1
|
Shen SY, Liang LF, Shi TL, Shen ZQ, Yin SY, Zhang JR, Li W, Mi WL, Wang YQ, Zhang YQ, Yu J. Microglia-Derived Interleukin-6 Triggers Astrocyte Apoptosis in the Hippocampus and Mediates Depression-Like Behavior. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412556. [PMID: 39888279 PMCID: PMC11923973 DOI: 10.1002/advs.202412556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/19/2025] [Indexed: 02/01/2025]
Abstract
In patients with major depressive disorder (MDD) and animal models of depression, key pathological hallmarks include activation of microglia as well as atrophy and loss of astrocytes. Under certain pathological conditions, microglia can inflict damage to neurons and astrocytes. However, the precise mechanisms underlying how activated microglia induced astrocyte atrophy and loss remain enigmatic. In this study, a depression model induced by chronic social defeat stress (CSDS) is utilized. The results show that CSDS induces significant anxiety- and depression-like behaviors, along with notable astrocyte atrophy and apoptosis, microglial activation, and elevated levels of microglial interleukin-6 (IL-6). Subsequent studies demonstrate that IL-6 released from activated microglia promotes astrocyte apoptosis. Furthermore, the knockdown of the P2X7 receptor (P2X7R) in microglia, which is implicated in the stress response, reduces stress-induced microglial activation, IL-6 release, and astrocyte apoptosis. Direct inhibition of microglia by minocycline corroborates these effects. The selective knockdown of IL-6 in microglia and IL-6 receptors in astrocytes effectively mitigates depression-like behaviors and reduces astrocyte atrophy. This study identifies microglial IL-6 as a key factor that contributes to astrocyte apoptosis and depressive symptoms. Consequently, the IL-6/IL-6R pathway has emerged as a promising target for the treatment of depression.
Collapse
Affiliation(s)
- Shi-Yu Shen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ling-Feng Liang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tian-Le Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zu-Qi Shen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shu-Yuan Yin
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jia-Rui Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wei Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wen-Li Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, 200433, China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, 200433, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, 200433, China
| |
Collapse
|
2
|
Snijders GJLJ, Gigase FAJ. Neuroglia in mood disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:287-302. [PMID: 40148049 DOI: 10.1016/b978-0-443-19102-2.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Multiple lines of evidence indicate that mood disorders, such as major depressive and bipolar disorder, are associated with abnormalities in neuroglial cells. This chapter discusses the existing literature investigating the potential role of astrocytes, oligodendrocytes, and microglia in mood pathology. We will describe evidence from in vivo imaging, postmortem, animal models based on (stress) paradigms that mimic depressive-like behavior, and biomarker studies in blood and cerebrospinal fluid in patients with mood disorders. The effect of medication used in the treatment of mood disorders, such as antidepressants and lithium, on glial function is discussed. Lastly, we highlight the most relevant findings about potential deficiencies in glia-glia crosstalk in mood disorders. Overall, decreased astrocyte and oligodendrocyte density and expression and microglial changes in homeostatic functions have frequently been put forward in MDD pathology. Studies of BD report similar findings to some extent; however, the evidence is less well established. Together, these findings are suggestive of reduced glial cell function leading to potential white matter abnormalities, glutamate dysregulation, disrupted neuronal functioning, and neurotransmission. However, more research is required to better understand the exact mechanisms underlying glial cell contributions to mood disorder development.
Collapse
Affiliation(s)
- Gijsje J L J Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Frederieke A J Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
3
|
Vivi E, Di Benedetto B. Brain stars take the lead during critical periods of early postnatal brain development: relevance of astrocytes in health and mental disorders. Mol Psychiatry 2024; 29:2821-2833. [PMID: 38553540 PMCID: PMC11420093 DOI: 10.1038/s41380-024-02534-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 09/25/2024]
Abstract
In the brain, astrocytes regulate shape and functions of the synaptic and vascular compartments through a variety of released factors and membrane-bound proteins. An imbalanced astrocyte activity can therefore have drastic negative impacts on brain development, leading to the onset of severe pathologies. Clinical and pre-clinical studies show alterations in astrocyte cell number, morphology, molecular makeup and astrocyte-dependent processes in different affected brain regions in neurodevelopmental (ND) and neuropsychiatric (NP) disorders. Astrocytes proliferate, differentiate and mature during the critical period of early postnatal brain development, a time window of elevated glia-dependent regulation of a proper balance between synapse formation/elimination, which is pivotal in refining synaptic connectivity. Therefore, any intrinsic and/or extrinsic factors altering these processes during the critical period may result in an aberrant synaptic remodeling and onset of mental disorders. The peculiar bridging position of astrocytes between synaptic and vascular compartments further allows them to "compute" the brain state and consequently secrete factors in the bloodstream, which may serve as diagnostic biomarkers of distinct healthy or disease conditions. Here, we collect recent advancements regarding astrogenesis and astrocyte-mediated regulation of neuronal network remodeling during early postnatal critical periods of brain development, focusing on synapse elimination. We then propose alternative hypotheses for an involvement of aberrancies in these processes in the onset of ND and NP disorders. In light of the well-known differential prevalence of certain brain disorders between males and females, we also discuss putative sex-dependent influences on these neurodevelopmental events. From a translational perspective, understanding age- and sex-dependent astrocyte-specific molecular and functional changes may help to identify biomarkers of distinct cellular (dys)functions in health and disease, favouring the development of diagnostic tools or the selection of tailored treatment options for male/female patients.
