1
|
Kobro-Flatmoen A, Omholt SW. Intraneuronal binding of amyloid beta with reelin-Implications for the onset of Alzheimer's disease. PLoS Comput Biol 2025; 21:e1012709. [PMID: 39775030 PMCID: PMC11741591 DOI: 10.1371/journal.pcbi.1012709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 01/17/2025] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Numerous studies of the human brain supported by experimental results from rodent and cell models point to a central role for intracellular amyloid beta (Aβ) in the onset of Alzheimer's disease (AD). In a rat model used to study AD, it was recently shown that in layer II neurons of the anteriolateral entorhinal cortex expressing high levels of the glycoprotein reelin (Re+alECLII neurons), reelin and Aβ engage in a direct protein-protein interaction. If reelin functions as a sink for intracellular Aβ and if the binding to reelin makes Aβ physiologically inert, it implies that reelin can prevent the neuron from being exposed to the harmful effects typically associated with increased levels of oligomeric Aβ. Considering that reelin expression is extraordinarily high in Re+alECLII neurons compared to most other cortical neurons, such a protective role appears to be very difficult to reconcile with the fact that this subset of ECLII neurons is clearly a major cradle for the onset of AD. Here, we show that this conundrum can be resolved if Re+alECLII neurons have a higher maximum production capacity of Aβ than neurons expressing low levels of reelin, and we provide a rationale for why this difference has evolved.
Collapse
Affiliation(s)
- Asgeir Kobro-Flatmoen
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- K. G. Jebsen Centre for Alzheimer’s Disease, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Stig W. Omholt
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
2
|
Hu Y, Zou Y, Zhang M, Yan J, Zheng Y, Chen Y. The relationship between major depressive disorder and dementia: A bidirectional two-sample Mendelian randomization study. J Affect Disord 2024; 355:167-174. [PMID: 38548196 DOI: 10.1016/j.jad.2024.03.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) and dementia psychiatric and neurological diseases that are clinically widespread, but whether there is a causal link between them is still unclear. In this study, bidirectional two-sample Mendelian randomization (MR) was used to investigate the potential causal relationship between MDD and dementia via a genome-wide association study (GWAS) database, containing samples from the European population. METHOD We collected data on MDD and common clinical dementia subtypes from GWAS, including Alzheimer's disease (AD), frontotemporal dementia (FTD), dementia with Lewy bodies (DLB), Parkinson's disease with dementia (PDD), and vascular dementia (VaD). A series of bidirectional two-sample MR studies and correlation sensitivity analysis were carried out. RESULTS In the study of the effect of MDD on dementia subtypes, no causal relationship was found between MDD and dementia subtypes other than VaD, inverse variance weighted (IVW) method: odds ratio (OR), 2.131; 95 % confidence interval (CI), 1.249-3.639, P = 0.006; MDD-AD: OR, 1.000; 95 % CI, 0.999-1.001, P = 0.537; MDD-FTD: OR, 1.476; 95 % CI, 0.471-4.627, P = 0.505; MDD-PDD: OR, 0.592; 95 % CI, 0.204-1.718, P = 0.335; MR-Egger method: MDD-DLB: OR, 0.582; 95 % CI, 0.021-15.962, P = 0.751. In reverse MR analyses, no dementia subtype was found to be a risk factor for MDD. LIMITATIONS The results of this study may not be generalizable to non-European populations. CONCLUSION MDD was identified as a potential risk factor for VaD, but no dementia subtype was found to be a risk factor for MDD. These results suggest a new avenue for the prevention of VaD.
Collapse
Affiliation(s)
- Yijun Hu
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Yuntao Zou
- Weifang Hospital of Traditional Chinese Medicine, Wei Fang, China
| | - Meng Zhang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Jinglan Yan
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Yuanjia Zheng
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Yongjun Chen
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, PR China.
| |
Collapse
|
3
|
Long J, Qin K, Wu Y, Li L, Zhou J. Gray matter abnormalities and associated familial risk endophenotype in individuals with first-episode bipolar disorder: Evidence from whole-brain voxel-wise meta-analysis. Asian J Psychiatr 2022; 74:103179. [PMID: 35691059 DOI: 10.1016/j.ajp.2022.103179] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/16/2022] [Accepted: 05/30/2022] [Indexed: 11/29/2022]
Abstract
Gray matter abnormalities have been widely reported in individuals with and at familial risk for bipolar disorder (BD). However, inconsistent findings were reported, and whether shared abnormalities exist between at-risk individuals and patients which can represent an endophenotype remained unclear. This meta-analysis aimed at identifying robust patterns of gray matter changes in patients with first-episode BD (FEBD) and associated risk endophenotype of BD. A systematic literature search was performed to identify eligible voxel-based morphometry studies comparing FEBD patients and healthy controls. Findings of included studies were integrated using the Seed-based d Mapping toolbox. Common and distinct patterns of gray matter abnormalities between FEBD patients and unaffected at-risk individuals were explored. A total of 16 VBM studies comparing 411 FEBD patients and 521 controls were included. FEBD patients showed increased gray matter volume in the cerebellum, posterior cingulate cortex and striatum, and decreased gray matter volume in the medial superior frontal gyrus and gyrus rectus. No common abnormalities were identified between FEBD patients and unaffected at-risk individuals. More gray matter loss in the medial superior frontal gyrus and insula were found in FEBD patients relative to unaffected at-risk individuals. These findings revealed robust gray matter abnormalities in the cortico-striato-cerebellar and default mode network regions in FEBD, and implicated that gray matter deficits may not represent a familial risk endophenotype of BD.
Collapse
Affiliation(s)
- Jingyi Long
- Department of Radiology, Wuhan Mental Health Center, Wuhan 430012, Hubei, China; Department of Radiology, Wuhan Hospital for Psychotherapy, Wuhan 430012, Hubei, China
| | - Kun Qin
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati 45219, OH, USA
| | - Yong Wu
- Department of Depression, Wuhan Hospital for Psychotherapy, Wuhan 430012, Hubei, China
| | - Lu Li
- Department of Interventional Radiology, Wuhan Jinyintan Hospital, Wuhan 430023, Hubei, China.
| | - Juan Zhou
- Department of Ultrasonography, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| |
Collapse
|
4
|
Zhu Z, Zhao Y, Wen K, Li Q, Pan N, Fu S, Li F, Radua J, Vieta E, Kemp GJ, Biswa BB, Gong Q. Cortical thickness abnormalities in patients with bipolar disorder: A systematic review and meta-analysis. J Affect Disord 2022; 300:209-218. [PMID: 34971699 DOI: 10.1016/j.jad.2021.12.080] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 10/10/2021] [Accepted: 12/19/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND An increasing number of neuroimaging studies report alterations of cortical thickness (CT) related to the neuropathology of bipolar disorder (BD). We provide here a whole-brain vertex-wise meta-analysis, which may help improve the spatial precision of these identifications. METHODS A comprehensive meta-analysis was performed to investigate the differences in CT between patients with BD and healthy controls (HCs) by using a newly developed mask for CT analysis in seed-based d mapping (SDM) meta-analytic software. We used meta-regression to explore the effects of demographics and clinical characteristics on CT. This meta-review was conducted in accordance with PRISMA guideline. RESULTS We identified 21 studies meeting criteria for the systematic review, of which 11 were eligible for meta-analysis. The meta-analysis comprising 649 BD patients and 818 HCs showed significant cortical thinning in the left insula extending to left Rolandic operculum and Heschl gyrus, the orbital part of left inferior frontal gyrus (IFG), the medial part of left superior frontal gyrus (SFG) as well as bilateral anterior cingulate cortex (ACC) in BD. In meta-regression analyses, mean patient age was negatively correlated with reduced CT in the left insula. LIMITATIONS All enrolled studies were cross-sectional; we could not explore the potential effects of medication and mood states due to the limited data. CONCLUSIONS Our results suggest that BD patients have significantly thinner frontoinsular cortex than HCs, and the results may be helpful in revealing specific neuroimaging biomarkers of BD patients.
Collapse
Affiliation(s)
- Ziyu Zhu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Youjin Zhao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
| | - Keren Wen
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qian Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Nanfang Pan
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Shiqin Fu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Fei Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
| | - Joaquim Radua
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, Chengdu, Sichuan, China; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Barcelona, Spain; Department of Clinical Neuroscience, Centre for Psychiatric Research and Education, Karolinska Institutet, Stockholm, Sweden; Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, Northern Ireland United Kingdom
| | - Eduard Vieta
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Barcelona, Spain; Barcelona Bipolar Disorders and Depressive Unit, Hospital Clinic, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Graham J Kemp
- Liverpool Magnetic Resonance Imaging Centre (LiMRIC) and Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Bharat B Biswa
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA; The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China.
| |
Collapse
|
5
|
Glial Cell Abnormalities in Major Psychiatric Diseases: A Systematic Review of Postmortem Brain Studies. Mol Neurobiol 2022; 59:1665-1692. [PMID: 35013935 DOI: 10.1007/s12035-021-02672-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/25/2021] [Indexed: 10/19/2022]
Abstract
There have been a large number of reports about glial cell dysfunction being related to major psychiatric diseases such as schizophrenia (SCZ), bipolar disorder (BD), and major depressive disorder (MDD). In this review, we provide an overview of postmortem studies analyzing the structural changes of glial cells in these three major psychiatric diseases, including the density, number and size of glial cells, and the expression of related markers. Up to May 1, 2021, 108 articles that met the inclusion criteria were identified by searching PubMed and Web of Science. Although most studies evaluating total glial cells did not show abnormalities in the brains of postmortem patients, astrocytes, microglial cells, and oligodendrocytes seem to have specific patterns of changes in each disease. For example, out of 20 studies that evaluated astrocyte markers in MDD, 11 studies found decreased astrocyte marker expression in MDD patients. Similarly, out of 25 studies evaluating oligodendrocyte markers in SCZ, 15 studies showed decreased expression of oligodendrocyte markers in different brain regions of SCZ patients. In addition, activated microglial cells were observed in patients with SCZ, BD, and MDD, but suicide may be a confounding factor for the observed effects. Although the data from the included studies were heterogeneous and this cannot be fully explained at present, it is likely that there are a variety of contributing factors, including the measured brain regions, methods of measurement, gender, age at time of death, and medications.
Collapse
|
6
|
Dudek KA, Dion‐Albert L, Kaufmann FN, Tuck E, Lebel M, Menard C. Neurobiology of resilience in depression: immune and vascular insights from human and animal studies. Eur J Neurosci 2021; 53:183-221. [PMID: 31421056 PMCID: PMC7891571 DOI: 10.1111/ejn.14547] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
Major depressive disorder (MDD) is a chronic and recurrent psychiatric condition characterized by depressed mood, social isolation and anhedonia. It will affect 20% of individuals with considerable economic impacts. Unfortunately, 30-50% of depressed individuals are resistant to current antidepressant treatments. MDD is twice as prevalent in women and associated symptoms are different. Depression's main environmental risk factor is chronic stress, and women report higher levels of stress in daily life. However, not every stressed individual becomes depressed, highlighting the need to identify biological determinants of stress vulnerability but also resilience. Based on a reverse translational approach, rodent models of depression were developed to study the mechanisms underlying susceptibility vs resilience. Indeed, a subpopulation of animals can display coping mechanisms and a set of biological alterations leading to stress resilience. The aetiology of MDD is multifactorial and involves several physiological systems. Exacerbation of endocrine and immune responses from both innate and adaptive systems are observed in depressed individuals and mice exhibiting depression-like behaviours. Increasing attention has been given to neurovascular health since higher prevalence of cardiovascular diseases is found in MDD patients and inflammatory conditions are associated with depression, treatment resistance and relapse. Here, we provide an overview of endocrine, immune and vascular changes associated with stress vulnerability vs. resilience in rodents and when available, in humans. Lack of treatment efficacy suggests that neuron-centric treatments do not address important causal biological factors and better understanding of stress-induced adaptations, including sex differences, could contribute to develop novel therapeutic strategies including personalized medicine approaches.
