1
|
Madeira MM, Hage Z, Kokkosis AG, Nnah K, Guzman R, Schappell LE, Koliatsis D, Resutov E, Nadkarni NA, Rahme GJ, Tsirka SE. Oligodendroglia Are Primed for Antigen Presentation in Response to Chronic Stress-Induced Microglial-Derived Inflammation. Glia 2025; 73:1130-1147. [PMID: 39719686 PMCID: PMC12014386 DOI: 10.1002/glia.24661] [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: 06/18/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024]
Abstract
Chronic stress is a major contributor to the development of major depressive disorder, one of the leading causes of disability worldwide. Using a model of repeated social defeat stress in mice, we and others have reported that neuroinflammation plays a dynamic role in the development of behavioral deficits consistent with social avoidance and impaired reward responses. Animals susceptible to the model also exhibit hypomyelination in the medial prefrontal cortex, indicative of changes in the differentiation pathway of cells of the oligodendroglial lineage (OLN). We computationally confirmed the presence of immune oligodendrocytes, a population of OLN cells, which express immune markers and myelination deficits. In the current study, we report that microglia are necessary to induce expression of antigen presentation markers (and other immune markers) on oligodendroglia. We further associate the appearance of these markers with changes in the OLN and confirm that microglial changes precede OLN changes. Using co-cultures of microglia and OLN, we show that under inflammatory conditions the processes of phagocytosis and expression of MHCII are linked, suggesting potential priming for antigen presentation by OLN cells. Our findings provide insights into the nature of these OLN cells with immune capabilities, their obligatory interaction with microglia, and identify them as a potential cellular contributor to the pathological manifestations of psychosocial stress.
Collapse
Affiliation(s)
- Miguel M. Madeira
- Molecular and Cellular Pharmacology Program
- Scholars in Biomedical Sciences Program
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Zachary Hage
- Molecular and Cellular Pharmacology Program
- Scholars in Biomedical Sciences Program
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Alexandros G. Kokkosis
- Molecular and Cellular Pharmacology Program
- Scholars in Biomedical Sciences Program
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Kimberly Nnah
- Scholars in Biomedical Sciences Program
- Program in Neuroscience
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Ryan Guzman
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Laurel E. Schappell
- Molecular and Cellular Pharmacology Program
- Medical Scientist Training Program
- Department of Neurology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Dimitris Koliatsis
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Emran Resutov
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Neil A. Nadkarni
- Molecular and Cellular Pharmacology Program
- Department of Neurology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Gilbert J. Rahme
- Molecular and Cellular Pharmacology Program
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Stella E. Tsirka
- Molecular and Cellular Pharmacology Program
- Scholars in Biomedical Sciences Program
- Program in Neuroscience
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
2
|
Coleman SL, Sharpley CF, Vessey KA, Evans ID, Williams RJ, Bitsika V. Gamma oscillations as correlates of depression: updating Fitzgerald and Watson (2018). Rev Neurosci 2025:revneuro-2025-0023. [PMID: 40317135 DOI: 10.1515/revneuro-2025-0023] [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: 02/10/2025] [Accepted: 04/11/2025] [Indexed: 05/07/2025]
Abstract
Depression remains one of the most common and debilitating neuropsychiatric conditions, with little consistency in treatment efficacy. Some of the lack of success in developing effective treatments has been the absence of a reliable biomarker of depression, despite many attempts. One such potential biomarker is the electrical activity of the brain that occurs in the gamma band (30-200 Hz). To evaluate the state of research into gamma as a biomarker of depression, a review of recent research literature was conducted. A total of 31 relevant papers was identified, 22 of which used resting-state studies, and nine included a stimulus-task. These studies were examined here in terms of their definition of gamma, sample sizes, research focus, brain region examined, and EEG methodologies used. Due to the range of methodologies, some inconsistent results emerged but several valuable findings remained, including that depressed patients usually had higher gamma power than their healthy controls (HC), that the imposition of a perceptual task into the research protocol also introduced a strong element of confound to the results, and that studies that sought to evaluate the role of gamma in treatment were yet to be established as reliable. Key issues for future research are discussed, and the potential for gamma as a biomarker of depression is evaluated as emerging.
Collapse
Affiliation(s)
- Sarah L Coleman
- Brain-Behaviour Research Group, University of New England, Armidale, NSW, 2351, Australia
| | - Christopher F Sharpley
- Brain-Behaviour Research Group, University of New England, Armidale, NSW, 2351, Australia
| | - Kirstan A Vessey
- Brain-Behaviour Research Group, University of New England, Armidale, NSW, 2351, Australia
| | - Ian D Evans
- Brain-Behaviour Research Group, University of New England, Armidale, NSW, 2351, Australia
| | - Rebecca J Williams
- Brain-Behaviour Research Group, University of New England, Armidale, NSW, 2351, Australia
| | - Vicki Bitsika
- Brain-Behaviour Research Group, University of New England, Armidale, NSW, 2351, Australia
| |
Collapse
|
3
|
Flinkenflügel K, Borgers T, Klug M, Mummendey MM, Leehr EJ, Meinert S, Gruber M, Repple J, Kircher T, Opel N, Bauer J, Zwiky E, König P, Küttner A, Schöniger K, Kamrla R, Dannlowski U, Enneking V, Redlich R. Longitudinal associations between white matter integrity, early life adversities, and treatment response following cognitive-behavioral therapy in depression. Neuropsychopharmacology 2025; 50:1000-1007. [PMID: 40011705 PMCID: PMC12032135 DOI: 10.1038/s41386-025-02070-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
Cognitive-behavioral therapy (CBT) is a primary treatment for depression. Although previous research has underscored the significant roles of white matter (WM) alterations and maladaptive parenting in depression risk, their associations with CBT response remain largely unknown. This longitudinal study investigated the interplay of WM integrity changes over time, treatment response, and parenting style in patients with depression. Diffusion-tensor-imaging and clinical data were assessed in n = 65 (55% female) patients with depression before and after 20 CBT sessions and n = 65 (68% female) healthy controls (HC) in a naturalistic design. Linear-mixed-effect models compared changes in fractional anisotropy (FA) between groups and tested associations between FA changes and symptom changes. It was investigated whether parenting style predicts depressive symptoms at follow-up and whether FA changes mediate this association. Patients showed differential FA changes over time in the corpus callosum and corona radiata compared to HC (ptfce-FWE = 0.008). Increases in FA in the corpus callosum, corona radiata and superior longitudinal fasciculus were linked to symptom improvement after CBT in patients (ptfce-FWE = 0.023). High parental care (pFDR = 0.010) and low maternal overprotection (pFDR = 0.001) predicted fewer depressive symptoms at follow-up. The association between maternal overprotection and depressive symptoms at follow-up was mediated by FA changes (pFDR = 0.044). Robustness checks-controlling for outliers, non-linear age effects, clinical characteristics, and patient subgroups-supported these results. Overall, patients with depression show changes in WM integrity following CBT, which are linked to treatment response. The results highlight the significance of early life adversities and related microstructural changes in the effectiveness of CBT for treating depression.
Collapse
Affiliation(s)
- Kira Flinkenflügel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tiana Borgers
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Melissa Klug
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Marie M Mummendey
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Elisabeth J Leehr
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Marius Gruber
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Jonathan Repple
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Nils Opel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry and Psychotherapy, University Hospital Jena, Jena, Germany
- German Center for Mental Health (DZPG), Halle-Jena-Magdeburg, Halle, Germany
| | - Jochen Bauer
- Department of Radiology, University of Münster, Münster, Germany
| | - Esther Zwiky
- German Center for Mental Health (DZPG), Halle-Jena-Magdeburg, Halle, Germany
- Department of Psychology, University of Halle, Halle, Germany
| | - Philine König
- Department of Psychology, University of Halle, Halle, Germany
| | - Antonia Küttner
- Department of Psychology, University of Halle, Halle, Germany
| | | | - Robin Kamrla
- Department of Psychology, University of Halle, Halle, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Verena Enneking
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Ronny Redlich
- Institute for Translational Psychiatry, University of Münster, Münster, Germany.
- German Center for Mental Health (DZPG), Halle-Jena-Magdeburg, Halle, Germany.
- Department of Psychology, University of Halle, Halle, Germany.
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Halle, Germany.
| |
Collapse
|
4
|
Li Q, Qi L, Zhang G, Hao J, Ren Q, Guan J, Zhan Y, Yu Y, Yang J, Wang K, Bai T. Disrupted interhemispheric functional and structural connectivity in patients with major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111374. [PMID: 40262672 DOI: 10.1016/j.pnpbp.2025.111374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is associated with disrupted interhemispheric cooperation. However, the relationship between structural and functional alterations in interhemispheric cooperation in patients with MDD remains unclear. We investigated the associations between voxel-mirrored homotopic connectivity (VMHC) and radial diffusivity (RD) within the corpus callosum (CC) and their links to depressive symptoms in patients with MDD. METHODS Sixty patients with MDD and 38 healthy controls (HCs) were assessed using resting-state functional MRI (rs-fMRI) and diffusion MRI (dMRI) to evaluate interhemispheric functional connectivity (VMHC) and structural integrity (RD) in the CC subregions. Group comparisons, correlation analyses, and mediation analyses were conducted to identify the significant differences, relationships, and indirect effects. RESULTS Patients with MDD showed significantly reduced VMHC in the bilateral postcentral gyrus and lingual gyrus and increased RD in the CC subregions CC3, CC4, and CC5, indicating impaired functional and structural connectivity. Lower VMHC in the lingual gyrus was negatively correlated with depressive severity, whereas increased RD in the CC4 and CC5 was positively correlated with depressive symptoms. Mediation analysis revealed that the VMHC in the lingual gyrus fully mediated the relationship between RD in CC5 and depressive symptoms, suggesting a pathway through which structural impairments may affect mood through abnormal functional connectivity. LIMITATIONS The cross-sectional design limits the assessment of changes over time, and focusing solely on interhemispheric connectivity may overlook other networks involved in MDD. CONCLUSION These findings provide preliminary evidence for disrupted interhemispheric coordination in MDD, with both functional and structural connectivity impairments linked to depressive symptoms. The mediating effect of the VMHC in the lingual gyrus highlights the potential role of interhemispheric connectivity in the pathophysiology of MDD. Our results provide an integrative perspective on the functional and microstructural organization of the brain in patients with MDD.
Collapse
Affiliation(s)
- Qianqian Li
- Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei 230032, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China
| | - Li Qi
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Gu Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Jiajia Hao
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qiufang Ren
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Jian Guan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yuqian Zhan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yue Yu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Jinying Yang
- Laboratory Center for Information Science, University of Science and Technology of China, Hefei 230026, China; Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei 230026, China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei 230032, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China; The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei 230032, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China.
| | - Tongjian Bai
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei 230032, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| |
Collapse
|
5
|
Lyall LM, Stolicyn A, Lyall DM, Zhu X, Sangha N, Ward J, Strawbridge RJ, Cullen B, Smith DJ. Lifetime depression, sleep disruption and brain structure in the UK Biobank cohort. J Affect Disord 2025; 374:247-257. [PMID: 39719181 DOI: 10.1016/j.jad.2024.12.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
Whether depression and poor sleep interact or have statistically independent associations with brain structure and its change over time is not known. Within a subset of UK Biobank participants with neuroimaging and subjective and/or objective sleep data (n = 28,351), we examined associations between lifetime depression and sleep disruption, and their interaction with structural neuroimaging measures, both cross-sectionally and longitudinally. Sleep variables were: self-reported insomnia and difficulty getting up; actigraphy-derived short sleep (<7 h); sustained inactivity bouts during daytime (SIBD); and sleep efficiency. Imaging measures were white matter microstructure, subcortical volumes, cortical thickness and surface area of 24 cortical regions of interest. Individuals with lifetime depression (self-reported, mental health questionnaire or health records) were contrasted with healthy controls. Interactions between depression and difficulty getting up for i) right nucleus accumbens volume and ii) mean diffusivity of forceps minor, reflected a larger negative association of poor sleep in the presence vs. absence of depression. Depression was associated with widespread reductions in white matter integrity. Depression, higher SIBD and difficulty getting up were individually associated with smaller cortical volumes and surface area, particularly in the frontal and parietal lobes. Many regions showed age-related decline, but this was not exacerbated by either depression or sleep disturbance. Overall, we identified widespread cross-sectional associations of both lifetime depression and sleep measures with brain structure. Findings were more consistent with additive rather than synergistic effects - although in some regions we observed greater magnitude of deleterious associations from poor sleep phenotypes in the presence of depression.
Collapse
Affiliation(s)
- Laura M Lyall
- School of Health and Wellbeing, University of Glasgow, Glasgow, UK; Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Aleks Stolicyn
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Donald M Lyall
- School of Health and Wellbeing, University of Glasgow, Glasgow, UK; Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Xingxing Zhu
- School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Natasha Sangha
- School of Health and Wellbeing, University of Glasgow, Glasgow, UK; Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Joey Ward
- School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Rona J Strawbridge
- School of Health and Wellbeing, University of Glasgow, Glasgow, UK; Health Data Research, Glasgow, UK; Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Breda Cullen
- School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Daniel J Smith
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
6
|
Yin SQ, Li YH. Advancing the diagnosis of major depressive disorder: Integrating neuroimaging and machine learning. World J Psychiatry 2025; 15:103321. [PMID: 40109992 PMCID: PMC11886342 DOI: 10.5498/wjp.v15.i3.103321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 02/26/2025] Open
Abstract
Major depressive disorder (MDD), a psychiatric disorder characterized by functional brain deficits, poses considerable diagnostic and treatment challenges, especially in adolescents owing to varying clinical presentations. Biomarkers hold substantial clinical potential in the field of mental health, enabling objective assessments of physiological and pathological states, facilitating early diagnosis, and enhancing clinical decision-making and patient outcomes. Recent breakthroughs combine neuroimaging with machine learning (ML) to distinguish brain activity patterns between MDD patients and healthy controls, paving the way for diagnostic support and personalized treatment. However, the accuracy of the results depends on the selection of neuroimaging features and algorithms. Ensuring privacy protection, ML model accuracy, and fostering trust are essential steps prior to clinical implementation. Future research should prioritize the establishment of comprehensive legal frameworks and regulatory mechanisms for using ML in MDD diagnosis while safeguarding patient privacy and rights. By doing so, we can advance accuracy and personalized care for MDD.
Collapse
Affiliation(s)
- Shi-Qi Yin
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Ying-Huan Li
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
7
|
Zhao M, Eguchi A, Murayama R, Xu D, Zhu T, Xu B, Liu G, Mori C, Yang J, Hashimoto K. Repeated intermittent administration of 3,4-methylenedioxymethamphetamine mitigates demyelination in the brain from cuprizone-treated mice. Eur J Pharmacol 2025; 991:177345. [PMID: 39904416 DOI: 10.1016/j.ejphar.2025.177345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/15/2025] [Accepted: 01/31/2025] [Indexed: 02/06/2025]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA), commonly known as a recreational drug, may also offer therapeutic benefits for mental health. Population-based studies suggest that MDMA users have a lower risk of demyelinating diseases, such as depression. Given the role of the gut microbiota in mediating MDMA's effects, we hypothesized that MDMA might confer mental health benefits via the gut-brain axis. Cuprizone (CPZ) induces demyelination by chelating copper, which leads to oligodendrocyte death and subsequent myelin loss. This study investigated the impact of MDMA on brain demyelination in CPZ-treated mice, focusing on the gut-brain axis. Repeated intermittent MDMA administration (10 mg/kg, three times weekly for 6 weeks) significantly reduced demyelination in the corpus callosum (CC) of CPZ-treated mice. Gut microbiota and non-targeted metabolomics analyses revealed notable differences in specific gut bacteria and plasma (β-D-allose and L-sorbose) or fecal metabolite (carnitine) levels between MDMA-treated and vehicle-treated CPZ-exposed mice. Negative correlations were found between the levels of metabolites (β-D-allose, L-sorbose, and carnitine) and the relative abundance of Romboutsia and Romboutsia timonensis. These findings suggest that intermittent MDMA administration may alleviate demyelination in the CC of CPZ-treated mice via the gut-brain axis. Further research is needed to elucidate the roles of gut microbiota and metabolites in MDMA's effects on brain demyelination and to investigate other demyelination models.
Collapse
Affiliation(s)
- Mingming Zhao
- Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Akifumi Eguchi
- Department of Sustainable Health Science, Chiba University Center for Preventive Medical Sciences, Chiba, 263-8522, Japan
| | - Rumi Murayama
- Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan; Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Dan Xu
- Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan; Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tingting Zhu
- Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Biao Xu
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453000, China
| | - Guiling Liu
- Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan; Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan; Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, 266100, China
| | - Chisato Mori
- Department of Sustainable Health Science, Chiba University Center for Preventive Medical Sciences, Chiba, 263-8522, Japan; Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
8
|
Liu X, Wei Z, Li L, Li J, Deng Y, Liu Y, Li H, Peng D, Wan X, Wu G. Effect of continuous esketamine infusion on brain white matter microstructure in patients with major depression: A diffusion tensor imaging study. J Affect Disord 2025; 372:173-181. [PMID: 39631703 DOI: 10.1016/j.jad.2024.12.002] [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: 06/10/2024] [Revised: 10/09/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Esketamine has demonstrated acute antidepressant effects in patients with major depressive disorder (MDD). This study investigated whether these effects associate with reversible white matter fiber integrity recovery using diffusion imaging. METHOD Twenty patients with MDD and 20 healthy controls received 2-week esketamine treatment. Patients received 0.25 mg/kg intravenous esketamine. Emotional and cognitive recovery were assessed. Diffusion tensor imaging and tract-based spatial statistics evaluated white matter fiber integrity pre/post-treatment. Correlation analyses examined associations between white matter changes and clinical scales. RESULTS Compared to controls, patients with MDD exhibited decreased fractional anisotropy (FA) values of cerebral white matter fibers involving the association fibers, the commissural fibers and projection fibers. Esketamine effectively reduced depression, anxiety, and suicidal ideation scores while improving cognitive function. However, no reversible recovery of compromised white matter integrity was observed after 2 weeks of esketamine treatment. FA reductions in projection fibers correlated with anxiety and suicidal ideation severity. LIMITATIONS Concurrent sertraline use and lack of placebo control limited our ability to isolate esketamine's effects. The wide age range may have introduced response variability. We used minimal effective dosages based on previous research. The small sample size limited statistical power. Larger, more controlled studies are needed to validate these preliminary findings. DISCUSSION This study enhances MDD neuropathological understanding, with widespread white matter impairment and associations between projection fibers and symptom severity. While producing significant antidepressant effects, short-term esketamine did not recover compromised white matter microstructure.
