:2405. [PMID: 40069198 PMCID: PMC11897166 DOI: 10.1038/s41467-025-57682-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
The heterogeneity of major depressive disorder (MDD) has hindered clinical translation and neuromarker identification. Biotyping facilitates solving the problems of heterogeneity, by dissecting MDD patients into discrete subgroups. However, interindividual variations suggest that depression may be conceptualized as a "continuum," rather than as a "category." We use a Bayesian model to decompose structural MRI features of MDD patients from a multisite cross-sectional cohort into three latent disease factors (spatial pattern) and continuum factor compositions (individual expression). The disease factors are associated with distinct neurotransmitter receptors/transporters obtained from open PET sources. Increases cortical thickness in sensory and decreases in orbitofrontal cortices (Factor 1) associate with norepinephrine and 5-HT2A density, decreases in the cingulo-opercular network and subcortex (Factor 2) associate with norepinephrine and 5-HTT density, and increases in social and affective brain systems (Factor 3) relate to 5-HTT density. Disease factor patterns can also be used to predict depressive symptom improvement in patients from the longitudinal cohort. Moreover, individual factor expressions in MDD are stable over time in a longitudinal cohort, with differentially expressed disease controls from a transdiagnostic cohort. Collectively, our data-driven disease factors reveal that patients with MDD organize along continuous dimensions that affect distinct sets of regions.
Collapse
Affiliation(s)
- Jiao Li
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Zhiliang Long
- School of Psychology, Southwest University, Chongqing, P.R. China
| | - Gong-Jun Ji
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, P.R. China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Yuan Chen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Guanqun Yao
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, P.R. China
| | - Yong Xu
- Department of Clinical Psychology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P.R. China
| | - Kerang Zhang
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, P.R. China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, P.R. China
| | - Huafu Chen
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Wei Liao
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China.
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China.
| |
Collapse
|
27
|
Hebda-Bauer EK, Hagenauer MH, Munro DB, Blandino P, Meng F, Arakawa K, Stead JDH, Chitre AS, Ozel AB, Mohammadi P, Watson SJ, Flagel SB, Li J, Palmer AA, Akil H. Bioenergetic-related gene expression in the hippocampus predicts internalizing vs. externalizing behavior in an animal model of temperament. Front Mol Neurosci 2025; 18:1469467. [PMID: 40103584 PMCID: PMC11913853 DOI: 10.3389/fnmol.2025.1469467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025] Open
Abstract
Externalizing and internalizing behavioral tendencies underlie many psychiatric and substance use disorders. These tendencies are associated with differences in temperament that emerge early in development via the interplay of genetic and environmental factors. To better understand the neurobiology of temperament, we have selectively bred rats for generations to produce two lines with highly divergent behavior: bred Low Responders (bLRs) are highly inhibited and anxious in novel environments, whereas bred High Responders (bHRs) are highly exploratory, sensation-seeking, and prone to drug-seeking behavior. Recently, we delineated these heritable differences by intercrossing bHRs and bLRs (F0-F1-F2) to produce a heterogeneous F2 sample with well-characterized lineage and behavior (exploratory locomotion, anxiety-like behavior, Pavlovian conditioning). The identified genetic loci encompassed variants that could influence behavior via many mechanisms, including proximal effects on gene expression. Here we measured gene expression in male and female F0s (n = 12 bHRs, 12 bLRs) and in a large sample of heterogeneous F2s (n = 250) using hippocampal RNA-Seq. This enabled triangulation of behavior with both genetic and functional genomic data to implicate specific genes and biological pathways. Our results show that bHR/bLR differential gene expression is robust, surpassing sex differences in expression, and predicts expression associated with F2 behavior. In F0 and F2 samples, gene sets related to growth/proliferation are upregulated with bHR-like behavior, whereas gene sets related to mitochondrial function, oxidative stress, and microglial activation are upregulated with bLR-like behavior. Integrating our F2 RNA-Seq data with previously-collected whole genome sequencing data identified genes with hippocampal expression correlated with proximal genetic variation (cis-expression quantitative trait loci or cis-eQTLs). These cis-eQTLs successfully predict bHR/bLR differential gene expression based on F0 genotype. Sixteen of these genes are associated with cis-eQTLs colocalized within loci we previously linked to behavior and are strong candidates for mediating the influence of genetic variation on behavioral temperament. Eight of these genes are related to bioenergetics. Convergence between our study and others targeting similar behavioral traits revealed five more genes consistently related to temperament. Overall, our results implicate hippocampal bioenergetic regulation of oxidative stress, microglial activation, and growth-related processes in shaping behavioral temperament, thereby modulating vulnerability to psychiatric and addictive disorders.
Collapse
Affiliation(s)
- Elaine K Hebda-Bauer
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Megan H Hagenauer
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Daniel B Munro
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
- Seattle Children's Research Institute, University of Washington, Seattle, WA, United States
| | - Peter Blandino
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Fan Meng
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Keiko Arakawa
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - John D H Stead
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Apurva S Chitre
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - A Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Pejman Mohammadi
- Seattle Children's Research Institute, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Stanley J Watson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Shelly B Flagel
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Jun Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, United States
| | - Huda Akil
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
28
|
Wei X, Li S, Yue W, Zhang Z, Wei J, Wang L, Hu B, Long N, Li C, Hou B, Feng J, Luo C. The mediating effects of depression in sedentary behavior and the metabolic syndrome with its components. BMC Public Health 2025; 25:851. [PMID: 40033385 PMCID: PMC11877762 DOI: 10.1186/s12889-025-22030-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Previous studies have shown a positive association between sedentary behavior (SB) and metabolic syndrome (MetS), but no studies have assessed the mediating effect of depressive symptoms in this process. METHODS Participants from the 2007-2018 National Health and Nutrition Examination Survey (NHANES) were included. Multiple logistic regression analyses and restricted cubic splines (RCSs) were adopted to assess the correlations among SB duration, depressed mood and MetS. A mediation effect model was constructed to analyze whether there was a mediating effect of depressed mood on the relationship between SB duration and MetS. RESULTS This study included 15,944 adults (7,268 patients with MetS in total). We identified high SB duration as an independent risk factor for MetS in a regression model adjusted for relevant confounders (odds ratio (OR) = 1.29, 95% CI [1.08, 1.53]). RCS analysis revealed a nonlinear relationship between SB duration and MetS. The mediation effect analysis revealed that depressive symptoms accounted for 6.70% of the mediation effect between SB duration and MetS. Depressive symptoms were also partially mediated in the analyses with MetS subcomponents. CONCLUSION SB is closely associated with MetS, and this association is not only caused by SB itself but also partially mediated by depressive symptoms.
Collapse
Affiliation(s)
- Xing Wei
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China
- The Fifth Clinical School of Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Siqi Li
- The Fifth Clinical School of Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
- Department of Endocrinology, The Second People's Hospital of Hefei, Hefei, 230011, China
| | - Wenwen Yue
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China
- The Fifth Clinical School of Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Zhipeng Zhang
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China
- The Fifth Clinical School of Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Jing Wei
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China
- The Fifth Clinical School of Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Liqi Wang
- The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China
| | - Ben Hu
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China
- The Fifth Clinical School of Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Nv Long
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China
- The Fifth Clinical School of Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Chao Li
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China
| | - Bingmei Hou
- The Fifth Clinical School of Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
- Department of Endocrinology, The Second People's Hospital of Hefei, Hefei, 230011, China
| | - Jun Feng
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China
| | - Chunmiao Luo
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, China.
| |
Collapse
|
29
|
Mucci F, Arone A, Gurrieri R, Weiss F, Russomanno G, Marazziti D. Third-Generation Antipsychotics: The Quest for the Key to Neurotrophism. Life (Basel) 2025; 15:391. [PMID: 40141736 PMCID: PMC11944073 DOI: 10.3390/life15030391] [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: 12/27/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Antipsychotic drugs (APs) have profoundly changed the treatment landscape for psychiatric disorders, yet their impact on neuroplasticity and neurotrophism remains only partially understood. While second-generation antipsychotics (SGAs) are associated with a better side effect profile than their predecessors, the emergence of third-generation antipsychotics (TGAs)-such as brexpiprazole, cariprazine, lurasidone, iloperidone, lumateperone, pimavanserin, and roluperidone-has prompted renewed interest in their potential neuroprotective and pro-cognitive effects. This review attempts to carefully examine the evidence on the neurotrophic properties of TGAs and their role in modulating brain plasticity by analyzing studies published between 2010 and 2024. Although data remain limited and focused primarily on earlier SGAs, emerging findings suggest that some TGAs may exert positive effects on neuroplastic processes, including the modulation of brain-derived neurotrophic factors (BDNFs) and synaptic architecture. However, robust clinical data on their long-term effects and comparative efficacy are lacking; therefore, further research is necessary to validate their role in preventing neurodegenerative changes and improving cognitive outcomes in patients with psychiatric conditions.
