1
|
Rabl U, Bartova L, Sezen P, Keller J, Schatzberg A, Pezawas L. HPA axis in psychotic and non-psychotic major depression: Cortisol plasma levels and hippocampal volume. J Affect Disord 2025; 377:14-22. [PMID: 39955073 DOI: 10.1016/j.jad.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Psychotic major depression (PMD) differs from non-psychotic MD (NPMD) in psychopathology and is linked to changes in brain volumetry and hypothalamic-pituitary-adrenal (HPA) axis function that can be reflected by its principal output - the glucocorticoid cortisol. NPMD patients exhibit smaller hippocampi than healthy controls (HC), purportedly representing exposure to chronic stress. However, the relationship between the individual clinical phenotype, hippocampal volume and diurnal cortisol signaling remains unclear. METHODS Since understanding the interplay among symptoms, neuroimaging and HPA function is crucial for discerning biological differences between PMD and NPMD, this study explored the link between clinical phenotype, hippocampal structural MRI and circadian plasma cortisol levels in 32 HC, 27 NPMD and 26 PMD patients. RESULTS PMD patients showed significantly elevated evening (6 p.m. - 1 a.m.) cortisol levels compared to NPMD and HC, while NPMD and HC did not differ. No group differences in hippocampal volume were observed, but a significant interaction effect emerged between overnight (1 a.m. - 9 a.m.) cortisol levels, hippocampal volume, and clinical phenotype. NPMD patients displayed a negative correlation between overnight cortisol levels and hippocampal volume, which was specific to the ascending cortisol curve (2 a.m. - 5 a.m.) and absent in PMD and HC. The hippocampus-cortisol interaction was associated with depressive symptom severity in NPMD but not PMD, where cortisol alone predicted greater severity. CONCLUSIONS These findings imply a time-dependent relationship between hippocampal volume and overnight cortisol in NPMD, which is absent in PMD and HC. In contrast, PMD patients exhibited increased evening cortisol levels. In an exploratory analysis, these effects were also related to symptom severity at similar timepoints. While correlational, these results point to distinct neurobiological mechanisms underlying NPMD and PMD, which are potentially related to the heterogeneous clinical manifestations.
Collapse
Affiliation(s)
- U Rabl
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - L Bartova
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - P Sezen
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - J Keller
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - A Schatzberg
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - L Pezawas
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
2
|
Malykhin N, Serrano J, Reiz B, Hegadoren K, Pietrasik W, Whittal R. Effects of Variations in Daily Cortisol Pattern and Long-Term Cortisol Output on Hippocampal Subfield Volumes in the Adult Human Brain. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100458. [PMID: 40201775 PMCID: PMC11978376 DOI: 10.1016/j.bpsgos.2025.100458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 04/10/2025] Open
Abstract
Background Animal models of adult chronic stress indicate that the cornu ammonis 1-3 (CA1-3) and dentate gyrus (DG) hippocampal subfields are most susceptible to cellular changes associated with prolonged psychogenic stressors and glucocorticoid overexposure. However, no study reported to date has examined associations between long-term cortisol output, chronic stress, and hippocampal subfield volumes in healthy adults experiencing different levels of chronic stress. The main goal of the current study was to test whether higher long-term cortisol output measured by hair cortisol concentration would be associated with atrophy of CA1-3 and DG hippocampal subfields. Methods We examined associations between short- and long-term cortisol output and hippocampal subfield volumes in healthy adults (N = 40). High-resolution structural magnetic resonance imaging datasets were acquired together with diurnal salivary cortisol and hair cortisol measures. Hair cortisol concentration was analyzed using the high-resolution liquid chromatography-mass spectrometry method. Results Higher hair cortisol concentration was associated with smaller volumes of all hippocampal subfields in the anterior hippocampus and smaller DG volumes in both the anterior and posterior hippocampus. We found that a larger increase in morning cortisol level after awakening was associated with smaller DG and CA1-3 volumes, while a smaller decrease in cortisol level in the afternoon from awakening was associated with smaller CA1-3 volume in the anterior hippocampus. The observed associations between cortisol and hippocampal subfield volumes were not predicted by individual chronic stress levels or history of childhood trauma. Conclusions Our results suggest that both increased hair cortisol concentration and daily cortisol fluctuations can have a negative impact on the CA1-3 and DG subfields.
Collapse
Affiliation(s)
- Nikolai Malykhin
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Joseph Serrano
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Béla Reiz
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Kathleen Hegadoren
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Nursing, University of Alberta, Edmonton, Alberta, Canada
| | - Wojciech Pietrasik
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Randy Whittal
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
van Diepen J, Hendriks GJ, Zuidersma M, Oude Voshaar R, Janssen N. Data-driven subtypes of late-life depression-secondary analysis of a cluster-randomized RCT. Aging Ment Health 2025:1-8. [PMID: 39987525 DOI: 10.1080/13607863.2025.2468406] [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: 10/25/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
OBJECTIVES Cognitive performance varies among depressed older patients and is known to affect treatment outcome. We used a data-driven approach to create subtypes of late-life depression and exploratively visualized the course of depression during either behavioural activation (BA) or treatment as usual (TAU). METHOD Among 161 depressed (PHQ ≥ 10) older (≥65 years) participants of a cluster randomized controlled trial in primary care (NL5436), we performed latent class analysis (LCA) on individual depressive symptoms and performance on several cognitive tests. The course of depressive symptoms during treatment was plotted to explore whether differences between the classes differed between BA and TAU. RESULTS Five classes best fitted the data: (1) mild depression without cognitive deficits, (2) moderate depression with insomnia and cognitive deficits, (3) severe depression with cognitive deficits, (4) moderately severe depression with hypersomnia and cognitive deficits, and (5) moderately severe depression with cognitive-affective symptoms but no cognitive deficits. Graphs showed that depressive symptoms of subgroups with severe depressive symptoms improved more during BA compared to TAU, regardless of cognitive deficits. CONCLUSIONS We identified five subgroups. Graphs suggest that effectiveness of BA is similar across all subgroups, whereas TAU seems less effective in the more severely depressed subgroups. Replication is warranted.
Collapse
Affiliation(s)
- Judith van Diepen
- Department of Primary and Community Care, Radboud University Medical Center, Research institute of Health Sciences, Nijmegen, Netherlands
- Pro Persona (Research), Pro Persona, Renkum, Netherlands
| | - Gert-Jan Hendriks
- Pro Persona (Research), Pro Persona, Renkum, Netherlands
- Behavioural Science Institute, Radboud University, Nijmegen, Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marij Zuidersma
- Department of Psychiatry, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Richard Oude Voshaar
- Department of Psychiatry, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Noortje Janssen
- Department of Primary and Community Care, Radboud University Medical Center, Research institute of Health Sciences, Nijmegen, Netherlands
- Pro Persona (Research), Pro Persona, Renkum, Netherlands
- Behavioural Science Institute, Radboud University, Nijmegen, Netherlands
| |
Collapse
|
4
|
Li X, Su W, Cai L. A bibliometric analysis of research on dementia comorbid with depression from 2005 to 2024. Front Neurosci 2025; 19:1508662. [PMID: 39981405 PMCID: PMC11841476 DOI: 10.3389/fnins.2025.1508662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction With the global rise in life expectancy, the incidence of dementia is increasing, often accompanied by depressive symptoms. Understanding the interplay between dementia and depression is crucial, as depression may not only co-occur with but also potentially exacerbate the progression of dementia. This study employs bibliometric analysis to map the global research landscape, identify prevailing themes, and discern future research directions. Methods We analyzed reviews and original research articles on dementia and depression extracted from the Web of Science Core Collection spanning from 2005 to 2024. Utilizing tools such as CiteSpace, VOSviewer, and an R-based bibliometric analysis package, we assessed trends in publication volume, citation frequency, contributing countries, leading institutions, predominant journals, influential authors, and emergent keywords. Results A total of 1972 publications were obtained, revealing a consistent increase in both the number of publications and their citation impact over the study period. The United States is the country with the most publications and the most extensive collaborations. The University of Toronto and the Journal of Alzheimer's Disease were identified as key contributors to this field. This research area is currently focused on cognitive impairments, the role of gut microbiota, and non-drug interventions. Future directions emphasize the importance of early detection and intervention, a deeper understanding of the gut-brain axis, and the integration of technology in treatment strategies. Additionally, there is a growing interest in the physiological and psychological interplays such as oxidative stress and its implications. Conclusion This study underscores pathogenesis, comorbid conditions, and non-drug interventions as primary research focal points, suggesting these areas as potential pathways for therapeutic innovation. These insights are intended to deepen our understanding, enhance diagnostics, and improve the management of dementia and depression, providing guidance for future research aimed at addressing these escalating global health challenges.
Collapse
Affiliation(s)
| | - Wei Su
- Department of Psychiatry, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou, China
| | | |
Collapse
|
5
|
Jalalvandi M, Batouli SAH. Comparative analysis of Voxel-based morphometry using T1 and T2-weighted magnetic resonance imaging to explore the relationship between brain structure and cognitive abilities. Exp Brain Res 2025; 243:50. [PMID: 39836225 DOI: 10.1007/s00221-025-07000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Voxel-based morphometry (VBM) of T1-weighted (T1-w) magnetic resonance imaging (MRI) is primarily used to study the association of brain structure with cognitive functions. However, in theory, T2-weighted (T2-w) MRI could also be used in VBM studies because of its sensitivity to pathology and tissue changes. We aimed to compare the T1-w and T2-w images to study brain structures in association with cognitive abilities. VBM analysis was applied to T1-w and T2-w MRI data of 120 healthy participants aged 20 to 40. The MRI data was collected using a 3T machine, and it was analyzed with CAT12 to extract maps of Gray matter(GM). We used six cognitive tasks to assess cognitive abilities, including the balloon analog risk task (BART), block design, forward and backward digit span (FDST and BDST), and trail-making tasks A and B. Compared to T2-w, T1-w data showed more brain voxels in the BART, block design, FDST, TMT-A, and TMT-B tasks. However, T2-w imaging identified a greater number of voxels in the BDST. T1-w images identified more correlated brain regions with cognitive scores in the FDST, TMT-A, and B tasks than T2-w. In BART and Block design tasks, both methods revealed the same number of correlated regions, and T2-w just showed more regions than T1-w in the BDST. Findings revealed distinct patterns of sensitivity between T1-w and T2-w imaging in detecting brain regions associated with cognition. The two approaches demonstrated different strengths in identifying areas correlated with cognitive abilities. This study provides valuable guidance for selecting appropriate methods for identifying the optimal approach for detecting brain regions that exhibit the strongest correlations with cognitive abilities.
Collapse
Affiliation(s)
- Maziar Jalalvandi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Hossein Batouli
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- BrainEE Research Group, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, No.88, Italia Street, Keshavarz Boulevard, Tehran, Iran.
| |
Collapse
|
6
|
Peng YY, Tang C, Wang HY, Ding Y, Yang H, Ma XM, Gao J, Li S, Long ZY, Lu XM, Wang YT. p75NTR mediated chronic restraint stress-induced depression-like behaviors in mice via hippocampal mTOR pathway. Life Sci 2024; 358:123175. [PMID: 39477145 DOI: 10.1016/j.lfs.2024.123175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
AIMS Major depressive disorder (MDD) is an enduring and severe mood disorder. Previous studies have indicated that p75NTR is involved in neuronal survival and death. However, the specific mechanism of p75NTR in depression remains unknown. The present study aimed to explore the role and mechanism of p75NTR in depression, and try to provide a new target for the treatment of MDD. MAIN METHODS The p75NTR knockout and overexpression mice were used to establish a mouse model of depression induced by chronic restraint stress (CRS), and the behavioral effects and potential mechanisms associated with p75NTR knockout/overexpression on CRS-induced depressive mice were investigated by animal behavior, histopathology, immunofluorescence and western blot, respectively. KEY FINDINGS The results demonstrate that p75NTR knockout/overexpression can ameliorate the depressive-like behaviors observed in CRS-induced depressive mice. Furthermore, p75NTR knockout/overexpression safeguards the tissue morphology of the hippocampus, inhibits the mTOR signaling pathway to restore autophagy, and modulates apoptosis-related proteins (Bcl-2 and Bax) to reestablish normal levels of autophagy and apoptosis in hippocampal neurons of depressed mice. Importantly, p75NTR knockout/overexpression can improve synaptic plasticity through protecting the dendritic structure and dendritic spines of hippocampal neurons, and upregulating the expression of hippocampal synaptic-related proteins (PSD95 and SYN1). SIGNIFICANCE These findings suggest that p75NTR knockout/overexpression can alleviate CRS-induced depression-like behaviors by reinstating autophagy and suppressing apoptosis in hippocampal neurons, and enhancing hippocampal synaptic plasticity via mTOR pathway. These insights may provide potential targets for clinical treatment of depression.
Collapse
MESH Headings
- Animals
- TOR Serine-Threonine Kinases/metabolism
- Mice
- Hippocampus/metabolism
- Hippocampus/pathology
- Stress, Psychological/complications
- Stress, Psychological/metabolism
- Depression/metabolism
- Depression/etiology
- Male
- Mice, Knockout
- Signal Transduction
- Restraint, Physical
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Nerve Growth Factor/genetics
- Mice, Inbred C57BL
- Behavior, Animal
- Neuronal Plasticity
- Apoptosis
- Disease Models, Animal
- Nerve Tissue Proteins/metabolism
- Nerve Tissue Proteins/genetics
- Autophagy/physiology
- Depressive Disorder, Major/metabolism
- Neurons/metabolism
- Neurons/pathology
Collapse
Affiliation(s)
- Yu-Yuan Peng
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China; College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Can Tang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yang Ding
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Huan Yang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Xin-Mei Ma
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Jie Gao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Sen Li
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zai-Yun Long
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| | - Yong-Tang Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
7
|
Mata-Bermudez A, Diaz-Ruiz A, Silva-García LR, Gines-Francisco EM, Noriega-Navarro R, Rios C, Romero-Sánchez HA, Arroyo D, Landa A, Navarro L. Mucuna pruriens, a Possible Treatment for Depressive Disorders. Neurol Int 2024; 16:1509-1527. [PMID: 39585071 PMCID: PMC11587415 DOI: 10.3390/neurolint16060112] [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: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024] Open
Abstract
Depression is a mental disorder that depicts a wide variety of symptoms, including mood and cognitive alterations, as well as recurrent thoughts of death or suicide. It could become the second leading cause of premature death or disability worldwide. Treatments with conventional antidepressants have several limitations in terms of effectiveness, side effects, and high costs. Therefore, medicinal plants such as Mucuna pruriens are potent candidates for treating depressive disorders. This review shows a compendium of evidence supporting the antidepressant effect of the Mucuna pruriens plant in diverse animal models. This includes the mechanisms of action underlying the antidepressant activity of the treatment concerning dopamine, serotonin, norepinephrine, reactive oxygen species, nitric oxide, cortisol, and inflammation. Clinical trials are needed to study the efficacy and safety of Mucuna pruriens for depression.
Collapse
Affiliation(s)
- Alfonso Mata-Bermudez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (A.M.-B.); (R.N.-N.); (D.A.)