Collapse
Affiliation(s)
- Xiang Liu
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Zhipeng Wei
- Department of Radiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Lifeng Li
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China; Department of Radiology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hunan, China
| | - Jiangping Li
- Department of Psychosomatic Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Yingke Deng
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Yumeng Liu
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Haijun Li
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Dechang Peng
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Xin Wan
- Department of TCM, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
| | - Guojiang Wu
- Department of Psychosomatic Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
| |
Collapse
|
9
|
Takahashi K, Noda Y, Hondo N, Shibukawa S, Kamagata K, Wada M, Honda S, Homma S, Tsukazaki A, Tsugawa S, Tobari Y, Moriyama S, Taniguchi K, Koike S, Cassidy C, Mimura M, Uchida H, Nakajima S. Abnormal neuritic microstructures in the anterior limb of internal capsules in treatment-resistant depression - A cross-sectional NODDI study. J Psychiatr Res 2025; 183:93-99. [PMID: 39954542 DOI: 10.1016/j.jpsychires.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Microstructural deficits of brain tissue are implicated in the pathophysiology of major depressive disorder (MDD). Recent studies have highlighted the neurotrophic mechanisms underlying effective treatments such as ketamine for treatment-resistant depression (TRD). However, little is known about microstructural changes in TRD. Neurite orientation dispersion and density imaging (NODDI) has enabled in vivo investigation of gray matter (GM) and white matter (WM) microstructure. This study sought to examine microstructural abnormalities in gray and white matter in patients with TRD using NODDI. METHODS This study compared the neurite density index (NDI) and orientation dispersion index (ODI) of neurites in 70 patients with TRD and 35 healthy controls. We fitted separate optimal NODDI models for gray and white matter. The locations of microstructural deficit were identified using region-based and voxel-based analysis. The affected white matter fibers were tracked with correlational tractography analysis. RESULTS An increase of ODI at the middle to the ventral part of the right anterior limb of the internal capsule (ALIC) was observed in patients with TRD compared with healthy controls. The quantitative anisotropy of frontothalamic fibers passing through the ALIC negatively correlated with the ODI increase in the TRD group. CONCLUSION The microstructural disorganization of the frontothalamic pathway could be linked to the pathophysiology and individual heterogeneity of TRD.
Collapse
Affiliation(s)
- Koki Takahashi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; Department of Psychiatry, International University of Health and Welfare, Mita Hospital, Tokyo, Japan.
| | - Nobuaki Hondo
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shuhei Shibukawa
- Department of Radiology, Tokyo Medical University, Tokyo, Japan; Faculty of Health Science, Department of Radiological Technology, Juntendo University, Tokyo, Japan; Center for Evolutionary Cognitive Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University, Tokyo, Japan
| | - Masataka Wada
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Shiori Honda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; Graduate School of Media and Governance, Keio University, Kanagawa, Japan; Department of Psychiatry and Behavioral Health, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Saki Homma
- Graduate School of Media and Governance, Keio University, Kanagawa, Japan
| | - Amaki Tsukazaki
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Sakiko Tsugawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yui Tobari
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Sotaro Moriyama
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Keita Taniguchi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shinsuke Koike
- Center for Evolutionary Cognitive Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Clifford Cassidy
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
10
|
Jadhav KK, Daouk J, Kurkinen K, Kraav SL, Eriksson P, Tolmunen T, Kanninen KM. Blood cytokines in major depressive disorder in drug-naïve adolescents: A systematic review and meta-analysis. J Affect Disord 2025; 372:48-55. [PMID: 39603515 DOI: 10.1016/j.jad.2024.11.071] [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: 09/06/2024] [Revised: 11/01/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is the most common mental health problem worldwide. Increased levels of inflammation are associated with MDD, though this relationship has been suggested to be bidirectional. The first incidence of a depressive episode usually occurs during adolescence. Hence, examining depressed, drug-naïve adolescents is important to understand the role of inflammation in the pathophysiology of MDD. Cytokines might play a crucial role in inflammation associated with MDD. Therefore, this article aims to investigate the changes in the levels of peripheral blood cytokines in adolescents with MDD. METHODS We conducted a systematic review and meta-analysis to assess the changes in peripheral blood cytokines in drug-naïve adolescents (10-18 years) with MDD. A comprehensive search across four databases was performed to identify original research articles. Studies in which the diagnosis of MDD was set by semi-structured interview were included. RESULTS Of 2291 articles, 12 met the inclusion criteria for the review, with seven suitable for meta-analysis & including up to five studies per cytokine. The meta-analysis revealed significant associations between tumor necrosis factor (TNF)-α (n = 222, Hedge's g = 0.51, p <0.01) and MDD in adolescents compared to healthy individuals. However, other blood cytokines, including interleukin (IL)-1β, IL-4, IL-6, IL-8, and interferon (IFN)-γ, did not significantly correlate with MDD in adolescents. CONCLUSION TNF-α was significantly elevated in drug-naïve adolescents with MDD. To further understand the role of TNF-α in MDD, a thorough investigation is required, taking into account the diversity, subtypes, chronicity, and severity of MDD.
Collapse
Affiliation(s)
- Kaustubh Kishor Jadhav
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Joud Daouk
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Karoliina Kurkinen
- Institute of Clinical Medicine, Department of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Siiri-Liisi Kraav
- Department of Social Sciences, Faculty of Social Sciences and Business Studies, University of Eastern Finland, Kuopio, Finland
| | - Päivi Eriksson
- Business School, Faculty of Social Sciences and Business Studies, University of Eastern Finland, Kuopio, Finland
| | - Tommi Tolmunen
- Institute of Clinical Medicine, Department of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland; Kuopio University Hospital, Department of Adolescent Psychiatry, Kuopio, Finland
| | - Katja M Kanninen
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
11
|
Zhang W, Zhang C, Zhao J, Cui J, Bai J, Deng X, Ji J, Li T, Wang Y, Li K, Qu Y, Li J. Microstructure Abnormalities of Diffusion Tensor Imaging Measures in First-Episode, Treatment-Naïve Adolescents With Major Depressive Disorder: An Integrated AFQ and TBSS Study. Brain Behav 2025; 15:e70416. [PMID: 40079635 PMCID: PMC11905106 DOI: 10.1002/brb3.70416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/15/2025] Open
Abstract
PURPOSE Structural changes during depressive episodes in adolescents with major depressive disorder (MDD) remains unclear due to participant heterogeneity, illness chronicity, and medication confounders. This study aimed to explore white matter (WM) microstructural changes in first-episode, treatment-naïve adolescents with MDD using an integrated diffusion tensor imaging (DTI) approach. METHOD We recruited 66 subjects, including 37 adolescents with MDD and 29 healthy controls. Two main DTI techniques, automated fiber quantification (AFQ) and tract-based spatial statistics (TBSS), were used to analyze fractional anisotropy (FA), axial diffusivity (AD), radial diffusivity (RD), and mean diffusivity (MD) in WM tracts. DTI measures were then correlated with the depressive symptoms evaluated by Hamilton Depression Rating Scale scores (HAMD-17). FINDINGS In AFQ, MDD patients showed significant segmental differences in WM tracts compared to controls, including a negative correlation between SLF AD values and depression severity. TBSS revealed reduced FA in the cingulum, forceps minor, inferior fronto-occipital fasciculus, inferior longitudinal fasciculus, SLF, and uncinate fasciculus in MDD. CONCLUSION Our integrated DTI analysis in a unique first-episode, medication-naïve cohort revealed microstructural changes in adolescent MDD not previously reported. These findings may provide imaging markers for early detection and enhance our understanding of depression pathology in youth.
Collapse
Affiliation(s)
- Wenjie Zhang
- Department of Radiology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Changzhi Key Lab of Functional Imaging for Brain Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Department of Radiology, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, Shandong, China
| | - Chan Zhang
- Department of Radiology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Changzhi Key Lab of Functional Imaging for Brain Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Jinyuan Zhao
- Department of Radiology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Changzhi Key Lab of Functional Imaging for Brain Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Jiajing Cui
- Department of Radiology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Changzhi Key Lab of Functional Imaging for Brain Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Jinji Bai
- Department of Radiology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Changzhi Key Lab of Functional Imaging for Brain Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Xuan Deng
- Department of Radiology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Changzhi Key Lab of Functional Imaging for Brain Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Junjun Ji
- Department of Radiology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Changzhi Key Lab of Functional Imaging for Brain Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Department of Psychiatry, Changzhi Mental Health Center, Changzhi, Shanxi, China
| | - Ting Li
- Department of Radiology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Changzhi Key Lab of Functional Imaging for Brain Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Yu Wang
- Department of Psychiatry, Changzhi Mental Health Center, Changzhi, Shanxi, China
| | - Kefeng Li
- Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR, China
| | - Yunhui Qu
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Junfeng Li
- Department of Radiology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Changzhi Key Lab of Functional Imaging for Brain Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| |
Collapse
|
12
|
Sun T, Jiang C, Zhang Y, Li Y, Chen G, Zhou Y, Xu W, You L, Kong Y, Jiang W, Yuan Y. Distinguished multimodal imaging features affected by COVID-19 in major depressive disorder patients. J Psychiatr Res 2025; 183:1-9. [PMID: 39908714 DOI: 10.1016/j.jpsychires.2025.01.053] [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: 09/07/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
OBJECTIVES Growing attention has been directed toward the structural and functional alterations among individuals infected with COVID-19. However, data on its impact on patients with Major Depressive Disorder (MDD) remain limited. METHODS This study investigates the effects of COVID-19 on patients with MDD and healthy controls (HCs) using MRI scans. Participants were categorized into four groups: MDD patients before (n = 165) and after COVID-19 infection (n = 70), HCs before (n = 108) and after COVID-19 infection (n = 57). All participants underwent T1-weighted imaging, diffusion tensor imaging (DTI), and resting-state functional MRI from January 2022 to August 2023. RESULTS Structural alterations associated with COVID-19 were predominantly observed in the white matter (WM) rather than the gray matter (GM), with specific involvement noted in the superior longitudinal fasciculus tract, Forceps minor tract, and cingulum-cingulate gyrus tract among patients with MDD. Functional changes were spread from GM to WM. The bilateral supplementary motor area, the left angular gyrus, the left subcortical regions (amygdala and parahippocampal gyrus), and various WM tracts showed significant infection-related changes across groups. CONCLUSION COVID-19 infection induces significant microstructural damage of WM in healthy individuals and exacerbates white matter microstructural injury of MDD. These findings suggest that WM might be more susceptible to COVID-19 effects than GM in both MDD patients and HCs.
Collapse
Affiliation(s)
- Taipeng Sun
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China; Department of Medical Psychology, Huai'an No.3 People's Hospital, Huaian, 223001, Jiangsu, China
| | - Chenguang Jiang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yubo Zhang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yueying Li
- Lab of Image Science and Technology, School of Computer Science and Engineering, Key Laboratory of Computer Network and Information Integration, Ministry of Education, Southeast University, Nanjing, China
| | - Gang Chen
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China; Department of Medical Psychology, Huai'an No.3 People's Hospital, Huaian, 223001, Jiangsu, China
| | - Yue Zhou
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Wei Xu
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Linlin You
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Youyong Kong
- Lab of Image Science and Technology, School of Computer Science and Engineering, Key Laboratory of Computer Network and Information Integration, Ministry of Education, Southeast University, Nanjing, China.
| | - Wenhao Jiang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China; Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
13
|
Zhao Q, Wang S, Xiong D, Liu M, Zhang Y, Zhao G, Zhao J, Shi Z, Zhang Z, Lei M, Zhai Y, Xu J, Hao X, Li S, Liu F. Genome-wide analysis identifies novel shared loci between depression and white matter microstructure. Mol Psychiatry 2025:10.1038/s41380-025-02932-2. [PMID: 39972055 DOI: 10.1038/s41380-025-02932-2] [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: 09/14/2024] [Revised: 01/09/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Depression, a complex and heritable psychiatric disorder, is associated with alterations in white matter microstructure, yet their shared genetic basis remains largely unclear. Utilizing the largest available genome-wide association study (GWAS) datasets for depression (N = 674,452) and white matter microstructure (N = 33,224), assessed through diffusion tensor imaging metrics such as fractional anisotropy (FA) and mean diffusivity (MD), we employed linkage disequilibrium score regression method to estimate global genetic correlations, local analysis of [co]variant association approach to pinpoint genomic regions with local genetic correlations, and conjunctional false discovery rate analysis to identify shared variants. Our findings revealed that depression showed significant local genetic correlations with FA in 37 genomic regions and with MD in 59 regions, while global genetic correlations were weak. Variant-level analysis identified 78 distinct loci jointly associated with depression (25 novel loci) and FA (35 novel loci), and 41 distinct loci associated with depression (17 novel loci) and MD (25 novel loci). Further analyses showed that these shared loci exhibited both concordant and discordant effect directions between depression and white matter traits, as well as distinct yet overlapping hemispheric patterns in their genetic architecture. Enrichment analysis of these shared loci implicated biological processes related to metabolism and regulation. This study provides evidence of a mixed-direction shared genetic architecture between depression and white matter microstructure. The identification of specific loci and pathways offers potential insights for developing targeted interventions to improve white matter integrity and alleviate depressive symptoms.
Collapse
Affiliation(s)
- Qiyu Zhao
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Shuo Wang
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, Jinan, 250013, Shandong, China.
| | - Di Xiong
- Department of Mathematics, Shanghai University, Shanghai, 200444, China
| | - Mengge Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yujie Zhang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Guoshu Zhao
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jiaxuan Zhao
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ziqing Shi
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zhihui Zhang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Minghuan Lei
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ying Zhai
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jinglei Xu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaoke Hao
- School of Artificial Intelligence, Hebei University of Technology, Tianjin, 300401, China.
| | - Shen Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China.
- Brain Assessment & Intervention Laboratory, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China.
| | - Feng Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
14
|
Xue K, Liu F, Liang S, Guo L, Shan Y, Xu H, Xue J, Jiang Y, Zhang Y, Lu J. Brain connectivity and transcriptomic similarity inform abnormal morphometric similarity patterns in first-episode, treatment-naïve major depressive disorder. J Affect Disord 2025; 370:519-531. [PMID: 39522735 DOI: 10.1016/j.jad.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is associated with disrupted brain structural integration. Morphometric similarity offers a means to capture the coordinated patterns of various structural features. However, it remains unknown whether MDD-related changes can be detected in cortical morphometric similarity through the Morphometric Inverse Divergence (MIND) network. Additionally, the role of brain connectivity in shaping these alterations, and their links to neuroreceptors and gene expression, have yet to be investigated. METHODS Using the T1-weighted MRI data from 71 patients with first-episode, treatment-naïve MDD and 69 healthy controls, we constructed the MIND network for all participants. We then performed between-group comparisons to investigate abnormalities in the network and spatial relationships between the observed patterns of MIND disruption and the patterns of neuroreceptors were estimated. Network-based spreading was utilized to explore the abnormalities constrained by brain connectivity based on structural, functional, and transcriptional connectome architecture and to further identify potential epicenters of MDD. In addition, partial least squares regression was conducted to examine the associations of gene expression profiles with MIND changes in MDD. RESULTS Patients with MDD showed significantly increased MIND in regions associated with sensation and cognition compared with healthy controls, with this altered pattern being influenced by a combination of transcriptional and structural connectivity, and potential epicenters of MDD were identified in the frontal, parietal, and paracentral cortices. Furthermore, the cortical map of case-control differences in MIND was spatially correlated with the cannabinoid CB1 receptor and the brain-wide expression of a weighted combination of genes. These genes were enriched for neurobiologically relevant pathways and preferentially expressed in different cell classes and cortical layers. CONCLUSION These results highlight the abnormal pattern of morphometric similarity observed in MDD, shedding light on the complex interplay between disrupted macroscale coordinated morphology and microscale molecular organization in MDD.
Collapse
Affiliation(s)
- Kaizhong Xue
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China; Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Sixiang Liang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China; Tianjin Anding Hospital, Tianjin 300222, China
| | - Lining Guo
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yi Shan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Huijuan Xu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Jiao Xue
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Yifan Jiang
- School of Nursing, Tianjin Medical University, Tianjin 300070, China
| | - Yong Zhang
- Tianjin Anding Hospital, Tianjin 300222, China.
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China.
| |
Collapse
|
15
|
Liu W, Heij J, Liu S, Liebrand L, Caan M, van der Zwaag W, Veltman DJ, Lu L, Aghajani M, van Wingen G. Structural connectivity of thalamic subnuclei in major depressive disorder: An ultra-high resolution diffusion MRI study at 7-Tesla. J Affect Disord 2025; 370:412-426. [PMID: 39505018 DOI: 10.1016/j.jad.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND The thalamus serves as a central relay station within the brain, and thalamic connectional anomalies are increasingly thought to be present in major depressive disorder (MDD). However, the use of conventional MRI scanners and acquisition techniques has prevented a thorough examination of the thalamus and its subnuclear connectional profile. We combined ultra-high field diffusion MRI acquired at 7.0 Tesla to map the white matter connectivity of thalamic subnuclei. METHODS Fifty-three MDD patients and 12 healthy controls (HCs) were involved in the final analysis. FreeSurfer was used to segment the thalamic subnuclei, and MRtrix was used to perform the preprocessing and tractography. Fractional anisotropy, axial diffusivity, mean diffusivity, radial diffusivity, and streamline count of thalamic subnuclear tracts were measured as proxies of white matter microstructure. Bayesian multilevel model was used to assess group differences in white matter metrics for each thalamic subnuclear tract and the association between these white matter metrics and clinical features in MDD. RESULTS Evidence was found for reduced whiter matter metrics of the tracts spanning from all thalamic subnuclei among MDD versus HC participants. Moreover, evidence was found that white matter in various thalamic subnuclear tracts is related to medication status, age of onset and recurrence in MDD. CONCLUSIONS Structural connectivity was generally reduced in thalamic subnuclei in MDD participants. Several clinical characteristics are related to perturbed subnuclear thalamic connectivity with cortical and subcortical circuits that govern sensory processing, emotional function, and goal-directed behavior.