Collapse
Affiliation(s)
- Federico Mucci
- Department of Psychiatry, Lucca Zone, Azienda USL Toscana Nord Ovest, 55100 Lucca, Italy;
| | - Alessandro Arone
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Riccardo Gurrieri
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Francesco Weiss
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Gerardo Russomanno
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| |
Collapse
|
30
|
Schwefel MK, Kaufmann C, Gutmann G, Henze R, Fydrich T, Rapp MA, Ströhle A, Heissel A, Heinzel S. Effect of physical exercise training on neural activity during working memory in major depressive disorder. J Affect Disord 2025; 372:269-278. [PMID: 39638060 DOI: 10.1016/j.jad.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Deficits in working memory (WM) are common in patients with Major Depression Disorder (MDD). Previous research mainly in healthy adults indicated that physical exercise training may improve cognitive functions by stimulating neuronal plasticity particularly in hippocampal structures. Thus, the goal of this functional Magnetic Resonance Imaging (fMRI) study was to examine alterations in neuronal activity during a WM task and to investigate changes in brain volume and functioning following a physical exercise training in patients with MDD with a specific focus on hippocampal structures. METHODS 86 (39 female) MDD outpatients (average age 37.3), diagnosed by clinical psychologists, were randomly assigned to one of three groups for a 12-week intervention: High intensity exercise training (HEX), low intensity exercise training (LEX) or waiting list control group (WL). An n-back task (with WM loads of 0, 1, 2, and 3) during fMRI was conducted before and after interventions/waiting period. RESULTS Both exercise groups showed better performance and shorter reaction times at higher WM loads after 12-weeks of physical exercise training. Specifically in the HEX, we found an improvement in physical fitness and an increase in neural activation in the left hippocampus as compared to the WL following the exercise training. Training-related structural volume changes in gray matter or hippocampus were not detected. CONCLUSIONS Our results partly support the hypothesis that physical exercise training positively affects WM functions by improving neuronal plasticity in hippocampal regions. Exercise training seems to be a promising intervention to improve deficient WM performance in patients with MDD. CLINICAL TRIALS REGISTRATION NAME Neurobiological correlates and mechanisms of the augmentation of psychotherapy with endurance exercise in mild to moderate depression - SPeED, http://apps.who.int/trialsearch/Trial2.aspx?TrialID=DRKS00008869, DRKS00008869.
Collapse
Affiliation(s)
- M K Schwefel
- Clinical Psychology and Psychotherapy, Department of Education and Psychology, Freie Universität Berlin, Berlin, Germany.
| | - C Kaufmann
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - G Gutmann
- Clinical Psychology and Psychotherapy, Department of Education and Psychology, Freie Universität Berlin, Berlin, Germany
| | - R Henze
- Clinical Psychology and Psychotherapy, Department of Education and Psychology, Freie Universität Berlin, Berlin, Germany; Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - T Fydrich
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - M A Rapp
- Social and Preventive Medicine, University of Potsdam, Potsdam, Germany
| | - A Ströhle
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - A Heissel
- Social and Preventive Medicine, University of Potsdam, Potsdam, Germany
| | - S Heinzel
- Clinical Psychology and Psychotherapy, Department of Education and Psychology, Freie Universität Berlin, Berlin, Germany; Institute of Psychology, Department of Educational Sciences and Psychology, TU Dortmund University, Dortmund, Germany
| |
Collapse
|
31
|
Hanson T, Spencer S, Harker SA, Barry F, Burton P, Beauchemin J, Mennenga SE, Braden BB, D'Sa V, Koinis-Mitchell D, Deoni SC, Lewis CR. Peripheral DNA Methylation of Cortisol- and Serotonin-Related Genes Predicts Hippocampal Volume in a Pediatric Population. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100421. [PMID: 39867566 PMCID: PMC11758844 DOI: 10.1016/j.bpsgos.2024.100421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/28/2025] Open
Abstract
Background Hippocampal volume increases throughout early development and is an important indicator of cognitive abilities and mental health. However, hippocampal development is highly vulnerable to exposures during development, as seen by smaller hippocampal volume and differential epigenetic programming in genes implicated in mental health. However, few studies have investigated hippocampal volume in relation to the peripheral epigenome across development, and even less is known about potential genetic moderators. Therefore, in this study, we explored relationships between hippocampal volume and peripheral DNA methylation of mental health-related genes, specifically NR3C1, FKBP5, and SLC6A4, throughout early development and whether these associations were moderated by age or genotype. Methods Bilateral hippocampal volume was computed from T2-weighted images through FreeSurfer, and DNA methylation was measured from saliva using the Illumina MethylationEPIC microarray in a pediatric population (N = 248, females = 112, meanage = 5.13 years, SDage = 3.60 years). Results Multiple linear regression and bootstrapping analyses revealed that DNA methylation of NR3C1, FKBP5, and SLC6A4 was associated with hippocampal volume and that these relationships were moderated by age and gene-specific variants. Conclusions These findings support the validity of peripheral DNA methylation profiles for indirectly assessing hippocampal volume and development and underscore the importance of genotype and age considerations in research. Therefore, peripheral epigenetic profiles may be a promising avenue for investigating the impacts of early-life stress on brain structure and subsequent mental health outcomes.
Collapse
Affiliation(s)
- Taena Hanson
- Department of Psychology, Arizona State University, Tempe, Arizona
| | - Sophia Spencer
- Department of Psychology, Arizona State University, Tempe, Arizona
| | | | - Fatoumata Barry
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Phoebe Burton
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | | | | | - B. Blair Braden
- College of Health Solutions, Arizona State University, Tempe, Arizona
| | - Viren D'Sa
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
| | - Daphne Koinis-Mitchell
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
| | - Sean C.L. Deoni
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
- Advanced Baby Imaging Laboratory, Rhode Island Hospital, Providence, Rhode Island
| | - Candace R. Lewis
- Department of Psychology, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Neurogenomics, Translational Genomics Research Institute, Phoenix, Arizona
| |
Collapse
|
32
|
Yang Q, Wang J, Lu Y. The impact of aerobic exercise on depression in young people: A meta regression and meta-analysis. PSYCHOLOGY OF SPORT AND EXERCISE 2025; 77:102803. [PMID: 39800178 DOI: 10.1016/j.psychsport.2025.102803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 12/22/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
OBJECTIVE This systematic review aimed to 1) update the existing evidence on the antidepressant effects of aerobic exercise (AE) in youth. 2) Investigate any potential dose‒response relationships between AE interventions and depressive symptom reduction. 3) Provide evidence-based insights to inform future research and clinical depression treatment. METHODS Employing the PRISMA and PERSiST guidelines, a comprehensive search across nine databases (Web of Science, PubMed, Scopus, PsycINFO, SportDiscus, CINAHL, Medline, Embase, and CNKI) yielded a total of 782 relevant studies. Following rigorous selection criteria, 26 eligible studies (comprising 22 different samples) were included in the analysis, featuring a combined sample size of 1308 participants. The meta-analysis was conducted via R. RESULTS AE notably decreased depressive symptoms among youth [g = -0.92; 95% CI (-1.16, -0.69); p < 0.01]. Significant dose‒response relationships were observed across age groups [β = -0.06; 95% CI (-0.12, 0.00), p < 0.05], the intensity [β = -0.03; 95% CI (-0.06, 0.00); p < 0.05], and the duration ^ 2 [β = 0.001, 95% CI (0.001, 0.002), p < 0.05] of the AE intervention. CONCLUSIONS Aerobic exercise interventions can effectively alleviate depressive symptoms in youth. A moderate-intensity AE program, lasting 25-40 min and conducted three times a week for 9-15 weeks, can increase alleviation of depression.