Collapse
Affiliation(s)
- Eugenia Vivi
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Barbara Di Benedetto
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany.
- Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
4
|
An L, Shen J. In vivo magnetic resonance spectroscopy by transverse relaxation encoding with narrowband decoupling. Sci Rep 2023; 13:12211. [PMID: 37500714 PMCID: PMC10374641 DOI: 10.1038/s41598-023-39375-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/25/2023] [Indexed: 07/29/2023] Open
Abstract
Cell pathology in neuropsychiatric disorders has mainly been accessible by analyzing postmortem tissue samples. Although molecular transverse relaxation informs local cellular microenvironment via molecule-environment interactions, precise determination of the transverse relaxation times of molecules with scalar couplings (J), such as glutamate and glutamine, has been difficult using in vivo magnetic resonance spectroscopy (MRS) technologies, whose approach to measuring transverse relaxation has not changed for decades. We introduce an in vivo MRS technique that utilizes frequency-selective editing pulses to achieve homonuclear decoupled chemical shift encoding in each column of the acquired two-dimensional dataset, freeing up the entire row dimension for transverse relaxation encoding with J-refocusing. This results in increased spectral resolution, minimized background signals, and markedly broadened dynamic range for transverse relaxation encoding. The in vivo within-subject coefficients of variation for the transverse relaxation times of glutamate and glutamine, measured using the proposed method in the human brain at 7 T, were found to be approximately 4%. Since glutamate predominantly resides in glutamatergic neurons and glutamine in glia in the brain, this noninvasive technique provides a way to probe cellular pathophysiology in neuropsychiatric disorders for characterizing disease progression and monitoring treatment response in a cell type-specific manner in vivo.
Collapse
Affiliation(s)
- Li An
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room 3D46, 10 Center Drive, MSC 1216, Bethesda, MD, 20892-1216, USA.
| | - Jun Shen
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
An L, Shen J. In Vivo Magnetic Resonance Spectroscopy by J-Locked Chemical Shift Encoding for Determination of Neurochemical Concentration and Transverse Relaxation Time. ARXIV 2023:arXiv:2303.14230v1. [PMID: 37064530 PMCID: PMC10104196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Cell pathology in neuropsychiatric disorders has mainly been accessible by analyzing postmortem tissue samples. Although molecular transverse relaxation informs local cellular microenvironment via molecule-environment interactions, precise determination of the transverse relaxation times of molecules with scalar couplings (J), such as glutamate and glutamine, is difficult using current in vivo magnetic resonance spectroscopy (MRS) technologies, whose approach to measuring transverse relaxation has not changed for decades. We introduce an in vivo MRS technique that achieves chemical shift encoding with selectively locked J-couplings in each column of the acquired two-dimensional dataset, freeing up the entire row dimension for transverse relaxation encoding. This results in increased spectral resolution, minimized background signals, and markedly broadened dynamic range for transverse relaxation encoding. This technique enables determination of the transverse relaxation times of glutamate and glutamine in vivo with unprecedented high precision. Since glutamate predominantly resides in glutamatergic neurons and glutamine in glia in the brain, this noninvasive technique provides a way to probe cellular pathophysiology in neuropsychiatric disorders for characterizing disease progression and monitoring treatment response in a cell type-specific manner in vivo.
Collapse
Affiliation(s)
- Li An
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Jun Shen
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
6
|
Breit S, Mazza E, Poletti S, Benedetti F. White matter integrity and pro-inflammatory cytokines as predictors of antidepressant response in MDD. J Psychiatr Res 2023; 159:22-32. [PMID: 36657311 DOI: 10.1016/j.jpsychires.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 11/12/2022] [Accepted: 12/10/2022] [Indexed: 01/13/2023]
Abstract
Major depressive disorder (MDD) is a multifactorial, serious and heterogeneous mental disorder that can lead to chronic recurrent symptoms, treatment resistance and suicidal behavior. MDD often involves immune dysregulation with high peripheral levels of inflammatory cytokines that might have an influence on the clinical course and treatment response. Moreover, patients with MDD show brain volume changes as well as white matter (WM) alterations that are already existing in the early stage of illness. Mounting evidence suggests that both neuroimaging markers, such as WM integrity and blood markers, such as inflammatory cytokines might serve as predictors of treatment response in MDD. However, the relationship between peripheral inflammation, WM structure and antidepressant response is not yet clearly understood. The aim of the present review is to elucidate the association between inflammation and WM integrity and its impact on the pathophysiology and progression of MDD as well as the role of possible novel biomarkers of treatment response to improve MDD prevention and treatment strategies.