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico; (L.R.S.-G.); (E.M.G.-F.); (H.A.R.-S.)
| | - Araceli Diaz-Ruiz
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México 14269, Mexico;
| | - Luis Ricardo Silva-García
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico; (L.R.S.-G.); (E.M.G.-F.); (H.A.R.-S.)
| | - Eduardo Manuel Gines-Francisco
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico; (L.R.S.-G.); (E.M.G.-F.); (H.A.R.-S.)
| | - Roxana Noriega-Navarro
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (A.M.-B.); (R.N.-N.); (D.A.)
| | - Camilo Rios
- Laboratorio de Neurofarmacología Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico;
- Dirección de Investigación, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra, Calzada Mexico-Xochimilco 289, Arenal de Guadalupe, Ciudad de México 14389, Mexico
| | - Héctor Alonso Romero-Sánchez
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico; (L.R.S.-G.); (E.M.G.-F.); (H.A.R.-S.)
| | - Diego Arroyo
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (A.M.-B.); (R.N.-N.); (D.A.)
| | - Abraham Landa
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Luz Navarro
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (A.M.-B.); (R.N.-N.); (D.A.)
| |
Collapse
|
8
|
Hu Y, Zhu T, Yuan M, Zhu H, Zhang W. Longitudinal association of depressive symptoms with cognition and neuroimaging biomarkers in cognitively unimpaired older adults, mild cognitive impairment, and Alzheimer's disease. Cereb Cortex 2024; 34:bhae423. [PMID: 39441024 DOI: 10.1093/cercor/bhae423] [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: 04/10/2024] [Revised: 09/11/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
We aimed to longitudinally examine the relationship between depression and cognitive function and investigate the mediating effects of imaging indicators in this relationship. 2,251 subjects with longitudinal assessment of geriatric depression scale, Mini-Mental State Examination, Montreal Cognitive Assessment, Clinical Dementia Rating-Sum of Boxes (CDRSB), Alzheimer's Disease Assessment Scale11, Alzheimer's Disease Assessment Scale13 and imaging of 3DT1, diffusion tensor imaging, fluid-attenuated inversion recovery, arterial spin labeling, fluorodeoxyglucose positron emission tomography, 18F-AV45-PET, and 18F-AV1451-PET were included from the Alzheimer's Disease Neuroimaging Initiative database. The multivariate mixed-effects models were employed to analyze the correlation between geriatric depression scale scores, cognitive function, and imaging indicators. The sgmediation software package was utilized to analyze the mediating effects of imaging indicators. The geriatric depression scale was negatively correlated with Mini-Mental State Examination and Montreal Cognitive Assessment, and positively correlated with CDRSB, Alzheimer's Disease Assessment Scale11, and Alzheimer's Disease Assessment Scale13 when the subjects were not grouped. The geriatric depression scale was negatively correlated with Montreal Cognitive Assessment and positively correlated with Alzheimer's Disease Assessment Scal13 in groups with baseline diagnosis of early mild cognitive impairment and late mild cognitive impairment. Furthermore, depression was associated with regional imaging indicators, while cognitive function was linked to broad imaging indicators. Some of these indicators were related to both depression and cognitive function, playing a mediating role in their relationship. Depression was related to cognitive function, especially in subjects with mild cognitive impairment. Some imaging indicators may represent the underlying basis for the association between depression and cognitive function.
Collapse
Affiliation(s)
- Ying Hu
- Department of Radiology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| | - Ting Zhu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| | - Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| | - Hongru Zhu
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| | - Wei Zhang
- West China Biomedical Big Data Center of West China Hospital, Med-X Center for Informatics, Mental Health Center of West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| |
Collapse
|
9
|
Aberizk K, Addington JM, Bearden CE, Cadenhead KS, Cannon TD, Cornblatt BA, Keshavan M, Mathalon DH, Perkins DO, Stone WS, Tsuang MT, Woods SW, Walker EF, Ku BS. Relations of Lifetime Perceived Stress and Basal Cortisol With Hippocampal Volume Among Healthy Adolescents and Those at Clinical High Risk for Psychosis: A Structural Equation Modeling Approach. Biol Psychiatry 2024; 96:401-411. [PMID: 38092185 PMCID: PMC11166888 DOI: 10.1016/j.biopsych.2023.11.027] [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: 06/29/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Hippocampal volume (HV) is sensitive to environmental influences. Under normative conditions in humans, HV increases linearly into childhood and asymptotes in early adulthood. Studies of humans and nonhuman animals have provided evidence of inverse relationships between several measures of stress and HV. METHODS Using structural equation modeling, this study aimed to characterize the relationships of age, basal cortisol, biological sex, and lifetime perceived stress with bilateral HV in a sample of healthy adolescents and adolescents at clinical high risk for psychosis (CHR-P) (N = 571, 43% female; age range = 12-19.9 years). This sample included 469 individuals at CHR-P and 102 healthy comparison participants from the combined baseline cohorts of the second and third NAPLS (North American Prodrome Longitudinal Study). RESULTS A structural model that constrained the individual effects of basal cortisol and perceived stress to single path coefficients, and freely estimated the effects of age and biological sex in group models, optimized model fit and parsimony relative to other candidate models. Significant inverse relationships between basal cortisol and bilateral HV were observed in adolescents at CHR-P and healthy comparison participants. Significant sex differences in bilateral HV were also observed, with females demonstrating smaller HV than males in both groups. CONCLUSIONS Multigroup structural equation modeling revealed heterogeneity in the relationships of age and biological sex with basal cortisol, lifetime perceived stress, and bilateral HV in individuals at CHR-P and healthy comparison participants. Moreover, the findings support previous literature indicating that elevated basal cortisol is a nonspecific risk factor for reduced HV.
Collapse
Affiliation(s)
- Katrina Aberizk
- Department of Psychology, Emory University, Atlanta, Georgia.
| | - Jean M Addington
- Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada
| | - Carrie E Bearden
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Psychology, University of California, Los Angeles, California
| | | | - Tyrone D Cannon
- Departments of Psychology and Psychiatry, Yale University, New Haven, Connecticut
| | | | - Matcheri Keshavan
- Department of Psychiatry, Harvard Medical School, Harvard University, Cambridge, Massachusetts
| | - Daniel H Mathalon
- Department of Psychiatry, University of California, San Francisco, California
| | - Diana O Perkins
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - William S Stone
- Department of Psychiatry, Harvard Medical School, Harvard University, Cambridge, Massachusetts
| | - Ming T Tsuang
- Department of Psychiatry, University of California, San Diego, California
| | - Scott W Woods
- Departments of Psychology and Psychiatry, Yale University, New Haven, Connecticut
| | - Elaine F Walker
- Department of Psychology, Emory University, Atlanta, Georgia
| | - Benson S Ku
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
10
|
Jin GN, Wang Y, Liu YM, Lu YN, Lu JM, Wang JH, Ma JW, Quan YZ, Gao HY, Cui YX, Xu X, Piao LX. Arctiin Mitigates Neuronal Injury by Modulating the P2X7R/NLPR3 Inflammasome Signaling Pathway. Inflammation 2024:10.1007/s10753-024-02117-z. [PMID: 39154088 DOI: 10.1007/s10753-024-02117-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/13/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
Depression, recognized globally as a primary cause of disability, has its pathogenesis closely related to neuroinflammation and neuronal damage. Arctiin (ARC), the major bioactive component of Fructus arctii, has various pharmacological activities, such as anti-inflammatory and neuroprotective effects. Building on previous findings that highlighted ARC's capability to mitigate depression by dampening microglial hyperactivation and thereby reducing neuroinflammatory responses and cortical neuronal damage in mice, the current study delves deeper into ARC's therapeutic potential by examining its impact on hippocampal neuronal damage in depression. Utilizing both chronic unpredictable mild stress (CUMS)-induced depression model in mice and corticosterone (CORT)-stimulated PC12 cell model of neuronal damage, the techniques including Nissl staining, immunohistochemistry, western blotting, ELISA, lactate dehydrogenase assays, colony formation assays, immunofluorescence staining and molecular docking were employed to unravel the mechanisms behind ARC's neuroprotective effects. The findings revealed that ARC not only mitigates hippocampal neuropathological damage and reduces serum CORT levels in CUMS-exposed mice but also enhances cell activity while reducing lactate dehydrogenase release in CORT-stimulated PC12 cells. ARC attenuated neuroinflammatory responses and neuronal apoptosis by inhibiting the overactivation of the P2X7 receptor (P2X7R)/NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome signaling pathway, similar to the effect of A438079 (P2X7R antagonist). Interestingly, pretreatment with A438079 blocked the neuroprotective effect of ARC. Computer modeling predicted that both ARC and A438079 have strong binding with P2X7R and they have the same binding site. These results suggested that ARC may exert a neuroprotective role by binding to P2X7R, thereby inhibiting the P2X7R/NLRP3 inflammasome signaling pathway.
Collapse
Affiliation(s)
- Guang-Nan Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yu Wang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yi-Ming Liu
- Department of Neurology, Yanbian University Hospital, Yanbian University, Yanji, 133000, Jilin Province, China
| | - Yu-Nan Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jing-Mei Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jing-He Wang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jing-Wen Ma
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yan-Zhu Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Hong-Yan Gao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yue-Xian Cui
- Department of Neurology, Yanbian University Hospital, Yanbian University, Yanji, 133000, Jilin Province, China.
| | - Xiang Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| | - Lian-Xun Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| |
Collapse
|
11
|
Kong SDX, Espinosa N, McKinnon AC, Gordon CJ, Wassing R, Hoyos CM, Hickie IB, Naismith SL. Different heart rate variability profile during sleep in mid-later life adults with remitted early-onset versus late-onset depression. J Affect Disord 2024; 358:175-182. [PMID: 38701901 DOI: 10.1016/j.jad.2024.04.054] [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: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND In mid-later life adults, early-onset and late-onset (i.e., onset ≥50 years) depression appear to be underpinned by different pathophysiology yet have not been examined in relation to autonomic function. Sleep provides an opportunity to examine the autonomic nervous system as the physiology changes across the night. Hence, we aimed to explore if autonomic profile is altered in mid-later life adults with remitted early-onset, late-onset and no history of lifetime depression. METHODS Participants aged 50-90 years (n = 188) from a specialised clinic underwent a comprehensive clinical assessment and completed an overnight polysomnography study. General Linear Models were used to examine the heart rate variability differences among the three groups for four distinct sleep stages and the wake after sleep onset. All analyses controlled for potential confounders - age, sex, current depressive symptoms and antidepressant usage. RESULTS For the wake after sleep onset, mid-later life adults with remitted early-onset depression had reduced standard deviation of Normal to Normal intervals (SDNN; p = .014, d = -0.64) and Shannon Entropy (p = .004, d = -0.46,) than those with no history of lifetime depression. Further, the late-onset group showed a reduction in high-frequency heart rate variability (HFn.u.) during non-rapid eye movement sleep stage 2 (N2; p = .005, d = -0.53) and non-rapid eye movement sleep stage 3 (N3; p = .009, d = -0.55) when compared to those with no lifetime history. LIMITATIONS Causality between heart rate variability and depression cannot be derived in this cross-sectional study. Longitudinal studies are needed to examine the effects remitted depressive episodes on autonomic function. CONCLUSION The findings suggest differential autonomic profile for remitted early-onset and late-onset mid-later life adults during sleep stages and wake periods. The differences could potentially serve as peripheral biomarkers in conjunction with more disease-specific markers of depression to improve diagnosis and prognosis.
Collapse
Affiliation(s)
- Shawn D X Kong
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Camperdown, NSW 2050, Australia; Charles Perkins Centre, University of Sydney, Camperdown, NSW 2050, Australia; CogSleep, Australian National Health and Medical Research Council Centre of Research Excellence, Australia.
| | - Nicole Espinosa
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Camperdown, NSW 2050, Australia
| | - Andrew C McKinnon
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Camperdown, NSW 2050, Australia; Charles Perkins Centre, University of Sydney, Camperdown, NSW 2050, Australia; Race Against Dementia, Dementia Australia Research Foundation Initiative, Dementia Australia, Australia
| | - Christopher J Gordon
- CogSleep, Australian National Health and Medical Research Council Centre of Research Excellence, Australia; Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Macquarie University, Glebe, NSW 2037, Australia; Faculty of Medicine and Health, Royal North Shore Hospital, Sydney 2050, Australia
| | - Rick Wassing
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Macquarie University, Glebe, NSW 2037, Australia; School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Sydney Local Health District, Sydney, NSW, Australia
| | - Camilla M Hoyos
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Macquarie University, Glebe, NSW 2037, Australia; School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Ian B Hickie
- Youth Mental Health Team, Brain and Mind Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Sharon L Naismith
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Camperdown, NSW 2050, Australia; Charles Perkins Centre, University of Sydney, Camperdown, NSW 2050, Australia; CogSleep, Australian National Health and Medical Research Council Centre of Research Excellence, Australia
| |
Collapse
|
12
|
Sallie FN, Pienaar L, Lubbe A, Xhakaza S, Manne SR, de la Torre BG, Albericio F, Mu Daniels W, Me Millen A, Baijnath S. Neurobehavioral and molecular changes in a rodent model of ACTH-induced HPA axis dysfunction. Brain Res 2024; 1834:148913. [PMID: 38580046 DOI: 10.1016/j.brainres.2024.148913] [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: 11/10/2023] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Hypothalamic-pituitary-adrenal (HPA) axis dysregulation is linked to the pathophysiology of depression. Although exogenous adrenocorticotropic hormone (ACTH) is associated with a depressive-like phenotype in rodents, comprehensive neurobehavioral and mechanistic evidence to support these findings are limited. Sprague-Dawley rats (male, n = 30; female, n = 10) were randomly assigned to the control (male, n = 10) or ACTH (male, n = 20; female n = 10) groups that received saline (0.1 ml, sc.) or ACTH (100 μg/day, sc.), respectively, for two weeks. Thereafter, rats in the ACTH group were subdivided to receive ACTH plus saline (ACTH_S; male, n = 10; female, n = 5; 0.2 ml, ip.) or ACTH plus imipramine (ACTH_I; male, n = 10; female, n = 5;10 mg/kg, ip.) for a further four weeks. Neurobehavioral changes were assessed using the forced swim test (FST), the sucrose preference test (SPT), and the open field test (OFT). Following termination, the brain regional mRNA expression of BDNF and CREB was determined using RT-PCR. After two-weeks, ACTH administration significantly increased immobility in the FST (p = 0.03), decreased interaction with the center of the OFT (p < 0.01), and increased sucrose consumption (p = 0.03) in male, but not female rats. ACTH administration significantly increased the expression of BDNF in the hippocampus and CREB in all brain regions in males (p < 0.05), but not in female rats. Imipramine treatment did not ameliorate these ACTH-induced neurobehavioral or molecular changes. In conclusion, ACTH administration resulted in a sex-specific onset of depressive-like symptoms and changes in brain regional expression of neurotrophic factors. These results suggest sex-specific mechanisms underlying the development of depressive-like behavior in a model of ACTH-induced HPA axis dysregulation.