Collapse
Affiliation(s)
- Qiushi Yang
- School of Physical Education, China University of Mining and Technology, Xuzhou, Jiangsu Province, 221116, China.
| | - Junli Wang
- School of Physical Education, China University of Mining and Technology, Xuzhou, Jiangsu Province, 221116, China.
| | - Yiyaochen Lu
- School of Physical Education, China University of Mining and Technology, Xuzhou, Jiangsu Province, 221116, China
| |
Collapse
|
33
|
Manelis A, Hu H, Miceli R, Satz S, Lau R, Iyengar S, Swartz HA. Lateral Ventricular Enlargement and Asymmetry and Myelin Content Imbalance in Individuals With Bipolar and Depressive Disorders: Clinical and Research Implications. Bipolar Disord 2025; 27:119-131. [PMID: 39981613 PMCID: PMC11950717 DOI: 10.1111/bdi.70012] [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: 07/23/2024] [Revised: 12/08/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND The link between ventricular enlargement and asymmetry with other indices of brain structure remains underexplored in individuals with bipolar (BD) and depressive (DD) disorders. Our study compared the lateral ventricular size, ventricular asymmetry, and cortical myelin content in individuals with BD versus those with DD versus healthy controls (HC). METHODS We obtained T1w and T2w images from 149 individuals (age = 27.7 (SD = 6.1) years, 78% female, BD = 38, DD = 57, HC = 54) using Magnetic Resonance Imaging (MRI). The BD group consisted of individuals with BD Type I (n = 11) and BD Type II (n = 27), while the DD group consisted of individuals with major depressive disorder (MDD, n = 38) and persistent depressive disorder (PDD, n = 19) Cortical myelin content was calculated using the T1w/T2w ratio. Elastic net regularized regression identified brain regions whose myelin content was associated with ventricular size and asymmetry. A post hoc linear regression examined how participants' diagnosis, illness duration, and current level of depression moderated the relationship between the size and asymmetry of the lateral ventricles and levels of cortical myelin in the selected brain regions. RESULTS Individuals with BD and DD had larger lateral ventricles than HC. Larger ventricles and lower asymmetry were observed in individuals with BD who had longer lifetime illness duration and more severe current depressive symptoms. A greater left asymmetry was observed in participants with DD than in those with BD (p < 0.01). Elastic net revealed that both ventricular enlargement and asymmetry were associated with altered myelin content in cingulate, frontal, and sensorimotor cortices. In BD, but not in other groups, ventricular enlargement was related to altered myelin content in the right insular regions. CONCLUSIONS Lateral ventricular enlargement and asymmetry are linked to myelin content imbalance, thus potentially leading to emotional and cognitive dysfunction in mood disorders.
Collapse
Affiliation(s)
- Anna Manelis
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Hang Hu
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rachel Miceli
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Skye Satz
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rachel Lau
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Satish Iyengar
- Department of StatisticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Holly A. Swartz
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
34
|
Corsi-Zuelli F, Donohoe G, Griffiths SL, Del-Ben CM, Watson AJ, Burke T, Lalousis PA, McKernan D, Morris D, Kelly J, McDonald C, Patlola SR, Pariante C, Barnes NM, Khandaker GM, Suckling J, Deakin B, Upthegrove R, Dauvermann MR. Depressive and Negative Symptoms in the Early and Established Stages of Schizophrenia: Integrating Structural Brain Alterations, Cognitive Performance, and Plasma Interleukin 6 Levels. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100429. [PMID: 39911538 PMCID: PMC11795630 DOI: 10.1016/j.bpsgos.2024.100429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 02/07/2025] Open
Abstract
Background Depressive and negative symptoms are related to poor functional outcomes in schizophrenia. Cognitive deficits, reduced brain cortical thickness and volumes, and inflammation may contribute to depressive and negative symptoms, but pharmacological treatment and disease progression may confound the associations. Methods We evaluated whether higher plasma interleukin 6 (IL-6) levels would be associated with more severe negative or depressive symptoms in schizophrenia and explored illness stage utilizing early (BeneMin [Benefit of Minocycline on Negative Symptoms of Psychosis: Extent and Mechanism], n = 201, 72.8% male) and established (iRELATE [Immune Response & Social Cognition in Schizophrenia], n = 94, 67.3% male) schizophrenia cohorts. Using structural equation modeling in a subsample (iRELATE: n = 42, 69.0% male; BeneMin: n = 102, 76.5% male) with data on structural brain metrics (cortical thickness and volume), general cognitive performance, and plasma IL-6 levels, we assessed the interrelationships between these variables on depressive and negative symptom severity in early and established schizophrenia samples combined and in early schizophrenia only. All analyses were adjusted for sex, age, and chlorpromazine equivalent dose. Results Higher plasma IL-6 levels were related to more severe depressive symptoms in early schizophrenia (p < .05) and negative symptoms in established schizophrenia (p < .05). Structural equation modeling findings in early and established schizophrenia samples combined and early schizophrenia only showed that the interrelationship between higher plasma IL-6 levels, structural brain metrics, and general cognitive performance did not predict the severity of depressive and negative symptoms (p > .05). Higher plasma IL-6 levels and lower general cognitive performance were associated with reduced brain metrics (p < .05). Conclusions Our results indicate that higher plasma IL-6 levels may be differently associated with the severity of depressive and negative symptoms dependent on the illness stage. Future work identifying elevated levels of inflammation in larger samples may allow stratification and personalized intervention by subgroups who are at risk of poor outcomes.
Collapse
Affiliation(s)
- Fabiana Corsi-Zuelli
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
- Department of Neurosciences and Behaviour, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Gary Donohoe
- Centre for Neuroimaging, Cognition and Genomics, School of Psychology, University of Galway, Galway, Ireland
| | - Siân Lowri Griffiths
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Cristina M. Del-Ben
- Department of Neurosciences and Behaviour, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Andrew J. Watson
- Department of Clinical and Motor Neuroscience, University College London, Queen Square Institute of Neurology, London, United Kingdom
| | - Tom Burke
- Centre for Neuroimaging, Cognition and Genomics, School of Psychology, University of Galway, Galway, Ireland
| | - Paris A. Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Declan McKernan
- Pharmacology & Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Derek Morris
- Centre for Neuroimaging, Cognition and Genomics, School of Psychology, University of Galway, Galway, Ireland
| | - John Kelly
- Pharmacology & Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Colm McDonald
- Centre for Neuroimaging, Cognition and Genomics, School of Psychology, University of Galway, Galway, Ireland
| | - Saahithh R. Patlola
- Pharmacology & Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Carmine Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Nicholas M. Barnes
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Golam M. Khandaker
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - John Suckling
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, United Kingdom
| | - Bill Deakin
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of the Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Rachel Upthegrove
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Maria R. Dauvermann
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
35
|
Sun F, Shuai Y, Wang J, Yan J, Lin B, Li X, Zhao Z. Hippocampal gray matter volume alterations in patients with first-episode and recurrent major depressive disorder and their associations with gene profiles. BMC Psychiatry 2025; 25:134. [PMID: 39955494 PMCID: PMC11829352 DOI: 10.1186/s12888-025-06562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Recent studies indicate that patients with first-episode drug-naïve (FEDN) and recurrent major depressive disorder (R-MDD) exhibit distinct atrophy patterns in the hippocampal subregions along the proximal-distal axis. However, it remains unclear whether such differences occur along the long axis and how they may relate to specific genes. METHODS In the present study, we analyzed T1-weighted images from 421 patients (FEDN: n = 232; R-MDD: n = 189) and 544 normal controls (NC) as part of the REST-meta-MDD consortium. Additionally, transcriptome maps and structural Magnetic Resonance Imaging (MRI) data of six donated brains were obtained from the Allen Human Brain Atlas (AHBA). We first identified changes in gray matter volume (GMV) within the hippocampus of both FEDN and R-MDD patients and then integrated these findings with AHBA transcriptome data to investigate the genes associated with hippocampal GMV changes. RESULTS Compared to NC, FEDN patients displayed reduced GMV in the left hippocampal tail, whereas R-MDD patients exhibited decreased GMV in the bilateral hippocampal body and increased GMV in the bilateral hippocampal tail. Further analysis revealed that expression levels of SYTL2 positively correlated with GMV changes in the hippocampus of FEDN patients, while SORCS3 and SLIT2 positively correlated with those in R-MDD. CONCLUSIONS Our results suggest that GMV alterations in hippocampal subfields along the long axis differ between FEDN and R-MDD, reflecting progressive hippocampal deterioration with prolonged depression, potentially supported by the expression of specific genes. These findings offer valuable insights into the distinct neural and genetic mechanisms underlying FEDN and R-MDD, which may aid in the development of more targeted and effective treatment strategies for MDD subtypes.
Collapse
Affiliation(s)
- Fenfen Sun
- Center for Brain, Mind and Education, Shaoxing University, Shaoxing, China
- Department of Psychology, Shaoxing University, Shaoxing, China
| | - Yifan Shuai
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Jingru Wang
- Department of Psychology, Shaoxing University, Shaoxing, China
| | - Jin Yan
- Department of Psychology, Shaoxing University, Shaoxing, China
| | - Bin Lin
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyun Li
- School of Rehabilitation, Hangzhou Medical College, Hangzhou, China
| | - Zhiyong Zhao
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Binjiang Campus, 3333 Binsheng Rd, Hangzhou, China.
| |
Collapse
|
36
|
Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium. Trans-ancestry genome-wide study of depression identifies 697 associations implicating cell types and pharmacotherapies. Cell 2025; 188:640-652.e9. [PMID: 39814019 PMCID: PMC11829167 DOI: 10.1016/j.cell.2024.12.002] [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: 04/22/2023] [Revised: 06/17/2024] [Accepted: 12/05/2024] [Indexed: 01/18/2025]
Abstract
In a genome-wide association study (GWAS) meta-analysis of 688,808 individuals with major depression (MD) and 4,364,225 controls from 29 countries across diverse and admixed ancestries, we identify 697 associations at 635 loci, 293 of which are novel. Using fine-mapping and functional tools, we find 308 high-confidence gene associations and enrichment of postsynaptic density and receptor clustering. A neural cell-type enrichment analysis utilizing single-cell data implicates excitatory, inhibitory, and medium spiny neurons and the involvement of amygdala neurons in both mouse and human single-cell analyses. The associations are enriched for antidepressant targets and provide potential repurposing opportunities. Polygenic scores trained using European or multi-ancestry data predicted MD status across all ancestries, explaining up to 5.8% of MD liability variance in Europeans. These findings advance our global understanding of MD and reveal biological targets that may be used to target and develop pharmacotherapies addressing the unmet need for effective treatment.