Collapse
Affiliation(s)
- Sigrid Breit
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy; Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland.
| | - Elena Mazza
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Sara Poletti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| |
Collapse
|
7
|
Understanding ayahuasca effects in major depressive disorder treatment through in vitro metabolomics and bioinformatics. Anal Bioanal Chem 2023:10.1007/s00216-023-04556-3. [PMID: 36717401 DOI: 10.1007/s00216-023-04556-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/27/2022] [Accepted: 01/19/2023] [Indexed: 02/01/2023]
Abstract
Emerging insights from metabolomic-based studies of major depression disorder (MDD) are mainly related to biochemical processes such as energy or oxidative stress, in addition to neurotransmission linked to specific metabolite intermediates. Hub metabolites represent nodes in the biochemical network playing a critical role in integrating the information flow in cells between metabolism and signaling pathways. Limited technical-scientific studies have been conducted to understand the effects of ayahuasca (Aya) administration in the metabolism considering MDD molecular context. Therefore, this work aims to investigate an in vitro primary astrocyte model by untargeted metabolomics of two cellular subfractions: secretome and intracellular content after pre-defined Aya treatments, based on DMT concentration. Mass spectrometry (MS)-based metabolomics data revealed significant hub metabolites, which were used to predict biochemical pathway alterations. Branched-chain amino acid (BCAA) metabolism, and vitamin B6 and B3 metabolism were associated to Aya treatment, as "housekeeping" pathways. Dopamine synthesis was overrepresented in the network results when considering the lowest tested DMT concentration (1 µmol L-1). Building reaction networks containing significant and differential metabolites, such as nicotinamide, L-DOPA, and L-leucine, is a useful approach to guide on dose decision and pathway selection in further analytical and molecular studies.
Collapse
|
8
|
Wu Y, Kong L, Yang A, Xin K, Lu Y, Yan X, Liu W, Zhu Y, Guo Y, Jiang X, Zhou Y, Sun Q, Tang Y, Wu F. Gray matter volume reduction in orbitofrontal cortex correlated with plasma glial cell line-derived neurotrophic factor (GDNF) levels within major depressive disorder. Neuroimage Clin 2023; 37:103341. [PMID: 36739789 PMCID: PMC9932451 DOI: 10.1016/j.nicl.2023.103341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) is a severe mental disorder characterized by reduced gray matter volume (GMV). To date, the pathogenesis of MDD remains unclear, but neurotrophic factors play an essential role in the pathophysiological alterations of MDD during disease development. In particular, plasma glial cell line-derived neurotrophic factor (GDNF) has been suggested as a potential biomarker that may be associated with disease activity and neurological progression in MDD. Our study investigated whether plasma GDNF levels in MDD patients and healthy controls (HCs) are correlated with GMV alterations. METHODS We studied 54 MDD patients and 48 HCs. The effect of different diagnoses on whole-brain GMV was investigated using ANOVA (Analysis of Variance). The threshold of significance was p < 0.05, and Gaussian random-field (GRF) correction for error was used. All analyses were controlled for covariates such as ethnicity, handedness, age, and gender that could affect GMV. RESULT Compared with the HC group, the GMV in the MDD group was significantly reduced in the right inferior orbitofrontal cortex (OFC), and plasma GDNF levels were significantly higher in the MDD group than in the HC group. In the right inferior OFC, the GDNF levels were positively correlated with GMV reduction in the MDD group, whereas in the HC group, a negative correlation was observed between GDNF levels and GMV reduction. CONCLUSION Although increased production of GDNF in MDD may help repair neural damage in brain regions associated with brain disease, its repairing effects may be interfered with and hindered by underlying neuroinflammatory processes.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Lingtao Kong
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Anqi Yang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Kaiqi Xin
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Yihui Lu
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Xintong Yan
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Wen Liu
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Yue Zhu
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Yingrui Guo
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Xiaowei Jiang
- Brain Function Research Section, Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Yifang Zhou
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Qikun Sun
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Yanqing Tang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China; Department of Geriatric Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Feng Wu
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
9
|
Liu J, Mo JW, Wang X, An Z, Zhang S, Zhang CY, Yi P, Leong ATL, Ren J, Chen LY, Mo R, Xie Y, Feng Q, Chen W, Gao TM, Wu EX, Feng Y, Cao X. Astrocyte dysfunction drives abnormal resting-state functional connectivity in depression. SCIENCE ADVANCES 2022; 8:eabo2098. [PMID: 36383661 PMCID: PMC9668300 DOI: 10.1126/sciadv.abo2098] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