Collapse
Affiliation(s)
- Farhanah N Sallie
- Wits Integrated Molecular Physiology Research Initiative, Wits Health Consortium (PTY) Ltd, School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa; School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa
| | - Leandrie Pienaar
- Wits Integrated Molecular Physiology Research Initiative, Wits Health Consortium (PTY) Ltd, School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa; School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa
| | - Andrea Lubbe
- Wits Integrated Molecular Physiology Research Initiative, Wits Health Consortium (PTY) Ltd, School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa; School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa
| | - Sanelisiwe Xhakaza
- Wits Integrated Molecular Physiology Research Initiative, Wits Health Consortium (PTY) Ltd, School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa; School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa
| | - Srinivasa R Manne
- Peptide Science Laboratory, School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | - Beatriz G de la Torre
- Peptide Science Laboratory, School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | - Fernando Albericio
- Peptide Science Laboratory, School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | - William Mu Daniels
- School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa
| | - Aletta Me Millen
- Wits Integrated Molecular Physiology Research Initiative, Wits Health Consortium (PTY) Ltd, School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa; School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa.
| | - Sooraj Baijnath
- Wits Integrated Molecular Physiology Research Initiative, Wits Health Consortium (PTY) Ltd, School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa; School of Physiology, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
13
|
Del Casale A, Mancino S, Arena JF, Spitoni GF, Campanini E, Adriani B, Tafaro L, Alcibiade A, Ciocca G, Romano A, Bozzao A, Ferracuti S. Neural Functioning in Late-Life Depression: An Activation Likelihood Estimation Meta-Analysis. Geriatrics (Basel) 2024; 9:87. [PMID: 39051251 PMCID: PMC11270429 DOI: 10.3390/geriatrics9040087] [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: 05/05/2024] [Revised: 06/14/2024] [Accepted: 06/23/2024] [Indexed: 07/27/2024] Open
Abstract
Late-life depression (LLD) is a relatively common and debilitating mental disorder, also associated with cognitive dysfunctions and an increased risk of mortality. Considering the growing elderly population worldwide, LLD is increasingly emerging as a significant public health issue, also due to the rise in direct and indirect costs borne by healthcare systems. Understanding the neuroanatomical and neurofunctional correlates of LLD is crucial for developing more targeted and effective interventions, both from a preventive and therapeutic standpoint. This ALE meta-analysis aims to evaluate the involvement of specific neurofunctional changes in the neurophysiopathology of LLD by analysing functional neuroimaging studies conducted on patients with LLD compared to healthy subjects (HCs). We included 19 studies conducted on 844 subjects, divided into 439 patients with LLD and 405 HCs. Patients with LLD, compared to HCs, showed significant hypoactivation of the right superior and medial frontal gyri (Brodmann areas (Bas) 8, 9), left cingulate cortex (BA 24), left putamen, and left caudate body. The same patients exhibited significant hyperactivation of the left superior temporal gyrus (BA 42), left inferior frontal gyrus (BA 45), right anterior cingulate cortex (BA 24), right cerebellar culmen, and left cerebellar declive. In summary, we found significant changes in activation patterns and brain functioning in areas encompassed in the cortico-limbic-striatal network in LLD. Furthermore, our results suggest a potential role for areas within the cortico-striatal-cerebellar network in the neurophysiopathology of LLD.
Collapse
Affiliation(s)
- Antonio Del Casale
- Department of Dynamic and Clinical Psychology and Health Studies, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
- Unit of Psychiatry, Emergency and Admissions Department, ‘Sant’Andrea’ University Hospital, 00189 Rome, Italy
| | - Serena Mancino
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
| | - Jan Francesco Arena
- Department of Dynamic and Clinical Psychology and Health Studies, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
| | - Grazia Fernanda Spitoni
- Department of Dynamic and Clinical Psychology and Health Studies, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
| | - Elisa Campanini
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
| | - Barbara Adriani
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
| | - Laura Tafaro
- Department of Clinical and Molecular Medicine, Sapienza University, 00189 Rome, Italy;
- Unit of Internal Medicine, ‘Sant’Andrea’ University Hospital, 00189 Rome, Italy
| | - Alessandro Alcibiade
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Marina Militare Italiana (Italian Navy), Ministry of Defence, Piazza della Marina, 4, 00196 Rome, Italy
| | - Giacomo Ciocca
- Department of Dynamic and Clinical Psychology and Health Studies, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
| | - Andrea Romano
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Neuroradiology, Department of Diagnostic Sciences, ‘Sant’Andrea’ University Hospital, 00189 Rome, Italy
| | - Alessandro Bozzao
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Neuroradiology, Department of Diagnostic Sciences, ‘Sant’Andrea’ University Hospital, 00189 Rome, Italy
| | - Stefano Ferracuti
- Department of Human Neuroscience, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00185 Rome, Italy
- Unit of Risk Management, ‘Sant’Andrea’ University Hospital, 00189 Rome, Italy
| |
Collapse
|
14
|
Chen CYA, Chiu CC, Huang CY, Cheng YC, Huang MC, Kuo PH, Chen WY. Cluster analysis dissecting cognitive deficits in older adults with major depressive disorder and the association with neurofilament light chain. BMC Geriatr 2024; 24:344. [PMID: 38627748 PMCID: PMC11020442 DOI: 10.1186/s12877-024-04960-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Cognitive impairment is a growing problem with increasing burden in global aging. Older adults with major depressive disorder (MDD) have higher risk of dementia. Neurofilament light chain (NfL) has been proven as a potential biomarker in neurodegenerative disease, including dementia. We aimed to investigate the association between cognitive deficits and NfL levels in older adults with MDD. METHODS In this cross-sectional study, we enrolled 39 MDD patients and 15 individuals with mild neurocognitive disorder or major neurocognitive disorder, Alzheimer's type, as controls, from a tertiary psychiatric hospital. Both groups were over age 65 and with matched Mini-Mental State Examination (MMSE) score. Demographic data, clinical variables, and plasma NfL levels were obtained. We used cluster analysis according to their cognitive profile and estimated the correlation between plasma NfL levels and each cognitive domain. RESULTS In the MDD group, participants had higher rate of family psychiatry history and current alcohol use habit compared with controls. Control group of neurocognitive disorders showed significantly lower score in total MMSE and higher plasma NfL levels. Part of the MDD patients presented cognitive deficits clustered with that of neurocognitive disorders (cluster A). In cluster A, the total MMSE score (r=-0.58277, p=0.0287) and the comprehension domain (r=-0.71717, p=0.0039) were negatively correlated to NfL levels after adjusting for age, while the associations had not been observed in the other cluster. CONCLUSIONS We noted the negative correlation between NfL levels and cognition in MDD patients clustered with neurodegenerative disorder, Alzheimer's type. NfL could be a promising candidate as a biomarker to predict subtype of patients in MDD to develop cognitive decline. Further longitudinal studies and within MDD cluster analysis are required to validate our findings for clinical implications.
Collapse
Affiliation(s)
- Cynthia Yi-An Chen
- Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chiang Chiu
- Department of Psychiatry, Taipei City Psychiatric Center, Songde branch, Taipei City Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cho-Yin Huang
- Department of Psychiatry, Taipei City Psychiatric Center, Songde branch, Taipei City Hospital, Taipei, Taiwan
| | - Ying-Chih Cheng
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Psychiatry, China Medical University Hsinchu Hospital, China Medical University, Hsinchu, Taiwan
| | - Ming-Chyi Huang
- Department of Psychiatry, Taipei City Psychiatric Center, Songde branch, Taipei City Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Hsiu Kuo
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Wen-Yin Chen
- Department of Psychiatry, Taipei City Psychiatric Center, Songde branch, Taipei City Hospital, Taipei, Taiwan.
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan.
| |
Collapse
|
15
|
Millen AME, Daniels WMU, Baijnath S. Depression, an unmet health need in Africa: Understanding the promise of ketamine. Heliyon 2024; 10:e28610. [PMID: 38601594 PMCID: PMC11004535 DOI: 10.1016/j.heliyon.2024.e28610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 04/12/2024] Open
Abstract
In Africa, there is currently a paucity of data on the epidemiology of depression, its treatment and management. The prevalence of depression is severely underestimated, with unique circumstances and societal risk factors associated with depression and its public awareness. Treating and managing depression is confounded by an inaccessibility to efficient and low-cost treatments for patients with depression. The aetiology of depression is multifactorial, with various theories implicating multiple neuronal networks. Despite this, the treatment of depression is one-dimensional focussing on outdated theories of depression and mainly targeting dysfunctional neurotransmitter pathways. Hence, it is not surprising that there is a significant increase in the prevalence of patients suffering from treatment resistant depression (TRD), with a large portion of patients deriving little clinical benefit from these traditional anti-depressant therapies. This highlights the need for more effective treatment strategies for depression, especially applicable to resource limited environments such as Africa, where there is little investment in public healthcare resources towards managing mental health disorders. The clinical potential of using ketamine in managing depression has received considerable attention in the past two decades, with the FDA approving esketamine for the management of TRD in 2019. This widespread attention has significantly increased ketamine's appeal as a novel antidepressant. Consequently, many ketamine infusion clinics have been established in Africa. However, there is little regulation or guidance for ketamine infusions. Furthermore, while esketamine is expensive and hence inaccessible to a large portion of the African population, racemic ketamine is significantly cheaper and has demonstrated clinical potential. However, there is currently a limited understanding of the neurological mechanisms of action of racemic ketamine in treating and managing depression, especially in a diverse African population. Therefore, this review aims to provide an African context of depression and the therapeutic potential of ketamine by highlighting aspects of its molecular mechanism of action.
Collapse
Affiliation(s)
- Aletta ME. Millen
- Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - William MU. Daniels
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sooraj Baijnath
- Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
16
|
Langella S, Lopera F, Baena A, Fox‐Fuller JT, Munera D, Martinez JE, Giudicessi A, Vannini P, Hanseeuw BJ, Marshall GA, Quiroz YT, Gatchel JR. Depressive symptoms and hippocampal volume in autosomal dominant Alzheimer's disease. Alzheimers Dement 2024; 20:986-994. [PMID: 37837524 PMCID: PMC10916972 DOI: 10.1002/alz.13501] [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: 04/17/2023] [Revised: 08/08/2023] [Accepted: 09/17/2023] [Indexed: 10/16/2023]
Abstract
INTRODUCTION Depressive symptoms are among early behavioral changes in Alzheimer's disease (AD); however, the relationship between neurodegeneration and depressive symptoms remains inconclusive. To better understand this relationship in preclinical AD, we examined hippocampal volume and depressive symptoms in cognitively unimpaired carriers of the presenilin-1 (PSEN1) E280A mutation for autosomal dominant AD. METHODS A total of 27 PSEN1 mutation carriers and 26 non-carrier family members were included. Linear regression was used to test the relationship between hippocampal volume and 15-item Geriatric Depression Scale. RESULTS Carriers and non-carriers did not differ in depressive symptoms or hippocampal volume. Within carriers, lower hippocampal volume was associated with greater depressive symptoms, which remained significant after adjusting for age and cognition. This relationship was not significant in non-carriers. DISCUSSION Hippocampal neurodegeneration may underlie depressive symptoms in preclinical autosomal dominant AD. These findings provide support for the utility of targeting depressive symptoms in AD prevention. HIGHLIGHTS We compared unimpaired autosomal dominant Alzheimer's disease (AD) mutation carriers and non-carriers. Carriers and non-carriers did not differ in severity of depressive symptoms. In carriers, hippocampal volume was inversely associated with depressive symptoms. Depressive symptoms may be a useful target in AD prevention.
Collapse
Affiliation(s)
- Stephanie Langella
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Francisco Lopera
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellinColombia
| | - Ana Baena
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellinColombia
| | - Joshua T. Fox‐Fuller
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of Psychological and Brain SciencesBoston UniversityBostonMassachusettsUSA
| | - Diana Munera
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Jairo E. Martinez
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of Psychological and Brain SciencesBoston UniversityBostonMassachusettsUSA
| | - Averi Giudicessi
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of Psychological and Brain SciencesBoston UniversityBostonMassachusettsUSA
| | - Patrizia Vannini
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Bernard J. Hanseeuw
- Gordon Center for Medical ImagingMassachusetts General HospitalBostonMassachusettsUSA
- Cliniques Universitaires Saint‐LucUniversité Catholique de LouvainBrusselsBelgium
| | - Gad A. Marshall
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Yakeel T. Quiroz
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellinColombia
| | - Jennifer R. Gatchel
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- McLean HospitalHarvard Medical SchoolBelmontMassachusettsUSA
- Department of PsychiatryBaylor College of MedicineHoustonTexasUSA
- Mental Health Care LineMEDVAMCHoustonTexasUSA
| |
Collapse
|
17
|
Ponsoni A, Branco LD, Cotrena C, Shansis FM, Fonseca RP. A longitudinal study of cognition, functional outcome and quality of life in bipolar disorder and major depression. APPLIED NEUROPSYCHOLOGY. ADULT 2023; 30:757-763. [PMID: 34597199 DOI: 10.1080/23279095.2021.1979551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Cognitive impairments are known to be a frequent cause of disability in bipolar disorder (BD) and major depression (MDD). Yet there is no consensus regarding the particular cognitive functions whose impairments can lead to disability in each domain of functioning. The aim of this study was to perform a longitudinal evaluation of working memory, inhibition, cognitive flexibility and attention in BD and MDD, investigate the relationship of these cognitive functions to disability and quality of life, and evaluate the impact of variables related to cognitive reserve (education and daily cognitive stimulation) on cognitive performance. METHOD 31 participants (MDD = 12; BD = 19) were evaluated at baseline and after an average time of 2 years. RESULTS the BD group showed improvements in attention while patients with MDD improved on measures of attention and working memory. In BD working memory performance was associated with the cognition and mobility domains of functioning, and with physical and environmental quality of life. In MDD, cognitive flexibility was related to social relationships and environmental quality of life. CONCLUSION working memory and cognitive flexibility may be an interesting target for interventions aiming to improve everyday functioning and quality of life in BD and MDD.
Collapse
Affiliation(s)
- André Ponsoni
- Department of Psychology, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Laura Damiani Branco
- Department of Psychology, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Charles Cotrena
- Department of Psychology, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Flávio Milman Shansis
- Medical Sciences Post Graduate Program, University of Vale do Taquari (Univates), Lajeado, Brazil
| | - Rochele Paz Fonseca
- Department of Psychology, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
18
|
Xu Y, Cui D, Zhao Y, Wu C, Mu Q, Fang Z, Hu S, Huang M, Zhang P, Lu S. Volumetric Alterations of the Hippocampal Subfields in Major Depressive Disorder with and without Suicidal Ideation. Behav Brain Res 2023; 457:114733. [PMID: 39491110 DOI: 10.1016/j.bbr.2023.114733] [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: 05/17/2023] [Revised: 08/29/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Major depressive disorder (MDD) is often accompanied with suicidal ideation (SI). Previous studies suggested that MDD patients who experienced suicidal attempts (SA) exhibited smaller hippocampal volume than those without SA. The hippocampus consists of several subfields that are histologically and functionally unique respectively. However, few studies have been designed to investigate the relationship between suicide and volumetric changes of the hippocampal subfields in MDD. A total of 92 participants, including 24 MDD patients with SI (MDD-SI), 30 MDD patients without SI (MDD-nSI), and 38 healthy controls (HC) were recruited in this study. High-resolution structural magnetic resonance images (MRI) were obtained and analyzed by using the automated hippocampal substructure module in FreeSurfer 7.3.2 Analyses of variance (ANOVA) were performed to obtain hippocampal subfields with significant differences among three groups and then post hoc tests were calculated for inter-group comparisons. Finally, the relationships between volumes of the hippocampal subfields and clinical characteristics were assessed using correlation analyses. The ANOVA revealed significant volumetric differences of the hippocampal subfields among three groups in the bilateral cornu ammonis (CA) 1 head, CA4, granule cell layer of the dentate gyrus (GC-ML-DG), molecular layer (ML), the right hippocampus-amygdala transition area (HATA), and the right subiculum (Sub) body. Relative to HC, both groups of MDD showed decreased volumes in the CA1 head, CA4, GC-ML-DG head, ML head, and whole hippocampal head of the left hippocampus, the right HATA, and bilateral whole hippocampal body and whole hippocampus. Meanwhile, the MDD-SI group further exhibited diminished volume in the CA1 head, GC-ML-DG head, ML head, CA4 body, Sub body, and whole hippocampal head of the right hippocampus, as well as bilateral GC-ML-DG body and ML body. Additionally, the MDD-SI group also showed decreased volumes in the right GC-ML-DG body, ML body, and Sub body when comparing to MDD-nSI group. However, no significant association was observed between hippocampal subfield volumes and clinical features in MDD. The present findings suggested that SI in MDD might be attributed to subfield abnormalities in the subiculum and DG-dominant circuit of the right hippocampus.