Collapse
|
37
|
Takamiya A, Radwan A, Christiaens D, Van Cauwenberge M, Vande Casteele T, Laroy M, Vansteelandt K, Sunaert S, Koole M, Van den Stock J, Van Laere K, Bouckaert F, Vandenbulcke M, Emsell L. Gray and white matter differences in the medial temporal lobe in late-life depression: a multimodal PET-MRI investigation. Psychol Med 2025; 55:e10. [PMID: 39901804 PMCID: PMC11968119 DOI: 10.1017/s0033291724003362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 02/05/2025]
Abstract
BACKGROUND Late-life depression (LLD) is characterized by medial temporal lobe (MTL) abnormalities. Although gray matter (GM) and white matter (WM) differences in LLD have been reported, few studies have investigated them concurrently. Moreover, the impact of aetiological factors, such as neurodegenerative and cerebrovascular burden, on tissue differences remains elusive. METHODS This prospective cross-sectional study involved 72 participants, including 33 patients with LLD (mean age 72.2 years, 23 female) and 39 healthy controls (HCs) (mean age 70.6 years, 24 female), who underwent clinical and positron emission tomography (PET)-magnetic resonance imaging (MRI) assessments. High-resolution 3D T1-weighted and T2-weighted FLAIR images were used to assess MTL GM volumes and white matter hyperintensities (WMHs), a proxy for cerebrovascular burden. Diffusion kurtosis imaging metrics derived from multishell diffusion MRI data were analyzed to assess WM microstructure in the following MTL bundles reconstructed using constrained spherical deconvolution tractography: uncinate fasciculus, fornix, and cingulum. Standardized uptake value ratio of 18F-MK-6240 in the MTL was used to assess Alzheimer's disease (AD) type tau accumulation as a proxy for neurodegenerative burden. RESULTS Compared to HCs, patients with LLD showed significantly lower bilateral MTL volumes and WM microstructural differences primarily in the uncinate fasciculi bilaterally and right fornix. In patients with LLD, higher vascular burden, but not tau, was associated with lower MTL volume and more pronounced WM differences. CONCLUSIONS LLD was associated with both GM and WM differences in the MTL. Cerebrovascular disease, rather than AD type tau-mediated neurodegenerative processes, may contribute to brain tissue differences in LLD.
Collapse
Affiliation(s)
- Akihiro Takamiya
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Hills Joint Research Laboratory for Future Preventive Medicine and Wellness, Keio University School of Medicine, TokyoJapan
| | - Ahmed Radwan
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
| | - Daan Christiaens
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Electrical Engineering, EST-PSI, KU Leuven, Leuven, Belgium
| | - Margot Van Cauwenberge
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Thomas Vande Casteele
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Maarten Laroy
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Kristof Vansteelandt
- Department of Neurosciences, Research Group Psychiatry, Neuropsychiatry, Academic Center for ECT and Neuromodulation (AcCENT), University Psychiatric Center KU Leuven, Kortenberg, Belgium
| | - Stefan Sunaert
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Michel Koole
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
| | - Jan Van den Stock
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
- Department of Nuclear Medicine and Molecular Imaging, University Hospitals Leuven, Leuven, Belgium
| | - Filip Bouckaert
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Louise Emsell
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Bravi B, Paolini M, Maccario M, Milano C, Raffaelli L, Melloni EMT, Zanardi R, Colombo C, Benedetti F. Abnormal choroid plexus, hippocampus, and lateral ventricles volumes as markers of treatment-resistant major depressive disorder. Psychiatry Clin Neurosci 2025; 79:69-77. [PMID: 39563010 PMCID: PMC11789456 DOI: 10.1111/pcn.13764] [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: 06/10/2024] [Revised: 10/09/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024]
Abstract
AIM One-third of patients with major depressive disorder (MDD) do not achieve full remission and have high relapse rates even after treatment, leading to increased medical costs and reduced quality of life and health status. The possible specificity of treatment-resistant depression (TRD) neurobiology is still under investigation, with risk factors such as higher inflammatory markers being identified. Given recent findings on the role of choroid plexus (ChP) in neuroinflammation and hippocampus in treatment response, the aim of the present study was to evaluate inflammatory- and trophic-related differences in these regions along with ventricular volumes among patients with treatment-sensitive depression (TSD), TRD, and healthy controls (HCs). METHODS ChP, hippocampal, and ventricular volumes were assessed in 197 patients with MDD and 58 age- and sex-matched HCs. Volumes were estimated using FreeSurfer 7.2. Treatment resistance status was defined as failure to respond to at least two separate antidepressant treatments. Region of interest volumes were then compared among groups. RESULTS We found higher ChP volumes in patients with TRD compared with patients with TSD and HCs. Our results also showed lower hippocampal volumes and higher lateral ventricular volumes in TRD compared with both patients without TRD and HCs. CONCLUSIONS These findings corroborate the link between TRD and neuroinflammation, as ChP volume could be considered a putative marker of central immune activity. The lack of significant differences in all of the region of interest volumes between patients with TSD and HCs may highlight the specificity of these features to TRD, possibly providing new insights into the specific neurobiological underpinnings of this condition.
Collapse
Affiliation(s)
- Beatrice Bravi
- Psychiatry & Clinical Psychobiology, Division of NeuroscienceIRCCS San Raffaele HospitalMilanItaly
- University Vita‐Salute San RaffaeleMilanItaly
| | - Marco Paolini
- Psychiatry & Clinical Psychobiology, Division of NeuroscienceIRCCS San Raffaele HospitalMilanItaly
| | - Melania Maccario
- University Vita‐Salute San RaffaeleMilanItaly
- Mood Disorders UnitIRCCS San Raffaele HospitalMilanItaly
| | - Chiara Milano
- Psychiatry & Clinical Psychobiology, Division of NeuroscienceIRCCS San Raffaele HospitalMilanItaly
| | - Laura Raffaelli
- Psychiatry & Clinical Psychobiology, Division of NeuroscienceIRCCS San Raffaele HospitalMilanItaly
- University Vita‐Salute San RaffaeleMilanItaly
| | | | - Raffaella Zanardi
- University Vita‐Salute San RaffaeleMilanItaly
- Mood Disorders UnitIRCCS San Raffaele HospitalMilanItaly
| | - Cristina Colombo
- University Vita‐Salute San RaffaeleMilanItaly
- Mood Disorders UnitIRCCS San Raffaele HospitalMilanItaly
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology, Division of NeuroscienceIRCCS San Raffaele HospitalMilanItaly
- University Vita‐Salute San RaffaeleMilanItaly
| |
Collapse
|
39
|
Sarkis RA, Orozco J, Lemus HN, Hankerson A, Liu L, Lam AD, Johnson E, Stufflebeam S, Viswanathan A, Amariglio RE, Purandare M, Trouten P, Young GS, Locascio JJ, Pennell PB, Marshall GA. Late-onset unexplained seizures are associated with cognitive impairment and lower amygdala volumes. Brain Commun 2025; 7:fcaf050. [PMID: 39944741 PMCID: PMC11815171 DOI: 10.1093/braincomms/fcaf050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Late-onset epilepsy has been linked with accelerated cognitive decline and a higher risk of dementia. In this study, we sought to characterize the cognitive profile of participants with late-onset unexplained epilepsy and compare their MRI findings to healthy controls, to better understand underlying disease mechanisms. We recruited participants with at least one new-onset unexplained seizure at age 55 or later, without cortical lesions on MRI, within 5 years of the first seizure. We administered a neuropsychological battery to generate Preclinical Alzheimer Cognitive Composite and composite scores for delayed verbal recall, processing speed and executive function. We held a consensus meeting to determine whether the participants fulfilled criteria for mild cognitive impairment. An MRI volumetric analysis of hippocampal, amygdalae, and white matter hyperintensity volume was performed and compared to 353 healthy controls from the Harvard Aging Brain Study. On late-onset unexplained epilepsy participants, we also obtained 24-h EEG recording. Seventy participants were recruited, mean age 71.0 ± 7.0 years, 49% female, 15.6 ± 3.0 years of education. Impaired cognition (z-score ≤ -1.5) for late-onset unexplained epilepsy included the following: 15.9% for Preclinical Alzheimer Cognitive Composite -5, 23.2% for delayed verbal recall, 15.6% for processing speed and 7.5% for executive function. Seventeen percent were found to have mild cognitive impairment. Late-onset unexplained epilepsy participants who were drug resistant were more likely to have cognitive impairment (50% vs. 9%). When controlling for age, sex and race, late-onset unexplained epilepsy group had lower left AV (%; β = -0.003, P = 0.0016), right AV (%) (β = -0.003, P = 0.01), and log-transformed WMV (mm3; β = -0.21, P = 0.03) compared with Harvard Aging Brain Study (HABS); there were no differences in left or right HV between groups. EEG captured epileptiform abnormalities in 49% late-onset unexplained epilepsy participants, with a left temporal predominance (54%). In this single-site study of prospectively enrolled participants with late-onset unexplained epilepsy, we show that individuals with late-onset unexplained epilepsy exhibit cognitive impairments, mostly in verbal memory, and temporal dysfunction with left-sided predominance. Neuroimaging, when compared with healthy controls, shows lower amygdalae and white matter hyperintensity but not hippocampal volumes suggesting that the amygdalae is one of the earliest sites involved in the disease. The results also highlight the importance of seizure control given the association between mild cognitive impairment and drug-resistant epilepsy. Future studies extending these findings to Alzheimer's disease biomarkers and longitudinal follow-up will inform predictors of cognitive decline.