Collapse
Affiliation(s)
- Katarzyna A. Dudek
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Laurence Dion‐Albert
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Fernanda Neutzling Kaufmann
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Ellen Tuck
- Smurfit Institute of GeneticsTrinity CollegeDublinIreland
| | - Manon Lebel
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Caroline Menard
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| |
Collapse
|
7
|
Harrison PJ, Colbourne L, Harrison CH. The neuropathology of bipolar disorder: systematic review and meta-analysis. Mol Psychiatry 2020; 25:1787-1808. [PMID: 30127470 PMCID: PMC6292507 DOI: 10.1038/s41380-018-0213-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/16/2018] [Accepted: 07/24/2018] [Indexed: 01/10/2023]
Abstract
Various neuropathological findings have been reported in bipolar disorder (BD). However, it is unclear which findings are well established. To address this gap, we carried out a systematic review of the literature. We searched over 5000 publications, identifying 103 data papers, of which 81 were eligible for inclusion. Our main findings can be summarised as follows. First, most studies have relied on a limited number of brain collections, and have used relatively small sample sizes (averaging 12 BD cases and 15 controls). Second, surprisingly few studies have attempted to replicate closely a previous one, precluding substantial meta-analyses, such that the latter were all limited to two studies each, and comprising 16-36 BD cases and 16-74 controls. As such, no neuropathological findings can be considered to have been established beyond reasonable doubt. Nevertheless, there are several replicated positive findings in BD, including decreased cortical thickness and glial density in subgenual anterior cingulate cortex, reduced neuronal density in some amygdalar nuclei, and decreased calbindin-positive neuron density in prefrontal cortex. Many other positive findings have also been reported, but with limited or contradictory evidence. As an important negative result, it can be concluded that gliosis is not a feature of BD; neither is there neuropathological evidence for an inflammatory process.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK.
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK.
| | - Lucy Colbourne
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Charlotte H Harrison
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
8
|
Han S, Cui Q, Wang X, Chen Y, Li D, Li L, Guo X, Fan YS, Guo J, Sheng W, Lu F, He Z, Chen H. The anhedonia is differently modulated by structural covariance network of NAc in bipolar disorder and major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2020; 99:109865. [PMID: 31962188 DOI: 10.1016/j.pnpbp.2020.109865] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/11/2020] [Accepted: 01/15/2020] [Indexed: 12/23/2022]
Abstract
During depressive episode, bipolar disorder (BD) patients share indistinguishable depression symptoms with major depressive disorder (MDD).However, whether neural correlates underlying the anhedonia, a core feature of depression, is different between BD and MDD remains unknown. To explore neural correlates underlying the anhedonia in BD and MDD, structural T1-weighted images from 36 depressed BD patients, 40 depressed MDD patients matched for depression severity and 34 health controls (HCs) were scanned. Considering the vital role of nucleus accumbens (NAc) in the anhedonia, we constructed the structural covariance network of NAc for each subject. Then, we explored altered structural covariance network of NAc and its interaction with the anhedonia severity in BD and MDD patients. As a result, BD and MDD patients shared decreased structural covariance of NAc connected to prefrontal gyrus, bilateral striatum extending to bilateral anterior insula. Apart from these regions, BD patients presented specifically increased structural covariance of NAc connected to left hippocampus extending to thalamus. The interaction between structural covariance network of NAc and the anhedonia severity in MDD was mainly associated anterior insula (AIC), amygdala, anterior cingulate cortex (ACC)and caudate while that in BD was mainly located in striatum and prefrontal cortex. Our results found that BD and MDD patients presented commonly and distinctly altered structural covariance network of NAc. What is more, the neural correlates underlying the anhedonia in BD and MDD might be different.
Collapse
Affiliation(s)
- Shaoqiang Han
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Qian Cui
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu 610054, PR China; School of Public Affairs and Administration, University of Electronic Science and Technology of China, Chengdu, PR China.
| | - Xiao Wang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Yuyan Chen
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Di Li
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Liang Li
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Xiaonan Guo
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Yun-Shuang Fan
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Jing Guo
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Wei Sheng
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Fengmei Lu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Zongling He
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China.
| | - Huafu Chen
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China; MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu 610054, PR China.
| |
Collapse
|
9
|
Postmortem evidence of brain inflammatory markers in bipolar disorder: a systematic review. Mol Psychiatry 2020; 25:94-113. [PMID: 31249382 DOI: 10.1038/s41380-019-0448-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 03/08/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Abstract
Bipolar disorder (BD) is a chronic affective disorder with extreme mood swings that include mania or hypomania and depression. Though the exact mechanism of BD is unknown, neuroinflammation is one of the numerous investigated etiopathophysiological causes of BD. This article presents a systematic review of the data regarding brain inflammation evaluating microglia, astrocytes, cytokines, chemokines, adhesion molecules, and other inflammatory markers in postmortem BD brain samples. This systematic review was performed according to PRISMA recommendations, and relevant studies were identified by searching the PubMed/MEDLINE, PsycINFO, EMBASE, LILACS, IBECS, and Web of Science databases for peer-reviewed journal articles published by March 2019. Quality of included studies appraised using the QUADAS-2 tool. Among the 1814 articles included in the primary screening, 51 articles measured inflammatory markers in postmortem BD brain samples. A number of studies have shown evidence of inflammation in BD postmortem brain samples. However, an absolute statement cannot be concluded whether neuroinflammation is present in BD due to the large number of studies did not evaluate the presence of infiltrating peripheral immune cells in the central nervous system (CNS) parenchyma, cytokines levels, and microglia activation in the same postmortem brain sample. For example, out of 15 studies that evaluated microglia cells markers, 8 studies found no effect of BD on these cells. Similarly, 17 out of 51 studies evaluating astrocytes markers, 9 studies did not find any effect of BD on astrocyte cells, whereas 8 studies found a decrease and 2 studies presented both increase and decrease in different brain regions. In addition, multiple factors account for the variability across the studies, including postmortem interval, brain area studied, age at diagnosis, undergoing treatment, and others. Future analyses should rectify these potential sources of heterogeneity and reach a consensus regarding the inflammatory markers in postmortem BD brain samples.
Collapse
|
10
|
Goodwill HL, Manzano-Nieves G, LaChance P, Teramoto S, Lin S, Lopez C, Stevenson RJ, Theyel BB, Moore CI, Connors BW, Bath KG. Early Life Stress Drives Sex-Selective Impairment in Reversal Learning by Affecting Parvalbumin Interneurons in Orbitofrontal Cortex of Mice. Cell Rep 2019; 25:2299-2307.e4. [PMID: 30485800 PMCID: PMC6310486 DOI: 10.1016/j.celrep.2018.11.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/18/2018] [Accepted: 10/31/2018] [Indexed: 01/25/2023] Open
Abstract
Poverty, displacement, and parental stress represent potent sources of early life stress (ELS). Stress disproportionately affects females, who are at increased risk for stress-related pathologies associated with cognitive impairment. Mechanisms underlying stress-associated cognitive impairment and enhanced risk of females remain unknown. Here, ELS is associated with impaired rule-reversal (RR) learning in females, but not males. Impaired performance was associated with decreased expression and density of interneurons expressing parvalbumin (PV+) in orbitofrontal cortex (OFC), but not other inter-neuron subtypes. Optogenetic silencing of PV+ inter-neuron activity in OFC of control mice phenocopied RR learning deficits observed in ELS females. Localization of reversal learning deficits to PV+ interneurons in OFC was confirmed by optogenetic studies in which neurons in medial prefrontal cortex (mPFC) were silenced and associated with select deficits in rule-shift learning. Sex-, cell-, and region-specific effects show altered PV+ interneuron development can be a driver of sex differences in cognitive dysfunction. Goodwill et al. investigate the effect of early life stress (ELS) on cognitive development in a mouse model. Using a combination of genetic, histological, optogenetic, and behavioral techniques, they find that ELS leads to female-selective impairments in the ability to engage in rule reversal, but not other forms of attentional learning. Impairments are associated with diminished parvalbumin (PV) expression and a decreased density of PV+ interneurons in the orbitofrontal cortex (OFC).
Collapse
Affiliation(s)
- Haley L Goodwill
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | | | - Patrick LaChance
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Sana Teramoto
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Shirley Lin
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Chelsea Lopez
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Rachel J Stevenson
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912, USA
| | - Brian B Theyel
- Department of Psychiatry and Human Behavior, Brown University, Providence, RI 02912, USA
| | | | - Barry W Connors
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Kevin G Bath
- Department of Cognitive, Linguistic and Psychological Sciences, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
11
|
Gigase FAJ, Snijders GJLJ, Boks MP, de Witte LD. Neurons and glial cells in bipolar disorder: A systematic review of postmortem brain studies of cell number and size. Neurosci Biobehav Rev 2019; 103:150-162. [PMID: 31163205 DOI: 10.1016/j.neubiorev.2019.05.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023]
Abstract
Bipolar disorder (BD) is a complex neurobiological disease. It is likely that both neurons and glial cells are affected in BD, yet how these cell types are changed at the structural and functional level is still largely unknown. In this review we provide an overview of postmortem studies analyzing structural cellular changes in BD, including the density, number and size of neurons and glia. We categorize the results per cell-type and validate outcome measures per brain region. Despite variations by brain region, outcome measure and methodology, several patterns could be identified. Total neuron, total glia, and cell subtypes astrocyte, microglia and oligodendrocyte presence appears unchanged in the BD brain. Interneuron density may be decreased across various cortical areas, yet findings of interneuron subpopulations show discrepancies. This structural review brings to light issues in validation and replication. Future research should therefore prioritize the validation of existing studies in order to increasingly refine the conceptual models of BD.
Collapse
Affiliation(s)
- Frederieke A J Gigase
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG Utrecht, the Netherlands
| | - Gijsje J L J Snijders
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG Utrecht, the Netherlands
| | - Marco P Boks
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG Utrecht, the Netherlands
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, USA; Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
12
|
Kim R, Healey KL, Sepulveda-Orengo MT, Reissner KJ. Astroglial correlates of neuropsychiatric disease: From astrocytopathy to astrogliosis. Prog Neuropsychopharmacol Biol Psychiatry 2018; 87:126-146. [PMID: 28989099 PMCID: PMC5889368 DOI: 10.1016/j.pnpbp.2017.10.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/24/2017] [Accepted: 10/04/2017] [Indexed: 01/22/2023]
Abstract
Complex roles for astrocytes in health and disease continue to emerge, highlighting this class of cells as integral to function and dysfunction of the nervous system. In particular, escalating evidence strongly implicates a range of changes in astrocyte structure and function associated with neuropsychiatric diseases including major depressive disorder, schizophrenia, and addiction. These changes can range from astrocytopathy, degeneration, and loss of function, to astrogliosis and hypertrophy, and can be either adaptive or maladaptive. Evidence from the literature indicates a myriad of changes observed in astrocytes from both human postmortem studies as well as preclinical animal models, including changes in expression of glial fibrillary protein, as well as changes in astrocyte morphology and astrocyte-mediated regulation of synaptic function. In this review, we seek to provide a comprehensive assessment of these findings and consequently evidence for common themes regarding adaptations in astrocytes associated with neuropsychiatric disease. While results are mixed across conditions and models, general findings indicate decreased astrocyte cellular features and gene expression in depression, chronic stress and anxiety, but increased inflammation in schizophrenia. Changes also vary widely in response to different drugs of abuse, with evidence reflective of features of astrocytopathy to astrogliosis, varying across drug classes, route of administration and length of withdrawal.