Collapse
Affiliation(s)
- Yuwei Xu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Mental Disorder's Management of Zhejiang Province, Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, Zhejiang, China; Faculty of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dong Cui
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong, China
| | - Yang Zhao
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Mental Disorder's Management of Zhejiang Province, Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, Zhejiang, China; Faculty of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Clinical Psychology, The Fifth Peoples' Hospital of Lin'an District, Hangzhou, Zhejiang, China
| | - Congchong Wu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Mental Disorder's Management of Zhejiang Province, Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, Zhejiang, China; Faculty of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingli Mu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Mental Disorder's Management of Zhejiang Province, Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, Zhejiang, China; Faculty of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhe Fang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Mental Disorder's Management of Zhejiang Province, Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, Zhejiang, China; Faculty of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Mental Disorder's Management of Zhejiang Province, Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, Zhejiang, China
| | - Manli Huang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Mental Disorder's Management of Zhejiang Province, Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, Zhejiang, China
| | - Peng Zhang
- Department of Psychiatry, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China.
| | - Shaojia Lu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Mental Disorder's Management of Zhejiang Province, Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, Zhejiang, China.
| |
Collapse
|
19
|
Mo M, Zacarias-Pons L, Hoang MT, Mostafaei S, Jurado PG, Stark I, Johnell K, Eriksdotter M, Xu H, Garcia-Ptacek S. Psychiatric Disorders Before and After Dementia Diagnosis. JAMA Netw Open 2023; 6:e2338080. [PMID: 37847498 PMCID: PMC10582787 DOI: 10.1001/jamanetworkopen.2023.38080] [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: 06/26/2023] [Accepted: 08/27/2023] [Indexed: 10/18/2023] Open
Abstract
Importance Little is known about the specific timing and sequence of incident psychiatric comorbidities at different stages of dementia diagnosis. Objectives To examine the temporal risk patterns of psychiatric disorders, including depression, anxiety, stress-related disorders, substance use disorders, sleep disorders, somatoform/conversion disorders, and psychotic disorders, among patients with dementia before, at the time of, and after receipt of a diagnosis. Design, Setting, and Participants This population-based, nationwide cohort study analyzed data from 796 505 participants obtained from 6 registers between January 1, 2000, and December 31, 2017, including the Swedish registry for cognitive/dementia disorders. Patients with dementia were matched on year of birth (±3 years), sex, and region of residence with up to 4 controls. Data were analyzed between March 1, 2023, and August 31, 2023. Exposures Any cause of dementia and dementia subtypes. Main Outcomes and Measures Flexible parametric survival models to determine the time-dependent risk of initial diagnosis of psychiatric disorders, from 7 years prior to dementia diagnosis to 10 years after diagnosis. Subgroup analysis was conducted for psychiatric drug use among persons receiving a diagnosis of dementia from January 1, 2011, to December 31, 2012. Results Of 796 505 patients included in the study (mean [SD] age at diagnosis, 80.2 [8.3] years; 448 869 (56.4%) female), 209 245 had dementia, whereas 587 260 did not, across 7 824 616 person-years. The relative risk of psychiatric disorders was consistently higher among patients with dementia compared with control participants and began to increase from 3 years before diagnosis (hazard ratio, [HR], 1.72; 95% CI, 1.67-1.76), peaked during the week after diagnosis (HR, 4.74; 95% CI, 4.21-5.34), and decreased rapidly thereafter. Decreased risk relative to controls was observed from 5 years after diagnosis (HR, 0.93; 95% CI, 0.87-0.98). The results were similar for Alzheimer disease, mixed dementia, vascular dementia and unspecified dementia. Among patients with dementia, markedly elevated use of psychiatric medications was observed in the year leading up to the dementia diagnosis and peaked 6 months after diagnosis. For example, antidepressant use was persistently higher among patients with dementia compared with controls, and the difference increased from 2 years before dementia diagnosis (15.9% vs 7.9%, P < .001), peaked approximately 6 months after dementia diagnosis (29.1% vs 9.7%, P < .001), and then decreased slowly from 3 years after diagnosis but remained higher than controls 5 years after diagnosis (16.4% vs 6.9%, P < .001). Conclusions and Relevance The findings of this cohort study that patients with dementia had markedly increased risks of psychiatric disorders both before and after dementia diagnosis highlight the significance of incorporating psychiatric preventative and management interventions for individuals with dementia across various diagnostic stages.
Collapse
Affiliation(s)
- Minjia Mo
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Lluis Zacarias-Pons
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Vascular Health Research Group of Girona (ISV-Girona), Institut Universitari d’Investigació en Atenció Primària Jordi Gol i Gurina (IDIAP Jordi Gol), Girona, Spain
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Spain
| | - Minh Tuan Hoang
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Shayan Mostafaei
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Pol Grau Jurado
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Isidora Stark
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Johnell
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Hong Xu
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Sara Garcia-Ptacek
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
20
|
Xu L, Gill MJ, Power C, Fujiwara E. Verbal Memory Performance and Depressive Symptoms in Persons with Treated HIV. AIDS Behav 2023; 27:2823-2833. [PMID: 36786939 DOI: 10.1007/s10461-023-04006-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 02/15/2023]
Abstract
The link between memory and comorbid depression in persons with HIV (PWH) is unclear based on evidence from published cohorts. We compared verbal memory in the HVLT-R in a well-characterized HIV cohort (n = 354) with (n = 102) or without (n = 252) comorbid depressive symptoms, and examined memory correlates in both scenarios. Memory fell within unimpaired ranges, but was lower in depressed than non-depressed PWH. Memory was related to quality of life, sociodemographic, and mental health factors, but not to assessed HIV-related or antiretroviral factors. However, longitudinally (n = 52) memory declined with presence and severity of depressive symptoms. In this treated cohort, verbal memory was unrelated to HIV-related variables but to quality of life and depressive symptoms. Greater performance decline over time also related to acute or ongoing depressive symptoms. These findings highlight the importance of addressing comorbid depressive symptoms to improve quality of life in persons with treated HIV.
Collapse
Affiliation(s)
- Lujie Xu
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - M John Gill
- Department of Medicine, University of Calgary, Calgary, AB, Canada
- Southern Alberta HIV Clinic, Alberta Health Services, Calgary, AB, Canada
| | - Christopher Power
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Esther Fujiwara
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
21
|
Sinclair LI, Ball HA, Bolea-Alamanac BM. Does depression in mid-life predispose to greater cognitive decline in later life in the Whitehall II cohort? J Affect Disord 2023; 335:111-119. [PMID: 37172658 DOI: 10.1016/j.jad.2023.05.014] [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: 10/19/2022] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Later-life depression appears to have different symptomatology and possibly underlying pathology to younger adults. Depression is linked to dementia but whether it is a risk factor or an early sign of dementia remains unclear. Neuroinflammation is increasingly recognised in both conditions. AIMS To investigate the link between depression, inflammation and dementia. We hypothesised that recurrent depression increases the rate of cognitive decline in older adults and that this effect is modified by anti-inflammatory medication. METHODS We used data from Whitehall II including cognitive test results and reliable measures to assess depression. Depression was defined as a self-reported diagnosis or a score of ≥20 on the CESD. The presence/absence of inflammatory illness was assessed using a standardised list of inflammatory conditions. Individuals with dementia, chronic neurological and psychotic conditions were excluded. Logistic and linear regression was used to examine the effect of depression on cognitive test performance and the effect of chronic inflammation. LIMITATIONS Lack of clinical diagnoses of depression. RESULTS There were 1063 individuals with and 2572 without depression. Depression did not affect deterioration in episodic memory, verbal fluency or the AH4 test at 15-year follow up. We found no evidence of an effect of anti-inflammatory medication. Depressed individuals had worse cross-sectional performance on the Mill Hill test and tests of abstract reasoning and verbal fluency at both baseline and 15-year follow-up. CONCLUSIONS Using a UK based study with a long follow-up interval we have shown that depression in individuals aged >50 is not associated with increased cognitive decline.
Collapse
Affiliation(s)
- Lindsey Isla Sinclair
- Department of Clinical Neuroscience, Bristol Medical School, University of Bristol, Learning & Research Building, Southmead Hospital, BS10 5NB, United Kingdom of Great Britain and Northern Ireland.
| | - Harriet Ann Ball
- Department of Clinical Neuroscience, Bristol Medical School, University of Bristol, Learning & Research Building, Southmead Hospital, BS10 5NB, United Kingdom of Great Britain and Northern Ireland
| | | |
Collapse
|
22
|
Zöllinger I, Bauer A, Blotenberg I, Brettschneider C, Buchholz M, Czock D, Döhring J, Escales C, Fankhaenel T, Frese T, Hoffmann W, Kaduszkiewicz H, König HH, Luppa M, Oey A, Pabst A, Sanftenberg L, Thyrian JR, Weiss J, Wendel F, Wiese B, Riedel-Heller SG, Gensichen J. Associations of Depressive Symptoms with Subjective Cognitive Decline in Elderly People-A Cross-Sectional Analysis from the AgeWell.de-Study. J Clin Med 2023; 12:5205. [PMID: 37629244 PMCID: PMC10455560 DOI: 10.3390/jcm12165205] [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: 07/05/2023] [Revised: 07/30/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
To develop effective dementia prevention strategies, it is necessary to understand risk factors, associated factors and early signs of dementia. Subjective cognitive decline (SCD) is the earliest form of dementia. The aim of this study is to assess depression as a factor that is significantly associated with SCD. The data of 1030 general practitioner patients from the AgeWell.de-study (60-77 years; CAIDE dementia risk score ≥ 9) were analysed. A descriptive analysis was conducted using validated instruments like the Geriatric depression scale (GDS), Lubben social network scale (LSNS-6) and education classes according to CASMIN (Comparative Analysis of Social Mobility in Industrial Nations). A multivariate regression model with the dependent variable SCD was calculated. Of the 1030 participants, 5.9% had depressive symptoms and 31.3% SCD. The group with depressive symptoms showed significantly higher body-mass-index (p = 0.005), lower education class (p = 0.022), lower LSNS-6 score (p < 0.001), higher sports activity (p < 0.001), and more sleeping problems (p = 0.026). In the regression model a higher GDS-score [Odds ratio (OR): 1.219 (p < 0.001)], more sleeping problems [OR: 1.550 (p = 0.017)] and higher education class [middle/high: OR: 1.474/1.875 (p = 0.037/0.004)] were significantly associated with SCD. This study identified depressive symptoms, sleeping problems, and higher education classes as factors associated with SCD, which can represent an early form of dementia.
Collapse
Affiliation(s)
- Isabel Zöllinger
- Institute of General Practice and Family Medicine, University Hospital of LMU Munich, 80336 Munich, Germany; (L.S.); (J.W.); (F.W.); (J.G.)
| | - Alexander Bauer
- Institute of General Practice and Family Medicine, Martin-Luther-University Halle-Wittenberg, 06112 Halle, Germany; (A.B.); (T.F.); (T.F.)
| | - Iris Blotenberg
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17489 Greifswald, Germany; (I.B.); (M.B.); (W.H.); (J.R.T.)
| | - Christian Brettschneider
- Department of Health Economics and Health Services Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (C.B.); (H.-H.K.)
| | - Maresa Buchholz
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17489 Greifswald, Germany; (I.B.); (M.B.); (W.H.); (J.R.T.)
| | - David Czock
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Juliane Döhring
- Institute of General Practice, University of Kiel, 24105 Kiel, Germany; (J.D.); (C.E.); (H.K.)
| | - Catharina Escales
- Institute of General Practice, University of Kiel, 24105 Kiel, Germany; (J.D.); (C.E.); (H.K.)
| | - Thomas Fankhaenel
- Institute of General Practice and Family Medicine, Martin-Luther-University Halle-Wittenberg, 06112 Halle, Germany; (A.B.); (T.F.); (T.F.)
| | - Thomas Frese
- Institute of General Practice and Family Medicine, Martin-Luther-University Halle-Wittenberg, 06112 Halle, Germany; (A.B.); (T.F.); (T.F.)
| | - Wolfgang Hoffmann
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17489 Greifswald, Germany; (I.B.); (M.B.); (W.H.); (J.R.T.)
- Institute for Community Medicine, University Medicine Greifswald, 17487 Greifswald, Germany
| | - Hanna Kaduszkiewicz
- Institute of General Practice, University of Kiel, 24105 Kiel, Germany; (J.D.); (C.E.); (H.K.)
| | - Hans-Helmut König
- Department of Health Economics and Health Services Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (C.B.); (H.-H.K.)
| | - Melanie Luppa
- Institute of Social Medicine, Occupational Health and Public Health (ISAP), Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (M.L.); (A.P.); (S.G.R.-H.)
| | - Anke Oey
- State Health Department of Lower Saxony, 30449 Hannover, Germany;
| | - Alexander Pabst
- Institute of Social Medicine, Occupational Health and Public Health (ISAP), Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (M.L.); (A.P.); (S.G.R.-H.)
| | - Linda Sanftenberg
- Institute of General Practice and Family Medicine, University Hospital of LMU Munich, 80336 Munich, Germany; (L.S.); (J.W.); (F.W.); (J.G.)
| | - Jochen René Thyrian
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17489 Greifswald, Germany; (I.B.); (M.B.); (W.H.); (J.R.T.)
- Institute for Community Medicine, University Medicine Greifswald, 17487 Greifswald, Germany
| | - Julian Weiss
- Institute of General Practice and Family Medicine, University Hospital of LMU Munich, 80336 Munich, Germany; (L.S.); (J.W.); (F.W.); (J.G.)
| | - Flora Wendel
- Institute of General Practice and Family Medicine, University Hospital of LMU Munich, 80336 Munich, Germany; (L.S.); (J.W.); (F.W.); (J.G.)
| | - Birgitt Wiese
- Work Group Medical Statistics and IT-Infrastructure, Institute for General Practice, Hannover Medical School, 30625 Hannover, Germany;
| | - Steffi G. Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health (ISAP), Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (M.L.); (A.P.); (S.G.R.-H.)
| | - Jochen Gensichen
- Institute of General Practice and Family Medicine, University Hospital of LMU Munich, 80336 Munich, Germany; (L.S.); (J.W.); (F.W.); (J.G.)
| |
Collapse
|
23
|
Akan O, Bierbrauer A, Kunz L, Gajewski PD, Getzmann S, Hengstler JG, Wascher E, Axmacher N, Wolf OT. Chronic stress is associated with specific path integration deficits. Behav Brain Res 2023; 442:114305. [PMID: 36682499 DOI: 10.1016/j.bbr.2023.114305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
Repeated exposure to stress (chronic stress) can cause excess levels of circulating cortisol and has detrimental influences on various cognitive functions including long-term memory and navigation. However, it remains an open question whether chronic stress affects path integration, a navigational strategy that presumably relies on the functioning of grid cells in the medial entorhinal cortex. The entorhinal cortex is a brain region in the medial temporal lobe, which contains multiple cell types involved in spatial navigation (and episodic memory), and a high number of corticosteroid receptors, predisposing it as a potential target of cortisol effects. Here, our goal was to investigate the association between chronic stress and path integration performance. We assessed chronic stress via hair cortisol concentration (physiological measure) and the Perceived Stress Questionnaire (subjective measure) in 52 female participants aged 22-65 years. Path integration was measured using a virtual homing task. Linear mixed models revealed selective impairments associated with chronic stress that depended on error type and environmental features. When focusing on distance estimations in the path integration task, we observed a significant relationship to hair cortisol concentrations indicating impaired path integration particularly during trials with higher difficulty in participants with high hair cortisol concentrations. This relationship especially emerged in the absence of spatial cues (a boundary or a landmark), and particularly in participants who reported high levels of subjectively experienced chronic stress. The findings are in line with the hypothesis that chronic stress compromises path integration, possibly via an effect on the entorhinal grid cell system.