Major depressive disorder (MDD) is a devastating mental disorder that affects up to 17% of the population worldwide. Although brain-wide network-level abnormalities in MDD patients via resting-state functional magnetic resonance imaging (rsfMRI) exist, the mechanisms underlying these network changes are unknown, despite their immense potential for depression diagnosis and management. Here, we show that the astrocytic calcium-deficient mice, inositol 1,4,5-trisphosphate-type-2 receptor knockout mice (Itpr2-/- mice), display abnormal rsfMRI functional connectivity (rsFC) in depression-related networks, especially decreased rsFC in medial prefrontal cortex (mPFC)-related pathways. We further uncover rsFC decreases in MDD patients highly consistent with those of Itpr2-/- mice, especially in mPFC-related pathways. Optogenetic activation of mPFC astrocytes partially enhances rsFC in depression-related networks in both Itpr2-/- and wild-type mice. Optogenetic activation of the mPFC neurons or mPFC-striatum pathway rescues disrupted rsFC and depressive-like behaviors in Itpr2-/- mice. Our results identify the previously unknown role of astrocyte dysfunction in driving rsFC abnormalities in depression.
Collapse
Affiliation(s)
- Jiaming Liu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Jia-Wen Mo
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xunda Wang
- Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Ziqi An
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Shuangyang Zhang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Can-Yuan Zhang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peiwei Yi
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Alex T. L. Leong
- Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jing Ren
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Liang-Yu Chen
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ran Mo
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuanyao Xie
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Qianjin Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Wufan Chen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Tian-Ming Gao
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ed X. Wu
- Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde, Foshan), Foshan, China
| | - Xiong Cao
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
10
|
Chen Y, Hao C, Chen W, Cheng W, Li P, Shen J, Tong T, Yan S, Huang S, He T, Huang Z, Meng X. Anti-depressant effects of acupuncture: The insights from NLRP3 mediated pyroptosis and inflammation. Neurosci Lett 2022; 785:136787. [PMID: 35820551 DOI: 10.1016/j.neulet.2022.136787] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/13/2022] [Accepted: 07/07/2022] [Indexed: 11/19/2022]
Abstract
The NLRP3-mediated pyroptosis, which could affect inflammation response, plays a key role in the development of depression. Acupuncture has been shown to be an effective treatment for depression. In this study, we aimed to determine whether acupuncture could confer antidepressant activity via decreasing NLRP3-mediated pyroptosis by reducing inflammation. Here, depression model of rats was induced by chronic unpredictable mild stress (CUMS) for 4 weeks. Acupuncture group was subjected to acupuncture at the Shangxing (GV23) and Fengfu (GV16) acupoints for 20 min every other day (a total of 14 times). Fluoxetine group was administered with fluoxetine (2.1 mg/kg with the concentration of 0.21 mg/mL) by oral gavage (1 mL/100 g) once a day for 28 days. Rats' depression-like phenotypes were reflected with behavioral tests and biological detection methods. Results showed that acupuncture significantly improved the depression-like behavior of CUMS rat, suppressed the expressions of NLRP3, ASC, caspase-1, GSDMD, IL-1β, IL-18, HMGB1, IFN-γ, IL-6 and TNF-α in the serum and hippocampus, restored the %area of microglia, astrocytes and neuronal cells in the hippocampus. These indicate that acupuncture can prevent CUMS-induced depression-like behaviors by reducing NLRP3-mediated pyroptosis and inflammation.
Collapse
Affiliation(s)
- Yiping Chen
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, China; Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chongyao Hao
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, China
| | - Wenjie Chen
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenjing Cheng
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Peng Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junliang Shen
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Tao Tong
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, China
| | - Simin Yan
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shuqiong Huang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Tianwei He
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhiyong Huang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xianjun Meng
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China; Shenzhen Research Institute of Xiamen University, Shenzhen, China.
| |
Collapse
|
11
|
Miyata S, Ishino Y, Shimizu S, Tohyama M. Involvement of inflammatory responses in the brain to the onset of major depressive disorder due to stress exposure. Front Aging Neurosci 2022; 14:934346. [PMID: 35936767 PMCID: PMC9354609 DOI: 10.3389/fnagi.2022.934346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
Major depressive disorder (MDD) is a multifactorial disease affected by several environmental factors. Although several potential onset hypotheses have been identified, the molecular mechanisms underlying the pathogenesis of this disorder remain unclear. Several recent studies have suggested that among many environmental factors, inflammation and immune abnormalities in the brain or the peripheral tissues are associated with the onset of MDDs. Furthermore, several stress-related hypotheses have been proposed to explain the onset of MDDs. Thus, inflammation or immune abnormalities can be considered stress responses that occur within the brain or other tissues and are regarded as one of the mechanisms underlying the stress hypothesis of MDDs. Therefore, we introduce several current advances in inflammation studies in the brain that might be related to the pathophysiology of MDD due to stress exposure in this review.