Collapse
Affiliation(s)
- Weijian Liu
- Amsterdam UMC location University of Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands; 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), Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing, China.
| | - Jurjen Heij
- Spinoza Centre for Neuroimaging, KNAW, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, KNAW, Amsterdam, the Netherlands
| | - Shu Liu
- Key Laboratory of Genetic Evolution & Animal Models, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Luka Liebrand
- Amsterdam Neuroscience, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiation Oncology, Amsterdam, the Netherlands
| | - Matthan Caan
- Amsterdam Neuroscience, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Biomedical Engineering & Physics, Amsterdam, the Netherlands
| | - Wietske van der Zwaag
- Spinoza Centre for Neuroimaging, KNAW, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, KNAW, Amsterdam, the Netherlands
| | - Dick J Veltman
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands
| | - Lin Lu
- 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), Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing, China; Peking-Tsinghua Centre for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.
| | - Moji Aghajani
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands; Institute of Education & Child Studies, Section Forensic Family & Youth Care, Leiden University, the Netherlands
| | - Guido van Wingen
- Amsterdam UMC location University of Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Murayama R, Liu G, Zhao MM, Xu D, Zhu TT, Cai Y, Yue Y, Nakamura H, Hashimoto K. Microbiome depletion by broad-spectrum antibiotics does not influence demyelination and remyelination in cuprizone-treated mice. Pharmacol Biochem Behav 2025; 247:173946. [PMID: 39672388 DOI: 10.1016/j.pbb.2024.173946] [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: 08/26/2024] [Revised: 11/10/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Demyelination in the central nervous system (CNS) is a feature of various psychiatric and neurological disorders. Emerging evidence suggests that the gut-brain axis may play a crucial role in CNS demyelination. The cuprizone (CPZ) model, which involves the administration of CPZ-containing food pellets, is commonly used to study the effects of different compounds on CNS demyelination and subsequent remyelination. This study aimed to evaluate the impact of microbiome depletion, induced by an antibiotic cocktail (ABX), on demyelination in CPZ-treated mice and the subsequent remyelination following CPZ withdrawal. Our findings indicate that a chronic 4-week oral ABX regimen, administered both during and after a 6-week CPZ exposure, does not affect demyelination or remyelination in the brains of CPZ-treated mice. Specifically, ABX treatment for 2 weeks before and 2 weeks after CPZ exposure, in the final 4 weeks before sacrifice, and for 4 weeks post-CPZ withdrawal, did not significantly alter these processes compared to control mice receiving water instead of ABX. These results indicate that despite effective microbiome depletion, a 4-week oral ABX regimen does not influence demyelination or remyelination in the CPZ model. Thus, it is unlikely that gut microbiota depletion by ABX plays a significant role in these processes. However, further research is needed to fully understand the role of the host microbiome on CPZ-induced demyelination.
Collapse
Affiliation(s)
- Rumi Murayama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Guilin Liu
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China
| | - Ming-Ming Zhao
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dan Xu
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ting-Ting Zhu
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yi Cai
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Yong Yue
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| |
Collapse
|
17
|
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. 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
|
18
|
Qi K, Li H, Tao J, Liu M, Zhang W, Liu Y, Liu Y, Gong H, Wei J, Wang A, Xu J, Li X. Glutamate chemical exchange saturation transfer (GluCEST) MRI to evaluate the relationship between demyelination and glutamate content in depressed mice. Behav Brain Res 2025; 476:115247. [PMID: 39277141 DOI: 10.1016/j.bbr.2024.115247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024]
Abstract
Glutamatergic alteration is one of the potential mechanisms of depression. However, there is no consensus on whether glutamate metabolism changes affect the myelin structure of depression in mouse models. Glutamate chemical exchange saturation transfer (GluCEST) is a novel and powerful molecular imaging technique that can visualize glutamate distribution. In this study, we used the GluCEST imaging technique to look at glutamate levels in mice under chronic unpredictable mild stress (CUMS) and how they relate to demyelination. The CUMS mice were exposed to different stress factors for 6 weeks. Evaluated of depression in CUMS mice by behavioral tests. MRI scans were then performed, including T2-mapping, GluCEST, and diffusion tensor imaging (DTI) sequences. Brain tissues were collected for Luxol Fast Blue staining and immunofluorescence staining to analyze the changes in the myelin sheath. Artificially sketched regions of interest (ROI) (corpus callosum, hippocampus, and thalamus) were used to calculate the GluCEST value, fractional anisotropy (FA), and T2 value. Compared with the control group, the GluCEST value in the ROIs of CUMS mice significantly decreased. Similarly, the FA value in ROIs was lower in the CUMS group than in the CTRL group, but the T2 value did not differ significantly between the two groups. The histological results showed that ROIs in the CUMS group had demyelination compared with the CTRL group, indicating that DTI was more sensitive than T2 mapping in detecting myelin abnormalities. Furthermore, the GluCEST value in the ROIs correlates positively with the FA value. These findings suggest that altered glutamate metabolism may be one of the important factors leading to demyelination in depression, and GluCEST is expected to serve as an imaging biological marker for the diagnosis of demyelination in depression.
Collapse
Affiliation(s)
- Kai Qi
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Hao Li
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Jin Tao
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Miaomiao Liu
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Wei Zhang
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Yan Liu
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Yuwei Liu
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - He Gong
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Junhui Wei
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Ailing Wang
- Department of Clinical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264100, China.
| | - Junhai Xu
- College of Intelligence and Computing, Tianjin University, Tianjin 300350, China.
| | - Xianglin Li
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
19
|
An L, Zhang C, Wulan N, Zhang S, Chen P, Ji F, Ng KK, Chen C, Zhou JH, Yeo BTT. DeepResBat: Deep residual batch harmonization accounting for covariate distribution differences. Med Image Anal 2025; 99:103354. [PMID: 39368279 DOI: 10.1016/j.media.2024.103354] [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: 01/18/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024]
Abstract
Pooling MRI data from multiple datasets requires harmonization to reduce undesired inter-site variabilities, while preserving effects of biological variables (or covariates). The popular harmonization approach ComBat uses a mixed effect regression framework that explicitly accounts for covariate distribution differences across datasets. There is also significant interest in developing harmonization approaches based on deep neural networks (DNNs), such as conditional variational autoencoder (cVAE). However, current DNN approaches do not explicitly account for covariate distribution differences across datasets. Here, we provide mathematical results, suggesting that not accounting for covariates can lead to suboptimal harmonization. We propose two DNN-based covariate-aware harmonization approaches: covariate VAE (coVAE) and DeepResBat. The coVAE approach is a natural extension of cVAE by concatenating covariates and site information with site- and covariate-invariant latent representations. DeepResBat adopts a residual framework inspired by ComBat. DeepResBat first removes the effects of covariates with nonlinear regression trees, followed by eliminating site differences with cVAE. Finally, covariate effects are added back to the harmonized residuals. Using three datasets from three continents with a total of 2787 participants and 10,085 anatomical T1 scans, we find that DeepResBat and coVAE outperformed ComBat, CovBat and cVAE in terms of removing dataset differences, while enhancing biological effects of interest. However, coVAE hallucinates spurious associations between anatomical MRI and covariates even when no association exists. Future studies proposing DNN-based harmonization approaches should be aware of this false positive pitfall. Overall, our results suggest that DeepResBat is an effective deep learning alternative to ComBat. Code for DeepResBat can be found here: https://github.com/ThomasYeoLab/CBIG/tree/master/stable_projects/harmonization/An2024_DeepResBat.
Collapse
Affiliation(s)
- Lijun An
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Chen Zhang
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Naren Wulan
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Shaoshi Zhang
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Pansheng Chen
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Fang Ji
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kwun Kei Ng
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
| | - B T Thomas Yeo
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore; Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore; Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
20
|
Xue L, Wang H, Wang X, Shao J, Sun Y, Zhu R, Yao Z, Lu Q. The relationship between demographic factors and brain hierarchical changes following antidepressant treatment in patients remitted from depression. J Psychiatr Res 2025; 181:425-432. [PMID: 39662329 DOI: 10.1016/j.jpsychires.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/25/2024] [Accepted: 12/01/2024] [Indexed: 12/13/2024]
Abstract
To investigate the associations between demographic factors and brain hierarchical changes following successful selective serotonin reuptake inhibitor (SSRI) treatment, 57 major depressive disorder (MDD) patients who achieved remission after a 12-week SSRI treatment and 39 healthy controls (HCs) were recruited. MDD patients underwent diffusion tensor imaging (DTI) scans before treatment and after the 12-week SSRI treatment. Depression severity was evaluated with the Hamilton Rating Scale for Depression (HAMD) using the total score and the subscales: retardation, cognitive impairment, anxiety, and sleep disturbance. All HCs also underwent DTI scans after enrollment. Building on gradient mapping techniques, we developed a set of measures to quantify the dispersion within functional communities and also studied demographic-relevant differences in the three-dimensional gradient space of remitted MDD patients. We defined the Z-scores of the gradients in the pre-treatment group relative to the HC group as the disease pattern, post-treatment group relative to the HC group as the recovery pattern. The results showed that the disease pattern of depression is associated with age, as older age groups exhibit more severe impairments in depression. A significant difference was detected in the dispersion of the frontoparietal network (FPN) between pre-treatment and post-treatment patients. With the moderating effect of the age of onset, the dispersion of the FPN was related to the improvement in cognitive impairment, the dorsal attention network (DAN) was related to the improvement in retardation symptoms. Our findings help clinicians be alert to the role of demographic effects on clinical efficacy when treating depressed patients.
Collapse
Affiliation(s)
- Li Xue
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, China
| | - Huan Wang
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, China
| | - Xinyi Wang
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, China
| | - Junneng Shao
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, China
| | - Yurong Sun
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, China
| | - Rongxin Zhu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China; Nanjing Brain Hospital, Medical School of Nanjing University, Nanjing, 210093, China
| | - Zhijian Yao
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China; Nanjing Brain Hospital, Medical School of Nanjing University, Nanjing, 210093, China.
| | - Qing Lu
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, China.
| |
Collapse
|
21
|
Xu Z, Zhou Z, Tao W, Lai W, Qian L, Cui W, Peng B, Zhang Y, Hou G. Altered topology in cortical morphometric similarity network in recurrent major depressive disorder. J Psychiatr Res 2025; 181:206-213. [PMID: 39616867 DOI: 10.1016/j.jpsychires.2024.11.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 10/11/2024] [Accepted: 11/21/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Recurrent major depressive disorder (RDD) is increasingly understood to be associated with a 'disconnection' within the brain areas. But, the true understanding of cortical connectivities remains challenging. Morphometric similarity network (MSN) with multi-modal magnetic resonance imaging (MRI) could provide more information about cortical micro-architecture changes in individuals with RDD. METHODS Here, we integrated multi-modal features from T1-weighted imaging, diffusion tensor imaging (DTI), and inhomogeneous magnetization transfer imaging (ihMT) to construct MSN. We used graph theory to calculate topological changes in MSN and explore their relationship with the severity and recurrence. The topological properties of 42 RDD patients were compared with 56 age, sex, and education-matched healthy controls. RESULTS RDD subjects showed significantly decreased global efficiency, increased characteristic path length, reduced nodal efficiencies in the parietal lobe, subcortical area, and temporal lobe, increased betweenness centrality in the left supplementary motor area (SMA), decreased intra-modular connections in the parietal module and decreased inter-modular connections between the parietal and prefrontal modules. Notably, the global efficiency, characteristic path length, local efficiency of the right superior parietal gyrus, and inter-modular connections between the parietal and prefrontal modules were significantly associated with the number of depressive episodes. The betweenness centrality in SMA and the intra-modular connections in the parietal module showed a positive relationship with 17-item Hamilton Rating Scale for Depression (HAMD) scores. CONCLUSIONS The altered topology of MSN may serve as potential underlying pathological mechanisms of RDD. The impaired information integration of the network, particularly the disconnection within the fronto-parietal network, may be associated with the recurrence of depression. The SMA and the fronto-parietal network may be related to the severity of depression.
Collapse
Affiliation(s)
- Ziyun Xu
- Neuropsychiatry Imaging Center, Department of Radiology, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, 518020, China
| | - Zhifeng Zhou
- Neuropsychiatry Imaging Center, Department of Radiology, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, 518020, China
| | - Weiqun Tao
- Department of Psychiatry, Acute Intervention Female Ward 1, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, 518000, China
| | - Wentao Lai
- Neuropsychiatry Imaging Center, Department of Radiology, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, 518020, China
| | - Long Qian
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Wei Cui
- MR Research, GE Healthcare, Beijing, 100176, China
| | - Bo Peng
- Department of Depressive Disorder, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, 518000, China
| | - Yingli Zhang
- Department of Depressive Disorder, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, 518000, China.
| | - Gangqiang Hou
- Neuropsychiatry Imaging Center, Department of Radiology, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, 518020, China.
| |
Collapse
|
22
|
Snijders GJLJ, Gigase FAJ. Neuroglia in mood disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:287-302. [PMID: 40148049 DOI: 10.1016/b978-0-443-19102-2.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Multiple lines of evidence indicate that mood disorders, such as major depressive and bipolar disorder, are associated with abnormalities in neuroglial cells. This chapter discusses the existing literature investigating the potential role of astrocytes, oligodendrocytes, and microglia in mood pathology. We will describe evidence from in vivo imaging, postmortem, animal models based on (stress) paradigms that mimic depressive-like behavior, and biomarker studies in blood and cerebrospinal fluid in patients with mood disorders. The effect of medication used in the treatment of mood disorders, such as antidepressants and lithium, on glial function is discussed. Lastly, we highlight the most relevant findings about potential deficiencies in glia-glia crosstalk in mood disorders. Overall, decreased astrocyte and oligodendrocyte density and expression and microglial changes in homeostatic functions have frequently been put forward in MDD pathology. Studies of BD report similar findings to some extent; however, the evidence is less well established. Together, these findings are suggestive of reduced glial cell function leading to potential white matter abnormalities, glutamate dysregulation, disrupted neuronal functioning, and neurotransmission. However, more research is required to better understand the exact mechanisms underlying glial cell contributions to mood disorder development.
Collapse
Affiliation(s)
- Gijsje J L J Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Frederieke A J Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
23
|
Xu D, Liu G, Zhao M, Wan X, Qu Y, Murayama R, Hashimoto K. Effects of arketamine on depression-like behaviors and demyelination in mice exposed to chronic restrain stress: A role of transforming growth factor-β1. J Affect Disord 2024; 367:745-755. [PMID: 39236893 DOI: 10.1016/j.jad.2024.08.222] [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: 05/30/2024] [Revised: 08/19/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Chronic restrain stress (CRS) induces depression-like behaviors and demyelination in the brain; however, the relationship between these depression-like behaviors and demyelination remains unclear. Arketamine, the (R)-enantiomer of ketamine, has shown rapid antidepressant-like effects in CRS-exposed mice. METHODS We examined whether arketamine can improve both depression-like behaviors and demyelination in the brains of CRS-exposed mice. Additionally, we investigated the role of transforming growth factor β1 (TGF-β1) in the beneficial effects of arketamine. RESULTS A single dose of arketamine (10 mg/kg) improved both depression-like behavior and demyelination in the corpus callosum of CRS-exposed mice. Correlations were found between depression-like behaviors and demyelination in this region. Furthermore, pretreatment with RepSox, an inhibitor of TGF-β1 receptor, significantly blocked the beneficial effects of arketamine on depression-like behaviors and demyelination in CRS-exposed mice. Finally, a single intranasal administration of TGF-β1 ameliorated both depression-like behaviors and demyelination in CRS-exposed mice. LIMITATIONS The precise mechanisms by which TGF-β1 contributes to the effects of arketamine remain unclear. CONCLUSIONS These data suggest that CRS-induced demyelination in the corpus callosum may contribute to depression-like behaviors, and that arketamine can mitigate these changes through a TGF-β1-dependent mechanism.
Collapse
Affiliation(s)
- Dan Xu
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Guilin Liu
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, PR China
| | - Mingming Zhao
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Xiayun Wan
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Youge Qu
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Rumi Murayama
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| |
Collapse
|
24
|
Liu S, Zhou J, Zhu X, Zhang Y, Zhou X, Zhang S, Yang Z, Wang Z, Wang R, Yuan Y, Fang X, Chen X, Wang Y, Zhang L, Wang G, Jin C. An objective quantitative diagnosis of depression using a local-to-global multimodal fusion graph neural network. PATTERNS (NEW YORK, N.Y.) 2024; 5:101081. [PMID: 39776853 PMCID: PMC11701859 DOI: 10.1016/j.patter.2024.101081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/09/2024] [Accepted: 10/07/2024] [Indexed: 01/11/2025]
Abstract
This study developed an artificial intelligence (AI) system using a local-global multimodal fusion graph neural network (LGMF-GNN) to address the challenge of diagnosing major depressive disorder (MDD), a complex disease influenced by social, psychological, and biological factors. Utilizing functional MRI, structural MRI, and electronic health records, the system offers an objective diagnostic method by integrating individual brain regions and population data. Tested across cohorts from China, Japan, and Russia with 1,182 healthy controls and 1,260 MDD patients from 24 institutions, it achieved a classification accuracy of 78.75%, an area under the receiver operating characteristic curve (AUROC) of 80.64%, and correctly identified MDD subtypes. The system further discovered distinct brain connectivity patterns in MDD, including reduced functional connectivity between the left gyrus rectus and right cerebellar lobule VIIB, and increased connectivity between the left Rolandic operculum and right hippocampus. Anatomically, MDD is associated with thickness changes of the gray and white matter interface, indicating potential neuropathological conditions or brain injuries.