Collapse
Affiliation(s)
- Osman Akan
- Department of Cognitive Psychology, Faculty of Psychology, Ruhr University Bochum, Germany.
| | - Anne Bierbrauer
- Institute for Systems Neuroscience, Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuropsychology, Faculty of Psychology, Ruhr University Bochum, Germany
| | - Lukas Kunz
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Patrick D Gajewski
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Germany
| | - Stephan Getzmann
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Germany
| | - Edmund Wascher
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Germany
| | - Nikolai Axmacher
- Department of Neuropsychology, Faculty of Psychology, Ruhr University Bochum, Germany
| | - Oliver T Wolf
- Department of Cognitive Psychology, Faculty of Psychology, Ruhr University Bochum, Germany
| |
Collapse
|
24
|
Rao RV, Subramaniam KG, Gregory J, Bredesen AL, Coward C, Okada S, Kelly L, Bredesen DE. Rationale for a Multi-Factorial Approach for the Reversal of Cognitive Decline in Alzheimer's Disease and MCI: A Review. Int J Mol Sci 2023; 24:ijms24021659. [PMID: 36675177 PMCID: PMC9865291 DOI: 10.3390/ijms24021659] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial, progressive, neurodegenerative disease typically characterized by memory loss, personality changes, and a decline in overall cognitive function. Usually manifesting in individuals over the age of 60, this is the most prevalent type of dementia and remains the fifth leading cause of death among Americans aged 65 and older. While the development of effective treatment and prevention for AD is a major healthcare goal, unfortunately, therapeutic approaches to date have yet to find a treatment plan that produces long-term cognitive improvement. Drugs that may be able to slow down the progression rate of AD are being introduced to the market; however, there has been no previous solution for preventing or reversing the disease-associated cognitive decline. Recent studies have identified several factors that contribute to the progression and severity of the disease: diet, lifestyle, stress, sleep, nutrient deficiencies, mental health, socialization, and toxins. Thus, increasing evidence supports dietary and other lifestyle changes as potentially effective ways to prevent, slow, or reverse AD progression. Studies also have demonstrated that a personalized, multi-therapeutic approach is needed to improve metabolic abnormalities and AD-associated cognitive decline. These studies suggest the effects of abnormalities, such as insulin resistance, chronic inflammation, hypovitaminosis D, hormonal deficiencies, and hyperhomocysteinemia, in the AD process. Therefore a personalized, multi-therapeutic program based on an individual's genetics and biochemistry may be preferable over a single-drug/mono-therapeutic approach. This article reviews these multi-therapeutic strategies that identify and attenuate all the risk factors specific to each affected individual. This article systematically reviews studies that have incorporated multiple strategies that target numerous factors simultaneously to reverse or treat cognitive decline. We included high-quality clinical trials and observational studies that focused on the cognitive effects of programs comprising lifestyle, physical, and mental activity, as well as nutritional aspects. Articles from PubMed Central, Scopus, and Google Scholar databases were collected, and abstracts were reviewed for relevance to the subject matter. Epidemiological, pathological, toxicological, genetic, and biochemical studies have all concluded that AD represents a complex network insufficiency. The research studies explored in this manuscript confirm the need for a multifactorial approach to target the various risk factors of AD. A single-drug approach may delay the progression of memory loss but, to date, has not prevented or reversed it. Diet, physical activity, sleep, stress, and environment all contribute to the progression of the disease, and, therefore, a multi-factorial optimization of network support and function offers a rational therapeutic strategy. Thus, a multi-therapeutic program that simultaneously targets multiple factors underlying the AD network may be more effective than a mono-therapeutic approach.
Collapse
Affiliation(s)
- Rammohan V. Rao
- Apollo Health, Burlingame, CA 94011, USA
- Correspondence: (R.V.R.); (D.E.B.)
| | | | | | | | | | - Sho Okada
- Apollo Health, Burlingame, CA 94011, USA
| | | | - Dale E. Bredesen
- Apollo Health, Burlingame, CA 94011, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90024, USA
- Correspondence: (R.V.R.); (D.E.B.)
| |
Collapse
|
25
|
Ikeda N, Yamada S, Yasuda K, Uenishi S, Tamaki A, Ishida T, Tabata M, Tsuji T, Kimoto S, Takahashi S. Structural connectivity between the hippocampus and cortical/subcortical area relates to cognitive impairment in schizophrenia but not in mood disorders. J Neuropsychol 2022. [DOI: 10.1111/jnp.12298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 08/10/2022] [Accepted: 09/11/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Natsuko Ikeda
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
- Department of Psychiatry Wakayama Prefectural Mental Health Care Center Wakayama Japan
| | - Shinichi Yamada
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
| | - Kasumi Yasuda
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
| | - Shinya Uenishi
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
- Department of Psychiatry Hidaka Hospital Gobo Japan
| | - Atsushi Tamaki
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
- Department of Psychiatry Hidaka Hospital Gobo Japan
| | - Takuya Ishida
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
| | - Michiyo Tabata
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
| | - Tomikimi Tsuji
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
| | - Sohei Kimoto
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
| | - Shun Takahashi
- Department of Neuropsychiatry Wakayama Medical University Wakayama Japan
- Clinical Research and Education Center Asakayama General Hospital Sakai Japan
- Graduate School of Rehabilitation Science Osaka Metropolitan University Habikino Japan
- Department of Psychiatry Osaka University Graduate School of Medicine Suita Japan
| |
Collapse
|
26
|
Chen RF, Cai Y, Zhu ZH, Hou WL, Chen P, Wang J, Jia YM, Zhu ZB, Zhang YH, Tao LY, Wu JW, Zhang J, Zhang H, Wang ZX, Jia QF, Hui L. Sleep disorder as a clinical risk factor of major depression: associated with cognitive impairment. Asian J Psychiatr 2022; 76:103228. [PMID: 35973338 DOI: 10.1016/j.ajp.2022.103228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/08/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND This research aims explored the sleep disorder (SD) role in major depressive disorder (MDD), and the SD influencing their cognition. METHODS 372 MDD patients and 457 healthy controls (HCs) were enrolled. RESULTS Patients increased a 38.88 times SD risk compared with HCs. In patients, visuospatial/constructional score was lower in SD than non-SD, and PSQI score was negatively associated with visuospatial/constructional score of SD. In SD and non-SD, RBANS scores were lower in MDD than HCs, excepted for visuospatial/constructional in non-SD. CONCLUSION The SD as a MDD risk factor, has more serious visuospatial/constructional impairment alleviated via improving sleep/depression in patients.
Collapse
Affiliation(s)
- Ru Feng Chen
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China
| | - Yuan Cai
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China
| | - Zhen Hua Zhu
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China
| | - Wen Long Hou
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China
| | - Peng Chen
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China
| | - Jing Wang
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China
| | - Yi Ming Jia
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu Province, PR China
| | - Zheng Bao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu Province, PR China
| | - Yong Hong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu Province, PR China
| | - Lu Yang Tao
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China
| | - Jia Wei Wu
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China
| | - Jianping Zhang
- Department of Psychiatry, Weill Cornell Medical College, Cornell University, New York, NY 10605, USA
| | - Huiping Zhang
- Departments of Psychiatry and Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02118-2526, USA
| | - Zhi Xin Wang
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China.
| | - Qiu Fang Jia
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China.
| | - Li Hui
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, School of Basic Medicine and Biological Sciences, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu Province, PR China.
| |
Collapse
|
27
|
Association between cortisol and aging-related hippocampus volume changes in community-dwelling older adults: a 7-year follow-up study. BMC Geriatr 2022; 22:765. [PMID: 36131257 PMCID: PMC9491648 DOI: 10.1186/s12877-022-03455-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Identifying peripheral biomarkers related to modifiable risk factors to prevent dementia at an early stage will be extremely beneficial. We have been studying how older adults can maintain their mental health and continue to live in a familiar community. The aim of this study is to investigate the association between serum cortisol levels and brain volume among older adults in rural Japan. METHODS This was a longitudinal study conducted in Kurokawa-cho, Imari, Saga Prefecture, Japan, among people aged 65 years and above, as reported previously. We conducted a survey twice. The first survey was conducted from October 2009 to March 2011 (Timepoint 1) and the second was conducted from November 2016 to September 2017 (Timepoint 2). Blood samples for serum cortisol levels analysis were collected from participants at Timepoint 1. Serum cortisol levels were measured using the enzyme-linked immunosorbent assay. The participants underwent brain MRI examinations, and Mini-Mental State Examination (MMSE) and Clinical Dementia Rating (CDR) for cognitive function assessment at Timepoint 1 and Timepoint 2. We obtained 70 participants (16 men, mean age 72.69 ± 3.18 years; 54 women, mean age 72.69 ± 4.60 years, at Timepoint 1) for analysis. Correlation analysis was performed between serum cortisol levels at baseline (Timepoint 1) and brain volume (Timepoint 1, Timepoint 2, and Timepoint 1-Timepoint 2 difference) using voxel-based morphometry method. RESULTS There was no significant difference in serum cortisol levels between men (72.32 ± 17.30 ng/ml) and women (76.60 ± 21.12 ng/ml) at baseline. Additionally, no effect of blood collection time on cortisol levels was observed in these participants. Small volume correction analysis at the cluster level by applying multiple comparison corrections (family-wise error; P < 0.05) showed a negative correlation between serum cortisol levels (Timepoint 1) and brain volume (Timepoint 2) within the region containing the left hippocampus. CONCLUSIONS Serum cortisol levels may serve as a peripheral biomarker of age-related volume changes involving the hippocampus in older adults aged 65 years and above.
Collapse
|
28
|
Ahmed R, Ryan C, Christman S, Elson D, Bermudez C, Landman BA, Szymkowicz SM, Boyd BD, Kang H, Taylor WD. Structural MRI-Based Measures of Accelerated Brain Aging do not Moderate the Acute Antidepressant Response in Late-Life Depression. Am J Geriatr Psychiatry 2022; 30:1015-1025. [PMID: 34949526 PMCID: PMC9142760 DOI: 10.1016/j.jagp.2021.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/14/2021] [Accepted: 11/21/2021] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Late-life depression (LLD) is characterized by accelerated biological aging. Accelerated brain aging, estimated from structural magnetic resonance imaging (sMRI) data by a machine learning algorithm, is associated with LLD diagnosis, poorer cognitive performance, and disability. We hypothesized that accelerated brain aging moderates the antidepressant response. DESIGN AND INTERVENTIONS Following MRI, participants entered an 8-week randomized, controlled trial of escitalopram. Nonremitting participants then entered an open-label 8-week trial of bupropion. PARTICIPANTS Ninety-five individuals with LLD. MEASUREMENTS A machine learning algorithm estimated each participant's brain age from sMRI data. This was used to calculate the brain-age gap (BAG), or how estimated age differed from chronological age. Secondary sMRI measures of aging pathology included white matter hyperintensity (WMH) volumes and hippocampal volumes. Mixed models examined the relationship between sMRI measures and change in depression severity. Initial analyses tested for a moderating effect of MRI measures on change in depression severity with escitalopram. Subsequent analyses tested for the effect of MRI measures on change in depression severity over time across trials. RESULTS In the blinded initial phase, BAG was not significantly associated with a differential response to escitalopram over time. BAG was also not associated with a change in depression severity over time across both arms in the blinded phase or in the subsequent open-label bupropion phase. We similarly did not observe effects of WMH volume or hippocampal volume on change in depression severity over time. CONCLUSION sMRI markers of accelerated brain aging were not associated with treatment response in this sequential antidepressant trial.
Collapse
Affiliation(s)
- Ryan Ahmed
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN
| | - Claire Ryan
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN
| | - Seth Christman
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN
| | - Damian Elson
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN
| | - Camilo Bermudez
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN
| | - Bennett A Landman
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN
| | - Sarah M Szymkowicz
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN
| | - Brian D Boyd
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN
| | - Hakmook Kang
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN
| | - Warren D Taylor
- School of Medicine (RA), Vanderbilt University, Nashville, TN; Department of Psychiatry and Behavioral Sciences (SC, DE, BAL, SMS, BDB, WDT), Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Engineering (CB, BAL), Vanderbilt University, Nashville TN; Department of Electrical Engineering and Computer Science (BAL), Vanderbilt University, Nashville, TN; Department of Biostatistics (HK), Vanderbilt University Medical Center, Nashville, TN; Geriatric Research, Education, and Clinical Center (WDT), Veterans Affairs Tennessee Valley Health System, Nashville, TN.
| |
Collapse
|
29
|
Thalamic abnormalities in older adults with remitted early-onset depression using structural magnetic resonance imaging. J Affect Disord 2022; 300:150-157. [PMID: 34942221 DOI: 10.1016/j.jad.2021.12.067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/16/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The thalamus is a key diencephalic structure involved in major depressive disorder (MDD). Studies have consistently revealed abnormalities in thalamic volumes in older adults with late-onset depression (LOD), however abnormalities in older adults with early-onset depression (EOD) have not yet been well-studied. METHODS Fifty-nine euthymic participants with a history of EOD and fifty-nine matched comparison participants without a lifetime history of depression underwent neuroimaging, medical and neuropsychological assessments. Thalamic volumes were compared between groups. To investigate the previously-proposed right hemispheric (RH) dominance theory of MDD, we explored the bilateral, right and left hemispheric (LH) thalamic volumes. Multiple regression analyses were used to evaluate between-group and within-group effects. Correlational analyses examined associations between group and cognitive performance. RESULTS Relative to the comparison group, those with EOD had significantly larger bilateral, LH and RH thalamic volumes. Those with EOD, those who were younger, and those who had fewer years of education demonstrated larger bilateral and LH thalamic volumes. For RH thalamic volumes, those with EOD and those who were younger demonstrated larger RH thalamic volumes. EOD within-group models were also run to assess associations between relevant depression variables. The results showed that only age was significant for bilateral and RH thalamic volumes. For the LH thalamic volumes, the model was not significant. No significant correlations were found between cognitive performance and EOD groups. CONCLUSION Older adults with a history of EOD showed significantly larger bilateral, RH and LH thalamic volumes. Further research is needed to delineate potential underlying mechanisms of this change.