Collapse
Affiliation(s)
- Shingo Miyata
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kindai University, Osaka, Japan
- *Correspondence: Shingo Miyata
| | - Yugo Ishino
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kindai University, Osaka, Japan
| | - Shoko Shimizu
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kindai University, Osaka, Japan
| | - Masaya Tohyama
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kindai University, Osaka, Japan
- Osaka Prefectural Hospital Organization, Osaka, Japan
| |
Collapse
|
12
|
Zhao F, Li B, Yang W, Ge T, Cui R. Brain-immune interaction mechanisms: Implications for cognitive dysfunction in psychiatric disorders. Cell Prolif 2022; 55:e13295. [PMID: 35860850 PMCID: PMC9528770 DOI: 10.1111/cpr.13295] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 04/28/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022] Open
Abstract
Objectives Cognitive dysfunction has been identified as a major symptom of a series of psychiatric disorders. Multidisciplinary studies have shown that cognitive dysfunction is monitored by a two‐way interaction between the neural and immune systems. However, the specific mechanisms of cognitive dysfunction in immune response and brain immune remain unclear. Materials and methods In this review, we summarized the relevant research to uncover our comprehension of the brain–immune interaction mechanisms underlying cognitive decline. Results The pathophysiological mechanisms of brain‐immune interactions in psychiatric‐based cognitive dysfunction involve several specific immune molecules and their associated signaling pathways, impairments in neural and synaptic plasticity, and the potential neuro‐immunological mechanism of stress. Conclusions Therefore, this review may provide a better theoretical basis for integrative therapeutic considerations for psychiatric disorders associated with cognitive dysfunction.
Collapse
Affiliation(s)
- Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Guo J, Qiu T, Wang L, Shi L, Ai M, Xia Z, Peng Z, Zheng A, Li X, Kuang L. Microglia Loss and Astrocyte Activation Cause Dynamic Changes in Hippocampal [18F]DPA-714 Uptake in Mouse Models of Depression. Front Cell Neurosci 2022; 16:802192. [PMID: 35250485 PMCID: PMC8896346 DOI: 10.3389/fncel.2022.802192] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/31/2022] [Indexed: 12/18/2022] Open
Abstract
Major depression is a serious and chronic mental illness. However, its etiology is poorly understood. Although glial cells have been increasingly implicated in the pathogenesis of depression, the specific role of microglia and astrocytes in stress-induced depression remains unclear. Translocator protein (TSPO) has long been considered a marker of neuroinflammation and microglial activation. However, this protein is also present on astrocytes. Thus, it is necessary to explore the relationships between TSPO, microglia, and astrocytes in the context of depression. In this study, C57BL/6J male mice were subjected to chronic unpredictable stress (CUS) for 5 weeks. Subsequently, sucrose preference and tail suspension tests (TSTs) were performed to assess anhedonia and despair in these mice. [18F]DPA-714 positron emission tomography (PET) was adopted to dynamically assess the changes in glial cells before and 2, 4, or 5 weeks after CUS exposure. The numbers of TSPO+ cells, ionized calcium-binding adaptor molecule (Iba)-1+ microglial cells, TSPO+/Iba-1+ cells, glial fibrillary acidic protein (GFAP)+ astrocytes, TSPO+/GFAP+ cells, and TUNEL-stained microglia were quantified using immunofluorescence staining. Real-time PCR was used to evaluate interleukin (IL)-1β, IL-4, and IL-18 expression in the hippocampus. We observed that hippocampal [18F]DPA-714 uptake significantly increased after 2 weeks of CUS. However, the signal significantly decreased after 5 weeks of CUS. CUS significantly reduced the number of Iba-1+, TSPO+, and TSPO+/Iba-1+ cells in the hippocampus, especially in the CA1 and dentate gyrus (DG) subregions. However, this intervention increased the number of GFAP+ astrocytes in the CA2/CA3 subregions of the hippocampus. In addition, microglial apoptosis in the early stage of CUS appeared to be involved in microglia loss. Further, the expression of pro-inflammatory cytokines (IL-1β and IL-18) was significantly decreased after CUS. In contrast, the expression of the anti-inflammatory cytokine IL-4 was significantly increased after 2 weeks of CUS. These results suggested that the CUS-induced dynamic changes in hippocampal [18F]DPA-714 uptake and several cytokines may be due to combined microglial and astrocyte action. These findings provide a theoretical reference for the future clinical applications of TSPO PET.