Collapse
Affiliation(s)
- Ronald Kim
- Department of Psychology and Neuroscience, CB 3270, UNC Chapel Hill, Chapel Hill, NC 27599, United States
| | - Kati L Healey
- Department of Psychology and Neuroscience, CB 3270, UNC Chapel Hill, Chapel Hill, NC 27599, United States
| | - Marian T Sepulveda-Orengo
- Department of Psychology and Neuroscience, CB 3270, UNC Chapel Hill, Chapel Hill, NC 27599, United States
| | - Kathryn J Reissner
- Department of Psychology and Neuroscience, CB 3270, UNC Chapel Hill, Chapel Hill, NC 27599, United States..
| |
Collapse
|
13
|
Layer-specific reduced neuronal density in the orbitofrontal cortex of older adults with obsessive–compulsive disorder. Brain Struct Funct 2018; 224:191-203. [DOI: 10.1007/s00429-018-1752-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 09/09/2018] [Indexed: 12/22/2022]
|
14
|
Rajkumar R, Dawe GS. OBscure but not OBsolete: Perturbations of the frontal cortex in common between rodent olfactory bulbectomy model and major depression. J Chem Neuroanat 2018; 91:63-100. [DOI: 10.1016/j.jchemneu.2018.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/02/2018] [Accepted: 04/04/2018] [Indexed: 02/08/2023]
|
15
|
Abstract
Traditional pharmacological treatments for depression have a delayed therapeutic onset, ranging from several weeks to months, and there is a high percentage of individuals who never respond to treatment. In contrast, ketamine produces rapid-onset antidepressant, anti-suicidal, and anti-anhedonic actions following a single administration to patients with depression. Proposed mechanisms of the antidepressant action of ketamine include N-methyl-D-aspartate receptor (NMDAR) modulation, gamma aminobutyric acid (GABA)-ergic interneuron disinhibition, and direct actions of its hydroxynorketamine (HNK) metabolites. Downstream actions include activation of the mechanistic target of rapamycin (mTOR), deactivation of glycogen synthase kinase-3 and eukaryotic elongation factor 2 (eEF2), enhanced brain-derived neurotrophic factor (BDNF) signaling, and activation of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors (AMPARs). These putative mechanisms of ketamine action are not mutually exclusive and may complement each other to induce potentiation of excitatory synapses in affective-regulating brain circuits, which results in amelioration of depression symptoms. We review these proposed mechanisms of ketamine action in the context of how such mechanisms are informing the development of novel putative rapid-acting antidepressant drugs. Such drugs that have undergone pre-clinical, and in some cases clinical, testing include the muscarinic acetylcholine receptor antagonist scopolamine, GluN2B-NMDAR antagonists (i.e., CP-101,606, MK-0657), (2R,6R)-HNK, NMDAR glycine site modulators (i.e., 4-chlorokynurenine, pro-drug of the glycineB NMDAR antagonist 7-chlorokynurenic acid), NMDAR agonists [i.e., GLYX-13 (rapastinel)], metabotropic glutamate receptor 2/3 (mGluR2/3) antagonists, GABAA receptor modulators, and drugs acting on various serotonin receptor subtypes. These ongoing studies suggest that the future acute treatment of depression will typically occur within hours, rather than months, of treatment initiation.
Collapse
Affiliation(s)
- Panos Zanos
- Department of Psychiatry, University of Maryland School of Medicine, Rm. 934F MSTF, 685 W. Baltimore St., Baltimore, MD, 21201, USA.
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, St. BRB 5-007, 655 W. Baltimore St., Baltimore, MD, 21201, USA, Baltimore, MD, 21201, USA
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D Gould
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, University of Maryland School of Medicine, Rm. 936 MSTF, 685 W. Baltimore St., Baltimore, MD, 21201, USA
| |
Collapse
|
16
|
Czéh B, Nagy SA. Clinical Findings Documenting Cellular and Molecular Abnormalities of Glia in Depressive Disorders. Front Mol Neurosci 2018. [PMID: 29535607 PMCID: PMC5835102 DOI: 10.3389/fnmol.2018.00056] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Depressive disorders are complex, multifactorial mental disorders with unknown neurobiology. Numerous theories aim to explain the pathophysiology. According to the “gliocentric theory”, glial abnormalities are responsible for the development of the disease. The aim of this review article is to summarize the rapidly growing number of cellular and molecular evidences indicating disturbed glial functioning in depressive disorders. We focus here exclusively on the clinical studies and present the in vivo neuroimaging findings together with the postmortem molecular and histopathological data. Postmortem studies demonstrate glial cell loss while the in vivo imaging data reveal disturbed glial functioning and altered white matter microstructure. Molecular studies report on altered gene expression of glial specific genes. In sum, the clinical findings provide ample evidences on glial pathology and demonstrate that all major glial cell types are affected. However, we still lack convincing theories explaining how the glial abnormalities develop and how exactly contribute to the emotional and cognitive disturbances. Abnormal astrocytic functioning may lead to disturbed metabolism affecting ion homeostasis and glutamate clearance, which in turn, affect synaptic communication. Abnormal oligodendrocyte functioning may disrupt the connectivity of neuronal networks, while microglial activation indicates neuroinflammatory processes. These cellular changes may relate to each other or they may indicate different endophenotypes. A theory has been put forward that the stress-induced inflammation—mediated by microglial activation—triggers a cascade of events leading to damaged astrocytes and oligodendroglia and consequently to their dysfunctions. The clinical data support the “gliocentric” theory, but future research should clarify whether these glial changes are truly the cause or simply the consequences of this devastating disorder.
Collapse
Affiliation(s)
- Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary.,Department of Laboratory Medicine, University of Pécs, Medical School, Pécs, Hungary
| | - Szilvia A Nagy
- Neurobiology of Stress Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary.,Department of Neurosurgery, University of Pécs, Medical School, Pécs, Hungary.,MTA-PTE, Clinical Neuroscience MR Research Group, Pécs, Hungary.,Pécs Diagnostic Centre, Pécs, Hungary
| |
Collapse
|
17
|
Hu J, Xu J, Pang L, Zhao H, Li F, Deng Y, Liu L, Lan Y, Zhang X, Zhao T, Xu C, Xu C, Xiao Y, Li X. Systematically characterizing dysfunctional long intergenic non-coding RNAs in multiple brain regions of major psychosis. Oncotarget 2018; 7:71087-71098. [PMID: 27661005 PMCID: PMC5342065 DOI: 10.18632/oncotarget.12122] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 09/12/2016] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SZ) and bipolar disorder (BD) are severe neuropsychiatric disorders with serious impact on patients, together termed “major psychosis”. Recently, long intergenic non-coding RNAs (lincRNAs) were reported to play important roles in mental diseases. However, little was known about their molecular mechanism in pathogenesis of SZ and BD. Here, we performed RNA sequencing on 82 post-mortem brain tissues from three brain regions (orbitofrontal cortex (BA11), anterior cingulate cortex (BA24) and dorsolateral prefrontal cortex (BA9)) of patients with SZ and BD and control subjects, generating over one billion reads. We characterized lincRNA transcriptome in the three brain regions and identified 20 differentially expressed lincRNAs (DELincRNAs) in BA11 for BD, 34 and 1 in BA24 and BA9 for SZ, respectively. Our results showed that these DELincRNAs exhibited brain region-specific patterns. Applying weighted gene co-expression network analysis, we revealed that DELincRNAs together with other genes can function as modules to perform different functions in different brain regions, such as immune system development in BA24 and oligodendrocyte differentiation in BA9. Additionally, we found that DNA methylation alteration could partly explain the dysregulation of lincRNAs, some of which could function as enhancers in the pathogenesis of major psychosis. Together, we performed systematical characterization of dysfunctional lincRNAs in multiple brain regions of major psychosis, which provided a valuable resource to understand their roles in SZ and BD pathology and helped to discover novel biomarkers.
Collapse
Affiliation(s)
- Jing Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jinyuan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongying Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yulan Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ling Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Tingting Zhao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chaohan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chun Xu
- Department of Psychiatry, Texas Tech University Health Science Center, El Paso, Texas, United States of America
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
18
|
Caletti E, Marotta G, Del Vecchio G, Paoli RA, Cigliobianco M, Prunas C, Zugno E, Bottinelli F, Brambilla P, Altamura AC. The metabolic basis of cognitive insight in psychosis: A positron emission tomography study. PLoS One 2017; 12:e0175803. [PMID: 28414766 PMCID: PMC5393874 DOI: 10.1371/journal.pone.0175803] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 03/31/2017] [Indexed: 01/23/2023] Open
Abstract
The purpose of this study was to investigate the relationship between cognitive insight and cerebral metabolism in patients suffering from psychosis. The Beck Cognitive Insight Scale (BCIS) was administered to 63 patients with psychosis undergoing Positron Emission Tomography investigation. The sample was divided into two groups considering the BCIS score. Data were analyzed using Statistical Parametric Mapping. RESULTS patients with low insight, compared to those with high insight, showed decreased metabolism in the right fusiform gyrus, left precuneus, superior temporal gyrus and insula bilaterally, as well as increased metabolism in the left orbito-frontal gyrus (all p<0.005). Our results suggest that reduced posterior (occipito-temporo-insulo-parietal) and increased anterior (orbitofrontal) cerebral metabolism may sustain low cognitive insight in psychosis.
Collapse
Affiliation(s)
- Elisabetta Caletti
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Marotta
- Section of Nuclear Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Riccardo A. Paoli
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Michela Cigliobianco
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia Prunas
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Zugno
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Bottinelli
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Brambilla
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Psychiatry and Behavioural Neurosciences, University of Texas at Houston, Houston, Texas, United States of America
| | - A. Carlo Altamura
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
19
|
Dossi E, Vasile F, Rouach N. Human astrocytes in the diseased brain. Brain Res Bull 2017; 136:139-156. [PMID: 28212850 PMCID: PMC5766741 DOI: 10.1016/j.brainresbull.2017.02.001] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 12/23/2022]
Abstract
Astrocytes are key active elements of the brain that contribute to information processing. They not only provide neurons with metabolic and structural support, but also regulate neurogenesis and brain wiring. Furthermore, astrocytes modulate synaptic activity and plasticity in part by controlling the extracellular space volume, as well as ion and neurotransmitter homeostasis. These findings, together with the discovery that human astrocytes display contrasting characteristics with their rodent counterparts, point to a role for astrocytes in higher cognitive functions. Dysfunction of astrocytes can thereby induce major alterations in neuronal functions, contributing to the pathogenesis of several brain disorders. In this review we summarize the current knowledge on the structural and functional alterations occurring in astrocytes from the human brain in pathological conditions such as epilepsy, primary tumours, Alzheimer's disease, major depressive disorder and Down syndrome. Compelling evidence thus shows that dysregulations of astrocyte functions and interplay with neurons contribute to the development and progression of various neurological diseases. Targeting astrocytes is thus a promising alternative approach that could contribute to the development of novel and effective therapies to treat brain disorders.