Collapse
Affiliation(s)
- Shuyu Liu
- Medical Robot Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jingjing Zhou
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xuequan Zhu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | - Ya Zhang
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China
- School of Electronic Information and Electronical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinzhu Zhou
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | - Shaoting Zhang
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China
| | - Zhi Yang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | - Ziji Wang
- Department of Cognitive Science, Swarthmore College, Philadelphia, PA 19081, USA
| | - Ruoxi Wang
- Medical Robot Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yizhe Yuan
- Medical Robot Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xin Fang
- Medical Robot Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiongying Chen
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | | | - Yanfeng Wang
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China
- School of Electronic Information and Electronical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ling Zhang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | - Gang Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Cheng Jin
- Medical Robot Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China
- Stanford University School of Medicine, Ground Floor, 875 Blake Wilbur Drive, Stanford, CA 94305-5847, USA
| |
Collapse
|
25
|
O'Connor EE, Salerno-Goncalves R, Rednam N, O'Brien R, Rock P, Levine AR, Zeffiro TA. Macro- and Microstructural White Matter Differences in Neurologic Postacute Sequelae of SARS-CoV-2 Infection. AJNR Am J Neuroradiol 2024; 45:1910-1918. [PMID: 39389778 PMCID: PMC11630878 DOI: 10.3174/ajnr.a8481] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/11/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND AND PURPOSE Neuropsychiatric complications of SARS-CoV-2 infection, also known as neurologic postacute sequelae of SARS-CoV-2 infection (NeuroPASC), affect 10%-60% of infected individuals. There is growing evidence that NeuroPASC is a multi system immune dysregulation disease affecting the brain. The behavioral manifestations of NeuroPASC, such as impaired processing speed, executive function, memory retrieval, and sustained attention, suggest widespread WM involvement. Although previous work has documented WM damage following acute SARS-CoV-2 infection, its involvement in NeuroPASC is less clear. We hypothesized that macrostructural and microstructural WM differences in NeuroPASC participants would accompany cognitive and immune system differences. MATERIALS AND METHODS In a cross-sectional study, we screened a total of 159 potential participants and enrolled 72 participants, with 41 asymptomatic controls (NoCOVID) and 31 NeuroPASC participants matched for age, sex, and education. Exclusion criteria included neurologic disorders unrelated to SARS-CoV-2 infection. Assessments included clinical symptom questionnaires, psychometric tests, brain MRI measures, and peripheral cytokine levels. Statistical modeling included separate multivariable regression analyses of GM/WM/CSF volume, WM microstructure, cognitive, and cytokine concentration between-group differences. RESULTS NeuroPASC participants had larger cerebral WM volume than NoCOVID controls (β = 0.229; 95% CI: 0.017-0.441; t = 2.16; P = .035). The most pronounced effects were in the prefrontal and anterior temporal WM. NeuroPASC participants also exhibited higher WM mean kurtosis, consistent with ongoing neuroinflammation. NeuroPASC participants had more self-reported symptoms, including headache, and lower performance on measures of attention, concentration, verbal learning, and processing speed. A multivariate profile analysis of the cytokine panel showed different group cytokine profiles (Wald-type-statistic = 44.6, P = .046), with interferon (IFN)-λ1 and IFN-λ2/3 levels higher in the NeuroPASC group. CONCLUSIONS NeuroPASC participants reported symptoms of lower concentration, higher fatigue, and impaired cognition compatible with WM syndrome. Psychometric testing confirmed these findings. NeuroPASC participants exhibited larger cerebral WM volume and higher WM mean kurtosis than NoCOVID controls. These findings suggest that immune dysregulation could influence WM properties to produce WM volume increases and consequent cognitive effects and headaches. Further work will be needed to establish mechanistic links among these variables.
Collapse
Affiliation(s)
- Erin E O'Connor
- From the Department of Diagnostic Radiology & Nuclear Medicine (E.E.O., N.R., T.A.Z.), University of Maryland School of Medicine, Baltimore, Maryland
| | | | - Nikita Rednam
- From the Department of Diagnostic Radiology & Nuclear Medicine (E.E.O., N.R., T.A.Z.), University of Maryland School of Medicine, Baltimore, Maryland
| | | | - Peter Rock
- Department of Anesthesiology (P.R.), University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrea R Levine
- Department of Medicine (A.R.L.), Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Thomas A Zeffiro
- From the Department of Diagnostic Radiology & Nuclear Medicine (E.E.O., N.R., T.A.Z.), University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
26
|
Sretavan K, Braun H, Liu Z, Bullock D, Palnitkar T, Patriat R, Chandrasekaran J, Brenny S, Johnson MD, Widge AS, Harel N, Heilbronner SR. A Reproducible Pipeline for Parcellation of the Anterior Limb of the Internal Capsule. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:1249-1261. [PMID: 39053578 DOI: 10.1016/j.bpsc.2024.07.008] [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: 06/04/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The anterior limb of the internal capsule (ALIC) is a white matter structure that connects the prefrontal cortex (PFC) to the brainstem, thalamus, and subthalamic nucleus. It is a target for deep brain stimulation for obsessive-compulsive disorder. There is strong interest in improving deep brain stimulation targeting by using diffusion tractography to reconstruct and target specific ALIC fiber pathways, but this methodology is susceptible to errors and lacks validation. To address these limitations, we developed a novel diffusion tractography pipeline that generates reliable and biologically validated ALIC white matter reconstructions. METHODS Following algorithm development and refinement, we analyzed 43 control participants, each with 2 sets of 3T magnetic resonance imaging data and a subset of 5 control participants with 7T data from the Human Connectome Project. We generated 22 segmented ALIC fiber bundles (11 per hemisphere) based on PFC regions of interest, and we analyzed the relationships among bundles. RESULTS We successfully reproduced the topographies established by previous anatomical work using images acquired at both 3T and 7T. Quantitative assessment demonstrated significantly smaller intraparticipant variability than interparticipant variability for both test and retest groups across all but one PFC region. We examined the overlap between fibers from different PFC regions and a response tract for obsessive-compulsive disorder deep brain stimulation, and we reconstructed the PFC hyperdirect pathway using a modified version of our pipeline. CONCLUSIONS Our diffusion magnetic resonance imaging algorithm reliably generates biologically validated ALIC white matter reconstructions, thereby allowing for more precise modeling of fibers for neuromodulation therapies.
Collapse
Affiliation(s)
- Karianne Sretavan
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota; Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| | - Henry Braun
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| | - Zoe Liu
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Daniel Bullock
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Tara Palnitkar
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| | - Remi Patriat
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| | - Jayashree Chandrasekaran
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| | - Samuel Brenny
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| | - Matthew D Johnson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Alik S Widge
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota
| | - Noam Harel
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota; Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota
| | | |
Collapse
|
27
|
Tura A, Promet L, Goya-Maldonado R. Structural-functional connectomics in major depressive disorder following aiTBS treatment. Psychiatry Res 2024; 342:116217. [PMID: 39369459 DOI: 10.1016/j.psychres.2024.116217] [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: 03/15/2024] [Revised: 08/16/2024] [Accepted: 09/22/2024] [Indexed: 10/08/2024]
Abstract
Major depressive disorder (MDD) has been associated with changes in the structural (SC) and functional connectivity (FC) of the brain. This study investigated the effects of accelerated intermittent theta burst stimulation (aiTBS) on SC-FC coupling and graph theory measures, focusing on the association between baseline SC-FC coupling of the dorsolateral prefrontal cortex (dlPFC) and clinical improvement. In a randomized, sham-controlled, quadruple-blind, crossover study, aiTBS was delivered to the left dlPFC of depressed patients with MDD, and diffusion tensor imaging (DTI) and resting-state functional magnetic resonance imaging (rsfMRI) data were acquired. In 77 MDD patients, significantly increased whole-brain SC-FC coupling was observed, primarily driven by default mode network (DMN) SC-FC coupling, along with increased somatomotor network FC, and decreased FC between the DMN hubs and limbic regions after active aiTBS. Furthermore, significant increases were observed in structural global and local efficiency measures that were not specific to the stimulation condition (active/sham aiTBS). However, these changes did not significantly correlate with clinical improvement. Notably, baseline SC-FC coupling of the left dlPFC was a significant predictor of clinical improvement. Our findings highlight the potential of left dlPFC SC-FC coupling as a predictor of aiTBS treatment outcomes, as well as the effect of aiTBS in enhancing SC-FC coupling.
Collapse
Affiliation(s)
- Asude Tura
- Laboratory of Systems Neuroscience and Imaging in Psychiatry (SNIP-Lab), Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), University of Göttingen, Göttingen, Germany
| | - Liisi Promet
- Laboratory of Systems Neuroscience and Imaging in Psychiatry (SNIP-Lab), Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), University of Göttingen, Göttingen, Germany
| | - Roberto Goya-Maldonado
- Laboratory of Systems Neuroscience and Imaging in Psychiatry (SNIP-Lab), Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
28
|
Neațu M, Ioniță I, Jugurt A, Davidescu EI, Popescu BO. Exploring the Complex Relationship Between Antidepressants, Depression and Neurocognitive Disorders. Biomedicines 2024; 12:2747. [PMID: 39767653 PMCID: PMC11727177 DOI: 10.3390/biomedicines12122747] [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: 10/29/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025] Open
Abstract
The coexistence of dementia and depression in older populations presents a complex clinical challenge, with each condition often exacerbating the other. Cognitive decline can intensify mood disturbances, and untreated or recurring depression accelerates neurodegenerative processes. As depression is a recognized risk factor for dementia, it is crucial to address both conditions concurrently to prevent further deterioration. Antidepressants are frequently used to manage depression in dementia patients, with some studies suggesting they offer neuroprotective benefits. These benefits include promoting neurogenesis, enhancing synaptic plasticity, and reducing neuroinflammation, potentially slowing cognitive decline. Additionally, antidepressants have shown promise in addressing Alzheimer's-related pathologies by reducing amyloid-beta accumulation and tau hyperphosphorylation. However, treatment-resistant depression remains a significant challenge, particularly in older adults with cognitive impairment. Many do not respond well to standard antidepressant therapies due to advanced neurodegenerative changes. Conflicting findings from studies add to the uncertainty, with some research suggesting that antidepressants may increase dementia risk, especially when used in patients with undiagnosed early-stage dementia. This article aims to explore the intricate relationship between depression and dementia, examining the benefits and risks of antidepressant use. We highlight the urgent need for personalized, comprehensive treatment strategies that balance mental health improvement with cognitive protection.
Collapse
Affiliation(s)
- Monica Neațu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Iulia Ioniță
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Ana Jugurt
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Eugenia Irene Davidescu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Bogdan Ovidiu Popescu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Department of Cell Biology, Neurosciences and Experimental Myology, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
29
|
Serrarens C, Kashyap S, Otter M, Campforts BCM, Stumpel CTRM, Linden DEJ, van Amelsvoort TAMJ, Vingerhoets C. White matter organization abnormalities in adults with 47,XXX: A 7 Tesla MRI study. Psychiatry Res Neuroimaging 2024; 345:111915. [PMID: 39546963 DOI: 10.1016/j.pscychresns.2024.111915] [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: 09/19/2024] [Revised: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
47,XXX (Triple X syndrome) is a sex chromosome aneuploidy characterized by the presence of a supernumerary X chromosome in affected females, and has been associated with a variable cognitive, behavioral, and psychiatric phenotype. Alterations in brain gray matter structure and function have been reported, but less is known about white matter (WM) organization in 47,XXX. Therefore, we conducted 7 T diffusion tensor imaging and characterized fractional anisotropy, mean diffusivity, axial diffusivity, and radial diffusivity of 22 adult women with 47,XXX and 22 age-matched typically developing females using tract-based spatial statistics. Relationships between phenotypic traits and WM organization characteristics in 47,XXX were also investigated. Adults with 47,XXX showed lower axial diffusivity in the body of the corpus callosum and the right superior longitudinal fasciculus. WM organization variability was not associated with IQ and social cognition and social functioning deficits in 47,XXX. Our findings indicate an effect of a supernumerary X chromosome in adult women on axonal integrity of the body of the corpus callosum and the right superior longitudinal fasciculus. These findings provide additional insight into the role of the X chromosome on WM organization. Future research is warranted to explore the clinical significant impact of altered WM organization in 47,XXX.
Collapse
Affiliation(s)
- Chaira Serrarens
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands.
| | - Sriranga Kashyap
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands; Krembil Brain Institute, University Health Network, Toronto, Canada
| | - Maarten Otter
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands; Medical Department, SIZA, Arnhem, the Netherlands
| | - Bea C M Campforts
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Constance T R M Stumpel
- Department of Clinical Genetics and School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - David E J Linden
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Thérèse A M J van Amelsvoort
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Claudia Vingerhoets
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands; 's Heeren Loo Zorggroep, Amersfoort, the Netherlands
| |
Collapse
|
30
|
van Velzen LS, Colic L, Ceja Z, Dauvermann MR, Villa LM, Savage HS, Toenders YJ, Dehestani N, Zhu AH, Campos AI, Salminen LE, Agartz I, Alexander N, Ayesa-Arriola R, Ballard ED, Banaj N, Barkhau C, Başgöze Z, Bauer J, Benedetti F, Berger K, Besteher B, Brosch K, Canal-Rivero M, Cervenka S, Colle R, Connolly CG, Corruble E, Courtet P, Couvy-Duchesne B, Crespo-Facorro B, Cullen KR, Dannlowski U, Deverdun J, Diaz-Zuluaga AM, Dietze LM, Evans JW, Fani N, Flinkenflügel K, Friedman NP, Gotlib IH, Groenewold NA, Grotegerd D, Hajek T, Hatoum AS, Hermesdorf M, Hickie IB, Hirano Y, Ho TC, Ikemizu Y, Iorfino F, Ipser JC, Isobe Y, Jackowski AP, Jollant F, Kircher T, Klug M, Koopowitz SM, Kraus A, Krug A, Le Bars E, Leehr EJ, Li M, Lippard ET, Lopez-Jaramillo C, Maximov II, McIntosh AM, McLaughlin KA, McWhinney SR, Meinert S, Melloni E, Mitchell PB, Mwangi B, Nenadić I, Nerland S, Olie E, Ortiz-García de la Foz V, Pan PM, Pereira F, Piras F, Piras F, Poletti S, Reineberg AE, Roberts G, Romero-García R, Sacchet MD, Salum GA, Sandu AL, Sellgren CM, Shimizu E, Smolker HR, Soares JC, Spalletta G, Douglas Steele J, Stein F, Stein DJ, Straube B, Teutenberg L, Thomas-Odenthal F, Usemann P, et alvan Velzen LS, Colic L, Ceja Z, Dauvermann MR, Villa LM, Savage HS, Toenders YJ, Dehestani N, Zhu AH, Campos AI, Salminen LE, Agartz I, Alexander N, Ayesa-Arriola R, Ballard ED, Banaj N, Barkhau C, Başgöze Z, Bauer J, Benedetti F, Berger K, Besteher B, Brosch K, Canal-Rivero M, Cervenka S, Colle R, Connolly CG, Corruble E, Courtet P, Couvy-Duchesne B, Crespo-Facorro B, Cullen KR, Dannlowski U, Deverdun J, Diaz-Zuluaga AM, Dietze LM, Evans JW, Fani N, Flinkenflügel K, Friedman NP, Gotlib IH, Groenewold NA, Grotegerd D, Hajek T, Hatoum AS, Hermesdorf M, Hickie IB, Hirano Y, Ho TC, Ikemizu Y, Iorfino F, Ipser JC, Isobe Y, Jackowski AP, Jollant F, Kircher T, Klug M, Koopowitz SM, Kraus A, Krug A, Le Bars E, Leehr EJ, Li M, Lippard ET, Lopez-Jaramillo C, Maximov II, McIntosh AM, McLaughlin KA, McWhinney SR, Meinert S, Melloni E, Mitchell PB, Mwangi B, Nenadić I, Nerland S, Olie E, Ortiz-García de la Foz V, Pan PM, Pereira F, Piras F, Piras F, Poletti S, Reineberg AE, Roberts G, Romero-García R, Sacchet MD, Salum GA, Sandu AL, Sellgren CM, Shimizu E, Smolker HR, Soares JC, Spalletta G, Douglas Steele J, Stein F, Stein DJ, Straube B, Teutenberg L, Thomas-Odenthal F, Usemann P, Valabregue R, Valencia-Echeverry J, Wagner G, Waiter G, Walter M, Whalley HC, Wu MJ, Yang TT, Zarate CA, Zugman A, Zunta-Soares GB, van Heeringen K, van Rooij SJ, van der Wee N, van der Werff S, Thompson PM, Blumberg HP, van Harmelen AL, Rentería ME, Jahanshad N, Schmaal L. Transdiagnostic alterations in white matter microstructure associated with suicidal thoughts and behaviours in the ENIGMA Suicidal Thoughts and Behaviours consortium. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.07.24316876. [PMID: 39802789 PMCID: PMC11722476 DOI: 10.1101/2024.11.07.24316876] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Previous studies have suggested that alterations in white matter (WM) microstructure are implicated in suicidal thoughts and behaviours (STBs). However, findings of diffusion tensor imaging (DTI) studies have been inconsistent. In this large-scale mega-analysis conducted by the ENIGMA Suicidal Thoughts and Behaviours (ENIGMA-STB) consortium, we examined WM alterations associated with STBs. Data processing was standardised across sites, and resulting WM microstructure measures (fractional anisotropy, axial diffusivity, mean diffusivity and radial diffusivity) for 25 WM tracts were pooled across 40 cohorts. We compared these measures among individuals with a psychiatric diagnosis and lifetime history of suicide attempt (n=652; mean age=35.4±14.7; female=71.8%), individuals with a psychiatric diagnosis but no STB (i.e., clinical controls; n=1871; mean age=34±14.8; female=59.8%), and individuals with no mental disorder diagnosis and no STB (i.e., healthy controls; n=642; mean age=29.6±13.1; female=62.9%). We also compared these measures among individuals with recent suicidal ideation (n=714; mean age=36.3±15.3; female=66.1%), clinical controls (n=1184; mean age=36.8±15.6; female=63.1%), and healthy controls (n=1240; mean age= 31.6±15.5; female=61.0%). We found subtle but statistically significant effects, such as lower fractional anisotropy associated with a history of suicide attempt, over and above the effect of psychiatric diagnoses. These effects were strongest in the corona radiata, thalamic radiation, fornix/stria terminalis, corpus callosum and superior longitudinal fasciculus. Effect sizes were small (Cohen's d < 0.25). Recent suicidal ideation was not associated with alterations in WM microstructure. This large-scale coordinated mega-analysis revealed subtle regional and global alterations in WM microstructure in individuals with a history of suicide attempt. Longitudinal studies are needed to confirm whether these alterations are a risk factor for suicidal behaviour.