Collapse
Affiliation(s)
- Rani A Sarkis
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Janet Orozco
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hernan Nicolas Lemus
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alexis Hankerson
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lei Liu
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alice D Lam
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Emily Johnson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MA 21205, USA
| | - Steven Stufflebeam
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Harvard-MIT Health Science and Technology, Massachusetts Institute of Technology, Boston, MA 02139, USA
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Rebecca E Amariglio
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Mallika Purandare
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Trouten
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Geoffrey S Young
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph J Locascio
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Page B Pennell
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Gad A Marshall
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
40
|
Goya-Maldonado R, Erwin-Grabner T, Zeng LL, Ching CRK, Aleman A, Amod AR, Basgoze Z, Benedetti F, Besteher B, Brosch K, Bülow R, Colle R, Connolly CG, Corruble E, Couvy-Duchesne B, Cullen K, Dannlowski U, Davey CG, Dols A, Ernsting J, Evans JW, Fisch L, Fuentes-Claramonte P, Gonul AS, Gotlib IH, Grabe HJ, Groenewold NA, Grotegerd D, Hahn T, Hamilton JP, Han LKM, Harrison BJ, Ho TC, Jahanshad N, Jamieson AJ, Karuk A, Kircher T, Klimes-Dougan B, Koopowitz SM, Lancaster T, Leenings R, Li M, Linden DEJ, MacMaster FP, Mehler DMA, Meinert S, Melloni E, Mueller BA, Mwangi B, Nenadić I, Ojha A, Okamoto Y, Oudega ML, Penninx BWJH, Poletti S, Pomarol-Clotet E, Portella MJ, Pozzi E, Radua J, Rodríguez-Cano E, Sacchet MD, Salvador R, Schrantee A, Sim K, Soares JC, Solanes A, Stein DJ, Stein F, Stolicyn A, Thomopoulos SI, Toenders YJ, Uyar-Demir A, Vieta E, Vives-Gilabert Y, Völzke H, Walter M, Whalley HC, Whittle S, Winter N, Wittfeld K, Wright MJ, Wu MJ, Yang TT, Zarate C, Veltman DJ, Schmaal L, Thompson PM, for the ENIGMA Major Depressive Disorder working group. Classification of Major Depressive Disorder Using Vertex-Wise Brain Sulcal Depth, Curvature, and Thickness with a Deep and a Shallow Learning Model. ARXIV 2025:arXiv:2311.11046v2. [PMID: 39975425 PMCID: PMC11838705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Major depressive disorder (MDD) is a complex psychiatric disorder that affects the lives of hundreds of millions of individuals around the globe. Even today, researchers debate if morphological alterations in the brain are linked to MDD, likely due to the heterogeneity of this disorder. The application of deep learning tools to neuroimaging data, capable of capturing complex non-linear patterns, has the potential to provide diagnostic and predictive biomarkers for MDD. However, previous attempts to demarcate MDD patients and healthy controls (HC) based on segmented cortical features via linear machine learning approaches have reported low accuracies. In this study, we used globally representative data from the ENIGMA-MDD working group containing 7,012 participants from 30 sites (N=2,772 MDD and N=4,240 HC), which allows a comprehensive analysis with generalizable results. Based on the hypothesis that integration of vertex-wise cortical features can improve classification performance, we evaluated the classification of a DenseNet and a Support Vector Machine (SVM), with the expectation that the former would outperform the latter. As we analyzed a multi-site sample, we additionally applied the ComBat harmonization tool to remove potential nuisance effects of site. We found that both classifiers exhibited close to chance performance (balanced accuracy DenseNet: 51%; SVM: 53%), when estimated on unseen sites. Slightly higher classification performance (balanced accuracy DenseNet: 58%; SVM: 55%) was found when the cross-validation folds contained subjects from all sites, indicating site effect. In conclusion, the integration of vertex-wise morphometric features and the use of the non-linear classifier did not lead to the differentiability between MDD and HC. Our results support the notion that MDD classification on this combination of features and classifiers is unfeasible. Future studies are needed to determine whether more sophisticated integration of information from other MRI modalities such as fMRI and DWI will lead to a higher performance in this diagnostic task.
Collapse
Affiliation(s)
- Roberto Goya-Maldonado
- Laboratory of Systems Neuroscience and Imaging in Psychiatry (SNIP-Lab), Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August University, Göttingen, Germany
| | - Tracy Erwin-Grabner
- Laboratory of Systems Neuroscience and Imaging in Psychiatry (SNIP-Lab), Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August University, Göttingen, Germany
| | - Ling-Li Zeng
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha 410073, China
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90274, USA
| | - Christopher R. K. Ching
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90274, USA
| | - Andre Aleman
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alyssa R. Amod
- Department of Psychiatry & Mental Health, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Zeynep Basgoze
- Department of Psychiatry and Behavioral Science, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Francesco Benedetti
- Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Bianca Besteher
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Rudolf Bultmann Str. 8, 35039 Marburg, Germany
| | - Robin Bülow
- Institute for Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Romain Colle
- MOODS Team, CESP, INSERM U1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre 94275, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux deParis, Hôpital de Bicêtre, Le Kremlin Bicêtre F-94275, France
| | - Colm G. Connolly
- Department of Biomedical Sciences, Florida State University, Tallahassee FL, USA
| | - Emmanuelle Corruble
- MOODS Team, CESP, INSERM U1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre 94275, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux deParis, Hôpital de Bicêtre, Le Kremlin Bicêtre F-94275, France
| | - Baptiste Couvy-Duchesne
- Sorbonne University, Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, F-75013, Paris, France
- Institute for Molecular Bioscience, the University of Queensland, St Lucia, QLD, Australia
| | - Kathryn Cullen
- Department of Psychiatry and Behavioral Science, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Christopher G. Davey
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, the University of Melbourne, Parkville, Victoria, Australia
| | - Annemiek Dols
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
- Department of Psychiatry, UMC Utrecht Brain Center, University Utrecht, Utrecht, the Netherlands
| | - Jan Ernsting
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Jennifer W. Evans
- Experimental Therapeutics and Pathophysiology Branch, National Institute for Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Lukas Fisch
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Paola Fuentes-Claramonte
- FIDMAG Germanes Hospitalàries Research Foundation, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Catalonia, Spain
| | - Ali Saffet Gonul
- SoCAT Lab, Department of Psychiatry, School of Medicine, Ege University, Izmir, Turkey
| | - Ian H. Gotlib
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Nynke A. Groenewold
- Department of Psychiatry & Mental Health, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - J. Paul Hamilton
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Laura K. M. Han
- Centre for Youth Mental Health, the University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Ben J. Harrison
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, the University of Melbourne, Parkville, Victoria, Australia
| | - Tiffany C. Ho
- Department of Psychiatry and Behavioral Sciences, Division of Child and Adolescent Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90274, USA
| | - Alec J. Jamieson
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, the University of Melbourne, Parkville, Victoria, Australia
| | - Andriana Karuk
- FIDMAG Germanes Hospitalàries Research Foundation, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Catalonia, Spain
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Rudolf Bultmann Str. 8, 35039 Marburg, Germany
| | | | - Sheri-Michelle Koopowitz
- Department of Psychiatry & Mental Health, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Thomas Lancaster
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Ramona Leenings
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Meng Li
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - David E. J. Linden
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, 6229 ER, the Netherlands
| | - Frank P. MacMaster
- Departments of Psychiatry and Pediatrics, University of Calgary, Calgary, AB, Canada
| | - David M. A. Mehler
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical School, RWTH Aachen University, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Germany
| | - Elisa Melloni
- Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Bryon A. Mueller
- Department of Psychiatry and Behavioral Science, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Benson Mwangi
- Center Of Excellence on Mood Disorders, Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences at McGovern Medical School, the University of Texas Health Science Center at Houston, USA
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Rudolf Bultmann Str. 8, 35039 Marburg, Germany
| | - Amar Ojha
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yasumasa Okamoto
- Department of Psychiatry and Neurosciences, Hiroshima University, Hiroshima, Japan
| | - Mardien L. Oudega
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
- GGZ inGeest Mental Health Care, Amsterdam, the Netherlands
| | - Brenda W. J. H. Penninx
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Sara Poletti
- Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Catalonia, Spain
| | - Maria J. Portella
- Sant Pau Mental Health Research Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain. CIBERSAM, Madrid, Spain
| | - Elena Pozzi
- Centre for Youth Mental Health, the University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Joaquim Radua