Collapse
Affiliation(s)
- Elena Dossi
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France.
| | - Flora Vasile
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France.
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France.
| |
Collapse
|
20
|
Niu M, Wang Y, Jia Y, Wang J, Zhong S, Lin J, Sun Y, Zhao L, Liu X, Huang L, Huang R. Common and Specific Abnormalities in Cortical Thickness in Patients with Major Depressive and Bipolar Disorders. EBioMedicine 2017; 16:162-171. [PMID: 28109831 PMCID: PMC5474436 DOI: 10.1016/j.ebiom.2017.01.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/08/2017] [Accepted: 01/09/2017] [Indexed: 01/10/2023] Open
Abstract
Major depressive disorder (MDD) and bipolar disorder (BD) are severe psychiatric diseases with overlapping symptomatology. Although previous studies reported abnormal brain structures in MDD or BD patients, the disorder-specific underlying neural mechanisms remain poorly understood. The purpose of this study was to investigate the whole-brain gray matter morphological patterns in unmedicated patients with MDD or BD and to identify the shared and disease-specific brain morphological alterations in these two disorders. We acquired high-resolution brain structural MRI data from a sample of 36 MDD patients, 32 BD patients, and 30 healthy controls. Using FreeSurfer, we estimated their brain cortical thickness (CT) and compared between-group difference in multiple locations across the continuous cortical surface. Compared to the healthy controls, both the MDD and BD patient groups showed significantly reduced CT in the left inferior temporal cortex (ITC). However, compared to the MDD patients, the BD patients showed a significantly thinner CT in the left rostral middle frontal region. In addition, compared to the healthy controls, the BD patients displayed thinner CT in the left ITC, left frontal pole (FPO), left superior frontal, right lateral occipital, right pars triangularis (PTRI) and right lateral orbitofrontal regions. Further analysis revealed a significantly positive correlation between the mean CT in the left FPO and the onset age, but a negative correlation between the mean CT in the right PTRI and the number of episodes, in the BD patients. Our findings revealed that the BD and MDD patients had variations in CT that were in common, but many more that were distinct, suggesting potential differences in their neural mechanisms. We found thinner CT in the left ITC in both MDD and BD groups compared to controls. We detected thinner CT in the left rMFC in the BD group compared to the MDD group. The BD group had more pronounced abnormality in CT primarily in the PFC than the MDD group. Clinical variables of BD group were associated with decreased CT in the left FPO and right PTRI.
This study aims to detect abnormal cortical thickness in patients with major depressive disorder (MDD) or bipolar disorder (BD), and to identify the shared and disease-specific brain morphological alterations in these two disorders. The two patient groups showed several common but more distinct variation patterns in cortical thickness, and the BD patients had lower cortical thickness in widespread brain areas than the MDD and the controls. These findings may have potential clinical implications for distinguishing BD from MDD patients.
Collapse
Affiliation(s)
- Meiqi Niu
- Center for the Study of Applied Psychology, Key Laboratory of Mental Health and Cognitive Science of Guangdong Province, School of Psychology, Brain Study Institute, South China Normal University, Guangzhou 510631, China
| | - Ying Wang
- Clinical Experimental Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Junjing Wang
- Center for the Study of Applied Psychology, Key Laboratory of Mental Health and Cognitive Science of Guangdong Province, School of Psychology, Brain Study Institute, South China Normal University, Guangzhou 510631, China
| | - Shuming Zhong
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jiabao Lin
- Center for the Study of Applied Psychology, Key Laboratory of Mental Health and Cognitive Science of Guangdong Province, School of Psychology, Brain Study Institute, South China Normal University, Guangzhou 510631, China
| | - Yao Sun
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ling Zhao
- Center for the Study of Applied Psychology, Key Laboratory of Mental Health and Cognitive Science of Guangdong Province, School of Psychology, Brain Study Institute, South China Normal University, Guangzhou 510631, China
| | - Xiaojin Liu
- Center for the Study of Applied Psychology, Key Laboratory of Mental Health and Cognitive Science of Guangdong Province, School of Psychology, Brain Study Institute, South China Normal University, Guangzhou 510631, China
| | - Li Huang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Ruiwang Huang
- Center for the Study of Applied Psychology, Key Laboratory of Mental Health and Cognitive Science of Guangdong Province, School of Psychology, Brain Study Institute, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
21
|
Smiley JF, Hackett TA, Bleiwas C, Petkova E, Stankov A, Mann JJ, Rosoklija G, Dwork AJ. Reduced GABA neuron density in auditory cerebral cortex of subjects with major depressive disorder. J Chem Neuroanat 2016; 76:108-121. [PMID: 26686292 PMCID: PMC4903945 DOI: 10.1016/j.jchemneu.2015.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 10/30/2015] [Indexed: 12/13/2022]
Abstract
Although major depressive disorder (MDD) and schizophrenia (SZ) are closely associated with disrupted functions in frontal and limbic areas of cerebral cortex, cellular pathology has also been found in other brain areas, including primary sensory cortex. Auditory cortex is of particular interest, given the prominence of auditory hallucinations in SZ, and sensory deficits in MDD. We used stereological sampling methods in auditory cortex to look for cellular differences between MDD, SZ and non-psychiatric subjects. Additionally, as all of our MDD subjects died of suicide, we evaluated the association of suicide with our measurements by selecting a SZ sample that was divided between suicide and non-suicide subjects. Measurements were done in primary auditory cortex (area A1) and auditory association cortex (area Tpt), two areas with distinct roles in sensory processing and obvious differences in neuron density and size. In MDD, densities of GABAergic interneurons immunolabeled for calretinin (CR) and calbindin (CB) were 23-29% lower than non-psychiatric controls in both areas. Parvalbumin (PV) interneurons (counted only in area Tpt) showed a nominally smaller (16%) reduction that was not statistically significant. Total neuron and glia densities measured in Nissl stained sections did not show corresponding reductions. Analysis of suicide in the SZ sample indicated that reduced CR cell density was associated with suicide, whereas the densities of CB and other cells were not. Our results are consistent with previous studies in MDD that found altered GABA-associated markers throughout the cerebral cortex including primary sensory areas.
Collapse
Affiliation(s)
- John F Smiley
- Program in Cognitive Neuroscience and Schizophrenia, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department of Child and Adolescent Psychiatry, New York University Langone Medical Center, New York, NY, USA.
| | - Troy A Hackett
- Department of Psychology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Cynthia Bleiwas
- Program in Cognitive Neuroscience and Schizophrenia, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Eva Petkova
- Program in Cognitive Neuroscience and Schizophrenia, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department of Child and Adolescent Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | | | - J John Mann
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute and Columbia University, New York, NY, USA; Department of Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Gorazd Rosoklija
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute and Columbia University, New York, NY, USA; Department of Neuroscience, New York State Psychiatric Institute, New York, NY, USA; Macedonian Academy of Sciences and Arts, Skopje, Macedonia
| | - Andrew J Dwork
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute and Columbia University, New York, NY, USA; Department of Neuroscience, New York State Psychiatric Institute, New York, NY, USA; Macedonian Academy of Sciences and Arts, Skopje, Macedonia
| |
Collapse
|
22
|
Varga Z, Csabai D, Miseta A, Wiborg O, Czéh B. Chronic stress affects the number of GABAergic neurons in the orbitofrontal cortex of rats. Behav Brain Res 2016; 316:104-114. [PMID: 27555539 DOI: 10.1016/j.bbr.2016.08.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/08/2016] [Accepted: 08/12/2016] [Indexed: 12/27/2022]
Abstract
Cortical GABAergic dysfunctions have been documented by clinical studies in major depression. We used here an animal model for depression and investigated whether long-term stress exposure can affect the number of GABAergic neurons in the orbitofrontal cortex (OFC). Adult male rats were subjected to 7-weeks of daily stress exposure and behaviorally phenotyped as anhedonic or stress-resilient animals. GABAergic interneurons were identified by immunohistochemistry and systematically quantified. We analyzed calbindin-(CB), calretinin-(CR), cholecystokinin-(CCK), parvalbumin-(PV), neuropeptide Y-(NPY) and somatostatin-positive (SST+) neurons in the following specific subareas of the OFC: medial orbital (MO), ventral orbital (VO), lateral orbital (LO) and dorsolateral orbital (DLO) cortex. For comparison, we also analyzed the primary motor cortex (M1) as a non-limbic cortical area. Stress had a pronounced effect on CB+ neurons and reduced their densities by 40-50% in the MO, VO and DLO. Stress had no effect on CCK+, CR+, PV+, NPY+ and SST+ neurons in any cortical areas. None of the investigated GABAergic neurons were affected by stress in the primary motor cortex. Interestingly, in the stress-resilient animals, we observed a significantly increased density of CCK+ neurons in the VO. NPY+ neuron densities were also significantly different between the anhedonic and stress-resilient rats, but only in the LO. Our present data demonstrate that chronic stress can specifically reduce the density of calbindin-positive GABAergic neurons in the orbitofrontal cortex and suggest that NPY and CCK expression in the OFC may relate to the stress resilience of the animals.
Collapse
Affiliation(s)
- Zsófia Varga
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Center, 7624 Pécs, Hungary
| | - Dávid Csabai
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Center, 7624 Pécs, Hungary
| | - Attila Miseta
- Department of Laboratory Medicine, University of Pécs, Medical School, 7624 Pécs, Hungary
| | - Ove Wiborg
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Boldizsár Czéh
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Center, 7624 Pécs, Hungary; Department of Laboratory Medicine, University of Pécs, Medical School, 7624 Pécs, Hungary; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark.
| |
Collapse
|
23
|
Trépanier MO, Hopperton KE, Mizrahi R, Mechawar N, Bazinet RP. Postmortem evidence of cerebral inflammation in schizophrenia: a systematic review. Mol Psychiatry 2016; 21:1009-26. [PMID: 27271499 PMCID: PMC4960446 DOI: 10.1038/mp.2016.90] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/29/2016] [Accepted: 04/20/2016] [Indexed: 12/13/2022]
Abstract
Schizophrenia is a psychiatric disorder which has a lifetime prevalence of ~1%. Multiple candidate mechanisms have been proposed in the pathogenesis of schizophrenia. One such mechanism is the involvement of neuroinflammation. Clinical studies, including neuroimaging, peripheral biomarkers and randomized control trials, have suggested the presence of neuroinflammation in schizophrenia. Many studies have also measured markers of neuroinflammation in postmortem brain samples from schizophrenia patients. The objective of this study was to conduct a systematic search of the literature on neuroinflammation in postmortem brains of schizophrenia patients indexed in MEDLINE, Embase and PsycINFO. Databases were searched up until 20th March 2016 for articles published on postmortem brains in schizophrenia evaluating microglia, astrocytes, glia, cytokines, the arachidonic cascade, substance P and other markers of neuroinflammation. Two independent reviewers extracted the data. Out of 5385 articles yielded by the search, 119 articles were identified that measured neuroinflammatory markers in schizophrenic postmortem brains. Glial fibrillary acidic protein expression was elevated, lower or unchanged in 6, 6 and 21 studies, respectively, and similar results were obtained for glial cell densities. On the other hand, microglial markers were increased, lower or unchanged in schizophrenia in 11, 3 and 8 studies, respectively. Results were variable across all other markers, but SERPINA3 and IFITM were consistently increased in 4 and 5 studies, respectively. Despite the variability, some studies evaluating neuroinflammation in postmortem brains in schizophrenia suggest an increase in microglial activity and other markers such as SERPINA3 and IFITM. Variability across studies is partially explained by multiple factors including brain region evaluated, source of the brain, diagnosis, age at time of death, age of onset and the presence of suicide victims in the cohort.