Collapse
Affiliation(s)
- Jiamei Guo
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tian Qiu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lixia Wang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Shi
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Ai
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhu Xia
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiping Peng
- College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Anhai Zheng
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Li
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Kuang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Li Kuang,
| |
Collapse
|
14
|
Verkhratsky A, Li B, Scuderi C, Parpura V. Principles of Astrogliopathology. ADVANCES IN NEUROBIOLOGY 2021; 26:55-73. [PMID: 34888830 DOI: 10.1007/978-3-030-77375-5_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The role of astrocytes in the nervous system pathology was early on embraced by neuroscientists at end of the nineteenth and the beginning of the twentieth century, only to be pushed aside by neurone-centric dogmas during most of the twentieth century. However, the last decade of the twentieth century and the twenty-first century have brought the astroglial "renaissance", which has put astroglial cells as key players in pathophysiology of most if not all disorders of the nervous system and has regarded astroglia as a fertile ground for therapeutic intervention.Astrocytic contribution to neuropathology can be primary, whereby cell-autonomous changes, such as mutations in gene encoding for glial fibrillary acidic protein, can drive the pathologic progression, in this example, Alexander disease. They can also be secondary, when astrocytes respond to a variety of insults to the nervous tissue. Regardless of their origin, being cell-autonomous or not, changes in astroglia that occur in pathology, that is, astrogliopathology, can be contemporary and arbitrary classified into four forms: (i) reactive astrogliosis, (ii) astrocytic atrophy with loss of function, (iii) pathological remodelling of astrocytes and (iv) astrodegeneration morphologically manifested as clasmatodendrosis. Inevitably, as with any other classification, this classification of astrogliopathology awaits its revision that shall be rooted in new discoveries and concepts.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| | - Baoman Li
- Practical Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
15
|
Wei SZ, Yao XY, Wang CT, Dong AQ, Li D, Zhang YT, Ren C, Zhang JB, Mao CJ, Wang F, Liu CF. Pramipexole regulates depression-like behavior via dopamine D3 receptor in a mouse model of Parkinson's disease. Brain Res Bull 2021; 177:363-372. [PMID: 34699917 DOI: 10.1016/j.brainresbull.2021.10.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 01/02/2023]
Abstract
Depression is one of the strongest predictors of quality of life in patients with Parkinson's disease (PD). Despite the high prevalence of depression, there is no clear guidance for its treatment in PD because the evidence for the efficacy of most antidepressants remains insufficient. Pramipexole, a dopamine agonist, is one of the few drugs that has proven to be clinically useful. However, the underlying mechanisms of antidepressive effects of pramipexole are still unknown. A 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model, dopamine D2 receptor (DRD2) and D3 receptor (DRD3) knockout mice were used in our study. Compared with other dopamine D2-like receptor agonists and madopar, pramipexole improved depression-like behavior and alleviate bradykinesia in an MPTP-induced mouse model of PD. Pramipexole significantly improved depression-like behavior in DRD2-/- mice but not in DRD3-/- mice. These results demonstrate that the antidepressive effect of pramipexole is mediated by DRD3 but not DRD2. Our findings highlight the need to develop novel dopamine agonists specifically targeting DRD3 for the treatment of depression in PD in the future.
Collapse
Affiliation(s)
- Shi-Zhuang Wei
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xiao-Yu Yao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chen-Tao Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - An-Qi Dong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Dan Li
- Department of Neurology, Suqian First Hospital, Suqian, China
| | - Yu-Ting Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chao Ren
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jin-Bao Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China; Department of Neurology, Suqian First Hospital, Suqian, China; Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
16
|
Chen B, Zhang M, Ji M, Gong W, Chen B, Zorec R, Stenovec M, Verkhratsky A, Li B. The Association Between Antidepressant Effect of SSRIs and Astrocytes: Conceptual Overview and Meta-analysis of the Literature. Neurochem Res 2021; 46:2731-2745. [PMID: 33527219 DOI: 10.1007/s11064-020-03225-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/19/2022]
Abstract
Major depressive disorders (MDD) a worldwide psychiatric disease, is yet to be adequately controlled by therapies; while the mechanisms of action of antidepressants are yet to be fully characterised. In the last two decades, an increasing number of studies have demonstrated the role of astrocytes in the pathophysiology and therapy of MDD. Selective serotonin reuptake inhibitors (SSRIs) are the most widely used antidepressants. It is generally acknowledged that SSRIs increase serotonin levels in the central nervous system by inhibiting serotonin transporters, although the SSRIs action is not ideal. The SSRIs antidepressant effect develops with considerable delay; their efficacy is low and frequent relapses are common. Neither cellular nor molecular pharmacological mechanisms of SSRIs are fully characterised; in particular their action on astrocytes remain underappreciated. In this paper we overview potential therapeutic mechanisms of SSRIs associated with astroglia and report the results of meta-analysis of studies dedicated to MDD, SSRIs and astrocytes. In particular, we argue that fluoxetine, the representative SSRI, improves depressive-like behaviours in animals treated with chronic mild stress and reverses depression-associated decrease in astrocytic glial fibrillary acidic protein (GFAP) expression. In addition, fluoxetine upregulates astrocytic mRNA expression of 5-hydroxytriptamin/serotonin2B receptors (5-HT2BR). In summary, we infer that SSRIs exert their anti-depressant effect by regulating several molecular and signalling pathways in astrocytes.