Collapse
Affiliation(s)
- Laura S. van Velzen
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Lejla Colic
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
- DZPG (German Center for Mental Health), partner site Halle/Jena/Magdeburg
| | - Zuriel Ceja
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Maria R. Dauvermann
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Luca M. Villa
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Hannah S. Savage
- Centre for Youth Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Victoria, Australia
| | - Yara J. Toenders
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Department of Psychology, Education and Child Studies, Erasmus University Rotterdam, the Netherlands
| | - Niousha Dehestani
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Victoria, Australia
- School of Psychology, Deakin University, Victoria, Australia
| | - Alyssa H. Zhu
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | | | - Lauren E. Salminen
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Ingrid Agartz
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Nina Alexander
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain, and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Rosa Ayesa-Arriola
- Department of Psychiatry. Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain
| | - Elizabeth D. Ballard
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda MD, USA
| | - Nerisa Banaj
- Laboratory of Neuropsychiatry, Clinical Neuroscience and Neurorehabilitation Department, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Carlotta Barkhau
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Zeynep Başgöze
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jochen Bauer
- Clinic for Radiology, University Hospital Münster, Münster, Germany
| | - Francesco Benedetti
- Division of Neuroscience, Psychiatry and Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Bianca Besteher
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
- DZPG (German Center for Mental Health), partner site Halle/Jena/Magdeburg
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain, and Behavior (CMBB), University of Marburg, Marburg, Germany
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Manuel Canal-Rivero
- Instituto de Biomedicina de Sevilla (IBiS)/HUVR/CSIC/Universidad de Sevilla. CIBERSAM (ISCIII)
- Mental Health Service, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Romain Colle
- MOODS Team, INSERM 1018, CESP, Université Paris-Saclay, Faculté de Médecine Paris-Saclay, Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Colm G. Connolly
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL
| | - Emmanuelle Corruble
- MOODS Team, INSERM 1018, CESP, Université Paris-Saclay, Faculté de Médecine Paris-Saclay, Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Philippe Courtet
- Department of Emergency Psychiatry and Acute Care, Lapeyronie Hospital, CHU Montpellier, Montpellier, France
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Baptiste Couvy-Duchesne
- Institute for Molecular Bioscience, the University of Queensland, St Lucia, QLD, Australia
- Sorbonne University, Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, F-75013, Paris, France
| | - Benedicto Crespo-Facorro
- Instituto de Biomedicina de Sevilla (IBiS)/HUVR/CSIC/Universidad de Sevilla. CIBERSAM (ISCIII)
- Mental Health Service, Hospital Universitario Virgen del Rocío, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM), Madrid, Spain
| | - Kathryn R Cullen
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Jeremy Deverdun
- Institut d’Imagerie Fonctionnelle Humaine, I2FH, Department of Neuroradiology, Gui de Chauliac Hospital and University of Montpellier, Montpellier, France
- Department of Neuroradiology, Gui de Chauliac Hospital and University of Montpellier, Montpellier, France
| | - Ana M. Diaz-Zuluaga
- Center for Neurobehavioral Genetics,Semel Institute for Neuroscience and Behavior David Geffen School of Medicine, University of California Los Angeles
- Research Group in Psychiatry GIPSI, Department of Psychiatry, Faculty of Medicine, Universidad de Antioquia, Medellin, Columbia
| | | | - Jennifer W Evans
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda MD, USA
| | - Negar Fani
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia, USA
| | - Kira Flinkenflügel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Naomi P. Friedman
- Institute for Behavioral Genetics and Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Ian H. Gotlib
- Department of Psychology, Stanford University, Stanford, CA 94305 USA
| | - Nynke A. Groenewold
- Department of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tomas Hajek
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Alexander S. Hatoum
- Department of Psychological & Brain Sciences, Washington University in St. Louis
| | - Marco Hermesdorf
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | | | - Yoshiyuki Hirano
- Research Center for Child Mental Development, Chiba University
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University, Chiba University and University of Fukui
| | - Tiffany C. Ho
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yuki Ikemizu
- Research Center for Child Mental Development, Chiba University
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine, Chiba University
| | | | - Jonathan C. Ipser
- Department of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Yuko Isobe
- Research Center for Child Mental Development, Chiba University
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University, Chiba University and University of Fukui
| | - Andrea P. Jackowski
- Østfold University College Department of Education, ICT and Learning, Halden, Norway
- Universidade Federal de São Paulo, Brazil
| | - Fabrice Jollant
- MOODS Team, INSERM 1018, CESP, Université Paris-Saclay, Faculté de Médecine Paris-Saclay, Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
- Faculty of medicine, University Paris-Saclay & Bicetre hospital, APHP, Le Kremlin-Bicetre, France
- Department of psychiatry, CHU Nîmes, Nîmes, France
- Department of psychiatry, McGill University, Montreal, Canada
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain, and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Melissa Klug
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Sheri-Michelle Koopowitz
- Department of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Anna Kraus
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Department of Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Emmanuelle Le Bars
- Institut d’Imagerie Fonctionnelle Humaine, I2FH, Department of Neuroradiology, Gui de Chauliac Hospital and University of Montpellier, Montpellier, France
- Department of Neuroradiology, Gui de Chauliac Hospital and University of Montpellier, Montpellier, France
| | - Elisabeth J. Leehr
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Meng Li
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
- DZPG (German Center for Mental Health), partner site Halle/Jena/Magdeburg
| | - Elizabeth T.C. Lippard
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, University of Texas at Austin
- University of Texas at Austin
| | - Carlos Lopez-Jaramillo
- Research Group in Psychiatry GIPSI, Department of Psychiatry, Faculty of Medicine, Universidad de Antioquia, Medellin, Columbia
| | - Ivan I. Maximov
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Health and Functioning, Western Norway University of Applied Sciences, Bergen, Norway
| | - Andrew M. McIntosh
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Katie A. McLaughlin
- Ballmer Institute for Children’s Behavioral Health, University of Oregon
- Department of Psychology, Harvard University
| | | | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Elisa Melloni
- Division of Neuroscience, Psychiatry and Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Philip B. Mitchell
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Benson Mwangi
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston
- UTHealth Houston School of Behavioral Health Sciences
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain, and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Stener Nerland
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Emilie Olie
- Department of Emergency Psychiatry and Acute Care, Lapeyronie Hospital, CHU Montpellier, Montpellier, France
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Victor Ortiz-García de la Foz
- Department of Psychiatry. Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain
| | | | - Fabricio Pereira
- MIPA, Université de Nîmes, Nimes, France
- Division for clinical research and innovation, University Hospital Center of Nimes, Nimes, France
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, Clinical Neuroscience and Neurorehabilitation Department, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Federica Piras
- Laboratory of Neuropsychiatry, Clinical Neuroscience and Neurorehabilitation Department, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Sara Poletti
- Division of Neuroscience, Psychiatry and Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrew E. Reineberg
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Gloria Roberts
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Rafael Romero-García
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Instituto de Biomedicina de Sevilla (IBiS)/HUVR/CSIC/Universidad de Sevilla. CIBERSAM (ISCIII)
| | - Matthew D. Sacchet
- Meditation Research Program, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Giovanni A. Salum
- Child Mind Institute, New York
- Universidade Federal do Rio Grande do Sul, Hospital de Clinicas de Porto Alegre - Porto Alegre, Brazil
| | - Anca-Larisa Sandu
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Carl M. Sellgren
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eiji Shimizu
- Research Center for Child Mental Development, Chiba University
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University, Chiba University and University of Fukui
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine, Chiba University
| | - Harry R. Smolker
- Institute for Cognitive Science, University of Colorado Boulder, Boulder, CO, USA
| | - Jair C. Soares
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston
- UTHealth Houston School of Behavioral Health Sciences
| | - Gianfranco Spalletta
- Laboratory of Neuropsychiatry, Clinical Neuroscience and Neurorehabilitation Department, Santa Lucia Foundation IRCCS, Rome, Italy
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - J. Douglas Steele
- Division of Imaging Science and Technology, Medical School, University of Dundee, Dundee UK
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain, and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Dan J. Stein
- SAMRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry & Neuroscience Institute, University of Cape Town
| | - Benjamin Straube
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain, and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Lea Teutenberg
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain, and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Florian Thomas-Odenthal
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain, and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Paula Usemann
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain, and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Romain Valabregue
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, France
- Centre de Neuro-Imagerie de Recherche, CENIR, ICM, Paris, France
| | - Johanna Valencia-Echeverry
- Research Group in Psychiatry GIPSI, Department of Psychiatry, Faculty of Medicine, Universidad de Antioquia, Medellin, Columbia
| | - Gerd Wagner
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Jena-Magdeburg-Halle, Jena, Germany
| | - Gordon Waiter
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
- DZPG (German Center for Mental Health), partner site Halle/Jena/Magdeburg
- Department of Psychiatry and Psychotherapy, University Tübingen, Tübingen, Germany
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Heather C. Whalley
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Mon-Ju Wu
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston
- UTHealth Houston School of Behavioral Health Sciences
| | - Tony T. Yang
- Department of Psychiatry and Behavioral Sciences, Division of Child and Adolescent Psychiatry, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda MD, USA
| | - Andre Zugman
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Giovana B. Zunta-Soares
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston
- UTHealth Houston School of Behavioral Health Sciences
| | | | - Sanne J.H. van Rooij
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia, USA
| | - Nic van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition. Leiden University Medical Center, Leiden, The Netherlands
| | - Steven van der Werff
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition. Leiden University Medical Center, Leiden, The Netherlands
| | - 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, CA, USA
| | - Hilary P. Blumberg
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Anne-Laura van Harmelen
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Education and Child Studies, Leiden University, Leiden, the Netherlands
| | - Miguel E. Rentería
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | | | - Lianne Schmaal
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
31
|
Baltazar VA, Demchenko I, Tassone VK, Sousa-Ho RL, Schweizer TA, Bhat V. Brain-based correlates of depression and traumatic brain injury: a systematic review of structural and functional magnetic resonance imaging studies. FRONTIERS IN NEUROIMAGING 2024; 3:1465612. [PMID: 39563730 PMCID: PMC11573519 DOI: 10.3389/fnimg.2024.1465612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024]
Abstract
Introduction Depression is prevalent after traumatic brain injury (TBI). However, there is a lack of understanding of the brain-based correlates of depression post-TBI. This systematic review aimed to synthesize findings of structural and functional magnetic resonance imaging (MRI) studies to identify consistently reported neural correlates of depression post-TBI. Methods A search for relevant published studies was conducted through OVID (MEDLINE, APA PsycINFO, and Embase), with an end date of August 3rd, 2023. Fourteen published studies were included in this review. Results TBI patients with depression exhibited distinct changes in diffusion- based white matter fractional anisotropy, with the direction of change depending on the acuteness or chronicity of TBI. Decreased functional connectivity (FC) of the salience and default mode networks was prominent alongside the decreased volume of gray matter within the insular, dorsomedial prefrontal, and ventromedial prefrontal cortices. Seven studies reported the correlation between observed neuroimaging and depression outcomes. Of these studies, 42% indicated that FC of the bilateral medial temporal lobe subregions was correlated with depression outcomes in TBI. Discussion This systematic review summarizes existing neuroimaging evidence and reports brain regions that can be leveraged as potential treatment targets in future studies examining depression post-TBI.
Collapse
Affiliation(s)
- Vanessa A Baltazar
- Interventional Psychiatry Program, St. Michael's Hospital, Toronto, ON, Canada
| | - Ilya Demchenko
- Interventional Psychiatry Program, St. Michael's Hospital, Toronto, ON, Canada
- Temerty Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Vanessa K Tassone
- Interventional Psychiatry Program, St. Michael's Hospital, Toronto, ON, Canada
- Temerty Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Rachel L Sousa-Ho
- Interventional Psychiatry Program, St. Michael's Hospital, Toronto, ON, Canada
| | - Tom A Schweizer
- Temerty Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Neuroscience Research Program, St. Michael's Hospital, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Venkat Bhat
- Interventional Psychiatry Program, St. Michael's Hospital, Toronto, ON, Canada
- Temerty Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Neuroscience Research Program, St. Michael's Hospital, Toronto, ON, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Ye Z, Pan Y, McCoy RG, Bi C, Mo C, Feng L, Yu J, Lu T, Liu S, Carson Smith J, Duan M, Gao S, Ma Y, Chen C, Mitchell BD, Thompson PM, Elliot Hong L, Kochunov P, Ma T, Chen S. Contrasting association pattern of plasma low-density lipoprotein with white matter integrity in APOE4 carriers versus non-carriers. Neurobiol Aging 2024; 143:41-52. [PMID: 39213809 PMCID: PMC11514318 DOI: 10.1016/j.neurobiolaging.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Apolipoprotein E ε4 (APOE4) is a strong genetic risk factor of Alzheimer's disease and metabolic dysfunction. However, whether APOE4 and markers of metabolic dysfunction synergistically impact the deterioration of white matter (WM) integrity in older adults remains unknown. In the UK Biobank data, we conducted a multivariate analysis to investigate the interactions between APOE4 and 249 plasma metabolites (measured using nuclear magnetic resonance spectroscopy) with whole-brain WM integrity (measured by diffusion-weighted magnetic resonance imaging) in a cohort of 1917 older adults (aged 65.0-81.0 years; 52.4 % female). Although no main association was observed between either APOE4 or metabolites with WM integrity (adjusted P > 0.05), significant interactions between APOE4 and metabolites with WM integrity were identified. Among the examined metabolites, higher concentrations of low-density lipoprotein and very low-density lipoprotein were associated with a lower level of WM integrity (b=-0.12, CI=-0.14,-0.10) among APOE4 carriers. Conversely, among non-carriers, they were associated with a higher level of WM integrity (b=0.05, CI=0.04,0.07), demonstrating a significant moderation role of APOE4 (b =-0.18, CI=-0.20,-0.15, P<0.00001).
Collapse
Affiliation(s)
- Zhenyao Ye
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, United States; Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Yezhi Pan
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Rozalina G McCoy
- Division of Endocrinology, Diabetes, & Nutrition, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD 21201, United States; University of Maryland Institute for Health Computing, Bethesda, MD 20852, United States
| | - Chuan Bi
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Chen Mo
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Li Feng
- Department of Nutrition and Food Science, College of Agriculture & Natural Resources, University of Maryland, College Park, MD 20742, United States
| | - Jiaao Yu
- Department of Mathematics, University of Maryland, College Park, MD 20742, United States
| | - Tong Lu
- Department of Mathematics, University of Maryland, College Park, MD 20742, United States
| | - Song Liu
- School of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong 250353, China
| | - J Carson Smith
- Department of Kinesiology, University of Maryland, College Park, MD 20742, United States
| | - Minxi Duan
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Si Gao
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, United States
| | - Yizhou Ma
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, United States
| | - Chixiang Chen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, United States; University of Maryland Institute for Health Computing, Bethesda, MD 20852, United States
| | - Braxton D Mitchell
- Division of Endocrinology, Diabetes, & Nutrition, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - Paul M Thompson
- Imaging Genetics Center, Keck School of Medicine, University of Southern California, Marina del Rey, CA 90033, United States
| | - L Elliot Hong
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, United States
| | - Peter Kochunov
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, United States
| | - Tianzhou Ma
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, United States; Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20742, United States.
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, United States; Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, United States; University of Maryland Institute for Health Computing, Bethesda, MD 20852, United States.
| |
Collapse
|
33
|
Lam J, Mårtensson J, Westergren H, Svensson P, Sundgren PC, Alstergren P. Structural MRI findings in the brain related to pain distribution in chronic overlapping pain conditions: An explorative case-control study in females with fibromyalgia, temporomandibular disorder-related chronic pain and pain-free controls. J Oral Rehabil 2024; 51:2415-2426. [PMID: 39152537 DOI: 10.1111/joor.13842] [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: 02/13/2023] [Revised: 11/03/2023] [Accepted: 08/03/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Few neuroimaging studies have investigated structural brain differences associated with variations in pain distribution. OBJECTIVE To explore structural differences of the brain in fibromyalgia (FM), temporomandibular disorder pain (TMD) and healthy pain-free controls (CON) using structural and diffusion MRI. METHODS A case-control exploratory study with three study groups with different pain distribution were recruited: FM (n = 16; mean age [standard deviation]: 44 [14] years), TMD (n = 17, 39 [14] years) and CON (n = 10, 37 [14] years). Participants were recruited at the University Dental Clinic in Malmö, Sweden. T1-weighted and diffusion MRIs were acquired, clinical and psychosocial measures were obtained. Main outcome measures were subcortical volume, cortical thickness, white matter microstructure and whole brain grey matter intensity. RESULTS Patients with FM had smaller volume in the right thalamus than patients with TMD (p = .020) and CON (p = .030). The right thalamus volume was negatively correlated to pain intensity (r = -0.37, p = .022) and pain-related disability (r = -0.45, p = .004). The FM group had lower cortical thickness in the right anterior prefrontal cortex than CON (p = .005). Cortical thickness in this area was negatively correlated to pain intensity (r [37] = - 0.48, p = .002). CONCLUSIONS This study suggests that thalamus grey matter alterations are associated with FM and TMD, and that anterior prefrontal cortex grey matter alterations are associated with FM but not TMD. Studies on chronic overlapping pain conditions are needed in relation to possible nociplastic pain mechanisms in the brain and central nervous system.
Collapse
Affiliation(s)
- Julia Lam
- Department of Orofacial Pain and Jaw Function, Faculty of Odontology, Malmö University, Malmö, Sweden
- General Dental Care, Folktandvården Skåne, Lund, Sweden
- Scandinavian Center for Orofacial Neurosciences, Malmö, Sweden
| | - Johan Mårtensson
- Division of Logopedics, Phoniatrics and Audiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Hans Westergren
- Department of Health Sciences, Lund University, Lund, Sweden
| | - Peter Svensson
- Department of Orofacial Pain and Jaw Function, Faculty of Odontology, Malmö University, Malmö, Sweden
- Scandinavian Center for Orofacial Neurosciences, Malmö, Sweden
- Section for Orofacial Pain and Jaw Function, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Pia C Sundgren
- Department of Medical Imaging and Physiology, Skåne University Hospital Lund University, Lund, Sweden
- Division of Radiology, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University BioImaging Center, Lund University, Lund, Sweden
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Per Alstergren
- Department of Orofacial Pain and Jaw Function, Faculty of Odontology, Malmö University, Malmö, Sweden
- Scandinavian Center for Orofacial Neurosciences, Malmö, Sweden
- Specialised Pain Rehabilitation, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
34
|
Al-Sharif NB, Zavaliangos-Petropulu A, Narr KL. A review of diffusion MRI in mood disorders: mechanisms and predictors of treatment response. Neuropsychopharmacology 2024; 50:211-229. [PMID: 38902355 PMCID: PMC11525636 DOI: 10.1038/s41386-024-01894-3] [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: 04/08/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/22/2024]
Abstract
By measuring the molecular diffusion of water molecules in brain tissue, diffusion MRI (dMRI) provides unique insight into the microstructure and structural connections of the brain in living subjects. Since its inception, the application of dMRI in clinical research has expanded our understanding of the possible biological bases of psychiatric disorders and successful responses to different therapeutic interventions. Here, we review the past decade of diffusion imaging-based investigations with a specific focus on studies examining the mechanisms and predictors of therapeutic response in people with mood disorders. We present a brief overview of the general application of dMRI and key methodological developments in the field that afford increasingly detailed information concerning the macro- and micro-structural properties and connectivity patterns of white matter (WM) pathways and their perturbation over time in patients followed prospectively while undergoing treatment. This is followed by a more in-depth summary of particular studies using dMRI approaches to examine mechanisms and predictors of clinical outcomes in patients with unipolar or bipolar depression receiving pharmacological, neurostimulation, or behavioral treatments. Limitations associated with dMRI research in general and with treatment studies in mood disorders specifically are discussed, as are directions for future research. Despite limitations and the associated discrepancies in findings across individual studies, evolving research strongly indicates that the field is on the precipice of identifying and validating dMRI biomarkers that could lead to more successful personalized treatment approaches and could serve as targets for evaluating the neural effects of novel treatments.