Collapse
Affiliation(s)
- M O Trépanier
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - K E Hopperton
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - R Mizrahi
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - N Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - R P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Kim HK, Nunes PV, Oliveira KC, Young LT, Lafer B. Neuropathological relationship between major depression and dementia: A hypothetical model and review. Prog Neuropsychopharmacol Biol Psychiatry 2016; 67:51-7. [PMID: 26780170 DOI: 10.1016/j.pnpbp.2016.01.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/10/2016] [Accepted: 01/13/2016] [Indexed: 01/18/2023]
Abstract
Major depression (MDD) is a chronic psychiatric condition in which patients often show increasing cognitive impairment with recurring episodes. Neurodegeneration may play an important component in the pathogenesis of MDD associated with cognitive complaints. In agreement with this, patients with MDD show decreased brain volumes in areas implicated in emotional regulation and cognition, neuronal and glial cell death as well as activation of various pathways that can contribute to cell death. Therefore, the aim of this review is to provide an integrative overview of potential contributing factors to neurodegeneration in MDD. Studies have reported increased neuronal and glial cell death in the frontal cortex, amygdala, and hippocampus of patients with MDD. This may be due to decreased neurogenesis from lower levels of brain-derived neurotrophic factor (BDNF), excitotoxicity from increased glutamate signaling, and lower levels of gamma-aminobutyric acid (GABA) signaling. In addition, mitochondrial dysfunction and oxidative stress are found in similar brain areas where evidence of excitotoxicity has been reported. Also, levels of antioxidant enzymes were reported to be increased in patients with MDD. Inflammation may also be a contributing factor, as levels of inflammatory cytokines were reported to be increased in the prefrontal cortex of patients with MDD. While preliminary, studies have also reported neuropathological alterations in patients with MDD. Together, these studies suggest that lower BDNF levels, mitochondrial dysfunction, oxidative stress, inflammation and excitotoxicity may be contributing to neuronal and glial cell death in MDD, leading to decreased brain volume and cognitive dysfunction with multiple recurrent episodes. This highlights the need to identify specific pathways involved in neurodegeneration in MDD, which may elucidate targets that can be treated to ameliorate the effects of disease progression in this disorder.
Collapse
Affiliation(s)
- Helena Kyunghee Kim
- Departments of Psychiatry and Pharmacology, University of Toronto, RM4204, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| | - Paula Villela Nunes
- Bipolar Disorder Program (PROMAN), Department of Psychiatry, University of São Paulo Medical School, Rua Dr. Ovídio Pires de Campos, 785, São Paulo, 3671, Brazil.
| | - Katia C Oliveira
- Bipolar Disorder Program (PROMAN), Department of Psychiatry, University of São Paulo Medical School, Rua Dr. Ovídio Pires de Campos, 785, São Paulo, 3671, Brazil.
| | - L Trevor Young
- Departments of Psychiatry and Pharmacology, University of Toronto, RM4204, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| | - Beny Lafer
- Bipolar Disorder Program (PROMAN), Department of Psychiatry, University of São Paulo Medical School, Rua Dr. Ovídio Pires de Campos, 785, São Paulo, 3671, Brazil.
| |
Collapse
|
25
|
Xu C, Ma XM, Chen HB, Zhou MH, Qiao H, An SC. Orbitofrontal cortex 5-HT2A receptor mediates chronic stress-induced depressive-like behaviors and alterations of spine density and Kalirin7. Neuropharmacology 2016; 109:7-17. [PMID: 26921771 DOI: 10.1016/j.neuropharm.2016.02.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 12/22/2022]
Abstract
Neuroimaging studies show that patients with major depression have reduced volume of the orbitofrontal cortex (OFC). Although the serotonin (5-HT) 2A receptor, which is abundant in the OFC, has been implicated in depression, the underlying mechanisms in the development of stress-induced depression remain unclear. Kalirin-7 (Kal7) is an essential component of mature excitatory synapses for maintaining dendritic spines density, size and synaptic functions. The aim of this study was to investigate the role of orbitofrontal 5-HT and 5-HT2A receptors in depressive-like behaviors and their associations with Kal7 and dendritic spines using chronic unpredictable mild stress (CUMS), an established animal model of depression. CUMS had no effect on the levels of 5-HT or the 5-HT2A receptor in the OFC. However, CUMS or microinjection of the 5-HT2A/2C receptor agonist (±)-1-(2, 5-Dimethoxy-4-iodophenyl)- 2-aminopropane hydrochloride (DOI, 5 μg/0.5 μL) into the OFC induced depressive-like behaviors, including anhedonia in the sucrose preference test and behavioral despair in the tail suspension test, a significant reduction in body weight gain and locomotor activity in the open field test, which were accompanied by decreased expression of Kal7 and PSD95 as well as decreased density of dendritic spines in the OFC. These alterations induced by CUMS were reversed by pretreatment with the 5-HT2A receptor antagonist Ketanserin (Ket, 5 μg/0.5 μL into the OFC). These results suggest that CUMS alters structural plasticity through activation of the orbital 5-HT2A receptor and is associated with decreased expression of Kal7, thereby resulting in depressive-like behaviors in rats, suggesting an important role of Kal7 in the OFC in depression.
Collapse
Affiliation(s)
- Chang Xu
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xin-Ming Ma
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China; University of Connecticut Health Center, Department of Neuroscience, Farmington, CT 06030, USA
| | - Hui-Bin Chen
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Meng-He Zhou
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Hui Qiao
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Shu-Cheng An
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
26
|
Dendritic Spines in Depression: What We Learned from Animal Models. Neural Plast 2016; 2016:8056370. [PMID: 26881133 PMCID: PMC4736982 DOI: 10.1155/2016/8056370] [Citation(s) in RCA: 301] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/26/2015] [Indexed: 02/07/2023] Open
Abstract
Depression, a severe psychiatric disorder, has been studied for decades, but the underlying mechanisms still remain largely unknown. Depression is closely associated with alterations in dendritic spine morphology and spine density. Therefore, understanding dendritic spines is vital for uncovering the mechanisms underlying depression. Several chronic stress models, including chronic restraint stress (CRS), chronic unpredictable mild stress (CUMS), and chronic social defeat stress (CSDS), have been used to recapitulate depression-like behaviors in rodents and study the underlying mechanisms. In comparison with CRS, CUMS overcomes the stress habituation and has been widely used to model depression-like behaviors. CSDS is one of the most frequently used models for depression, but it is limited to the study of male mice. Generally, chronic stress causes dendritic atrophy and spine loss in the neurons of the hippocampus and prefrontal cortex. Meanwhile, neurons of the amygdala and nucleus accumbens exhibit an increase in spine density. These alterations induced by chronic stress are often accompanied by depression-like behaviors. However, the underlying mechanisms are poorly understood. This review summarizes our current understanding of the chronic stress-induced remodeling of dendritic spines in the hippocampus, prefrontal cortex, orbitofrontal cortex, amygdala, and nucleus accumbens and also discusses the putative underlying mechanisms.
Collapse
|
27
|
Mamah D, Wen J, Luo J, Ulrich X, Barch DM, Yablonskiy D. Subcomponents of brain T2* relaxation in schizophrenia, bipolar disorder and siblings: A Gradient Echo Plural Contrast Imaging (GEPCI) study. Schizophr Res 2015; 169:36-45. [PMID: 26603058 PMCID: PMC4681636 DOI: 10.1016/j.schres.2015.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/02/2015] [Accepted: 10/06/2015] [Indexed: 11/30/2022]
Abstract
Investigating brain tissue T2* relaxation properties in vivo can potentially guide the uncovering of neuropathology in psychiatric illness, which is traditionally examined post mortem. We use an MRI-based Gradient Echo Plural Contrast Imaging (GEPCI) technique that produces inherently co-registered images allowing quantitative assessment of tissue cellular and hemodynamic properties. Usually described as R2* (=1/T2*) relaxation rate constant, recent developments in GEPCI allow the separation of cellular-specific (R2*C) and hemodynamic (BOLD) contributions to the MRI signal decay. We characterize BOLD effect in terms of tissue concentration of deoxyhemoglobin, i.e. CDEOXY, which reflects brain activity. 17 control (CON), 17 bipolar disorder (BPD), 16 schizophrenia (SCZ), and 12 unaffected schizophrenia sibling (SIB) participants were scanned and post-processed using GEPCI protocols. A MANOVA of 38gray matter regions ROIs showed significant group effects for CDEOXY but not for R2*C. In the three non-control groups, 71-92% of brain regions had increased CDEOXY. Group effects were observed in the superior temporal cortex and the thalamus. Increased superior temporal cortex CDEOXY was found in SCZ (p=0.01), BPD (p=0.01) and SIB (p=0.02), with bilateral effects in SCZ and only left hemisphere effects in BPD and SIB. Thalamic CDEOXY abnormalities were observed in SCZ (p=0.003), BPD (p=0.03) and SIB (p=0.02). Our results suggest that increased activity in certain brain regions is part of the underlying pathophysiology of specific psychiatric disorders. High CDEOXY in the superior temporal cortex suggests abnormal activity with auditory, language and/or social cognitive processing. Larger studies are needed to clarify the clinical significance of relaxometric abnormalities.
Collapse
Affiliation(s)
- Daniel Mamah
- Department of Psychiatry, Washington University Medical School, St. Louis, United States.
| | - Jie Wen
- Department of Radiology, Washington University Medical School, St. Louis, United States
| | - Jie Luo
- Department of Radiology, Washington University Medical School, St. Louis, United States
| | - Xialing Ulrich
- Department of Radiology, Washington University Medical School, St. Louis, United States
| | - Deanna M. Barch
- Department of Psychiatry, Washington University Medical School, St. Louis, United States, Department of Psychology, Washington University in St. Louis, United States, Department of Anatomy and Neurobiology, Washington University in St. Louis, United States
| | - Dmitriy Yablonskiy
- Department of Radiology, Washington University Medical School, St. Louis, United States
| |
Collapse
|
28
|
O'Shea KS, McInnis MG. Neurodevelopmental origins of bipolar disorder: iPSC models. Mol Cell Neurosci 2015; 73:63-83. [PMID: 26608002 DOI: 10.1016/j.mcn.2015.11.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 10/14/2015] [Accepted: 11/18/2015] [Indexed: 12/22/2022] Open
Abstract
Bipolar disorder (BP) is a chronic neuropsychiatric condition characterized by pathological fluctuations in mood from mania to depression. Adoption, twin and family studies have consistently identified a significant hereditary component to BP, yet there is no clear genetic event or consistent neuropathology. BP has been suggested to have a developmental origin, although this hypothesis has been difficult to test since there are no viable neurons or glial cells to analyze, and research has relied largely on postmortem brain, behavioral and imaging studies, or has examined proxy tissues including saliva, olfactory epithelium and blood cells. Neurodevelopmental factors, particularly pathways related to nervous system development, cell migration, extracellular matrix, H3K4 methylation, and calcium signaling have been identified in large gene expression and GWAS studies as altered in BP. Recent advances in stem cell biology, particularly the ability to reprogram adult somatic tissues to a pluripotent state, now make it possible to interrogate these pathways in viable cell models. A number of induced pluripotent stem cell (iPSC) lines from BP patient and healthy control (C) individuals have been derived in several laboratories, and their ability to form cortical neurons examined. Early studies suggest differences in activity, calcium signaling, blocks to neuronal differentiation, and changes in neuronal, and possibly glial, lineage specification. Initial observations suggest that differentiation of BP patient-derived neurons to dorsal telencephalic derivatives may be impaired, possibly due to alterations in WNT, Hedgehog or Nodal pathway signaling. These investigations strongly support a developmental contribution to BP and identify novel pathways, mechanisms and opportunities for improved treatments.