- Imaging of Mood- and Anxiety-Related Disorders (IMARD) Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elena Rodríguez-Cano
- FIDMAG Germanes Hospitalàries Research Foundation, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Catalonia, Spain
| | - Matthew D. Sacchet
- Meditation Research Program, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research Foundation, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Catalonia, Spain
| | - Anouk Schrantee
- Amsterdam University Medical Centers, location AMC, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands
| | - Kang Sim
- West Region, Institute of Mental Health, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jair C. Soares
- Center Of Excellence on Mood Disorders, Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences at McGovern Medical School, the University of Texas Health Science Center at Houston, USA
| | - Aleix Solanes
- Imaging of Mood- and Anxiety-Related Disorders (IMARD) Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Dan J. Stein
- Department of Psychiatry & Mental Health, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Rudolf Bultmann Str. 8, 35039 Marburg, Germany
| | - Aleks Stolicyn
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Scotland, UK
| | - Sophia I. Thomopoulos
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90274, USA
| | - Yara J. Toenders
- Centre for Youth Mental Health, the University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
- Developmental and Educational Psychology, Leiden University, the Netherlands
- Erasmus School of Social and Behavioral Sciences, Erasmus University Rotterdam, the Netherlands
| | - Aslihan Uyar-Demir
- SoCAT Lab, Department of Psychiatry, School of Medicine, Ege University, Izmir, Turkey
| | - Eduard Vieta
- Hospital Clinic, Institute of Neuroscience, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Yolanda Vives-Gilabert
- Intelligent Data Analysis Laboratory (IDAL), Department of Electronic Engineering, Universitat de València, Valencia, Spain
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
- Clinical Affective Neuroimaging Laboratory, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Heather C. Whalley
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Scotland, UK
| | - Sarah Whittle
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, the University of Melbourne, Parkville, Victoria, Australia
| | - Nils Winter
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/ Greifswald, Greifswald, Germany
| | - Margaret J. Wright
- Clinical Affective Neuroimaging Laboratory, Leibniz Institute for Neurobiology, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/ Greifswald, Greifswald, Germany
| | - Mon-Ju Wu
- Center Of Excellence on Mood Disorders, Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences at McGovern Medical School, the University of Texas Health Science Center at Houston, USA
| | - Tony T. Yang
- Department of Psychiatry and Behavioral Sciences, Division of Child and Adolescent Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Carlos Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, Bethesda, MD, USA
| | - Dick J. Veltman
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Lianne Schmaal
- Centre for Youth Mental Health, the University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Paul M. Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90274, USA
| | | |
Collapse
|
41
|
Gui Y, Zhou G, Cui S, Li H, Lu H, Zhao H. The left amygdala is genetically sexually-dimorphic: multi-omics analysis of structural MRI volumes. Transl Psychiatry 2025; 15:17. [PMID: 39843917 PMCID: PMC11754786 DOI: 10.1038/s41398-025-03223-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Brain anatomy plays a key role in complex behaviors and mental disorders that are sexually divergent. While our understanding of the sex differences in the brain anatomy remains relatively limited, particularly of the underlying genetic and molecular mechanisms that contribute to these differences. We performed the largest study of sex differences in brain volumes (N = 33,208) by examining sex differences both in the raw brain volumes and after controlling the whole brain volumes. Genetic correlation analysis revealed sex differences only in the left amygdala. We compared transcriptome differences between males and females using data from GTEx and characterized cell-type compositions using GTEx bulk amygdala RNA-seq data and LIBD amygdala single-cell reference profiles. We also constructed polygenic risk scores (PRS) to investigate sex-specific genetic correlations between left amygdala volume and mental disorders (N = 25,576~105,318) of Psychiatric Genomics Consortium and other traits of UKB (N = 347,996). Although there were pronounced sex differences in brain volumes, there was no difference in the heritability between sexes. There was a significant sex-specific genetic correlation between male and female left amygdala. We identified sex-differentiated genetic effects of PRSs for schizophrenia on left amygdala volume, as well as significant sex-differentiated genetic correlations between PRSs of left amygdala and six traits in UKB. We also found several sex-differentially expressed genes in the amygdala. These findings not only advanced the current knowledge of genetic basis of sex differences in brain anatomy, but also presented an important clue for future research on the mechanism of sex differences in mental disorders and targeted treatments.
Collapse
Affiliation(s)
- Yuanyuan Gui
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Geyu Zhou
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Shuya Cui
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Hongyu Li
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Hui Lu
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Hongyu Zhao
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA.
| |
Collapse
|
42
|
See CR, Tan AX, Valmaggia LR, Kempton MJ. The association between recent stressful life events and brain structure: a UK Biobank longitudinal MRI study. Eur Psychiatry 2025; 68:e18. [PMID: 39834263 PMCID: PMC11822962 DOI: 10.1192/j.eurpsy.2025.2] [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: 11/08/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Recent stressful life events (SLEs) are an established risk factor for a range of psychiatric disorders. Animal studies have shown evidence of gray matter (GM) reductions associated with stress, and previous work has found similar associations in humans. However longitudinal studies investigating the association between stress and changes in brain structure are limited. METHODS The current study uses longitudinal data from the UK Biobank and comprises 4,543 participants with structural neuroimaging and recent SLE data (mean age = 61.5 years). We analyzed the association between recent SLEs and changes in brain structure, determined using the longitudinal FreeSurfer pipeline, focusing on total GM volume and five a priori brain regions: the hippocampus, amygdala, anterior cingulate cortex, orbitofrontal cortex, and insula. We also examined if depression and childhood adversity moderated the relationship between SLEs and brain structure. RESULTS Individuals who had experienced recent SLEs exhibited a slower rate of hippocampal decrease over time compared to individuals who did not report any SLEs. Individuals with depression exhibited smaller GM volumes when exposed to recent SLEs. There was no effect of childhood adversity on the relationship between SLEs and brain structure. CONCLUSIONS Our findings suggest recent SLEs are not directly associated with an accelerated decline in brain volumes in a population sample of older adults, but instead may alter brain structure via affective disorder psychopathology. Further work is needed to investigate the effects of stress in younger populations who may be more vulnerable to stress-induced changes, and may yet pinpoint brain regions linked to stress-related disorders.
Collapse
Affiliation(s)
- Cheryl R.Z. See
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Annabel X. Tan
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
| | - Lucia R. Valmaggia
- Centre for Youth Mental Health, University of Melbourne, Parkville, VIC, Australia
- Department of Psychology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Department of Psychiatry, KU Leuven, Leuven, Belgium
| | - Matthew J. Kempton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| |
Collapse
|
43
|
Doretto VF, Salto ABR, Schivoletto S, Zugman A, Oliveira MC, Brañas M, Croci M, Ito LT, Santoro M, Jackowski AP, Bressan RA, Rohde LA, Salum G, Miguel EC, Pan PM. Childhood maltreatment and the structural development of hippocampus across childhood and adolescence. Psychol Med 2025; 54:1-9. [PMID: 39773537 PMCID: PMC11769901 DOI: 10.1017/s0033291724001636] [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: 08/02/2023] [Revised: 03/30/2024] [Accepted: 07/08/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Prior studies suggest that childhood maltreatment is associated with altered hippocampal volume. However, longitudinal studies are currently scarce, making it difficult to determine how alterations in hippocampal volume evolve over time. The current study examined the relationship between childhood maltreatment and hippocampal volumetric development across childhood and adolescence in a community sample. METHODS In this longitudinal study, a community sample of 795 participants underwent brain magnetic resonance imaging (MRI) in three waves spanning ages 6-21 years. Childhood maltreatment was assessed using parent-report and children´s self-report at baseline (6-12 years old). Mixed models were used to examine the relationship between childhood maltreatment and hippocampal volume across time. RESULTS The quadratic term of age was significantly associated with both right and left hippocampal volume development. High exposure to childhood maltreatment was associated with reduced offset of right hippocampal volume and persistent reduced volume throughout adolescence.Critically, the relationship between childhood maltreatment and reduced right hippocampal volume remained significant after adjusting for the presence of any depressive disorder during late childhood and adolescence and hippocampal volume polygenic risk scores. Time-by-CM and Sex-by-CM interactions were not statistically significant. CONCLUSIONS The present study showed that childhood maltreatment is associated with persistent reduction of hippocampal volume in children and adolescents, even after adjusting for the presence of major depressive disorder and genetic determinants of hippocampal structure.