Collapse
Affiliation(s)
- Noor B Al-Sharif
- Departments of Neurology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Artemis Zavaliangos-Petropulu
- Departments of Neurology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Katherine L Narr
- Departments of Neurology and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
35
|
Flinkenflügel K, Meinert S, Hirtsiefer C, Grotegerd D, Gruber M, Goltermann J, Winter NR, Stein F, Brosch K, Leehr EJ, Böhnlein J, Dohm K, Bauer J, Redlich R, Hahn T, Repple J, Opel N, Nitsch R, Jamalabadi H, Straube B, Alexander N, Jansen A, Nenadić I, van den Heuvel MP, Kircher T, Dannlowski U. Associations between white matter microstructure and cognitive decline in major depressive disorder versus controls in Germany: a prospective case-control cohort study. Lancet Psychiatry 2024; 11:899-909. [PMID: 39419563 DOI: 10.1016/s2215-0366(24)00291-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/18/2024] [Accepted: 09/04/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Cognitive deficits are a key source of disability in individuals with major depressive disorder (MDD) and worsen with disease progression. Despite their clinical relevance, the underlying mechanisms of cognitive deficits remain poorly elucidated, hampering effective treatment strategies. Emerging evidence suggests that alterations in white matter microstructure might contribute to cognitive dysfunction in MDD. We aimed to investigate the complex association between changes in white matter integrity, cognitive decline, and disease course in MDD in a comprehensive longitudinal dataset. METHODS In the naturalistic, observational, prospective, case-control Marburg-Münster Affective Disorders Cohort Study, individuals aged 18-65 years and of Caucasian ancestry were recruited from local psychiatric hospitals in Münster and Marburg, Germany, and newspaper advertisements. Individuals diagnosed with MDD and individuals without any history of psychiatric disorder (ie, healthy controls) were included in this subsample analysis. Participants had diffusion-weighted imaging, a battery of neuropsychological tests, and detailed clinical data collected at baseline and at 2 years of follow-up. We used linear mixed-effect models to compare changes in cognitive performance and white matter integrity between participants with MDD and healthy controls. Diffusion-weighted imaging analyses were conducted using tract-based spatial statistics. To correct for multiple comparisons, threshold free cluster enhancement (TFCE) was used to correct α-values at the family-wise error rate (FWE; ptfce-FWE). Effect sizes were estimated by conditional, partial R2 values (sr2) following the Nakagawa and Schielzeth method to quantify explained variance. The association between changes in cognitive performance and changes in white matter integrity was analysed. Finally, we examined whether the depressive disease course between assessments predicted cognitive performance at follow-up and whether white matter integrity mediated this association. People with lived experience were not involved in the research and writing process. FINDINGS 881 participants were selected for our study, of whom 418 (47%) had MDD (mean age 36·8 years [SD 13·4], 274 [66%] were female, and 144 [34%] were male) and 463 (53%) were healthy controls (mean age 35·6 years [13·5], 295 [64%] were female, and 168 [36%] were male). Baseline assessments were done between Sept 11, 2014, and June 3, 2019, and after a mean follow-up of 2·20 years (SD 0·19), follow-up assessments were done between Oct 6, 2016, and May 31, 2021. Participants with MDD had lower cognitive performance than did healthy controls (p<0·0001, sr2=0·056), regardless of timepoint. Analyses of diffusion-weighted imaging indicated a significant diagnosis × time interaction with a steeper decline in white matter integrity of the superior longitudinal fasciculus over time in participants with MDD than in healthy controls (ptfce-FWE=0·026, sr2=0·002). Furthermore, cognitive decline was robustly associated with the decline in white matter integrity over time across both groups (ptfce-FWE<0·0001, sr2=0·004). In participants with MDD, changes in white matter integrity (p=0·0040, β=0·071) and adverse depressive disease course (p=0·0022, β=-0·073) independently predicted lower cognitive performance at follow-up. INTERPRETATION Alterations of white matter integrity occurred over time to a greater extent in participants with MDD than in healthy controls, and decline in white matter integrity was associated with a decline in cognitive performance across groups. Our findings emphasise the crucial role of white matter microstructure and disease progression in depression-related cognitive dysfunction, making both priority targets for future treatment development. FUNDING German Research Foundation (DFG).
Collapse
Affiliation(s)
- Kira Flinkenflügel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany; Institute for Translational Neuroscience, University of Münster, Münster, Germany.
| | | | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Marius Gruber
- Institute for Translational Psychiatry, University of Münster, Münster, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Nils R Winter
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany; Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Elisabeth J Leehr
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Joscha Böhnlein
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Dohm
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Jochen Bauer
- Department of Radiology, University of Münster, Münster, Germany
| | - Ronny Redlich
- Institute for Translational Psychiatry, University of Münster, Münster, Germany; Department of Psychology, University of Halle, Halle, Germany; German Center for Mental Health (DZPG), Halle-Jena-Magdeburg, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Jonathan Repple
- Institute for Translational Psychiatry, University of Münster, Münster, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Nils Opel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany; German Center for Mental Health (DZPG), Halle-Jena-Magdeburg, Germany; Department of Psychiatry, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Robert Nitsch
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Hamidreza Jamalabadi
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany; Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Benjamin Straube
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany; Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Nina Alexander
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany; Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany; Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany; Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany; Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Martijn P van den Heuvel
- Connectome Laboratory, Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands; Department of Child Psychiatry, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany; Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| |
Collapse
|
36
|
Guo Y, Liu Y, Zhang T, Ruan J, Liu S, Ren Z. Intrinsic disruption of white matter microarchitecture in major depressive disorder: A voxel-based meta analysis of diffusion tensor imaging. J Affect Disord 2024; 363:161-173. [PMID: 39032713 DOI: 10.1016/j.jad.2024.07.050] [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: 12/12/2023] [Revised: 06/17/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is a prevalent and disabling mood disorder, thought to be linked with brain white matter (WM) alterations. Prior diffusion tensor imaging (DTI) studies have reported inconsistent changes in fractional anisotropy (FA) across different brain regions in MDD patients. However, none of these studies utilized raw t-map data for WM meta-analysis in MDD. Our study aims to address this gap by conducting a whole-brain-based meta-analysis of FA in MDD using Seed-based d mapping via permutation of subject images (SDM-PSI), combining reported peak coordinates and raw statistical parametric maps. OBJECTIVES Following PRISMA guidelines, we performed a systematic search and meta-analysis to compare FA in MDD patients with healthy controls (HC). Our goal was to identify WM abnormalities in MDD, using SDM, which could shed light on the disorder's pathogenesis. RESULTS The meta-analysis included 39 studies with 3696 participants (2094 with MDD, 1602HC). It revealed that MDD patients, in comparison to HC, have lower FA in the corpus callosum (CC) and anterior thalamic projections (ATP). Subgroup analyses indicated that the CC is a more stable pathogenic factor in MDD. Meta-regression analyses showed no linear correlation between the mean age, percentage of female patients, duration of depression, and FA abnormalities. This suggests that WM impairments in interhemispheric connections and anterior thalamocortical circuits are significant in the pathogenesis of MDD.
Collapse
Affiliation(s)
- Yunxiao Guo
- Key Laboratory of Adolescent Cyberpsychology and Behavior (Ministry of Education), School of Psychology, Central China Normal University, Wuhan, China; Key Laboratory of Human Development and Mental Health of Hubei Province, National Intelligent Society Governance Experiment Base (Education), School of Psychology, Central China Normal University, Wuhan, China
| | - Yinong Liu
- Key Laboratory of Adolescent Cyberpsychology and Behavior (Ministry of Education), School of Psychology, Central China Normal University, Wuhan, China; Key Laboratory of Human Development and Mental Health of Hubei Province, National Intelligent Society Governance Experiment Base (Education), School of Psychology, Central China Normal University, Wuhan, China
| | - Tao Zhang
- Key Laboratory of Adolescent Cyberpsychology and Behavior (Ministry of Education), School of Psychology, Central China Normal University, Wuhan, China; Key Laboratory of Human Development and Mental Health of Hubei Province, National Intelligent Society Governance Experiment Base (Education), School of Psychology, Central China Normal University, Wuhan, China
| | - Jun Ruan
- Key Laboratory of Adolescent Cyberpsychology and Behavior (Ministry of Education), School of Psychology, Central China Normal University, Wuhan, China; Key Laboratory of Human Development and Mental Health of Hubei Province, National Intelligent Society Governance Experiment Base (Education), School of Psychology, Central China Normal University, Wuhan, China
| | - Sijun Liu
- Key Laboratory of Adolescent Cyberpsychology and Behavior (Ministry of Education), School of Psychology, Central China Normal University, Wuhan, China; Key Laboratory of Human Development and Mental Health of Hubei Province, National Intelligent Society Governance Experiment Base (Education), School of Psychology, Central China Normal University, Wuhan, China
| | - Zhihong Ren
- Key Laboratory of Adolescent Cyberpsychology and Behavior (Ministry of Education), School of Psychology, Central China Normal University, Wuhan, China; Key Laboratory of Human Development and Mental Health of Hubei Province, National Intelligent Society Governance Experiment Base (Education), School of Psychology, Central China Normal University, Wuhan, China.
| |
Collapse
|
37
|
Zhu Y, Wei Y, Yu X, Liu J, Lan R, Guo X, Luo Y. Altered sleep onset transition in depression: Evidence from EEG activity and EEG functional connectivity analyses. Clin Neurophysiol 2024; 166:129-141. [PMID: 39163676 DOI: 10.1016/j.clinph.2024.08.002] [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: 08/14/2023] [Revised: 08/01/2024] [Accepted: 08/03/2024] [Indexed: 08/22/2024]
Abstract
OBJECTIVE Sleep disorders constitute a principal diagnostic criterion for depression, potentially reflecting the abnormal persistence of brain activity during the sleep onset (SO) transition. Here, we sought to explore the differences in the dynamic changes in the EEG activity and the EEG functional connectivity (FC) during the SO transition in depressed patients. METHODS Overnight polysomnography recordings were obtained from thirty-two depressed patients and thirty-three healthy controls. The multiscale permutation entropy (MSPE) and EEG relative power were extracted to characterize EEG activity, and weighted phase lag index (WPLI) was calculated to characterize EEG FC. RESULTS The intergroup differences in EEG activity of relative power and MSPE were reversed near SO, which attributed to slower rates of change among depressed patients. Regarding the characteristics of the EEG FC network, depressed patients exhibited significantly higher inter-hemispheric and interregional WPLI values in both delta and alpha bands throughout the SO transition, concomitant with different dynamic properties in the delta band FC. During the process after SO, patients exhibited increased inter-hemispheric long-range links, whereas controls showed more intra-hemispheric ones. Finally, we found significant correlations in the dynamic changes between different EEG measures. CONCLUSIONS Our research revealed that the abnormal changes during the SO transition in depressed patients were manifested in both homeostatic and dynamic aspects, which were reflected in EEG FC and EEG activity, respectively. SIGNIFICANCE These findings may elucidate the mechanism underlying sleep disorders in depression from the perspective of neural activity.
Collapse
Affiliation(s)
- Yongpeng Zhu
- School of Biomedical Engineering, Sun Yat-sen University-Shenzhen Campus, Shenzhen 518000, China
| | - Yu Wei
- School of Biomedical Engineering, Sun Yat-sen University-Shenzhen Campus, Shenzhen 518000, China
| | - Xiaokang Yu
- School of Biomedical Engineering, Sun Yat-sen University-Shenzhen Campus, Shenzhen 518000, China
| | - Jiahao Liu
- School of Biomedical Engineering, Sun Yat-sen University-Shenzhen Campus, Shenzhen 518000, China
| | - Rongxi Lan
- School of Biomedical Engineering, Sun Yat-sen University-Shenzhen Campus, Shenzhen 518000, China
| | - Xinwen Guo
- The Seventh Affiliated Hospital of Southern Medical University, Foshan 528000, China.
| | - Yuxi Luo
- School of Biomedical Engineering, Sun Yat-sen University-Shenzhen Campus, Shenzhen 518000, China; Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, Sun Yat-sen University-Shenzhen Campus, Shenzhen 518000, China.
| |
Collapse
|
38
|
Flinkenflügel K, Gruber M, Meinert S, Thiel K, Winter A, Goltermann J, Usemann P, Brosch K, Stein F, Thomas-Odenthal F, Wroblewski A, Pfarr JK, David FS, Beins EC, Grotegerd D, Hahn T, Leehr EJ, Dohm K, Bauer J, Forstner AJ, Nöthen MM, Jamalabadi H, Straube B, Alexander N, Jansen A, Witt SH, Rietschel M, Nenadić I, van den Heuvel MP, Kircher T, Repple J, Dannlowski U. The interplay between polygenic score for tumor necrosis factor-α, brain structural connectivity, and processing speed in major depression. Mol Psychiatry 2024; 29:3151-3159. [PMID: 38693319 PMCID: PMC11449800 DOI: 10.1038/s41380-024-02577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024]
Abstract
Reduced processing speed is a core deficit in major depressive disorder (MDD) and has been linked to altered structural brain network connectivity. Ample evidence highlights the involvement of genetic-immunological processes in MDD and specific depressive symptoms. Here, we extended these findings by examining associations between polygenic scores for tumor necrosis factor-α blood levels (TNF-α PGS), structural brain connectivity, and processing speed in a large sample of MDD patients. Processing speed performance of n = 284 acutely depressed, n = 177 partially and n = 198 fully remitted patients, and n = 743 healthy controls (HC) was estimated based on five neuropsychological tests. Network-based statistic was used to identify a brain network associated with processing speed. We employed general linear models to examine the association between TNF-α PGS and processing speed. We investigated whether network connectivity mediates the association between TNF-α PGS and processing speed. We identified a structural network positively associated with processing speed in the whole sample. We observed a significant negative association between TNF-α PGS and processing speed in acutely depressed patients, whereas no association was found in remitted patients and HC. The mediation analysis revealed that brain connectivity partially mediated the association between TNF-α PGS and processing speed in acute MDD. The present study provides evidence that TNF-α PGS is associated with decreased processing speed exclusively in patients with acute depression. This association was partially mediated by structural brain connectivity. Using multimodal data, the current findings advance our understanding of cognitive dysfunction in MDD and highlight the involvement of genetic-immunological processes in its pathomechanisms.
Collapse
Grants
- WI 3439/3-1, WI 3439/3-2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- RI 908/11-1, RI 908/11-2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- JA 1890/7-1, JA 1890/7-2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- EP-C-16-015 EPA
- DA1151/5-1, DA1151/5-2, DA1151/11‑1 DA1151/6-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- NO 246/10-1, NO 246/10-2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- HA7070/2-2, HA7070/3, HA7070/4 Deutsche Forschungsgemeinschaft (German Research Foundation)
- STR 1146/18-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- ERC-COG 101001062, VIDI-452-16-015 Nederlandse Organisatie voor Wetenschappelijk Onderzoek (Netherlands Organisation for Scientific Research)
- KI 588/14-1, KI 588/14-2, KI 588/22-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- Interdisziplinäres Zentrum für Klinische Forschung, medizinische Fakultät, Münster (Dan3/012/17)
- Innovative medizinische Forschung Münster (IMF): RE111604, RE111722, RE 221707
Collapse
Affiliation(s)
- Kira Flinkenflügel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Marius Gruber
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Katharina Thiel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Alexandra Winter
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Paula Usemann
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), 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, Marburg, Germany
| | - Florian Thomas-Odenthal
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Adrian Wroblewski
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Julia-Katharina Pfarr
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Friederike S David
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Eva C Beins
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Elisabeth J Leehr
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Dohm
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Jochen Bauer
- Department of Radiology, University of Münster, Münster, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Hamidreza Jamalabadi
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Benjamin Straube
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Nina Alexander
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
- Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Martijn P van den Heuvel
- Connectome Lab, Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Child Psychiatry, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
| | - Jonathan Repple
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany.
| |
Collapse
|
39
|
Feng L, Milleson HS, Ye Z, Canida T, Ke H, Liang M, Gao S, Chen S, Hong LE, Kochunov P, Lei DKY, Ma T. Nongenetic and Genetic Factors Associated with White Matter Brain Aging: Exposome-Wide and Genome-Wide Association Study. Genes (Basel) 2024; 15:1285. [PMID: 39457408 PMCID: PMC11507416 DOI: 10.3390/genes15101285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Human brain aging is a complex process that affects various aspects of brain function and structure, increasing susceptibility to neurological and psychiatric disorders. A number of nongenetic (e.g., environmental and lifestyle) and genetic risk factors are found to contribute to the varying rates at which the brain ages among individuals. METHODS In this paper, we conducted both an exposome-wide association study (XWAS) and a genome-wide association study (GWAS) on white matter brain aging in the UK Biobank, revealing the multifactorial nature of brain aging. We applied a machine learning algorithm and leveraged fractional anisotropy tract measurements from diffusion tensor imaging data to predict the white matter brain age gap (BAG) and treated it as the marker of brain aging. For XWAS, we included 107 variables encompassing five major categories of modifiable exposures that potentially impact brain aging and performed both univariate and multivariate analysis to select the final set of nongenetic risk factors. RESULTS We found current tobacco smoking, dietary habits including oily fish, beef, lamb, cereal, and coffee intake, length of mobile phone use, use of UV protection, and frequency of solarium/sunlamp use were associated with the BAG. In genetic analysis, we identified several SNPs on chromosome 3 mapped to genes IP6K1, GMNC, OSTN, and SLC25A20 significantly associated with the BAG, showing the high heritability and polygenic architecture of human brain aging. CONCLUSIONS The critical nongenetic and genetic risk factors identified in our study provide insights into the causal relationship between white matter brain aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Li Feng
- Department of Nutrition and Food Science, College of Agriculture & Natural Resources, University of Maryland, College Park, MD 20740, USA; (L.F.); (D.K.Y.L.)