Collapse
Affiliation(s)
- K Sue O'Shea
- Department of Cell and Developmental Biology, University of Michigan, 3051 BSRB, 109 Zina Pitcher PL, Ann Arbor, MI 48109-2200, United States; Department of Psychiatry, University of Michigan, 4250 Plymouth Rd, Ann Arbor, MI 48109-5765, United States.
| | - Melvin G McInnis
- Department of Psychiatry, University of Michigan, 4250 Plymouth Rd, Ann Arbor, MI 48109-5765, United States
| |
Collapse
|
29
|
Xu H. Neuroinflammation in Schizophrenia Focused on the Pharmacological and Therapeutic Evidence. ACTA ACUST UNITED AC 2015. [DOI: 10.5567/pharmacologia.2015.438.453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
30
|
Long Z, Duan X, Wang Y, Liu F, Zeng L, Zhao JP, Chen H. Disrupted structural connectivity network in treatment-naive depression. Prog Neuropsychopharmacol Biol Psychiatry 2015; 56:18-26. [PMID: 25092218 DOI: 10.1016/j.pnpbp.2014.07.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 07/12/2014] [Accepted: 07/23/2014] [Indexed: 01/15/2023]
Abstract
BACKGROUND Neuroimaging studies suggest that treatment-naive depression (TD) is characterized by abnormal functional connectivity between specific brain regions. However, the question surrounding the structural basis of functional aberrations in TD patients still remains. METHODS In the present study, diffusion tensor imaging tractography was employed to construct structural connectivity networks in 22 early adult-onset, first-episode TD patients and 19 healthy controls (HC). Graph theory and network-based statistic (NBS) were then employed to investigate systematically the alteration of whole brain structural topological organization and structural connectivity in TD patients. RESULTS Graph theoretical analysis revealed that, compared with HC, TD patients exhibited altered structural topological measures, including decreased shortest path length, normalized clustering coefficient, normalized shortest path length, and small-worldness, as well as increased global and local efficiency. NBS results further revealed that TD patients showed two altered structural sub-networks. One sub-network mainly involved connections between the right orbitofrontal cortex (OFC) and the right insula, putamen, caudate, hippocampus, fusiform gyrus, inferior temporal gyrus and lingual gyrus. The other sub-network mainly included connections between the left OFC and the left gyrus rectus, insula, putamen, caudate, thalamus, pallidum and middle occipital gyrus. CONCLUSIONS The findings suggest that TD patients exhibit a disruption in the topological organization of structural brain networks. The altered orbitofrontal connectivity may particularly contribute to the manifestation of symptoms in TD patients. The abnormalities may facilitate understanding of the functional disturbances of mood and cognition in the disease.
Collapse
Affiliation(s)
- Zhiliang Long
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Xujun Duan
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Yifeng Wang
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Feng Liu
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Ling Zeng
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China
| | - Jing-Ping Zhao
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China.
| | - Huafu Chen
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, PR China.
| |
Collapse
|
31
|
Frontal cortical and subcortical projections provide a basis for segmenting the cingulum bundle: implications for neuroimaging and psychiatric disorders. J Neurosci 2014; 34:10041-54. [PMID: 25057206 DOI: 10.1523/jneurosci.5459-13.2014] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The cingulum bundle (CB) is one of the brain's major white matter pathways, linking regions associated with executive function, decision-making, and emotion. Neuroimaging has revealed that abnormalities in particular locations within the CB are associated with specific psychiatric disorders, including depression and bipolar disorder. However, the fibers using each portion of the CB remain unknown. In this study, we used anatomical tract-tracing in nonhuman primates (Macaca nemestrina, Macaca fascicularis, Macaca mulatta) to examine the organization of specific cingulate, noncingulate frontal, and subcortical pathways through the CB. The goals were as follows: (1) to determine connections that use the CB, (2) to establish through which parts of the CB these fibers travel, and (3) to relate the CB fiber pathways to the portions of the CB identified in humans as neurosurgical targets for amelioration of psychiatric disorders. Results indicate that cingulate, noncingulate frontal, and subcortical fibers all travel through the CB to reach both cingulate and noncingulate targets. However, many brain regions send projections through only part, not all, of the CB. For example, amygdala fibers are not present in the caudal portion of the dorsal CB. These results allow segmentation of the CB into four unique zones. We identify the specific connections that are abnormal in psychiatric disorders and affected by neurosurgical interventions, such as deep brain stimulation and cingulotomy.
Collapse
|
32
|
Foster V, Oakley AE, Slade JY, Hall R, Polvikoski TM, Burke M, Thomas AJ, Khundakar A, Allan LM, Kalaria RN. Pyramidal neurons of the prefrontal cortex in post-stroke, vascular and other ageing-related dementias. ACTA ACUST UNITED AC 2014; 137:2509-21. [PMID: 24974383 DOI: 10.1093/brain/awu172] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Dementia associated with cerebrovascular disease is common. It has been reported that ∼30% of elderly patients who survive stroke develop delayed dementia (post-stroke dementia), with most cases being diagnosed as vascular dementia. The pathological substrates associated with post-stroke or vascular dementia are poorly understood, particularly those associated with executive dysfunction. Three separate yet interconnecting circuits control executive function within the frontal lobe involving the dorsolateral prefrontal cortex, anterior cingulate cortex and the orbitofrontal cortex. We used stereological methods, along with immunohistological and related cell morphometric analysis, to examine densities and volumes of pyramidal neurons of the dorsolateral prefrontal cortex, anterior cingulate cortex and orbitofrontal cortex in the frontal lobe from a total of 90 elderly subjects (age range 71-98 years). Post-mortem brain tissues from post-stroke dementia and post-stroke patients with no dementia were derived from our prospective Cognitive Function After Stroke study. We also examined, in parallel, samples from ageing controls and similar age subjects pathologically diagnosed with Alzheimer's disease, mixed Alzheimer's disease and vascular dementia, and vascular dementia. We found pyramidal cell volumes in layers III and V in the dorsolateral prefrontal cortex of post-stroke and vascular dementia and, of mixed and Alzheimer's disease subjects to be reduced by 30-40% compared to post-stroke patients with no dementia and controls. There were no significant changes in neuronal volumes in either the anterior cingulate or orbitofrontal cortices. Remarkably, pyramidal neurons within the orbitofrontal cortex were also found to be smaller in size when compared to those in the other two neocortical regions. To relate the cell changes to cognitive function, we noted significant correlations between neuronal volumes and total CAMCOG, orientation and memory scores and clinical dementia ratings. Total estimated neuronal densities were not significantly changed between patients with post-stroke dementia and post-stroke patients with no dementia groups or ageing controls in any of the three frontal regions. In further morphometric analysis of the dorsolateral prefrontal cortex, we showed that neither diffuse cerebral atrophy nor neocortical thickness explained the selective neuronal volume effects. We also noted that neurofilament protein SMI31 immunoreactivity was increased in post-stroke and vascular dementia compared with post-stroke patients with no dementia and correlated with decreased neuronal volumes in subjects with post-stroke dementia and vascular dementia. Our findings suggest selective regional pyramidal cell atrophy in the dorsolateral prefrontal cortex-rather than neuronal density changes per se-are associated with dementia and executive dysfunction in post-stroke dementia and vascular dementia. The changes in dorsolateral prefrontal cortex pyramidal cells were not associated with neurofibrillary pathology suggesting there is a vascular basis for the observed highly selective neuronal atrophy.
Collapse
Affiliation(s)
- Vincent Foster
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Arthur E Oakley
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Janet Y Slade
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Roslyn Hall
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Tuomo M Polvikoski
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Matthew Burke
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Alan J Thomas
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Ahmad Khundakar
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Louise M Allan
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Raj N Kalaria
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| |
Collapse
|
33
|
Griffiths KR, Morris RW, Balleine BW. Translational studies of goal-directed action as a framework for classifying deficits across psychiatric disorders. Front Syst Neurosci 2014; 8:101. [PMID: 24904322 PMCID: PMC4033402 DOI: 10.3389/fnsys.2014.00101] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/09/2014] [Indexed: 11/13/2022] Open
Abstract
The ability to learn contingencies between actions and outcomes in a dynamic environment is critical for flexible, adaptive behavior. Goal-directed actions adapt to changes in action-outcome contingencies as well as to changes in the reward-value of the outcome. When networks involved in reward processing and contingency learning are maladaptive, this fundamental ability can be lost, with detrimental consequences for decision-making. Impaired decision-making is a core feature in a number of psychiatric disorders, ranging from depression to schizophrenia. The argument can be developed, therefore, that seemingly disparate symptoms across psychiatric disorders can be explained by dysfunction within common decision-making circuitry. From this perspective, gaining a better understanding of the neural processes involved in goal-directed action, will allow a comparison of deficits observed across traditional diagnostic boundaries within a unified theoretical framework. This review describes the key processes and neural circuits involved in goal-directed decision-making using evidence from animal studies and human neuroimaging. Select studies are discussed to outline what we currently know about causal judgments regarding actions and their consequences, action-related reward evaluation, and, most importantly, how these processes are integrated in goal-directed learning and performance. Finally, we look at how adaptive decision-making is impaired across a range of psychiatric disorders and how deepening our understanding of this circuitry may offer insights into phenotypes and more targeted interventions.