Collapse
Affiliation(s)
- Victoria Fogaça Doretto
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Ana Beatriz Ravagnani Salto
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Sandra Schivoletto
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Andre Zugman
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Melaine Cristina Oliveira
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marcelo Brañas
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Marcos Croci
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Lucas Toshio Ito
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marcos Santoro
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Andrea P. Jackowski
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
- Department of Education, Information and Communications Technology (ICT) and Learning, Østfold University College, Halden, Norway
| | - Rodrigo A. Bressan
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luis Augusto Rohde
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Attention-Deficit/Hyperactivity Disorder and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Giovanni Salum
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eurípedes Constantino Miguel
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Pedro Mario Pan
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
44
|
Zhang W, Dutt R, Lew D, Barch DM, Bijsterbosch JD. Higher amplitudes of visual networks are associated with trait- but not state-depression. Psychol Med 2025; 54:1-12. [PMID: 39757726 PMCID: PMC11769906 DOI: 10.1017/s0033291724003167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/09/2024] [Accepted: 11/07/2024] [Indexed: 01/07/2025]
Abstract
Despite depression being a leading cause of global disability, neuroimaging studies have struggled to identify replicable neural correlates of depression or explain limited variance. This challenge may, in part, stem from the intertwined state (current symptoms; variable) and trait (general propensity; stable) experiences of depression.Here, we sought to disentangle state from trait experiences of depression by leveraging a longitudinal cohort and stratifying individuals into four groups: those in remission ('trait depression group'), those with large longitudinal severity changes in depression symptomatology ('state depression group'), and their respective matched control groups (total analytic n = 1030). We hypothesized that spatial network organization would be linked to trait depression due to its temporal stability, whereas functional connectivity between networks would be more sensitive to state-dependent depression symptoms due to its capacity to fluctuate.We identified 15 large-scale probabilistic functional networks from resting-state fMRI data and performed group comparisons on the amplitude, connectivity, and spatial overlap between these networks, using matched control participants as reference. Our findings revealed higher amplitude in visual networks for the trait depression group at the time of remission, in contrast to controls. This observation may suggest altered visual processing in individuals predisposed to developing depression over time. No significant group differences were observed in any other network measures for the trait-control comparison, nor in any measures for the state-control comparison. These results underscore the overlooked contribution of visual networks to the psychopathology of depression and provide evidence for distinct neural correlates between state and trait experiences of depression.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rosie Dutt
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Biological Sciences Collegiate Division, University of Chicago, Chicago, IL, USA
| | - Daphne Lew
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Deanna M. Barch
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | | |
Collapse
|
45
|
Meier TB, Savitz J, España LY, Goeckner BD, Kent Teague T, van der Horn HJ, Tugan Muftuler L, Mayer AR, Brett BL. Association of concussion history with psychiatric symptoms, limbic system structure, and kynurenine pathway metabolites in healthy, collegiate-aged athletes. Brain Behav Immun 2025; 123:619-630. [PMID: 39414174 PMCID: PMC11624060 DOI: 10.1016/j.bbi.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Psychiatric outcomes are commonly observed in individuals with repeated concussions, though their underlying mechanism is unknown. One potential mechanism linking concussion with psychiatric symptoms is inflammation-induced activation of the kynurenine pathway, which is thought to play a role in the pathogenesis of mood disorders. Here, we investigated the association of prior concussion with multiple psychiatric-related outcomes in otherwise healthy male and female collegiate-aged athletes (N = 212) with varying histories of concussion recruited from the community. Specially, we tested the hypotheses that concussion history is associated with worse psychiatric symptoms, limbic system structural abnormalities (hippocampal volume, white matter microstructure assessed using neurite orientation dispersion and density imaging; NODDI), and elevations in kynurenine pathway (KP) metabolites (e.g., Quinolinic acid; QuinA). Given known sex-effects on concussion risk and recovery, psychiatric outcomes, and the kynurenine pathway, the moderating effect of sex was considered for all analyses. More concussions were associated with greater depression, anxiety, and anhedonia symptoms in female athletes (ps ≤ 0.005) and greater depression symptoms in male athletes (p = 0.011). More concussions were associated with smaller bilateral hippocampal tail (ps < 0.010) and left hippocampal body (p < 0.001) volumes across male and female athletes. Prior concussion was also associated with elevations in the orientation dispersion index (ODI) and lower intracellular volume fraction in several white matter tracts including the in uncinate fasciculus, cingulum-gyrus, and forceps major and minor, with evidence of female-specific associations in select regions. Regarding serum KP metabolites, more concussions were associated with elevated QuinA in females and lower tryptophan in males (ps ≤ 0.010). Finally, serum levels of QuinA were associated with elevated ODI (male and female athletes) and worse anxiety symptoms (females only), while higher ODI in female athletes and smaller hippocampal volumes in male athletes were associated with more severe anxiety and depression symptoms (ps ≤ 0.05). These data suggest that cumulative concussion is associated with psychiatric symptoms and limbic system structure in healthy athletes, with increased susceptibility to these effects in female athletes. Moreover, the associations of outcomes with serum KP metabolites highlight the KP as one potential molecular pathway underlying these observations.
Collapse
Affiliation(s)
- Timothy B Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, the United States of America; Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, the United States of America; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, the United States of America.
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK 74136, the United States of America; Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK 74119, the United States of America
| | - Lezlie Y España
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, the United States of America
| | - Bryna D Goeckner
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, the United States of America
| | - T Kent Teague
- Department of Psychiatry, The University of Oklahoma School of Community Medicine, Tulsa, OK 74135, the United States of America; Department of Surgery, The University of Oklahoma School of Community Medicine, Tulsa, OK 74135, the United States of America; Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, Tulsa, OK 74135, the United States of America
| | - Harm Jan van der Horn
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, the United States of America; University of Groningen, University Medical Center Groningen, the Netherlands
| | - L Tugan Muftuler
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, the United States of America
| | - Andrew R Mayer
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, the United States of America; Departments of Neurology and Psychiatry, University of New Mexico School of Medicine, Albuquerque, NM, the United States of America; Department of Psychology, University of New Mexico, Albuquerque, NM, the United States of America
| | - Benjamin L Brett
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, the United States of America; Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, the United States of America
| |
Collapse
|
46
|
Hagenberg J, Brückl TM, Erhart M, Kopf-Beck J, Ködel M, Rehawi G, Röh-Karamihalev S, Sauer S, Yusupov N, Rex-Haffner M, Spoormaker VI, Sämann P, Binder E, Knauer-Arloth J. Dissecting depression symptoms: Multi-omics clustering uncovers immune-related subgroups and cell-type specific dysregulation. Brain Behav Immun 2025; 123:353-369. [PMID: 39303816 DOI: 10.1016/j.bbi.2024.09.013] [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: 03/06/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
In a subset of patients with mental disorders, such as depression, low-grade inflammation and altered immune marker concentrations are observed. However, these immune alterations are often assessed by only one data type and small marker panels. Here, we used a transdiagnostic approach and combined data from two cohorts to define subgroups of depression symptoms across the diagnostic spectrum through a large-scale multi-omics clustering approach in 237 individuals. The method incorporated age, body mass index (BMI), 43 plasma immune markers and RNA-seq data from peripheral mononuclear blood cells (PBMCs). Our initial clustering revealed four clusters, including two immune-related depression symptom clusters characterized by elevated BMI, higher depression severity and elevated levels of immune markers such as interleukin-1 receptor antagonist (IL-1RA), C-reactive protein (CRP) and C-C motif chemokine 2 (CCL2 or MCP-1). In contrast, the RNA-seq data mostly differentiated a cluster with low depression severity, enriched in brain related gene sets. This cluster was also distinguished by electrocardiography data, while structural imaging data revealed differences in ventricle volumes across the clusters. Incorporating predicted cell type proportions into the clustering resulted in three clusters, with one showing elevated immune marker concentrations. The cell type proportion and genes related to cell types were most pronounced in an intermediate depression symptoms cluster, suggesting that RNA-seq and immune markers measure different aspects of immune dysregulation. Lastly, we found a dysregulation of the SERPINF1/VEGF-A pathway that was specific to dendritic cells by integrating immune marker and RNA-seq data. This shows the advantages of combining different data modalities and highlights possible markers for further stratification research of depression symptoms.