Collapse
Affiliation(s)
- Beina Chen
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, No. 77, Puhe Street, Shenbei District, Shenyang, 110177, People's Republic of China
| | - Manman Zhang
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, No. 77, Puhe Street, Shenbei District, Shenyang, 110177, People's Republic of China
| | - Ming Ji
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, No. 77, Puhe Street, Shenbei District, Shenyang, 110177, People's Republic of China
| | - Wenliang Gong
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, No. 77, Puhe Street, Shenbei District, Shenyang, 110177, People's Republic of China
| | - Binjie Chen
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, No. 77, Puhe Street, Shenbei District, Shenyang, 110177, People's Republic of China
| | - Robert Zorec
- Celica BIOMEDICAL, Tehnološki park 24, 1000, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia
| | - Matjaž Stenovec
- Celica BIOMEDICAL, Tehnološki park 24, 1000, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achucarro Center for Neuroscience, IKERBASQUE, 48011, Bilbao, Spain.
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
| | - Baoman Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, No. 77, Puhe Street, Shenbei District, Shenyang, 110177, People's Republic of China.
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
| |
Collapse
|
17
|
Bonvento G, Bolaños JP. Astrocyte-neuron metabolic cooperation shapes brain activity. Cell Metab 2021; 33:1546-1564. [PMID: 34348099 DOI: 10.1016/j.cmet.2021.07.006] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/11/2021] [Accepted: 07/03/2021] [Indexed: 12/12/2022]
Abstract
The brain has almost no energy reserve, but its activity coordinates organismal function, a burden that requires precise coupling between neurotransmission and energy metabolism. Deciphering how the brain accomplishes this complex task is crucial to understand central facets of human physiology and disease mechanisms. Each type of neural cell displays a peculiar metabolic signature, forcing the intercellular exchange of metabolites that serve as both energy precursors and paracrine signals. The paradigm of this biological feature is the astrocyte-neuron couple, in which the glycolytic metabolism of astrocytes contrasts with the mitochondrial oxidative activity of neurons. Astrocytes generate abundant mitochondrial reactive oxygen species and shuttle to neurons glycolytically derived metabolites, such as L-lactate and L-serine, which sustain energy needs, conserve redox status, and modulate neurotransmitter-receptor activity. Conversely, early disruption of this metabolic cooperation may contribute to the initiation or progression of several neurological diseases, thus requiring innovative therapies to preserve brain energetics.
Collapse
Affiliation(s)
- Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France.
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Institute of Biomedical Research of Salamanca, Salamanca, Spain
| |
Collapse
|
18
|
Nishiyama A, Serwanski DR, Pfeiffer F. Many roles for oligodendrocyte precursor cells in physiology and pathology. Neuropathology 2021; 41:161-173. [PMID: 33913208 DOI: 10.1111/neup.12732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) are a fourth resident glial cell population in the mammalian central nervous system. They are evenly distributed throughout the gray and white matter and continue to proliferate and generate new oligodendrocytes (OLs) throughout life. They were understudied until a few decades ago when immunolabeling for NG2 and platelet-derived growth factor receptor alpha revealed cells that are distinct from mature OLs, astrocytes, neurons, and microglia. In this review, we provide a summary of the known properties of OPCs with some historical background, followed by highlights from recent studies that suggest new roles for OPCs in certain pathological conditions.