Collapse
Affiliation(s)
- Kristi R Griffiths
- Behavioural Neuroscience Laboratory, Brain and Mind Research Institute, University of Sydney Camperdown, Sydney, NSW, Australia
| | - Richard W Morris
- Behavioural Neuroscience Laboratory, Brain and Mind Research Institute, University of Sydney Camperdown, Sydney, NSW, Australia
| | - Bernard W Balleine
- Behavioural Neuroscience Laboratory, Brain and Mind Research Institute, University of Sydney Camperdown, Sydney, NSW, Australia
| |
Collapse
|
34
|
Eker C, Simsek F, Yılmazer EE, Kitis O, Cinar C, Eker OD, Coburn K, Gonul AS. Brain regions associated with risk and resistance for bipolar I disorder: a voxel-based MRI study of patients with bipolar disorder and their healthy siblings. Bipolar Disord 2014; 16:249-61. [PMID: 24589068 DOI: 10.1111/bdi.12181] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 08/30/2013] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Bipolar I disorder is a highly heritable disorder but not all siblings manifest with the illness, even though they may share similar genetic and environmental risk factors. Thus, sibling studies may help to identify brain structural endophenotypes associated with risk and resistance for the disorder. METHODS Structural magnetic resonance imaging (MRI) scans were acquired for 28 euthymic patients with bipolar disorder, their healthy siblings, and 30 unrelated healthy controls. Statistical Parametric Mapping 8 (SPM8) was used to identify group differences in regional gray matter volume by voxel-based morphometry (VBM). RESULTS Using analysis of covariance, gray matter analysis of the groups revealed a group effect indicating that the left orbitofrontal cortex [Brodmann area (BA) 11] was smaller in patients with bipolar disorder than in unrelated healthy controls [F = 14.83, p < 0.05 (family-wise error); 7 mm(3) ]. Paired t-tests indicated that the orbitofrontal cortex of patients with bipolar disorder [t = 5.19, p < 0.05 (family-wise error); 37 mm(3) ] and their healthy siblings [t = 3.89, p < 0.001 (uncorrected); 63 mm(3) ] was smaller than in unrelated healthy controls, and that the left dorsolateral prefrontal cortex was larger in healthy siblings than in patients with bipolar disorder [t = 4.28, p < 0.001 (uncorrected); 323 mm(3) ] and unrelated healthy controls [t = 4.36, p < 0.001 (uncorrected); 245 mm(3) ]. Additional region-of-interest analyses also found volume deficits in the right cerebellum of patients with bipolar disorder [t = 3.92, p < 0.001 (uncorrected); 178 mm(3) ] and their healthy siblings [t = 4.23, p < 0.001 (uncorrected); 489 mm(3) ], and in the left precentral gyrus of patients with bipolar disorder [t = 3.61, p < 0.001 (uncorrected); 115 mm(3) ] compared to unrelated healthy controls. CONCLUSIONS The results of this study suggest that a reduction in the volume of the orbitofrontal cortex, which plays a role in the automatic regulation of emotions and is a part of the medial prefrontal network, is associated with the heritability of bipolar disorder. Conversely, increased dorsolateral prefrontal cortex volume may be a neural marker of a resistance factor as it is part of a network of voluntary emotion regulation and balances the effects of the disrupted automatic emotion regulation system.
Collapse
Affiliation(s)
- Cagdas Eker
- Department of Psychiatry, Ege University School of Medicine, Bornova, Izmir, Turkey; SoCAT Lab and Affective Disorders Unit, Ege University School of Medicine, Bornova, Izmir, Turkey; Department of Neuroscience, Ege University Institute of Health Sciences, Bornova, Izmir, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Ray MT, Shannon Weickert C, Webster MJ. Decreased BDNF and TrkB mRNA expression in multiple cortical areas of patients with schizophrenia and mood disorders. Transl Psychiatry 2014; 4:e389. [PMID: 24802307 PMCID: PMC4035720 DOI: 10.1038/tp.2014.26] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 03/09/2014] [Indexed: 02/07/2023] Open
Abstract
Abnormalities in brain-derived neurotrophic factor (BDNF)/trkB signaling have been implicated in the etiology of schizophrenia and mood disorders. Patients with schizophrenia, bipolar disorder (BPD) and major depression (MDD) have reduced levels of neurotrophins in their brains when compared with normal unaffected individuals; however, only a few brain areas have been examined to date. Owing to the broad range of symptoms manifested in these disorders, we hypothesized that multiple associative areas of the neocortex may be implicated and that the degree of change in BDNF and trkB-TK+ mRNA expression and the cortical region or layers involved may vary according to Diagnostic and Statistical Manual of Mental Disorders (DSM) diagnosis. We compared BDNF and trkB-TK+ mRNA levels across all layers of the prefrontal cortex (dorsolateral prefrontal cortex, DLPFC), orbital frontal cortex (OFC), anterior cingulate cortex (ACC), inferior temporal gyrus (ITG) and superior temporal gyrus (STG) in four groups: schizophrenia, BPD, MDD and unaffected controls (n=60). BDNF mRNA levels were significantly decreased in layers IV and V of DLPFC in schizophrenia patients, in layer VI of ACC in schizophrenia and MDD and in layer VI of ITG in schizophrenia, BPD and MDD. BDNF mRNA levels were also significantly decreased in layer V and/or VI of STG in schizophrenia, BPD and MDD. TrkB-TK+ mRNA levels were only significantly decreased in the cortical layer VI of OFC in BPD. The shared and distinct patterns of neurotrophin transcript reductions, with some specific to each group, may compromise the function and plasticity of distinct cortical areas to various degrees in the different groups and contribute to the range and overlap of symptoms manifested across the diagnoses.
Collapse
Affiliation(s)
- M T Ray
- Stanley Medical Research Institute, Laboratory of Brain Research, Rockville, MD, USA,Trinity Washington University, NE Washington, DC, USA
| | - C Shannon Weickert
- Schizophrenia Research Institute, Sydney, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - M J Webster
- Stanley Medical Research Institute, Laboratory of Brain Research, Rockville, MD, USA,Stanley Laboratory of Brain Research, 9800 Medical Center Drive, Rockville, MD 20850, USA. E-mail:
| |
Collapse
|
36
|
Savitz JB, Price JL, Drevets WC. Neuropathological and neuromorphometric abnormalities in bipolar disorder: view from the medial prefrontal cortical network. Neurosci Biobehav Rev 2014; 42:132-47. [PMID: 24603026 DOI: 10.1016/j.neubiorev.2014.02.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 01/20/2014] [Accepted: 02/19/2014] [Indexed: 12/15/2022]
Abstract
The question of whether BD is primarily a developmental disorder or a progressive, neurodegenerative disorder remains unresolved. Here, we review the morphometric postmortem and neuroimaging literature relevant to the neuropathology of bipolar disorder (BD). We focus on the medial prefrontal cortex (mPFC) network, a key system in the regulation of emotional, behavioral, endocrine, and innate immunological responses to stress. We draw four main conclusions: the mPFC is characterized by (1) a decrease in volume, (2) reductions in neuronal size, and/or changes in neuronal density, (3) reductions in glial cell density, and (4) changes in gene expression. These data suggest the presence of dendritic atrophy of neurons and the loss of oligodendroglial cells in BD, although some data additionally suggest a reduction in the cell counts of specific subpopulations of GABAergic interneurons. Based on the weight of the postmortem and neuroimaging literature discussed herein, we favor a complex hypothesis that BD primarily constitutes a developmental disorder, but that additional, progressive, histopathological processes also are associated with recurrent or chronic illness. Conceivably BD may be best conceptualized as a progressive neurodevelopmental disorder.
Collapse
Affiliation(s)
- Jonathan B Savitz
- Laureate Institute for Brain Research, Tulsa, OK, USA; Faculty of Community Medicine, University of Tulsa, Tulsa, OK, USA.
| | - Joseph L Price
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wayne C Drevets
- Laureate Institute for Brain Research, Tulsa, OK, USA; Janssen Pharmaceuticals of Johnson & Johnson, Inc., Titusville, NJ, USA
| |
Collapse
|
37
|
Khundakar AA, Thomas AJ. Cellular morphometry in late-life depression: a review of postmortem studies. Am J Geriatr Psychiatry 2014; 22:122-32. [PMID: 24012224 DOI: 10.1016/j.jagp.2013.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 05/21/2013] [Accepted: 06/13/2013] [Indexed: 11/20/2022]
Abstract
The impact of major depression in late life is considerable and set to intensify with a worldwide shift in demographic profile toward an elderly population. Although the precise neurobiological mechanisms are not fully understood, a significant body of clinical, epidemiological, and imaging data have suggested divergent pathophysiological pathways underlie depression in late life, when compared with younger patients. Neuroimaging studies have demonstrated significant increases in white matter hyperintensities in late-life depression in several key areas involved in affective circuitry. Postmortem cellular morphometry studies have played a vital role in the identification of discrete changes in the brain microstructure in depression. This review draws together such postmortem studies, which have utilized tissue from younger/mixed age and late-life depressed patients. These findings have suggested varying neuronal and glial cell pathology in depression between different age cohorts. This age-related disparity may suggest different pathophysiological basis for depression, with vascular factors playing a potentially greater role in late life.
Collapse
Affiliation(s)
- Ahmad A Khundakar
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Alan J Thomas
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
38
|
Abstract
Multiple lines of evidence indicate that mood disorders are associated with abnormalities in the brain's cellular composition, especially in glial cells. Considered inert support cells in the past, glial cells are now known to be important for brain function. Treatments for mood disorders enhance glial cell proliferation, and experimental stimulation of cell growth has antidepressant effects in animal models of mood disorders. These findings suggest that the proliferation and survival of glial cells may be important in the pathogenesis of mood disorders and may be possible targets for the development of new treatments. In this article we review the evidence for glial abnormalities in mood disorders, and we discuss glial cell biology and evidence from postmortem studies of mood disorders. The goal is not to carry out a comprehensive review but to selectively discuss existing evidence in support of an argument for the role of glial cells in mood disorders.
Collapse
|
39
|
Najjar S, Pearlman DM, Devinsky O, Najjar A, Zagzag D. Neurovascular unit dysfunction with blood-brain barrier hyperpermeability contributes to major depressive disorder: a review of clinical and experimental evidence. J Neuroinflammation 2013; 10:142. [PMID: 24289502 PMCID: PMC4220803 DOI: 10.1186/1742-2094-10-142] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 11/15/2013] [Indexed: 12/27/2022] Open
Abstract
About one-third of people with major depressive disorder (MDD) fail at least two antidepressant drug trials at 1 year. Together with clinical and experimental evidence indicating that the pathophysiology of MDD is multifactorial, this observation underscores the importance of elucidating mechanisms beyond monoaminergic dysregulation that can contribute to the genesis and persistence of MDD. Oxidative stress and neuroinflammation are mechanistically linked to the presence of neurovascular dysfunction with blood-brain barrier (BBB) hyperpermeability in selected neurological disorders, such as stroke, epilepsy, multiple sclerosis, traumatic brain injury, and Alzheimer’s disease. In contrast to other major psychiatric disorders, MDD is frequently comorbid with such neurological disorders and constitutes an independent risk factor for morbidity and mortality in disorders characterized by vascular endothelial dysfunction (cardiovascular disease and diabetes mellitus). Oxidative stress and neuroinflammation are implicated in the neurobiology of MDD. More recent evidence links neurovascular dysfunction with BBB hyperpermeability to MDD without neurological comorbidity. We review this emerging literature and present a theoretical integration between these abnormalities to those involving oxidative stress and neuroinflammation in MDD. We discuss our hypothesis that alterations in endothelial nitric oxide levels and endothelial nitric oxide synthase uncoupling are central mechanistic links in this regard. Understanding the contribution of neurovascular dysfunction with BBB hyperpermeability to the pathophysiology of MDD may help to identify novel therapeutic and preventative approaches.