Collapse
|
30
|
Dobielska M, Bartosik NK, Zyzik KA, Kowalczyk E, Karbownik MS. Mechanisms of Cognitive Impairment in Depression. May Probiotics Help? Front Psychiatry 2022; 13:904426. [PMID: 35757204 PMCID: PMC9218185 DOI: 10.3389/fpsyt.2022.904426] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022] Open
Abstract
Depression is the major cause of disability globally. Apart from lowered mood and accompanying symptoms, it leads to cognitive impairment that altogether predicts disadvantaged social functioning. Reduced cognitive function in depression appears a bit neglected in the field of clinical and molecular psychiatry, while it is estimated to occur in two-thirds of depressed patients and persist in at least one third of remitted patients. This problem, therefore, requires elucidation at the biomolecular and system levels and calls for improvement in therapeutic approach. In this review study, we address the above-mentioned issues by discussing putative mechanisms of cognitive decline in depression: (1) increased oxidative stress and (2) inflammation, (3) disturbed hypothalamus-pituitary-adrenals axis, and (4) reduced monoamines functionality. Moreover, we acknowledge additional underpinnings of cognitive impairment in depressed elderly: (5) vascular-originated brain ischemia and (6) amyloid-beta plaque accumulation. Additionally, by reviewing molecular, pre-clinical and clinical evidence, we propose gut microbiota-targeted strategies as potential adjuvant therapeutics. The study provides a consolidated source of knowledge regarding mechanisms of cognitive impairment in depression and may path the way toward improved treatment options.
Collapse
Affiliation(s)
- Maria Dobielska
- Students' Research Club, Department of Pharmacology and Toxicology, Medical University of Łódź, Łódź, Poland
| | - Natalia Karina Bartosik
- Students' Research Club, Department of Pharmacology and Toxicology, Medical University of Łódź, Łódź, Poland
| | - Kamil A Zyzik
- Institute of Sociology, Jagiellonian University, Kraków, Poland
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Łódź, Łódź, Poland
| | | |
Collapse
|
31
|
Brosch K, Stein F, Schmitt S, Pfarr JK, Ringwald KG, Thomas-Odenthal F, Meller T, Steinsträter O, Waltemate L, Lemke H, Meinert S, Winter A, Breuer F, Thiel K, Grotegerd D, Hahn T, Jansen A, Dannlowski U, Krug A, Nenadić I, Kircher T. Reduced hippocampal gray matter volume is a common feature of patients with major depression, bipolar disorder, and schizophrenia spectrum disorders. Mol Psychiatry 2022; 27:4234-4243. [PMID: 35840798 PMCID: PMC9718668 DOI: 10.1038/s41380-022-01687-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023]
Abstract
Major depressive disorder (MDD), bipolar disorder (BD), and schizophrenia spectrum disorder (SSD, schizophrenia, and schizoaffective disorder) overlap in symptomatology, risk factors, genetics, and other biological measures. Based on previous findings, it remains unclear what transdiagnostic regional gray matter volume (GMV) alterations exist across these disorders, and with which factors they are associated. GMV (3-T magnetic resonance imaging) was compared between healthy controls (HC; n = 110), DSM-IV-TR diagnosed MDD (n = 110), BD (n = 110), and SSD patients (n = 110), matched for age and sex. We applied a conjunction analysis to identify shared GMV alterations across the disorders. To identify potential origins of identified GMV clusters, we associated them with early and current risk and protective factors, psychopathology, and neuropsychology, applying multiple regression models. Common to all diagnoses (vs. HC), we identified GMV reductions in the left hippocampus. This cluster was associated with the neuropsychology factor working memory/executive functioning, stressful life events, and with global assessment of functioning. Differential effects between groups were present in the left and right frontal operculae and left insula, with volume variances across groups highly overlapping. Our study is the first with a large, matched, transdiagnostic sample to yield shared GMV alterations in the left hippocampus across major mental disorders. The hippocampus is a major network hub, orchestrating a range of mental functions. Our findings underscore the need for a novel stratification of mental disorders, other than categorical diagnoses.
Collapse
Affiliation(s)
- Katharina Brosch
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany. .,Center for Mind, Brain and Behavior (CMBB), Marburg, Germany.
| | - Frederike Stein
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.513205.0Center for Mind, Brain and Behavior (CMBB), Marburg, Germany
| | - Simon Schmitt
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.513205.0Center for Mind, Brain and Behavior (CMBB), Marburg, Germany ,grid.10423.340000 0000 9529 9877Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Julia-Katharina Pfarr
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.513205.0Center for Mind, Brain and Behavior (CMBB), Marburg, Germany
| | - Kai G. Ringwald
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.513205.0Center for Mind, Brain and Behavior (CMBB), Marburg, Germany
| | - Florian Thomas-Odenthal
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.513205.0Center for Mind, Brain and Behavior (CMBB), Marburg, Germany
| | - Tina Meller
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.513205.0Center for Mind, Brain and Behavior (CMBB), Marburg, Germany
| | - Olaf Steinsträter
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.10253.350000 0004 1936 9756Core-Facility BrainImaging, Faculty of Medicine, Philipps-University Marburg, Marburg, Germany
| | - Lena Waltemate
- grid.5949.10000 0001 2172 9288Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Hannah Lemke
- grid.5949.10000 0001 2172 9288Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- grid.5949.10000 0001 2172 9288Institute for Translational Psychiatry, University of Münster, Münster, Germany ,grid.5949.10000 0001 2172 9288Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Alexandra Winter
- grid.5949.10000 0001 2172 9288Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Fabian Breuer
- grid.5949.10000 0001 2172 9288Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Thiel
- grid.5949.10000 0001 2172 9288Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dominik Grotegerd
- grid.5949.10000 0001 2172 9288Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tim Hahn
- grid.5949.10000 0001 2172 9288Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Andreas Jansen
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.513205.0Center for Mind, Brain and Behavior (CMBB), Marburg, Germany ,grid.10253.350000 0004 1936 9756Core-Facility BrainImaging, Faculty of Medicine, Philipps-University Marburg, Marburg, Germany
| | - Udo Dannlowski
- grid.5949.10000 0001 2172 9288Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Axel Krug
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.10388.320000 0001 2240 3300Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Igor Nenadić
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.513205.0Center for Mind, Brain and Behavior (CMBB), Marburg, Germany
| | - Tilo Kircher
- grid.10253.350000 0004 1936 9756Department of Psychiatry and Psychotherapy, Philipps-University Marburg, University Hospital Marburg, UKGM, Marburg, Germany ,grid.513205.0Center for Mind, Brain and Behavior (CMBB), Marburg, Germany
| |
Collapse
|
32
|
Kim IB, Park SC. The Entorhinal Cortex and Adult Neurogenesis in Major Depression. Int J Mol Sci 2021; 22:11725. [PMID: 34769155 PMCID: PMC8583901 DOI: 10.3390/ijms222111725] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022] Open
Abstract
Depression is characterized by impairments in adult neurogenesis. Reduced hippocampal function, which is suggestive of neurogenesis impairments, is associated with depression-related phenotypes. As adult neurogenesis operates in an activity-dependent manner, disruption of hippocampal neurogenesis in depression may be a consequence of neural circuitry impairments. In particular, the entorhinal cortex is known to have a regulatory effect on the neural circuitry related to hippocampal function and adult neurogenesis. However, a comprehensive understanding of how disruption of the neural circuitry can lead to neurogenesis impairments in depression remains unclear with respect to the regulatory role of the entorhinal cortex. This review highlights recent findings suggesting neural circuitry-regulated neurogenesis, with a focus on the potential role of the entorhinal cortex in hippocampal neurogenesis in depression-related cognitive and emotional phenotypes. Taken together, these findings may provide a better understanding of the entorhinal cortex-regulated hippocampal neurogenesis model of depression.
Collapse
Affiliation(s)
- Il Bin Kim
- Department of Psychiatry, Hanyang University Guri Hospital, Guri 11923, Korea;
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seon-Cheol Park
- Department of Psychiatry, Hanyang University Guri Hospital, Guri 11923, Korea;
- Department of Psychiatry, Hanyang University College of Medicine, Seoul 04763, Korea
| |
Collapse
|
33
|
Kim HK, Zai G, Hennings JM, Müller DJ, Kloiber S. Changes in RNA expression levels during antidepressant treatment: a systematic review. J Neural Transm (Vienna) 2021; 128:1461-1477. [PMID: 34415438 DOI: 10.1007/s00702-021-02394-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/26/2021] [Indexed: 12/28/2022]
Abstract
More than a third of patients treated with antidepressants experience treatment resistance. Furthermore, molecular pathways involved in antidepressant effect have yet to be fully understood. Therefore, we performed a systematic review of clinical studies that examined changes in RNA expression levels produced by antidepressant treatment. Literature search was performed through April 2021 for peer-reviewed studies measuring changes in mRNA or non-coding RNA levels before and after antidepressant treatment in human participants following PRISMA guidelines. Thirty-one studies were included in qualitative synthesis. We identified a large amount of heterogeneity between the studies for genes/RNAs measured, antidepressants used, and treatment duration. Of the six RNAs examined by more than one study, expression of the brain-derived neurotrophic factor (BDNF) gene and genes in the inflammation pathway, particularly IL-1β, were consistently reported to be altered by antidepressant treatment. Limitations of this review include heterogeneity of the studies, possibility of positive publication bias, and risk of false-negative findings secondary to small sample sizes. In conclusion, our systematic review provides an updated synthesis of RNA expression changes produced by antidepressant treatment in human participants, where genes in the BDNF and inflammatory pathways were identified as potential targets of antidepressant effect. Importantly, these findings also highlight the need for replication of the included studies in multiple strong, placebo-controlled studies for the identification of evidence-based markers that can be targeted to improve treatment outcomes.
Collapse
Affiliation(s)
| | - Gwyneth Zai
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 100 Stokes Street, Toronto, ON, M6H 1J4, Canada.,Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Daniel J Müller
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 100 Stokes Street, Toronto, ON, M6H 1J4, Canada.,Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Stefan Kloiber
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada. .,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 100 Stokes Street, Toronto, ON, M6H 1J4, Canada. .,Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
34
|
Takamiya A, Vande Casteele T, Koole M, De Winter FL, Bouckaert F, Van den Stock J, Sunaert S, Dupont P, Vandenberghe R, Van Laere K, Vandenbulcke M, Emsell L. Lower regional gray matter volume in the absence of higher cortical amyloid burden in late-life depression. Sci Rep 2021; 11:15981. [PMID: 34354136 PMCID: PMC8342521 DOI: 10.1038/s41598-021-95206-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
Late-life depression (LLD) is associated with a risk of developing Alzheimer's disease (AD). However, the role of AD-pathophysiology in LLD, and its association with clinical symptoms and cognitive function are elusive. In this study, one hundred subjects underwent amyloid positron emission tomography (PET) imaging with [18F]-flutemetamol and structural MRI: 48 severely depressed elderly subjects (age 74.1 ± 7.5 years, 33 female) and 52 age-/gender-matched healthy controls (72.4 ± 6.4 years, 37 female). The Geriatric Depression Scale (GDS) and Rey Auditory Verbal Learning Test (RAVLT) were used to assess the severity of depressive symptoms and episodic memory function respectively. Amyloid deposition was quantified using the standardized uptake value ratio. Whole-brain voxel-wise comparisons of amyloid deposition and gray matter volume (GMV) between LLD and controls were performed. Multivariate analysis of covariance was conducted to investigate the association of regional differences in amyloid deposition and GMV with clinical factors, including GDS and RAVLT. As a result, there were no significant group differences in amyloid deposition. In contrast, LLD showed significant lower GMV in the left temporal and parietal region. GMV reduction in the left temporal region was associated with episodic memory dysfunction, but not with depression severity. Regional GMV reduction was not associated with amyloid deposition. LLD is associated with lower GMV in regions that overlap with AD-pathophysiology, and which are associated with episodic memory function. The lack of corresponding associations with amyloid suggests that lower GMV driven by non-amyloid pathology may play a central role in the neurobiology of LLD presenting as a psychiatric disorder.Trial registration: European Union Drug Regulating Authorities Clinical Trials identifier: EudraCT 2009-018064-95.
Collapse
Affiliation(s)
- Akihiro Takamiya
- grid.5596.f0000 0001 0668 7884Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium ,grid.26091.3c0000 0004 1936 9959Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Thomas Vande Casteele
- grid.5596.f0000 0001 0668 7884Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Michel Koole
- grid.5596.f0000 0001 0668 7884Nuclear Medicine and Molecular Imaging, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - François-Laurent De Winter
- grid.5596.f0000 0001 0668 7884Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Filip Bouckaert
- grid.5596.f0000 0001 0668 7884Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Jan Van den Stock
- grid.5596.f0000 0001 0668 7884Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Stefan Sunaert
- grid.5596.f0000 0001 0668 7884Department of Imaging & Pathology, Translational MRI, KU Leuven, Leuven, Belgium ,grid.410569.f0000 0004 0626 3338Department of Radiology, University Hospitals Leuven (UZ Leuven), Leuven, Belgium
| | - Patrick Dupont
- grid.5596.f0000 0001 0668 7884Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Alzheimer Research Centre KU Leuven, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- grid.5596.f0000 0001 0668 7884Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Alzheimer Research Centre KU Leuven, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.410569.f0000 0004 0626 3338Neurology Department, University Hospitals Leuven (UZ Leuven), Leuven, Belgium
| | - Koen Van Laere
- grid.5596.f0000 0001 0668 7884Nuclear Medicine and Molecular Imaging, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- grid.5596.f0000 0001 0668 7884Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Louise Emsell
- grid.5596.f0000 0001 0668 7884Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Department of Imaging & Pathology, Translational MRI, KU Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Leung IHK, Broadhouse KM, Mowszowski L, LaMonica HM, Palmer JR, Hickie IB, Naismith SL, Duffy SL. Association between lifetime depression history, hippocampal volume and memory in non-amnestic mild cognitive impairment. Eur J Neurosci 2021; 54:4953-4970. [PMID: 33765347 DOI: 10.1111/ejn.15207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/01/2021] [Accepted: 03/14/2021] [Indexed: 11/28/2022]
Abstract
Hippocampal subfield volume loss in older adults with amnestic mild cognitive impairment (aMCI) and depression history are associated with amyloid beta and tau pathology, thereby increasing the risk for Alzheimer's disease (AD). However, no studies have exclusively examined distinct alterations in hippocampal subfields in non-amnestic MCI (naMCI) in relation to depression history. Here, we used both longitudinal and transverse hippocampal segmentation methods using the automated FreeSurfer software to examine whether a lifetime depression history is associated with differences in hippocampal head/body/tail (H/B/T) and key subfield volumes (CA1, subiculum, dentate gyrus) in older adults with naMCI. Further, we explored whether differences in hippocampal H/B/T and subfield volumes were associated with structured and unstructured verbal encoding and retention, comparing those with and without a depression history. The naMCI with a depression history group demonstrated larger or relatively preserved right CA1 volumes, which were associated with better unstructured verbal encoding and as well as structured verbal memory retention. This association between memory encoding and hippocampal CA1 and total head volume was significantly different to those with no depression history. The relationship between right CA1 volume and memory retention was also moderated by depression history status F (5,143) = 7.84, p < 0.001, R2 = 0.22. Those participants taking antidepressants had significantly larger hippocampal subiculum (p = 0.008), and right hippocampal body (p = 0.004) and better performance on structured encoding (p = 0.011) and unstructured memory retention (p = 0.009). These findings highlight the importance of lifetime depression history and antidepressant use on the hippocampus and encoding and memory retention in naMCI.