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
| | - Halley S. Milleson
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
- Department of Mathematics, The College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, MD 20740, USA
| | - Zhenyao Ye
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21228, USA; (Z.Y.); (S.G.); (S.C.)
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Travis Canida
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
- Department of Mathematics, The College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, MD 20740, USA
| | - Hongjie Ke
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
| | - Menglu Liang
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
| | - Si Gao
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21228, USA; (Z.Y.); (S.G.); (S.C.)
- Louis A. Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.E.H.); (P.K.)
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21228, USA; (Z.Y.); (S.G.); (S.C.)
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - L. Elliot Hong
- Louis A. Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.E.H.); (P.K.)
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.E.H.); (P.K.)
| | - David K. Y. Lei
- Department of Nutrition and Food Science, College of Agriculture & Natural Resources, University of Maryland, College Park, MD 20740, USA; (L.F.); (D.K.Y.L.)
| | - Tianzhou Ma
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
| |
Collapse
|
40
|
Roelofs EF, Bas-Hoogendam JM, Winkler AM, van der Wee NJ, Vermeiren RRM. Longitudinal development of resting-state functional connectivity in adolescents with and without internalizing disorders. NEUROSCIENCE APPLIED 2024; 3:104090. [PMID: 39634556 PMCID: PMC11615185 DOI: 10.1016/j.nsa.2024.104090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Longitudinal studies using resting-state functional magnetic resonance imaging (rs-fMRI) focused on adolescent internalizing psychopathology are scarce and have mostly investigated standardized treatment effects on functional connectivity (FC) of the full amygdala. The role of amygdala subregions and large resting-state networks had yet to be elucidated, and treatment is in practice often personalized. Here, longitudinal FC development of amygdala subregions and whole-brain networks are investigated in a clinically representative sample. Treatment-naïve adolescents with clinical depression and comorbid anxiety who started care-as-usual (n = 23; INT) and healthy controls (n = 24; HC) participated in rs-fMRI scans and questionnaires at baseline (before treatment) and after three months. Changes between and within groups over time in FC of the laterobasal amygdala (LBA), centromedial amygdala (CMA) and whole-brain networks derived from independent component analysis (ICA) were investigated. Groups differed significantly in FC development of the right LBA to the postcentral gyrus and the left LBA to the frontal pole. Within INT, FC to the frontal pole and postcentral gyrus changed over time while changes in FC of the right LBA were also linked to symptom change. No significant interactions were observed when considering FC from CMA bilateral seeds or within ICA-derived networks. Results in this cohort suggest divergent longitudinal development of FC from bilateral LBA subregions in adolescents with internalizing disorders compared to healthy peers, possibly reflecting nonspecific treatment effects. Moreover, associations were found with symptom change. These results highlight the importance of differentiation of amygdala subregions in neuroimaging research in adolescents.
Collapse
Affiliation(s)
- Eline F. Roelofs
- LUMC-Curium, Department of Child and Adolescent Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden, the Netherlands
| | - Janna Marie Bas-Hoogendam
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden, the Netherlands
- Developmental and Educational Psychology, Institute of Psychology, Leiden University, Leiden, the Netherlands
| | - Anderson M. Winkler
- Section on Development and Affective Neuroscience (SDAN), Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Nic J.A. van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden, the Netherlands
| | - Robert R.J. M. Vermeiren
- LUMC-Curium, Department of Child and Adolescent Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden, the Netherlands
| |
Collapse
|
41
|
Xu Y, Cheng X, Li Y, Shen H, Wan Y, Ping L, Yu H, Cheng Y, Xu X, Cui J, Zhou C. Shared and Distinct White Matter Alterations in Major Depression and Bipolar Disorder: A Systematic Review and Meta-Analysis. J Integr Neurosci 2024; 23:170. [PMID: 39344242 DOI: 10.31083/j.jin2309170] [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: 04/21/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Identifying white matter (WM) microstructural similarities and differences between major depressive disorder (MDD) and bipolar disorder (BD) is an important way to understand the potential neuropathological mechanism in emotional disorders. Numerous diffusion tensor imaging (DTI) studies over recent decades have confirmed the presence of WM anomalies in these two affective disorders, but the results were inconsistent. This study aimed to determine the statistical consistency of DTI findings for BD and MDD by using the coordinate-based meta-analysis (CBMA) approach. METHODS We performed a systematic search of tract-based spatial statistics (TBSS) studies comparing MDD or BD with healthy controls (HC) as of June 30, 2024. The seed-based d-mapping (SDM) was applied to investigate fractional anisotropy (FA) changes. Meta-regression was then used to analyze the potential correlations between demographics and neuroimaging alterations. RESULTS Regional FA reductions in the body of the corpus callosum (CC) were identified in both of these two diseases. Besides, MDD patients also exhibited decreased FA in the genu and splenium of the CC, as well as the left anterior thalamic projections (ATP), while BD patients showed FA reduction in the left median network, and cingulum in addition to the CC. CONCLUSIONS The results highlighted that altered integrity in the body of CC served as the shared basis of MDD and BD, and distinct microstructural WM abnormalities also existed, which might induce the various clinical manifestations of these two affective disorders. The study was registered on PROSPERO (http://www.crd.york.ac.uk/PROSPERO), registration number: CRD42022301929.
Collapse
Affiliation(s)
- Yinghong Xu
- Department of Psychiatry, Shandong Daizhuang Hospital, 272075 Jining, Shandong, China
- School of Mental Health, Jining Medical University, 272002 Jining, Shandong, China
| | - Xiaodong Cheng
- Department of Psychiatry, Shandong Daizhuang Hospital, 272075 Jining, Shandong, China
| | - Ying Li
- School of Mental Health, Jining Medical University, 272002 Jining, Shandong, China
| | - Hailong Shen
- School of Mental Health, Jining Medical University, 272002 Jining, Shandong, China
| | - Yu Wan
- School of Mental Health, Jining Medical University, 272002 Jining, Shandong, China
| | - Liangliang Ping
- Department of Psychiatry, Xiamen Xianyue Hospital, 361012 Xiamen, Fujian, China
| | - Hao Yu
- School of Mental Health, Jining Medical University, 272002 Jining, Shandong, China
| | - Yuqi Cheng
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan, China
| | - Xiufeng Xu
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan, China
| | - Jian Cui
- Department of Psychiatry, Shandong Daizhuang Hospital, 272075 Jining, Shandong, China
| | - Cong Zhou
- School of Mental Health, Jining Medical University, 272002 Jining, Shandong, China
- Department of Psychology, Affiliated Hospital of Jining Medical University, 272067 Jining, Shandong, China
| |
Collapse
|
42
|
Rojczyk P, Seitz-Holland J, Heller C, Marcolini S, Marshall AD, Sydnor VJ, Kaufmann E, Jung LB, Bonke EM, Berger L, Umminger LF, Wiegand TLT, Cho KIK, Rathi Y, Bouix S, Pasternak O, Hinds SR, Fortier CB, Salat D, Milberg WP, Shenton ME, Koerte IK. Posttraumatic survivor guilt is associated with white matter microstructure alterations. J Affect Disord 2024; 361:768-777. [PMID: 38897303 DOI: 10.1016/j.jad.2024.06.047] [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: 10/27/2023] [Revised: 05/31/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Military veterans with posttraumatic stress disorder (PTSD) commonly experience posttraumatic guilt. Guilt over commission or omission evolves when responsibility is assumed for an unfortunate outcome (e.g., the death of a fellow combatant). Survivor guilt is a state of intense emotional distress experienced by the weight of knowing that one survived while others did not. METHODS This study of the Translational Research Center for TBI and Stress Disorders (TRACTS) analyzed structural and diffusion-weighted magnetic resonance imaging data from 132 male Iraq/Afghanistan veterans with PTSD. The Clinician-Administered PTSD Scale for DSM-IV (CAPS-IV) was employed to classify guilt. Thirty (22.7 %) veterans experienced guilt over acts of commission or omission, 34 (25.8 %) experienced survivor guilt, and 68 (51.5 %) had no posttraumatic guilt. White matter microstructure (fractional anisotropy, FA), cortical thickness, and cortical volume were compared between veterans with guilt over acts of commission or omission, veterans with survivor guilt, and veterans without guilt. RESULTS Veterans with survivor guilt had significantly lower white matter FA compared to veterans who did not experience guilt (p < .001), affecting several regions of major white matter fiber bundles. There were no significant differences in white matter FA, cortical thickness, or volumes between veterans with guilt over acts of commission or omission and veterans without guilt (p > .050). LIMITATIONS This cross-sectional study with exclusively male veterans precludes inferences of causality between the studied variables and generalizability to the larger veteran population that includes women. CONCLUSION Survivor guilt may be a particularly impactful form of posttraumatic guilt that requires specific treatment efforts targeting brain health.
Collapse
Affiliation(s)
- Philine Rojczyk
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | - Johanna Seitz-Holland
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carina Heller
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany; Department of Clinical Psychology, Friedrich Schiller University Jena, Jena, Germany; German Center for Mental Health (DZPG), Partner Site Jena-Magdeburg-Halle, Jena, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Partner Site Jena-Magdeburg-Halle, Jena, Germany
| | - Sofia Marcolini
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, Alzheimer Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Amy D Marshall
- Department of Psychology, The Pennsylvania State University, PA, USA
| | - Valerie J Sydnor
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elisabeth Kaufmann
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Leonard B Jung
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | - Elena M Bonke
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | - Luisa Berger
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | - Lisa F Umminger
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | - Tim L T Wiegand
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | - Kang Ik K Cho
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yogesh Rathi
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sylvain Bouix
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Software Engineering and Information Technology, École de technologie supérieure, Université du Québec, Montréal, QC, Canada
| | - Ofer Pasternak
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sidney R Hinds
- Department of Neurology, Uniformed Services University, Bethesda, MD, USA
| | - Catherine B Fortier
- Translational Research Center for TBI and Stress Disorders (TRACTS) and Geriatric Research, Education and Clinical Center (GRECC), VA Boston Healthcare System, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - David Salat
- Translational Research Center for TBI and Stress Disorders (TRACTS) and Geriatric Research, Education and Clinical Center (GRECC), VA Boston Healthcare System, Boston, MA, USA; Neuroimaging Research for Veterans (NeRVe) Center, VA Boston Healthcare System, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital Department of Radiology, Boston, MA, USA
| | - William P Milberg
- Translational Research Center for TBI and Stress Disorders (TRACTS) and Geriatric Research, Education and Clinical Center (GRECC), VA Boston Healthcare System, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Neuroimaging Research for Veterans (NeRVe) Center, VA Boston Healthcare System, Boston, MA, USA
| | - Martha E Shenton
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, VA Boston Healthcare System, Brockton, MA, USA
| | - Inga K Koerte
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, VA Boston Healthcare System, Brockton, MA, USA; Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
43
|
Owens-Walton C, Nir TM, Al-Bachari S, Ambrogi S, Anderson TJ, Aventurato ÍK, Cendes F, Chen YL, Ciullo V, Cook P, Dalrymple-Alford JC, Dirkx MF, Druzgal J, Emsley HCA, Guimarães R, Haroon HA, Helmich RC, Hu MT, Johansson ME, Kim HB, Klein JC, Laansma M, Lawrence KE, Lochner C, Mackay C, McMillan CT, Melzer TR, Nabulsi L, Newman B, Opriessnig P, Parkes LM, Pellicano C, Piras F, Piras F, Pirpamer L, Pitcher TL, Poston KL, Roos A, Silva LS, Schmidt R, Schwingenschuh P, Shahid-Besanti M, Spalletta G, Stein DJ, Thomopoulos SI, Tosun D, Tsai CC, van den Heuvel OA, van Heese E, Vecchio D, Villalón-Reina JE, Vriend C, Wang JJ, Wu YR, Yasuda CL, Thompson PM, Jahanshad N, van der Werf Y. A worldwide study of white matter microstructural alterations in people living with Parkinson's disease. NPJ Parkinsons Dis 2024; 10:151. [PMID: 39128907 PMCID: PMC11317500 DOI: 10.1038/s41531-024-00758-3] [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: 01/17/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024] Open
Abstract
The progression of Parkinson's disease (PD) is associated with microstructural alterations in neural pathways, contributing to both motor and cognitive decline. However, conflicting findings have emerged due to the use of heterogeneous methods in small studies. Here we performed a large diffusion MRI study in PD, integrating data from 17 cohorts worldwide, to identify stage-specific profiles of white matter differences. Diffusion-weighted MRI data from 1654 participants diagnosed with PD (age: 20-89 years; 33% female) and 885 controls (age: 19-84 years; 47% female) were analyzed using the ENIGMA-DTI protocol to evaluate white matter microstructure. Skeletonized maps of fractional anisotropy (FA) and mean diffusivity (MD) were compared across Hoehn and Yahr (HY) disease groups and controls to reveal the profile of white matter alterations at different stages. We found an enhanced, more widespread pattern of microstructural alterations with each stage of PD, with eventually lower FA and higher MD in almost all regions of interest: Cohen's d effect sizes reached d = -1.01 for FA differences in the fornix at PD HY Stage 4/5. The early PD signature in HY stage 1 included higher FA and lower MD across the entire white matter skeleton, in a direction opposite to that typical of other neurodegenerative diseases. FA and MD were associated with motor and non-motor clinical dysfunction. While overridden by degenerative changes in the later stages of PD, early PD is associated with paradoxically higher FA and lower MD in PD, consistent with early compensatory changes associated with the disorder.
Collapse
Grants
- R01 AG058854 NIA NIH HHS
- P41 EB015922 NIBIB NIH HHS
- R01NS107513 U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01 MH117601 NIMH NIH HHS
- R01 NS107513 NINDS NIH HHS
- U19 AG062418 NIA NIH HHS
- F32 MH122057 NIMH NIH HHS
- R01 AG059874 NIA NIH HHS
- U.S. Alzheimer’s Association (AARG-23-1149996)
- Health Research Council of New Zealand (20/538; 21/165)
- São Paulo Research Foundation FAPESP-BRAINN Grants# 2013-07559-3 / FAPESP #2022-1178-4
- São Paulo Research Foundation FAPESP-BRAINN Grant # 2013–07559-3.
- Health Research Council of New Zealand (20/538); Marsden Fund New Zealand (UOC2105); Neurological Foundation of New Zealand (2232 PRG); Research and Education Trust Pacific Radiology (MRIJDA).
- Grant from ParkinsonNL (P2023-14); Honoraria from Movement Disorders Society Quebec.
- NINDS R01NS107513
- Engineering and Physical Sciences Research Council (EPSRC) UK
- Parkinson's UK, Cure Parkinsons Trust, Oxford Biomedical Research Centre, GSK-Oxford IMCM.
- JK is supported by the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), and the NIHR Oxford Health Clinical Research Facility. The views expressed are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health.
- NIMH 32MH122057
- U19 AG062418
- Health Research Council of New Zealand (20/538); Neurological Foundation of New Zealand (2232 PRG); Research and Education Trust Pacific Radiology (MRIJDA).
- EPSRC UK, MRC UK, GE medical systems, Academy of Medical Sciences UK
- Italian Ministry of Health, grant number RF-2019-12370182
- Health Research Council of New Zealand (21/165)
- Personal fees from Bial, AbbVie and Boston Scientific.
- NIH/NIA
- São Paulo Research Foundation FAPESP-BRAINN Grant # 2013–07559-3; CNPQ (#315953/2021-7) National Council for Scientific and Technological Development
- U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01AG059874, R01MH117601, R01NS107513, R01AG058854, P41EB015922
Collapse
Affiliation(s)
- Conor Owens-Walton
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA.
| | - Talia M Nir
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | | | - Sonia Ambrogi
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Tim J Anderson
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Neurology Department, Te Whatu Ora-Health New Zealand Waitaha Canterbury, Christchurch, New Zealand
| | - Ítalo Karmann Aventurato
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Fernando Cendes
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Yao-Liang Chen
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung, Taiwan, ROC
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan, ROC
| | - Valentina Ciullo
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Phil Cook
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John C Dalrymple-Alford
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Te Kura Mahi ā- Hirikapo | School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - Michiel F Dirkx
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jason Druzgal
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Hedley C A Emsley
- Lancaster Medical School, Lancaster University, Lancaster, UK
- Department of Neurology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| | - Rachel Guimarães
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Hamied A Haroon
- Division of Psychology, Communication & Human Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Rick C Helmich
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Michele T Hu
- Oxford Parkinson's Disease Centre, Nuffield, Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK
| | - Martin E Johansson
- Department of Neurology and Center of Expertise for Parkinson & Movement Disorders, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Ho Bin Kim
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Johannes C Klein
- Oxford Parkinson's Disease Centre, Nuffield, Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK
| | - Max Laansma
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Katherine E Lawrence
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Christine Lochner
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Cape Town, South Africa
| | - Clare Mackay
- Oxford Parkinson's Disease Centre, Nuffield, Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford, UK
| | - Corey T McMillan
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Tracy R Melzer
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Te Kura Mahi ā- Hirikapo | School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - Leila Nabulsi
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Ben Newman
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Peter Opriessnig
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Laura M Parkes
- Division of Psychology, Communication & Human Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Clelia Pellicano
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Federica Piras
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Lukas Pirpamer
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Toni L Pitcher
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Kathleen L Poston
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Annerine Roos
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Lucas Scárdua Silva
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Reinhold Schmidt
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Petra Schwingenschuh
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Marian Shahid-Besanti
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | | | - Dan J Stein
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Chih-Chien Tsai
- Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan, ROC
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Odile A van den Heuvel
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eva van Heese
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Daniela Vecchio
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Julio E Villalón-Reina
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Chris Vriend
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Amsterdam UMC, Department of Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging program, Amsterdam, The Netherlands
| | - Jiun-Jie Wang
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung, Taiwan, ROC
- Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan, ROC
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan City, Taiwan, ROC
- Department of Chemical Engineering, Ming-Chi University of Technology, New Taipei City, Taiwan, ROC
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan, ROC
- Department of Neurology, College of Medicine, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Clarissa Lin Yasuda
- Department of Neurology, University of Campinas-UNICAMP, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - 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, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Ysbrand van der Werf
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Amsterdam UMC, Dept. Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Khodanovich M, Svetlik M, Kamaeva D, Usova A, Kudabaeva M, Anan’ina T, Vasserlauf I, Pashkevich V, Moshkina M, Obukhovskaya V, Kataeva N, Levina A, Tumentceva Y, Vasilieva S, Schastnyy E, Naumova A. Demyelination in Patients with POST-COVID Depression. J Clin Med 2024; 13:4692. [PMID: 39200834 PMCID: PMC11355865 DOI: 10.3390/jcm13164692] [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: 06/14/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Depression is one of the most severe sequelae of COVID-19, with major depressive disorder often characterized by disruption in white matter (WM) connectivity stemming from changes in brain myelination. This study aimed to quantitatively assess brain myelination in clinically diagnosed post-COVID depression (PCD) using the recently proposed MRI method, macromolecular proton fraction (MPF) mapping. Methods: The study involved 63 recovered COVID-19 patients (52 mild, 11 moderate, and 2 severe) at 13.5 ± 10.0 months post-recovery, with matched controls without prior COVID-19 history (n = 19). A post-COVID depression group (PCD, n = 25) was identified based on psychiatric diagnosis, while a comparison group (noPCD, n = 38) included participants with neurological COVID-19 complications, excluding clinical depression. Results: Fast MPF mapping revealed extensive demyelination in PCD patients, particularly in juxtacortical WM (predominantly occipital lobe and medial surface), WM tracts (inferior fronto-occipital fasciculus (IFOF), posterior thalamic radiation, external capsule, sagittal stratum, tapetum), and grey matter (GM) structures (hippocampus, putamen, globus pallidus, and amygdala). The noPCD group also displayed notable demyelination, but with less magnitude and propagation. Multiple regression analysis highlighted IFOF demyelination as the primary predictor of Hamilton scores, PCD presence, and severity. The number of post-COVID symptoms was a significant predictor of PCD presence, while the number of acute symptoms was a significant predictor of PCD severity. Conclusions: This study, for the first time, reveals extensive demyelination in numerous WM and GM structures in PCD, outlining IFOF demyelination as a key biomarker.