Collapse
Affiliation(s)
- Souhel Najjar
- Department of Neurology, Neuroinflammation Research Group, Epilepsy Center Division, NYU School of Medicine, New York, NY 10016, USA.
| | | | | | | | | |
Collapse
|
40
|
Olfactory bulbectomy induces neuronal rearrangement in the entorhinal cortex in the rat. J Chem Neuroanat 2013; 52:80-6. [DOI: 10.1016/j.jchemneu.2013.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/07/2013] [Accepted: 07/07/2013] [Indexed: 12/20/2022]
|
41
|
Dwyer JM, Duman RS. Activation of mammalian target of rapamycin and synaptogenesis: role in the actions of rapid-acting antidepressants. Biol Psychiatry 2013; 73:1189-98. [PMID: 23295207 PMCID: PMC3622786 DOI: 10.1016/j.biopsych.2012.11.011] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 11/05/2012] [Accepted: 11/18/2012] [Indexed: 01/01/2023]
Abstract
Antidepressants that produce rapid and robust effects, particularly for severely ill patients, represent one of the largest unmet medical needs for the treatment of depression. Currently available drugs that modulate monoamine neurotransmission provide relief for only a subset of patients, and this minimal efficacy requires several weeks of chronic treatment. The recent discovery that the glutamatergic agent ketamine produces rapid antidepressant responses within hours has opened a new area of research to explore the molecular mechanisms through which ketamine produces these surprising responses. Clinical and preclinical findings have exposed some of the unique actions of ketamine and identified a cell-signaling pathway known as the mammalian target of rapamycin. Activation of mammalian target of rapamycin and increased synaptogenesis in the prefrontal cortex are crucial in mediating the antidepressant effects of ketamine. Importantly, the synaptic actions of ketamine allow rapid recovery from the insults produced by exposure to repeated stress that cause neuronal atrophy and loss of synaptic connections. In the following review, we explore some of the clinical and preclinical findings that have thrust ketamine to the forefront of rapid antidepressant research and unveiled some of its unique molecular and cellular actions.
Collapse
Affiliation(s)
- Jason M Dwyer
- Laboratory of Molecular Psychiatry, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
42
|
Najjar S, Pearlman DM, Alper K, Najjar A, Devinsky O. Neuroinflammation and psychiatric illness. J Neuroinflammation 2013; 10:43. [PMID: 23547920 PMCID: PMC3626880 DOI: 10.1186/1742-2094-10-43] [Citation(s) in RCA: 500] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/28/2013] [Indexed: 12/19/2022] Open
Abstract
Multiple lines of evidence support the pathogenic role of neuroinflammation in psychiatric illness. While systemic autoimmune diseases are well-documented causes of neuropsychiatric disorders, synaptic autoimmune encephalitides with psychotic symptoms often go under-recognized. Parallel to the link between psychiatric symptoms and autoimmunity in autoimmune diseases, neuroimmunological abnormalities occur in classical psychiatric disorders (for example, major depressive, bipolar, schizophrenia, and obsessive-compulsive disorders). Investigations into the pathophysiology of these conditions traditionally stressed dysregulation of the glutamatergic and monoaminergic systems, but the mechanisms causing these neurotransmitter abnormalities remained elusive. We review the link between autoimmunity and neuropsychiatric disorders, and the human and experimental evidence supporting the pathogenic role of neuroinflammation in selected classical psychiatric disorders. Understanding how psychosocial, genetic, immunological and neurotransmitter systems interact can reveal pathogenic clues and help target new preventive and symptomatic therapies.
Collapse
Affiliation(s)
- Souhel Najjar
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| | | | | | | | | |
Collapse
|
43
|
A systematic review of brain frontal lobe parcellation techniques in magnetic resonance imaging. Brain Struct Funct 2013; 219:1-22. [DOI: 10.1007/s00429-013-0527-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 02/14/2013] [Indexed: 01/06/2023]
|
44
|
Guintivano J, Aryee MJ, Kaminsky ZA. A cell epigenotype specific model for the correction of brain cellular heterogeneity bias and its application to age, brain region and major depression. Epigenetics 2013; 8:290-302. [PMID: 23426267 PMCID: PMC3669121 DOI: 10.4161/epi.23924] [Citation(s) in RCA: 287] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Brain cellular heterogeneity may bias cell type specific DNA methylation patterns, influencing findings in psychiatric epigenetic studies. We performed fluorescence activated cell sorting (FACS) of neuronal nuclei and Illumina HM450 DNA methylation profiling in post mortem frontal cortex of 29 major depression and 29 matched controls. We identify genomic features and ontologies enriched for cell type specific epigenetic variation. Using the top cell epigenotype specific (CETS) marks, we generated a publically available R package, “CETS,” capable of quantifying neuronal proportions and generating in silico neuronal profiles from DNA methylation data. We demonstrate a significant overlap in major depression DNA methylation associations between FACS separated and CETS model generated neuronal profiles relative to bulk profiles. CETS derived neuronal proportions correlated significantly with age in the frontal cortex and cerebellum and accounted for epigenetic variation between brain regions. CETS based control of cellular heterogeneity will enable more robust hypothesis testing in the brain.
Collapse
Affiliation(s)
- Jerry Guintivano
- The Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
45
|
Williams MR, Hampton T, Pearce RKB, Hirsch SR, Ansorge O, Thom M, Maier M. Astrocyte decrease in the subgenual cingulate and callosal genu in schizophrenia. Eur Arch Psychiatry Clin Neurosci 2013; 263:41-52. [PMID: 22660922 DOI: 10.1007/s00406-012-0328-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 05/05/2012] [Indexed: 10/28/2022]
Abstract
Decreases in glial cell density and in GFAP mRNA in the anterior cingulate cortex have been reported in schizophrenia, bipolar disorder and major depressive disorder. Our study examines astrocyte and oligodendrocyte density in the white and grey matter of the subgenual cingulate cortex, and at the midline of the genu of the corpus callosum, in schizophrenia, bipolar disorder, depression and normal control cases. Serial coronal sections were stained with H and E for anatomical guidance, cresyl haematoxylin for oligodendrocyte identification and GFAP immunohistochemistry for astrocyte identification. Oligodendrocyte and astrocyte density was measured using systematic anatomical distinctions and randomised counting methods. A significant decrease in astrocyte density was observed in schizophrenia compared with normal controls in the cingulate grey matter, cingulate white matter and the midline of the corpus callosum (p = 0.025). Bipolar disorder and depression cases showed no significant changes in astrocyte density. Oligodendrocytes did not show any changes between diagnostic groups. In subgenual cingulate cortex, the ratio of oligodendrocytes to astrocytes was decreased between the controls and the three disease groups, suggesting a specific glial cell type specific change in schizophrenia.
Collapse
Affiliation(s)
- Matthew Roy Williams
- Neuropathology Unit, Department of Experimental Medicine, Imperial College London, Charing Cross Hospital, UK.
| | | | | | | | | | | | | |
Collapse
|
46
|
Impaired structural hippocampal plasticity is associated with emotional and memory deficits in the olfactory bulbectomized rat. Neuroscience 2013; 236:233-43. [PMID: 23357118 DOI: 10.1016/j.neuroscience.2013.01.037] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/15/2013] [Accepted: 01/16/2013] [Indexed: 12/12/2022]
Abstract
Disturbances in olfactory circuitry have been associated with depression in humans. The olfactory bulbectomized (OBX lesion) has been largely used as a model of depression-like behavior in the rat. However, quantitative neuronal rearrangements in key brain regions in this animal model have not been evaluated yet. Accordingly, we investigated changes in hippocampal plasticity as well as behavioral deficits in this animal model. OBX-induced behavioral deficits were studied in a battery of tests, namely the open field test (OFT), forced swim test (FST), and spatial memory disturbances in the Morris water maze (MWM). To characterize the neuronal remodeling, neuroanatomical rearrangements were investigated in the CA1 hippocampus and piriform cortex (PirC), brain regions receiving inputs from the olfactory bulbs and associated with emotional or olfactory processes. Additionally, cell proliferation and survival of newborn cells in the adult dentate gyrus (DG) of the hippocampus were also determined. OBX induced hyperlocomotion and enhanced rearing and grooming in the OFT, increased immobility in the FST as well as required a longer time to find the hidden platform in the MWM. OBX also induced dendritic atrophy in the hippocampus and PirC. In addition, cell proliferation was decreased while the survival remained unchanged in the DG of these animals. These various features are also observed in depressed subjects, adding further support to the validity and usefulness of this model to evaluate potential novel antidepressants.
Collapse
|
47
|
McNamara RK. Deciphering the role of docosahexaenoic acid in brain maturation and pathology with magnetic resonance imaging. Prostaglandins Leukot Essent Fatty Acids 2013; 88:33-42. [PMID: 22521863 PMCID: PMC3458176 DOI: 10.1016/j.plefa.2012.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 03/30/2012] [Accepted: 03/31/2012] [Indexed: 01/11/2023]
Abstract
Animal studies have found that deficits in brain docosahexaenoic acid (DHA, 22:6n-3) accrual during perinatal development leads to transient and enduring abnormalities in brain development and function. Determining the relevance of this evidence to brain disorders in humans has been hampered by an inability to determine antimortem brain DHA levels and limitations associated with a postmortem approach. Accordingly, there is a need for alternate or complementary approaches to better understand the role of DHA in cortical function and pathology, and conventional magnetic resonance imaging (MRI) techniques may be ideally suited for this application. A major advantage of neuroimaging is that it permits prospective evaluation of the effects of manipulating DHA status on both clinical and neuroimaging variables. Emerging evidence from MRI studies suggest that greater DHA status is associated with cortical structural and functional integrity, and suggest that reduced DHA status and abnormalities in cortical function observed in psychiatric disorders may be interrelated phenomenon. Preliminary evidence from animal MRI studies support a critical role of DHA in normal brain development. Neuroimaging research in both human and animals therefore holds tremendous promise for developing a better understanding of the role of DHA status in cortical function, as well as for elucidating the impact of DHA deficiency on neuropathological processes implicated in the etiology and progression of neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Robert K McNamara
- Department of Psychiatry and Behavioral Neuroscience, Division of Bipolar Disorders Research, University of Cincinnati College of Medicine, Cincinnati, OH 45219, USA.
| |
Collapse
|
48
|
Abstract
Late-life depression (LLD) has a complex and multifactoral etiology. There is growing interest in elucidating how glia, acting alone or as part of a glial-neuronal network, may contribute to the pathophysiology of depression. In this paper, we explore results from neuroimaging studies showing gray-matter volume loss in key frontal and subcortical structures implicated in LLD, and present the few histological studies that have examined neuronal and glial densities in these regions. Compared to results in younger people with depression, there appear to be age-dependent differences in neuronal pathology but the changes in glial pathology may be more subtle, perhaps reflecting a longer-term compensatory gliosis to earlier damage. We then consider the mechanisms by which both astrocytes and microglia may mediate and modulate neuronal dysfunction and possible degeneration in depression. These include a critical role in the response to peripheral inflammation and central microglial activation, as well as a key role in glutamate metabolism. Advances in our understanding of glia are highlighted, including the role of microglia as "electricians" of the brain and astrocytes as key communicating cells, an integral part of the tripartite synapse. Finally, implications for clinicians are discussed, including the consideration of glia as biomarkers for LLD and incorporation of glia into future therapeutic strategies.
Collapse
Affiliation(s)
- Matt Bennett Paradise
- Brain and Mind Research Institute, University of Sydney, Camperdown, New South Wales, Australia.
| | | | | | | | | |
Collapse
|
49
|
Naismith SL, Norrie LM, Mowszowski L, Hickie IB. The neurobiology of depression in later-life: Clinical, neuropsychological, neuroimaging and pathophysiological features. Prog Neurobiol 2012; 98:99-143. [DOI: 10.1016/j.pneurobio.2012.05.009] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 05/03/2012] [Accepted: 05/09/2012] [Indexed: 02/07/2023]
|
50
|
Jackowski AP, de Araújo Filho GM, de Almeida AG, de Araújo CM, Reis M, Nery F, Batista IR, Silva I, Lacerda AL. The involvement of the orbitofrontal cortex in psychiatric disorders: an update of neuroimaging findings. BRAZILIAN JOURNAL OF PSYCHIATRY 2012; 34:207-12. [DOI: 10.1590/s1516-44462012000200014] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 02/17/2012] [Indexed: 02/07/2023]
|