Collapse
Affiliation(s)
- Jonas Hagenberg
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany; International Max Planck Research School for Translational Psychiatry, 80804 Munich, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| | - Tanja M Brückl
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.
| | - Mira Erhart
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany; International Max Planck Research School for Translational Psychiatry, 80804 Munich, Germany.
| | - Johannes Kopf-Beck
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany; Department of Psychology, LMU Munich, Leopoldstr. 13, 80802 Munich, Germany.
| | - Maik Ködel
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.
| | - Ghalia Rehawi
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| | | | - Susann Sauer
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.
| | - Natan Yusupov
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany; International Max Planck Research School for Translational Psychiatry, 80804 Munich, Germany.
| | - Monika Rex-Haffner
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.
| | - Victor I Spoormaker
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.
| | - Philipp Sämann
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.
| | - Elisabeth Binder
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 100 Woodruff Circle, Atlanta GA 30322, USA.
| | - Janine Knauer-Arloth
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| |
Collapse
|
47
|
Jensen KHR, Dam VH, Köhler-Forsberg K, Ozenne B, Stenbæk DS, Ganz M, Fisher PM, Frokjaer VG, Knudsen GM, Jørgensen MB. Changes in hippocampal volume, 5-HT 4 receptor binding, and verbal memory over the course of antidepressant treatment in major depressive disorder. J Psychiatr Res 2025; 181:197-205. [PMID: 39616866 DOI: 10.1016/j.jpsychires.2024.11.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/17/2024] [Accepted: 11/21/2024] [Indexed: 01/22/2025]
Abstract
Serotonin reuptake inhibitors have been reported to increase hippocampal volume and improve memory function in patients with Major depressive disorder (MDD). The postsynaptic 5-HT4 receptor (5-HT4R) is involved in hippocampal development, familial risk for depression and depressive pathology. In an open-label trial with 91 patients (72% female, mean 27.2 years) with MDD, we investigated the relation between changes in hippocampal volume, 5-HT4R, and verbal memory during 12 weeks treatment with 10-20 mg escitalopram. Depression severity, verbal memory, MRI-determined hippocampus volume and PET-determined 5-HT4R were measured pretreatment. Forty-three patients were rescanned at week 8. HAMD17 was reassessed at week 8 and together with verbal memory at week 12. We used mixed-effects models and linear regressions. We estimated a 27 mm3 (p = 0.086) reduction in mean hippocampus volume over the course of eight weeks. In patients clinically responding to treatment, we estimated a 45 mm3 reduction (p = 0.019), 8 mm3 increase in non-responders (p = 0.78), and a 52 mm3 group difference (p = 0.12). Hippocampal 5-HT4 receptor binding before treatment and at week eight was negatively associated with hippocampal volume in females, regardless of treatment response (p-values≤0.006). However, no clear evidence for an association in males or sex interaction could be established (p-values≥0.16). Although the hippocampus volume did not increase with treatment, we found a decrease in clinically responsive patients. Our findings suggest an association between 5-HT4R signalling and changes in hippocampal volume in females with MDD during antidepressant treatment, highlighting the need for further investigation into the role of serotonergic mechanisms in hippocampal plasticity.
Collapse
Affiliation(s)
- Kristian H Reveles Jensen
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Vibeke H Dam
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Kristin Köhler-Forsberg
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark
| | - Brice Ozenne
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Dea S Stenbæk
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Melanie Ganz
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Patrick MacDonald Fisher
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Vibe Gedsoe Frokjaer
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Martin Balslev Jørgensen
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Yao M, Qu Y, Zheng Y, Guo H. The effect of exercise on depression and gut microbiota: Possible mechanisms. Brain Res Bull 2025; 220:111130. [PMID: 39557221 DOI: 10.1016/j.brainresbull.2024.111130] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Exercise can effectively prevent and treat depression and anxiety, with gut microbiota playing a crucial role in this process. Studies have shown that exercise can influence the diversity and composition of gut microbiota, which in turn affects depression through immune, endocrine, and neural pathways in the gut-brain axis. The effectiveness of exercise varies based on its type, intensity, and duration, largely due to the different changes in gut microbiota. This article summarizes the possible mechanisms by which exercise affects gut microbiota and how gut microbiota influences depression. Additionally, we reviewed literature on the effects of exercise on depression at different intensities, types, and durations to provide a reference for future exercise-based therapies for depression.
Collapse
Affiliation(s)
- Mingchen Yao
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yaqi Qu
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yalin Zheng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Hao Guo
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China.
| |
Collapse
|
49
|
Fu W, Xu R, Bian P, Li X, Yang K, Wang X. Exploring the shared genetic basis of major depressive disorder and frailty. J Affect Disord 2024; 366:386-394. [PMID: 39214376 DOI: 10.1016/j.jad.2024.08.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) and frailty impose substantial health and economic burdens. MDD is recognized as a significant risk factor for frailty, but the genetic associations between these conditions remain unclear. This study investigates the genetic correlation, shared pleiotropic loci, causal relationships, and comorbid genes between MDD and frailty. METHODS The genetic correlation between MDD and frailty was assessed using linkage disequilibrium score regression (LDSC) based on data from genome-wide association studies (GWAS). A detailed analysis was performed to identify shared pleiotropic loci and causal relationships through cross-phenotype association tests and Mendelian randomization. Additionally, tissue enrichment analysis was conducted using stratified LDSC, gene-based associations with both conditions were assessed using Multimarker Analysis of Genomic Annotation (MAGMA), and pathway analysis of comorbid genes was performed using the g: GOSt tool. RESULTS Our findings revealed a significant positive genetic correlation between MDD and frailty (rg = 0.65, P = 1.49E-219). We identified 57 shared risk SNPs between the two conditions, including 6 novel SNPs. Mendelian randomization analyses indicated robust causal effects of MDD on frailty and vice versa. Furthermore, we observed tissue-specific heritability enrichment in 9 brain tissues. By combining MAGMA and CPASSOC analyses, we identified 10 comorbid genes associated with both MDD and frailty, primarily involved in synapse formation, modulation, plasticity, and desaturase activity. CONCLUSION This study provides strong evidence for a shared genetic basis between MDD and frailty. The identification of comorbid genes offers new insights into the mechanisms underlying the relationship between these conditions.
Collapse
Affiliation(s)
- Wei Fu
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, Shaanxi 710032, China
| | - Rong Xu
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, Shaanxi 710032, China
| | - Peiyu Bian
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, Shaanxi 710032, China
| | - Xu Li
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, Shaanxi 710032, China
| | - Kaikai Yang
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, Shaanxi 710032, China
| | - Xiaoming Wang
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
50
|
Chen Z, Adegboro AA, Gu L, Li X. Constructing and exploring neuroimaging projects: a survey from clinical practice to scientific research. Insights Imaging 2024; 15:272. [PMID: 39546176 PMCID: PMC11568082 DOI: 10.1186/s13244-024-01848-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/13/2024] [Indexed: 11/17/2024] Open
Abstract
Over the past decades, numerous large-scale neuroimaging projects that involved the collection and release of multimodal data have been conducted globally. Distinguished initiatives such as the Human Connectome Project, UK Biobank, and Alzheimer's Disease Neuroimaging Initiative, among others, stand as remarkable international collaborations that have significantly advanced our understanding of the brain. With the advancement of big data technology, changes in healthcare models, and continuous development in biomedical research, various types of large-scale projects are being established and promoted worldwide. For project leaders, there is a need to refer to common principles in project construction and management. Users must also adhere strictly to rules and guidelines, ensuring data safety and privacy protection. Organizations must maintain data integrity, protect individual privacy, and foster stakeholders' trust. Regular updates to legislation and policies are necessary to keep pace with evolving technologies and emerging data-related challenges. CRITICAL RELEVANCE STATEMENT: By reviewing global large-scale neuroimaging projects, we have summarized the standards and norms for establishing and utilizing their data, and provided suggestions and opinions on some ethical issues, aiming to promote higher-quality neuroimaging data development. KEY POINTS: Global neuroimaging projects are increasingly advancing but still face challenges. Constructing and utilizing neuroimaging projects should follow set rules and guidelines. Effective data management and governance should be developed to support neuroimaging projects.
Collapse
Affiliation(s)
- Ziyan Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Abraham Ayodeji Adegboro
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Lan Gu
- School of Foreign Languages, Central South University, Changsha, China.
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China.
- Xiangya School of Medicine, Central South University, Changsha, China.
| |
Collapse
|