Collapse
Affiliation(s)
- Marina Khodanovich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Mikhail Svetlik
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Daria Kamaeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Anna Usova
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 12/1 Savinykh Street, Tomsk 634028, Russia
| | - Marina Kudabaeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Tatyana Anan’ina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Irina Vasserlauf
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Valentina Pashkevich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Marina Moshkina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Victoria Obukhovskaya
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Department of Fundamental Psychology and Behavioral Medicine, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 634050, Russia
| | - Nadezhda Kataeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Department of Neurology and Neurosurgery, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 634028, Russia
| | - Anastasia Levina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Medica Diagnostic and Treatment Center, 86 Sovetskaya Street, Tomsk 634510, Russia
| | - Yana Tumentceva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Svetlana Vasilieva
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Evgeny Schastnyy
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Anna Naumova
- Department of Radiology, School of Medicine, South Lake Union Campus, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| |
Collapse
|
45
|
Hédouin R, Roy JC, Desmidt T, Robert G, Coloigner J. Microstructural brain assessment in late-life depression and apathy using diffusion MRI multi-compartments models and tractometry. Sci Rep 2024; 14:18193. [PMID: 39107406 PMCID: PMC11303796 DOI: 10.1038/s41598-024-67535-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Late-life depression (LLD) is both common and disabling and doubles the risk of dementia onset. Apathy might constitute an additional risk of cognitive decline but clear understanding of its pathophysiology is lacking. While white matter (WM) alterations have been assessed using diffusion tensor imaging (DTI), this model cannot accurately represent WM microstructure. We hypothesized that a more complex multi-compartment model would provide new biomarkers of LLD and apathy. Fifty-six individuals (LLD n = 35, 26 females, 75.2 ± 6.4 years, apathy evaluation scale scores (41.8 ± 8.7) and Healthy controls, n = 21, 16 females, 74.7 ± 5.2 years) were included. In this article, a tract-based approach was conducted to investigate novel diffusion model biomarkers of LLD and apathy by interpolating microstructural metrics directly along the fiber bundle. We performed multivariate statistical analysis, combined with principal component analysis for dimensional data reduction. We then tested the utility of our framework by demonstrating classically reported from the literature modifications in LDD while reporting new results of biological-basis of apathy in LLD. Finally, we aimed to investigate the relationship between apathy and microstructure in different fiber bundles. Our study suggests that new fiber bundles, such as the striato-premotor tracts, may be involved in LLD and apathy, which bring new light of apathy mechanisms in major depression. We also identified statistical changes in diffusion MRI metrics in 5 different tracts, previously reported in major cognitive disorders dementia, suggesting that these alterations among these tracts are both involved in motivation and cognition and might explain how apathy is a prodromal phase of degenerative disorders.
Collapse
Affiliation(s)
- Renaud Hédouin
- Univ Rennes, INRIA, CNRS, INSERM, IRISA UMR 6074, Empenn ERL U 1228, 35000, Rennes, France
| | - Jean-Charles Roy
- Univ Rennes, INRIA, CNRS, INSERM, IRISA UMR 6074, Empenn ERL U 1228, 35000, Rennes, France
- CIC 1414, CHU de Rennes, INSERM, Rennes, France
- Adult University Psychiatry Department, Guillaume Régnier Hospital, Rennes, France
| | - Thomas Desmidt
- CHU de Tours, Tours, France
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
- CIC 1415, CHU de Tours, INSERM, Tours, France
| | - Gabriel Robert
- Univ Rennes, INRIA, CNRS, INSERM, IRISA UMR 6074, Empenn ERL U 1228, 35000, Rennes, France
- CIC 1414, CHU de Rennes, INSERM, Rennes, France
- Adult University Psychiatry Department, Guillaume Régnier Hospital, Rennes, France
| | - Julie Coloigner
- Univ Rennes, INRIA, CNRS, INSERM, IRISA UMR 6074, Empenn ERL U 1228, 35000, Rennes, France.
| |
Collapse
|
46
|
Colombo F, Calesella F, Bravi B, Fortaner-Uyà L, Monopoli C, Tassi E, Carminati M, Zanardi R, Bollettini I, Poletti S, Lorenzi C, Spadini S, Brambilla P, Serretti A, Maggioni E, Fabbri C, Benedetti F, Vai B. Multimodal brain-derived subtypes of Major depressive disorder differentiate patients for anergic symptoms, immune-inflammatory markers, history of childhood trauma and treatment-resistance. Eur Neuropsychopharmacol 2024; 85:45-57. [PMID: 38936143 DOI: 10.1016/j.euroneuro.2024.05.015] [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: 11/21/2023] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/29/2024]
Abstract
An estimated 30 % of Major Depressive Disorder (MDD) patients exhibit resistance to conventional antidepressant treatments. Identifying reliable biomarkers of treatment-resistant depression (TRD) represents a major goal of precision psychiatry, which is hampered by the clinical and biological heterogeneity. To uncover biologically-driven subtypes of MDD, we applied an unsupervised data-driven framework to stratify 102 MDD patients on their neuroimaging signature, including extracted measures of cortical thickness, grey matter volumes, and white matter fractional anisotropy. Our novel analytical pipeline integrated different machine learning algorithms to harmonize data, perform data dimensionality reduction, and provide a stability-based relative clustering validation. The obtained clusters were characterized for immune-inflammatory peripheral biomarkers, TRD, history of childhood trauma and depressive symptoms. Our results indicated two different clusters of patients, differentiable with 67 % of accuracy: one cluster (n = 59) was associated with a higher proportion of TRD, and higher scores of energy-related depressive symptoms, history of childhood abuse and emotional neglect; this cluster showed a widespread reduction in cortical thickness (d = 0.43-1.80) and volumes (d = 0.45-1.05), along with fractional anisotropy in the fronto-occipital fasciculus, stria terminalis, and corpus callosum (d = 0.46-0.52); the second cluster (n = 43) was associated with cognitive and affective depressive symptoms, thicker cortices and wider volumes. Multivariate analyses revealed distinct brain-inflammation relationships between the two clusters, with increase in pro-inflammatory markers being associated with decreased cortical thickness and volumes. Our stratification of MDD patients based on structural neuroimaging identified clinically-relevant subgroups of MDD with specific symptomatic and immune-inflammatory profiles, which can contribute to the development of tailored personalized interventions for MDD.
Collapse
Affiliation(s)
- Federica Colombo
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy.
| | - Federico Calesella
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Beatrice Bravi
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Lidia Fortaner-Uyà
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Camilla Monopoli
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Emma Tassi
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Politecnico di Milano, Department of Electronics, Information and Bioengineering, Milan, Italy
| | | | - Raffaella Zanardi
- University Vita-Salute San Raffaele, Milano, Italy; Mood Disorders Unit, Scientific Institute IRCCS San Raffaele Hospital, Milan, Italy
| | - Irene Bollettini
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Sara Poletti
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Sara Spadini
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Eleonora Maggioni
- Politecnico di Milano, Department of Electronics, Information and Bioengineering, Milan, Italy
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Francesco Benedetti
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Benedetta Vai
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| |
Collapse
|
47
|
Li Q, Zhao Y, Hu Y, Liu Y, Wang Y, Zhang Q, Long F, Chen Y, Wang Y, Li H, Poels EMP, Kamperman AM, Sweeney JA, Kuang W, Li F, Gong Q. Linked patterns of symptoms and cognitive covariation with functional brain controllability in major depressive disorder. EBioMedicine 2024; 106:105255. [PMID: 39032426 PMCID: PMC11324849 DOI: 10.1016/j.ebiom.2024.105255] [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: 12/25/2023] [Revised: 06/14/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Controllability analysis is an approach developed for evaluating the ability of a brain region to modulate function in other regions, which has been found to be altered in major depressive disorder (MDD). Both depressive symptoms and cognitive impairments are prominent features of MDD, but the case-control differences of controllability between MDD and controls can not fully interpret the contribution of both clinical symptoms and cognition to brain controllability and linked patterns among them in MDD. METHODS Sparse canonical correlation analysis was used to investigate the associations between resting-state functional brain controllability at the network level and clinical symptoms and cognition in 99 first-episode medication-naïve patients with MDD. FINDINGS Average controllability was significantly correlated with clinical features. The average controllability of the dorsal attention network (DAN) and visual network had the highest correlations with clinical variables. Among clinical variables, depressed mood, suicidal ideation and behaviour, impaired work and activities, and gastrointestinal symptoms were significantly negatively associated with average controllability, and reduced cognitive flexibility was associated with reduced average controllability. INTERPRETATION These findings highlight the importance of brain regions in modulating activity across brain networks in MDD, given their associations with symptoms and cognitive impairments observed in our study. Disrupted control of brain reconfiguration of DAN and visual network during their state transitions may represent a core brain mechanism for the behavioural impairments observed in MDD. FUNDING National Natural Science Foundation of China (82001795 and 82027808), National Key R&D Program (2022YFC2009900), and Sichuan Science and Technology Program (2024NSFSC0653).
Collapse
Affiliation(s)
- Qian Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China
| | - Youjin Zhao
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China
| | - Yongbo Hu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China
| | - Yang Liu
- Academy of Mathematics and Systems Science Chinese, Academy of Science, Beijing, China
| | - Yaxuan Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China
| | - Qian Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China
| | - Fenghua Long
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China
| | - Yufei Chen
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China
| | - Yitian Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China
| | - Haoran Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China
| | - Eline M P Poels
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Astrid M Kamperman
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - John A Sweeney
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Department of Psychiatry and Behavioural Neuroscience, University of Cincinnati, Cincinnati, OH, 45219, USA
| | - Weihong Kuang
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Fei Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China.
| | - Qiyong Gong
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, PR China; Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China.
| |
Collapse
|
48
|
Liu Y, Yin J, Li X, Yang J, Liu Y. Examining the connection between weekend catch-up sleep and depression: Insights from 2017 to 2020 NHANES information. J Affect Disord 2024; 358:61-69. [PMID: 38705524 DOI: 10.1016/j.jad.2024.05.022] [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: 01/28/2024] [Revised: 04/07/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Depression, a prevalent mental disorder, has shown an increasing trend in recent years, imposing a significant burden on health and society. Adequate sleep has been proven to reduce the incidence of depression. This study seeks to explore how Weekend Catch-up Sleep (WCS) is connected with the prevalence of depression in the American population. METHODS The National Health and Nutrition Examination Survey (NHANES) provides representative data for the U.S. POPULATION We utilized data from the 2017-2018 and 2019-2020 cycles. Depression was operationally defined as a PHQ-9 score exceeding 10. WCS duration was categorized into five groups: no change in sleep duration (=0 h), decreased sleep duration (<0), short catch-up sleep duration (>0 h, ≤1 h), moderate catch-up sleep duration (>1 h, <2 h), and long catch-up sleep duration (≥2 h). RESULTS Among the 8039 individuals, the distribution of WCS duration was as follows: no change (WCS = 0 h) in 2999 individuals (37.3 %), decreased sleep (WCS < 0 h) in 1199 individuals (14.9 %), short catch-up sleep (0 h < WCS ≤ 1 h) in 1602 individuals (19.9 %), moderate catch-up sleep (1 h < WCS < 2 h) in 479 individuals (6.0 %), and long catch-up sleep (WCS ≥ 2 h) in 1760 individuals (21.9 %). Acting by adjustment for all covariates in a multiple regression analysis, we discovered that persons with 1 to 2 h of weekend catch-up sleep had a substantially low prevalence of depression concerning those with WCS = 0 (OR 0.22, 95 % CI 0.08-0.59, P = 0.007). CONCLUSION The prevalence of depression in individuals engaging in weekend catch-up sleep for 1 to 2 h is lower than those who do not catch up on weekends. This discovery on the treatment and prevention of depression provides a new perspective. However, further prospective research and clinical trials are needed for a comprehensive investigation.
Collapse
Affiliation(s)
- Yecun Liu
- College of First Clinical Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiahui Yin
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuhao Li
- College of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiguo Yang
- College of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yuanxiang Liu
- Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, China.
| |
Collapse
|
49
|
Lei X, Fang X, Ren J, Teng X, Guo C, Wu Z, Yu L, Wang D, Chen Y, Zhou Y, Wu Y, Zhang Y, Zhang C. Plasma Apo-E mediated corticospinal tract abnormalities and suicidality in patients with major depressive disorder. Eur Arch Psychiatry Clin Neurosci 2024; 274:1167-1175. [PMID: 38265467 DOI: 10.1007/s00406-023-01749-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024]
Abstract
This study aims to explore the link between Apo-E, brain white matter, and suicide in patients with major depressive disorder (MDD) to investigate the potential neuroimmune mechanisms of Apo-E that may lead to suicide. Thirty-nine patients with MDD (22 patients with suicidality) and 57 age, gender, and education-matched healthy controls participated in this study, provided plasma Apo-E samples, and underwent diffusion tensor imaging scans. Plasma Apo-E levels and white matter microstructure were analyzed among the MDD with suicidality, MDD without suicidality, and HC groups using analysis of variance with post hoc Bonferroni correction and tract-based spatial statistics (TBSS) with threshold-free cluster enhancement correction. Mediation analysis investigated the relationship between Apo-E, brain white matter, and suicidality in MDD. The MDD with suicidality subgroup had higher depressive and suicide scores, longer disease course, and lower plasma Apo-E levels than MDD without suicidality. TBSS revealed that the MDD non-suicide subgroup showed significantly increased mean diffusivity in the left corticospinal tract and body of the left corpus callosum, as well as increased axial diffusivity in the left anterior corona radiata and the right posterior thalamic radiation compared to the suicidal MDD group. The main finding was that the increased MD of the left corticospinal tract contributed to the elevated suicide score, with Apo-E mediating the effect. Preliminary result that Apo-E's mediating role between the left corticospinal tract and the suicide factor suggests the neuroimmune mechanism of suicide in MDD. The study was registered on ClinicalTrials.gov (NCT03790085).
Collapse
Affiliation(s)
- Xiaoxia Lei
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyu Fang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Ren
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyue Teng
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaoyue Guo
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zenan Wu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingfang Yu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dandan Wang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Chen
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunshan Zhou
- Department of Psychiatry, Huaian No. 3 People's Hospital, Huaian, Jiangsu, China
| | - Yujie Wu
- Department of Psychiatry, Shanghai Xuhui Mental Health Center, Shanghai, China
| | - Yi Zhang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai, 200030, China.
| | - Chen Zhang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai, 200030, China.
| |
Collapse
|
50
|
Bavato F, Barro C, Schnider LK, Simrén J, Zetterberg H, Seifritz E, Quednow BB. Introducing neurofilament light chain measure in psychiatry: current evidence, opportunities, and pitfalls. Mol Psychiatry 2024; 29:2543-2559. [PMID: 38503931 PMCID: PMC11412913 DOI: 10.1038/s41380-024-02524-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
The recent introduction of new-generation immunoassay methods allows the reliable quantification of structural brain markers in peripheral matrices. Neurofilament light chain (NfL), a neuron-specific cytoskeletal component released in extracellular matrices after neuroaxonal impairment, is considered a promising blood marker of active brain pathology. Given its sensitivity to a wide range of neuropathological alterations, NfL has been suggested for the use in clinical practice as a highly sensitive, but unspecific tool to quantify active brain pathology. While large efforts have been put in characterizing its clinical profile in many neurological conditions, NfL has received far less attention as a potential biomarker in major psychiatric disorders. Therefore, we briefly introduce NfL as a marker of neuroaxonal injury, systematically review recent findings on cerebrospinal fluid and blood NfL levels in patients with primary psychiatric conditions and highlight the opportunities and pitfalls. Current evidence suggests an elevation of blood NfL levels in patients with major depression, bipolar disorder, psychotic disorders, anorexia nervosa, and substance use disorders compared to physiological states. However, blood NfL levels strongly vary across diagnostic entities, clinical stage, and patient subgroups, and are influenced by several demographic, clinical, and analytical factors, which require accurate characterization. Potential clinical applications of NfL measure in psychiatry are seen in diagnostic and prognostic algorithms, to exclude neurodegenerative disease, in the assessment of brain toxicity for different pharmacological compounds, and in the longitudinal monitoring of treatment response. The high inter-individual variability of NfL levels and the lack of neurobiological understanding of its release are some of the main current limitations. Overall, this primer aims to introduce researchers and clinicians to NfL measure in the psychiatric field and to provide a conceptual framework for future research directions.
Collapse
Affiliation(s)
- Francesco Bavato
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy and Psychosomatics; Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Christian Barro
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura K Schnider
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy and Psychosomatics; Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics; Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Boris B Quednow
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy and Psychosomatics; Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|