Collapse
Affiliation(s)
- Isabella Hoi Kei Leung
- Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, Central Clinical School, University of Sydney, Sydney, NSW, Australia
| | - Kathryn Mary Broadhouse
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Science and Engineering, University of the Sunshine Coast, Sunshine Coast, QLD, Australia
| | - Loren Mowszowski
- Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Faculty of Science, School of Psychology, University of Sydney, Sydney, NSW, Australia
| | - Haley M LaMonica
- Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, Central Clinical School, University of Sydney, Sydney, NSW, Australia
| | - Jake Robert Palmer
- Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Department of Psychology, Macquarie University, Sydney, NSW, Australia
| | - Ian B Hickie
- Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Sharon L Naismith
- Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Faculty of Science, School of Psychology, University of Sydney, Sydney, NSW, Australia
| | - Shantel Leigh Duffy
- Healthy Brain Ageing Program, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, Discipline of Exercise and Sport Science, Faculty of Health Sciences, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
36
|
Kim IB, Park SC. Neural Circuitry-Neurogenesis Coupling Model of Depression. Int J Mol Sci 2021; 22:2468. [PMID: 33671109 PMCID: PMC7957816 DOI: 10.3390/ijms22052468] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Depression is characterized by the disruption of both neural circuitry and neurogenesis. Defects in hippocampal activity and volume, indicative of reduced neurogenesis, are associated with depression-related behaviors in both humans and animals. Neurogenesis in adulthood is considered an activity-dependent process; therefore, hippocampal neurogenesis defects in depression can be a result of defective neural circuitry activity. However, the mechanistic understanding of how defective neural circuitry can induce neurogenesis defects in depression remains unclear. This review highlights the current findings supporting the neural circuitry-regulated neurogenesis, especially focusing on hippocampal neurogenesis regulated by the entorhinal cortex, with regard to memory, pattern separation, and mood. Taken together, these findings may pave the way for future progress in neural circuitry-neurogenesis coupling studies of depression.
Collapse
Affiliation(s)
- Il Bin Kim
- Department of Psychiatry, Hanyang University Guri Hospital, Guri 11923, Korea;
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Seon-Cheol Park
- Department of Psychiatry, Hanyang University Guri Hospital, Guri 11923, Korea;
| |
Collapse
|
37
|
Geraets AFJ, Schram MT, Jansen JFA, Koster A, Dagnelie PC, van Greevenbroek MMJ, Stehouwer CDA, Verhey FRJ, Köhler S. The relation of depression with structural brain abnormalities and cognitive functioning: the Maastricht study. Psychol Med 2021; 52:1-10. [PMID: 33634767 PMCID: PMC9772903 DOI: 10.1017/s0033291721000222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/02/2020] [Accepted: 01/18/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Individuals with depression often experience widespread and persistent cognitive deficits, which might be due to brain atrophy and cerebral small vessel disease (CSVD). We therefore studied the associations between depression, markers of brain atrophy and CSVD, and cognitive functioning. METHODS We used cross-sectional data from the population-based Maastricht study (n = 4734; mean age 59.1 ± 8.6 years, 50.2% women), which focuses on type 2 diabetes. A current episode of major depressive disorder (MDD, n = 151) was assessed by the Mini-International Neuropsychiatric Interview. Volumes of cerebral spinal fluid, white matter, gray matter and white matter hyperintensities, presence of lacunar infarcts and cerebral microbleeds, and total CSVD burden were assessed by 3 T magnetic resonance imaging. Multiple linear and logistic regression analyses tested the associations between MDD, brain markers and cognitive functioning in memory, information processing speed, and executive functioning & attention, and presence of cognitive impairment. Structural equation modeling was used to test mediation. RESULTS In fully adjusted models, MDD was associated with lower scores in information processing speed [mean difference = -0.18(-0.28;-0.08)], executive functioning & attention [mean difference = -0.13(-0.25;-0.02)], and with higher odds of cognitive impairment [odds ratio (OR) = 1.60(1.06;2.40)]. MDD was associated with CSVD in participants without type 2 diabetes [OR = 1.65(1.06;2.56)], but CSVD or other markers of brain atrophy or CSVD did not mediate the association with cognitive functioning. CONCLUSIONS MDD is associated with more impaired information processing speed and executive functioning & attention, and overall cognitive impairment. Furthermore, MDD was associated with CSVD in participants without type 2 diabetes, but this association did not explain an impaired cognitive profile.
Collapse
Affiliation(s)
- Anouk F. J. Geraets
- Alzheimer Centrum Limburg, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Maastricht, the Netherlands
- Department of Internal Medicine, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht, the Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, the Netherlands
| | - Miranda T. Schram
- Department of Psychiatry and Neuropsychology, Maastricht, the Netherlands
- Department of Internal Medicine, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht, the Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, the Netherlands
- Heart and Vascular Centre, Maastricht, the Netherlands
| | - Jacobus F. A. Jansen
- School for Mental Health and Neuroscience, Maastricht, the Netherlands
- Department of Radiology, Maastricht, the Netherlands
| | - Annemarie Koster
- Department of Social Medicine, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
- Faculty of Health, Medicine & Life Sciences, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| | - Pieter C. Dagnelie
- Department of Internal Medicine, Maastricht, the Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, the Netherlands
| | - Marleen M. J. van Greevenbroek
- Department of Internal Medicine, Maastricht, the Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, the Netherlands
| | - Coen D. A. Stehouwer
- Department of Internal Medicine, Maastricht, the Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, the Netherlands
| | - Frans R. J. Verhey
- Alzheimer Centrum Limburg, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht, the Netherlands
| | - Sebastian Köhler
- Alzheimer Centrum Limburg, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht, the Netherlands
| |
Collapse
|
38
|
Associations between the quality of life in sarcopenia measured with the SarQoL® and nutritional status. Health Qual Life Outcomes 2021; 19:28. [PMID: 33482832 PMCID: PMC7821480 DOI: 10.1186/s12955-020-01619-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 11/04/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The purpose of this study was to evaluate the relationship between nutritional status and health-related quality of life after adjusting for essential factors of muscle mass, calf circumference, grip strength, and the timed up and go (TUG) test for diagnosis of sarcopenia. METHODS The subjects of this study were those who visited a health care center or a senior welfare center among the aged 65 years or older living in a community in two counties (Jinju, Sacheon), and the survey was conducted from April to August 2019. Among them, those with cardiovascular disease, cognitive disorder, or malignancy were excluded. To determine the nutritional status of the elderly subjects, a questionnaire-based screening tool called DETERMINE was used. Developed as a health-related quality of life tool for sarcopenia, the Sarcopenia-specific Quality of Life (SarQoL) questionnaire was used. For screening of sarcopenia, a rapid questionnaire based on self-reported information about falls, mobility, and strength known as the SARC-F questionnaire was used. Assessment of sarcopenia included skeletal muscle mass, calf circumference, grip strength, and the TUG test. RESULTS A total of 324 elderly people living in rural villages who were able to move to senior and welfare centers was surveyed. As a result of evaluating the association between SarQoL and nutritional risk in elderly subjects, the association was statistically significant in the moderate-risk group (B = - 5.542, p = 0.001) and in the high-risk group (B = - 8.136, p < 0.001) in comparison to the low-risk group. Significant correlations were found in all seven domains of SarQoL, except the fear domain. CONCLUSIONS This study confirms an association between quality of life dimensions surveyed by the SarQoL questionnaire and nutritional status in elderly subjects. Therefore, appropriate interventions are needed following brief evaluation of sarcopenia and nutritional deficiency among elderly people in communities.
Collapse
|
39
|
Kim YK, Han KM. Neural substrates for late-life depression: A selective review of structural neuroimaging studies. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110010. [PMID: 32544600 DOI: 10.1016/j.pnpbp.2020.110010] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 12/15/2022]
Abstract
Recent neuroimaging studies have characterized the pathophysiology of late-life depression (LLD) as a dysfunction of the brain networks involved in the regulation of emotion, motivational behavior, cognitive control, executive function, and self-referential thinking. In this article, we reviewed LLD-associated structural neuroimaging markers such as white matter hyperintensity (WMH), white matter integrity measured by diffusion tensor imaging, cortical and subcortical volumes, and cortical thickness, which may provide a structural basis for brain network dysfunction in LLD. LLD was associated with greater severity or volumes of deep, periventricular, or overall WMH and with decreased white matter integrity in the brain regions belonging to the fronto-striatal-limbic circuits and reduced white matter tract integrity which connects these circuits, such as the cingulum, corpus callosum, or uncinate fasciculus. Decreased volumes or cortical thickness in the prefrontal cortex, orbitofrontal cortex, anterior and posterior cingulate cortex, several temporal and parietal regions, hippocampus, amygdala, striatum, thalamus, and the insula were associated with LLD. These structural neuroimaging findings were also associated with cognitive dysfunction, which is a prominent clinical feature in LLD. Several structural neuroimaging markers including the WMH burden, white matter integrity, and cortical and subcortical volumes predicted antidepressant response in LLD. These structural neuroimaging findings support the hypothesis that disruption of the brain networks involved in emotion regulation and cognitive processing by impaired structural connectivity is strongly associated with the pathophysiology of LLD.
Collapse
Affiliation(s)
- Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Kyu-Man Han
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
40
|
Brenowitz WD, Al Hazzouri AZ, Vittinghoff E, Golden SH, Fitzpatrick AL, Yaffe K. Depressive Symptoms Imputed Across the Life Course Are Associated with Cognitive Impairment and Cognitive Decline. J Alzheimers Dis 2021; 83:1379-1389. [PMID: 34420969 PMCID: PMC9095065 DOI: 10.3233/jad-210588] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Depressive symptoms may increase risk for dementia, but findings are controversial because late-life depression may be a prodromal dementia symptom. Life course data on depression and dementia risk may clarify this association; however, data is limited. OBJECTIVE To impute adult depressive symptoms trajectories across adult life stages and estimate the association with cognitive impairment and decline. METHODS Using a pooled study of 4 prospective cohorts (ages 20-89), we imputed adult life course depressive symptoms trajectories based on Center for Epidemiologic Studies Depression Scale-10 (CESD-10) and calculated time-weighted averages for early adulthood (ages 20-49), mid-life (ages 50-69), and late-life (ages 70-89) for 6,122 older participants. Adjusted pooled logistic and mixed-effects models estimated associations of imputed depressive symptoms with two cognitive outcomes: cognitive impairment defined by established criteria and a composite cognitive score. RESULTS In separate models, elevated depressive symptoms in each life stage were associated with cognitive outcomes: early adulthood OR for cognitive impairment = 1.59 (95%CI: 1.35,1.87); mid-life OR = 1.94 (95%CI:1.16, 3.26); and late-life OR = 1.77 (95%CI:1.42, 2.21). When adjusted for depressive symptoms in the other life-stages, elevated depressive symptoms in early adulthood (OR = 1.73; 95%CI: 1.42,2.11) and late-life (OR = 1.43; 95%CI: 1.08,1.89) remained associated with cognitive impairment and were also associated with faster rates of cognitive decline (p < 0.05). CONCLUSION Imputing depressive symptom trajectories from pooled cohorts may help expand data across the life course. Our findings suggest early adulthood depressive symptoms may be a risk factor for cognitive impairment independent of mid- or late-life depressive symptoms.
Collapse
Affiliation(s)
- Willa D. Brenowitz
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, CA, USA
| | - Adina Zeki Al Hazzouri
- Department of Epidemiology, Mailman School of Public Health, Columbia University, NY, USA
| | - Eric Vittinghoff
- Department of Epidemiology and Biostatistics, University of California San Francisco, CA, USA
| | - Sherita H. Golden
- Department of Medicine Johns Hopkins University School of Medicine and Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Annette L. Fitzpatrick
- Departments of Family Medicine, Epidemiology, and Global Health, School of Public Health, University of Washington, WA, USA
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, CA, USA
- San Francisco VA Health Care System, San Francisco, CA, USA
| |
Collapse
|
41
|
Park MJ, Kim H, Kim EJ, Yook V, Chung IW, Lee SM, Jeon HJ. Recent Updates on Electro-Convulsive Therapy in Patients with Depression. Psychiatry Investig 2021; 18:1-10. [PMID: 33321557 PMCID: PMC7897863 DOI: 10.30773/pi.2020.0350] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Electro-convulsive therapy (ECT) has been established as a treatment modality for patients with treatment-resistant depression and with some specific subtypes of depression. This narrative review intends to provide psychiatrists with the latest findings on the use of ECT in depression, devided into total eight sub-topics. METHODS We searched PubMed for English-language articles using combined keywords and tried to analyze journals published from 1995-2020. RESULTS Pharmacotherapy such as antidepressants or maintenance ECT is more effective than a placebo as prevention of recurrence after ECT. The use of ECT in treatment-resistant depression, depressed patients with suicidal risks, elderly depression, bipolar depression, psychotic depression, and depression during pregnancy or postpartum have therapeutic benefits. As possible mechanisms of ECT, the role of neurotransmitters such as serotonin, dopamine, gamma-aminobutyric acid (GABA), and other findings in the field of neurophysiology, neuro-immunology, and neurogenesis are also supported. CONCLUSION ECT is evolving toward reducing cognitive side effects and maximizing therapeutic effects. If robust evidence for ECT through randomized controlled studies are more established and the mechanism of ECT gets further clarified, the scope of its use in the treatment of depression will be more expanded in the future.
Collapse
Affiliation(s)
- Mi Jin Park
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyewon Kim
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun Ji Kim
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Vidal Yook
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - In Won Chung
- Department of Psychiatry and Electroconvulsive Therapy Center, Dongguk University International Hospital, Goyang, Republic of Korea
| | - Sang Min Lee
- Department of Psychiatry, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hong Jin Jeon
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Health Sciences & Technology, Department of Medical Device Management & Research, and Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
42
|
Hirtz R, Libuda L, Hinney A, Föcker M, Bühlmeier J, Antel J, Holterhus PM, Kulle A, Kiewert C, Hebebrand J, Grasemann C. Lack of Evidence for a Relationship Between the Hypothalamus-Pituitary-Adrenal and the Hypothalamus-Pituitary-Thyroid Axis in Adolescent Depression. Front Endocrinol (Lausanne) 2021; 12:662243. [PMID: 34108936 PMCID: PMC8181732 DOI: 10.3389/fendo.2021.662243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/21/2021] [Indexed: 12/23/2022] Open
Abstract
In adults with major depressive disorder (MDD), a dysfunction between the hypothalamus-pituitary-adrenal (HPA) and the hypothalamus-pituitary-thyroid (HPT) axis has been shown, but the interaction of both axes has not yet been studied in adolescent major depressive disorder (MDD). Data from 273 adolescents diagnosed with MDD from two single center cross-sectional studies were used for analysis. Serum levels of thyrotropin (TSH), free levothyroxine (fT4), and cortisol were determined as indicators of basal HPT and HPA axis functioning and compared to that of adolescent controls by t-tests. Quantile regression was employed in the sample of adolescents with MDD to investigate the relationship between both axes in the normal as well as the pathological range of cortisol levels, considering confounders of both axes. In adolescent MDD, cortisol levels and TSH levels were significantly elevated in comparison to controls (p = <.001, d = 1.35, large effect size, and p = <.001, d = 0.79, moderate effect size, respectively). There was a positive linear relationship between TSH and cortisol (p = .003, d = 0.25, small effect size) at the median of cortisol levels (50th percentile). However, no relationship between TSH and cortisol was found in hypercortisolemia (cortisol levels at the 97.5th percentile). These findings imply that HPT and HPA axis dysfunction is common in adolescents with MDD and that function of both axes is only loosely related. Moreover, the regulation of the HPA and HPT axis are likely subjected to age-related maturational adjustments since findings of this study differ from those reported in adults.
Collapse
Affiliation(s)
- Raphael Hirtz
- Department of Pediatrics II, Division of Pediatric Endocrinology and Diabetology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Correspondence: Raphael Hirtz,
| | - Lars Libuda
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Exercise and Health, Institute of Nutrition, Consumption and Health Faculty of Natural Sciences, University Paderborn, Paderborn, Germany
| | - Anke Hinney
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Manuel Föcker
- Department of Child and Adolescent Psychiatry, University Hospital Münster, Münster, Germany
| | - Judith Bühlmeier
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jochen Antel
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Paul-Martin Holterhus
- Department of Paediatrics I, Paediatric Endocrinology and Diabetes, University Hospital of Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Alexandra Kulle
- Department of Paediatrics I, Paediatric Endocrinology and Diabetes, University Hospital of Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Cordula Kiewert
- Department of Pediatrics II, Division of Pediatric Endocrinology and Diabetology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Johannes Hebebrand
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Corinna Grasemann
- Department of Pediatrics, Division of Rare Diseases, St Josef-Hospital, and CeSER, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
43
|
Keeler J, Patsalos O, Thuret S, Ehrlich S, Tchanturia K, Himmerich H, Treasure J. Hippocampal volume, function, and related molecular activity in anorexia nervosa: A scoping review. Expert Rev Clin Pharmacol 2020; 13:1367-1387. [PMID: 33176113 DOI: 10.1080/17512433.2020.1850256] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Anorexia nervosa (AN) is a serious and persistent eating disorder, characterized by severe dietary restriction and weight loss, with a third of patients developing a severe-enduring form. The factors contributing to this progression are poorly understood, although there is evidence for impairments in neural structures such as the hippocampus, an area particularly affected by malnutrition and chronic stress. AREAS COVERED This study aimed to map the evidence for alterations in hippocampal volume, function, and related molecular activity in anorexia nervosa. PubMed, PsycINFO, and Web of Science were searched for studies related to hippocampal function and integrity using a range of methodologies, such as neuropsychological paradigms, structural and functional magnetic resonance imaging, and analysis of blood components. EXPERT OPINION Thirty-nine studies were included in this review. The majority were neuroimaging studies, which found hippocampus-specific volumetric and functional impairments. Neuropsychological studies showed evidence for a specific memory and learning impairments. There was some evidence for molecular abnormalities (e.g. cortisol), although these were few studies. Taken together, our review suggests that the hippocampus might be a particular region of interest when considering neurobiological approaches to understanding AN. These findings warrant further investigation and may lead to novel treatment approaches.
Collapse
Affiliation(s)
- Johanna Keeler
- Department of Psychological Medicine, King's College London, Institute of Psychiatry, Psychology & Neuroscience ,UK
| | - Olivia Patsalos
- Department of Psychological Medicine, King's College London, Institute of Psychiatry, Psychology & Neuroscience ,UK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience , UK
| | - Stefan Ehrlich
- Faculty of Medicine, Technische Universitat Dresden, Division of Psychological and Social Medicine and Developmental Neurosciences , Germany
| | - Kate Tchanturia
- Department of Psychological Medicine, King's College London, Institute of Psychiatry, Psychology & Neuroscience ,UK
| | - Hubertus Himmerich
- Department of Psychological Medicine, King's College London, Institute of Psychiatry, Psychology & Neuroscience ,UK
| | - Janet Treasure
- Department of Psychological Medicine, King's College London, Institute of Psychiatry, Psychology & Neuroscience ,UK
| |
Collapse
|
44
|
Taylor WD, Deng Y, Boyd BD, Donahue MJ, Albert K, McHugo M, Gandelman JA, Landman BA. Medial temporal lobe volumes in late-life depression: effects of age and vascular risk factors. Brain Imaging Behav 2020; 14:19-29. [PMID: 30251182 DOI: 10.1007/s11682-018-9969-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Substantial work associates late-life depression with hippocampal pathology. However, there is less information about differences in hippocampal subfields and other connected temporal lobe regions and how these regions may be influenced by vascular factors. Individuals aged 60 years or older with and without a DSM-IV diagnosis of Major Depressive Disorder completed clinical assessments and 3 T cranial MRI using a protocol allowing for automated measurement of medial temporal lobe subfield volumes. A subset also completed pseudo-continuous arterial spin labeling, allowing for the measurement of hippocampal cerebral blood flow. In 59 depressed and 21 never-depressed elders (mean age = 66.4 years, SD = 5.8y, range 60-86y), the depressed group did not exhibit statistically significant volumetric differences for the total hippocampus or hippocampal subfields but did exhibit significantly smaller volumes of the perirhinal cortex, specifically in the BA36 region. Additionally, age had a greater effect in the depressed group on volumes of the cornu ammonis, entorhinal cortex, and BA36 region. Finally, both clinical and radiological markers of vascular risk were associated with smaller BA36 volumes, while reduced hippocampal blood flow was associated with smaller hippocampal and cornu ammonis volumes. In conclusion, while we did not observe group differences in hippocampal regions, we observed group differences and an effect of vascular pathology on the BA36 region, part of the perirhinal cortex. This is a critical region exhibiting atrophy in prodromal Alzheimer's disease. Moreover, the observed greater effect of age in the depressed groups is concordant with past longitudinal studies reporting greater hippocampal atrophy in late-life depression.
Collapse
Affiliation(s)
- Warren D Taylor
- The Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Avenue South, Nashville, TN, 37212, USA. .,Geriatric Research, Education and Clinical Center, Department of Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN, 37212, USA.
| | - Yi Deng
- The Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Avenue South, Nashville, TN, 37212, USA
| | - Brian D Boyd
- The Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Avenue South, Nashville, TN, 37212, USA
| | - Manus J Donahue
- The Department of Radiology and Radiological Science, Vanderbilt University Medical Center, Nashville, TN, 37212, USA
| | - Kimberly Albert
- The Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Avenue South, Nashville, TN, 37212, USA
| | - Maureen McHugo
- The Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Avenue South, Nashville, TN, 37212, USA
| | | | - Bennett A Landman
- The Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Avenue South, Nashville, TN, 37212, USA.,The Department of Radiology and Radiological Science, Vanderbilt University Medical Center, Nashville, TN, 37212, USA.,The Department of Electrical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| |
Collapse
|
45
|
Performances on individual neurocognitive tests by people experiencing a current major depression episode: A systematic review and meta-analysis. J Affect Disord 2020; 276:249-259. [PMID: 32697706 DOI: 10.1016/j.jad.2020.07.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 06/12/2020] [Accepted: 07/11/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND PURPOSES Previous meta-analyses on neurocognitive test performances in depression have provided effect sizes for cognitive domains. Most domain effect sizes have medium to high variance heterogeneity. Restriction to each test performance could reduce variance and clarify differing test effect sizes. This systematic review and meta-analysis were done to 1. provide effect sizes for cognitive performances (test, subtest, or multiple measures within tests), and 2. investigate age as an effect modifier. METHODS Inclusion criteria were: 1. active major depression episode (MD), 2. a control group, 3 reported means and standard deviations, 4. non-computerized tests previously studied at least 3 times. Meta-analyses were performed using Cochrane Review Manager. Age under versus over 45 was investigated as an effect moderator. RESULTS Twenty-seven studies met criteria. MD patients performed significantly poorer on 16 of 16 neurocognitive measures (random effects d = -0.47 to -0.92 across tests). Variance was heterogeneous for 11 of 16 measures. Differences between cognitive measures were largely absent based on overlapping 95% confidence intervals. Effect sizes did not differ under versus over 45 years. LIMITATIONS Bias risk assessment showed limited control for subject selection, comparability of depressed and control groups, pre-morbid intelligence, drug treatment, and effort in testing. CONCLUSIONS The depression - cognition effect was in the moderate to large range regardless of test type. Variance heterogeneity was substantial despite exclusion of inactive depression and the absence of test pooling. The size of the depression - cognition effect was not a function of age.
Collapse
|
46
|
Shaw TB, York A, Barth M, Bollmann S. Towards Optimising MRI Characterisation of Tissue (TOMCAT) Dataset including all Longitudinal Automatic Segmentation of Hippocampal Subfields (LASHiS) data. Data Brief 2020; 32:106043. [PMID: 32793772 PMCID: PMC7415822 DOI: 10.1016/j.dib.2020.106043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/30/2020] [Accepted: 07/14/2020] [Indexed: 12/04/2022] Open
Abstract
Seven healthy participants were scanned using a Siemens Magnetom 7 Tesla (T) whole-body research MRI scanner (Siemens Healthcare, Erlangen, Germany). The first scan session was acquired in 2016 (time point one), the second and third session in 2019 (time point two and three, respectively) with the third session acquired 45 min following the second as a scan-rescan condition. The following scans were acquired for all time points: structural T1 weighted (T1w) MP2RAGE, high in-plane resolution Turbo-Spin Echo (TSE) dedicated for hippocampus subfield segmentation. The data were used in three projects to date, for more insight see: 1) Non-linear realignment for Turbo-Spin Echo retrospective motion correction and hippocampus segmentation improvement [1] 2) Longitudinal Automatic Segmentation of Hippocampal Subfields (LASHiS) using multi-contrast MRI [2]. 3) The challenge of bias-free coil combination for quantitative susceptibility mapping at ultra-high field [3]. Data were converted from DICOM to nifti format following the Brain Imaging Data Structure (BIDS) [4]. Data were analysed for the accompanying manuscript "Longitudinal Automatic Segmentation of Hippocampal Subfields (LASHiS) using multi-contrast MRI" including test-retest reliability and longitudinal Bayesian Linear Mixed Effects (LME) modelling.
Collapse
Affiliation(s)
- Thomas B Shaw
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Australia
| | - Ashley York
- School of Psychology, The University of Queensland, Brisbane, Australia
| | - Markus Barth
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Australia
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Australia
- ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, QLD, Australia
| | - Steffen Bollmann
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Australia
- ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
47
|
Rothe N, Steffen J, Penz M, Kirschbaum C, Walther A. Examination of peripheral basal and reactive cortisol levels in major depressive disorder and the burnout syndrome: A systematic review. Neurosci Biobehav Rev 2020; 114:232-270. [DOI: 10.1016/j.neubiorev.2020.02.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/27/2020] [Accepted: 02/19/2020] [Indexed: 12/15/2022]
|
48
|
Terock J, Van der Auwera S, Janowitz D, Wittfeld K, Teumer A, Grabe HJ. Functional polymorphisms of the mineralocorticoid receptor gene NR3C2 are associated with diminished memory decline: Results from a longitudinal general-population study. Mol Genet Genomic Med 2020; 8:e1345. [PMID: 32558353 PMCID: PMC7507013 DOI: 10.1002/mgg3.1345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The mineralocorticoid receptor (MR) in the brain has a key role in the regulation of the central stress response and is associated with memory performance. We investigated whether the genetic polymorphisms rs5522 and rs2070951 of NR3C2 showed main and interactive effects with childhood trauma on memory decline. METHODS Declarative memory was longitudinally assessed in 1,318 participants from the community-dwelling Study of Health in Pomerania using the Verbal Learning and Memory Test (VLMT). In a subsample of 377 participants aged 60 and older, the Mini-Mental Status Examination (MMSE) was additionally applied. Mean follow-up time for the VLMT and MMSE were 6.4 and 10.7 years, respectively. RESULTS Homozygous carriers of the G allele of rs2070951 (p < .01) and of the A allele of rs5522 (p < .001) showed higher immediate recall of words as compared to carriers of C allele (rs2070951) or the G allele (rs5522). The CG haplotype was associated with decreased recall (p < .001). Likewise, in the subsample of older patients, the AA genotype of rs5522 was associated with higher MMSE scores (p < .05). CG haplotypes showed significantly reduced MMSE scores in comparison to the reference haplotype (β = -0.60; p < .01). CONCLUSIONS Our results indicate that the GG genotype of rs2070951 as well as the AA genotype of rs5522 are associated with diminished memory decline.
Collapse
Affiliation(s)
- Jan Terock
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.,Department of Psychiatry and Psychotherapy, Helios Hanseklinikum Stralsund, Stralsund, Germany
| | - Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Deborah Janowitz
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.,German Center for Neurodegenerative Diseases (DZNE), Greifswald, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
49
|
Han X, Wu Y, Zhong Y, Becker S. Recovery of High Interference Memory in Spite of Lingering Cognitive Deficits in a Longitudinal Pilot Study of Hospitalized Depressed Patients. Front Psychiatry 2020; 11:736. [PMID: 32848916 PMCID: PMC7396669 DOI: 10.3389/fpsyt.2020.00736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/14/2020] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Major depressive disorder has deleterious impacts on mood, cognition, and many functions of daily life. Even after remission of mood symptoms, patients frequently report persistent cognitive deficits. By contrast, the neurogenic theory of depression posits that recovery from depression is dependent upon a restoration of neurogenesis. The present study was designed to test this prediction by assessing performance in MDD in-patients on a broad battery of cognitive tasks including the Mnemonic Similarity Task, a high interference memory test that is a putative correlate of neurogenesis. We predicted that remitted patients should exhibit recovery of function on this task, even though they may show residual deficits on other cognitive tasks. METHODS 18 hospitalized patients diagnosed with MDD and 22 healthy control participants matched for age, sex, and education completed a battery of mood and cognitive tests at two time points. Patients completed their baseline assessments when first admitted to hospital and repeated the same assessments upon remission, typically 4-5 weeks later and just prior to their release from hospital. Control participants were tested at baseline and 4-5 weeks later on the same assessment battery, which included the BDI-II, BAI, Cohen's PSS, Mnemonic Similarity Task, and several sub-tests adapted from the CANTAB. RESULTS At baseline, MDD patients were impaired relative to controls on the MST and many other cognitive tasks. Upon remission, patients' MST scores did not differ from those of healthy controls, although patients were still impaired on Pattern Recognition Memory, Spatial Recognition Memory, Delayed Matching to Sample and Paired Associates Learning relative to healthy control participants. CONCLUSION The lingering memory deficits observed in remitted patients with MDD observed here are broadly consistent with findings in the literature. Importantly, however, remitted patients showed recovery of cognitive function on the Mnemonic Similarity Task. This is the first study that we are aware of to report recovery of function on a high interference, putatively neurogenesis-dependent memory test in a longitudinal sample of hospitalized MDD patients from admission to remission. Our findings are consistent with the neurogenic theory of depression, which posits that a restoration of neurogenesis is linked to recovery from depression.
Collapse
Affiliation(s)
- Xue Han
- School of Psychology, Northeast Normal University, Changchun, China
| | - Yingga Wu
- Tongliao Mongolian High School, Tongliao, China
| | - Yanfeng Zhong
- Changchun Mental Hospital (Changchun Sixth Hospital), Changchun, China
| | - Suzanna Becker
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
50
|
Umschweif G, Greengard P, Sagi Y. The dentate gyrus in depression. Eur J Neurosci 2019; 53:39-64. [DOI: 10.1111/ejn.14640] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 11/05/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Gali Umschweif
- Laboratory for Molecular and Cellular Neuroscience Rockefeller University New York NY USA
| | - Paul Greengard
- Laboratory for Molecular and Cellular Neuroscience Rockefeller University New York NY USA
| | - Yotam Sagi
- Laboratory for Molecular and Cellular Neuroscience Rockefeller University New York NY USA
| |
Collapse
|