Collapse
Affiliation(s)
- Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA.,Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, USA.,The Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - David R Serwanski
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | - Friederike Pfeiffer
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA.,Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
19
|
Zheng H, Bergamino M, Ford BN, Kuplicki R, Yeh FC, Bodurka J, Burrows K, Hunt PW, Teague TK, Irwin MR, Yolken RH, Paulus MP, Savitz J. Replicable association between human cytomegalovirus infection and reduced white matter fractional anisotropy in major depressive disorder. Neuropsychopharmacology 2021; 46:928-938. [PMID: 33500556 PMCID: PMC8115597 DOI: 10.1038/s41386-021-00971-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/23/2020] [Accepted: 01/12/2021] [Indexed: 01/30/2023]
Abstract
Major depressive disorder (MDD) is associated with reductions in white matter microstructural integrity as measured by fractional anisotropy (FA), an index derived from diffusion tensor imaging (DTI). The neurotropic herpesvirus, human cytomegalovirus (HCMV), is a major cause of white matter pathology in immunosuppressed populations but its relationship with FA has never been tested in MDD despite the presence of inflammation and weakened antiviral immunity in a subset of depressed patients. We tested the relationship between FA and HCMV infection in two independent samples consisting of 176 individuals with MDD and 44 healthy controls (HC) (Discovery sample) and 88 participants with MDD and 48 HCs (Replication sample). Equal numbers of HCMV positive (HCMV+) and HCMV negative (HCMV-) groups within each sample were balanced on ten different clinical/demographic variables using propensity score matching. Anti-HCMV IgG antibodies were measured using a solid-phase ELISA. In the Discovery sample, significantly lower FA was observed in the right inferior fronto-occipital fasciculus (IFOF) in HCMV+ participants with MDD compared to HCMV- participants with MDD (cluster size 1316 mm3; pFWE < 0.05, d = -0.58). This association was confirmed in the replication sample by extracting the mean FA from this exact cluster and applying the identical statistical model (p < 0.05, d = -0.45). There was no significant effect of diagnosis or interaction between diagnosis and HCMV in either sample. The effect of chronic HCMV infection on white matter integrity may-in at-risk individuals-contribute to the psychopathology of depression. These findings may provide a novel target of intervention for a subgroup of patients with MDD.
Collapse
Affiliation(s)
- Haixia Zheng
- Laureate Institute for Brain Research, Tulsa, OK, USA.
| | - Maurizio Bergamino
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Bart N Ford
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | | | - Fang-Cheng Yeh
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jerzy Bodurka
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | | | - Peter W Hunt
- Department of Medicine, School of Medicine, The University of California, San Francisco, San Francisco, CA, USA
| | - T Kent Teague
- Department of Surgery, University of Oklahoma School of Community Medicine, Tulsa, OK, USA
- Department of Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK, USA
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Michael R Irwin
- Cousins Center for Psychoneuroimmunology at UCLA, Los Angeles, CA, USA
- Semel Institute for Neuroscience at UCLA, Los Angeles, CA, USA
- David Geffen School of Medicine, Los Angeles, CA, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Martin P Paulus
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK, USA
| |
Collapse
|
20
|
Czéh B, Simon M. Benefits of animal models to understand the pathophysiology of depressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110049. [PMID: 32735913 DOI: 10.1016/j.pnpbp.2020.110049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022]
Abstract
Major depressive disorder (MDD) is a potentially life-threatening mental disorder imposing severe social and economic burden worldwide. Despite the existence of effective antidepressant treatment strategies the exact pathophysiology of the disease is still unknown. Large number of animal models of MDD have been developed over the years, but all of them suffer from significant shortcomings. Despite their limitations these models have been extensively used in academic research and drug development. The aim of this review is to highlight the benefits of animal models of MDD. We focus here on recent experimental data where animal models were used to examine current theories of this complex disease. We argue, that despite their evident imperfections, these models provide invaluable help to understand cellular and molecular mechanisms contributing to the development of MDD. Furthermore, animal models are utilized in research to find clinically useful biomarkers. We discuss recent neuroimaging and microRNA studies since these investigations yielded promising candidates for biomarkers. Finally, we briefly summarize recent progresses in drug development, i.e. the FDA approval of two novel antidepressant drugs: S-ketamine and brexanolone (allopregnanolone). Deeper understanding of the exact molecular and cellular mechanisms of action responsible for the antidepressant efficacy of these rapid acting drugs could aid us to design further compounds with similar effectiveness, but less side effects. Animal studies are likely to provide valuable help in this endeavor.
Collapse
Affiliation(s)
- Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary; Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary.
| | - Maria Simon
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary; Department of Psychiatry and Psychotherapy, Medical School, University of Pécs, Hungary
| |
Collapse
|
21
|
Abstract
In the twentieth century, neuropsychiatric disorders have been perceived solely from a neurone-centric point of view, which considers neurones as the key cellular elements of pathological processes. This dogma has been challenged thanks to the better comprehension of the brain functioning, which, even if far from being complete, has revealed the complexity of interactions that exist between neurones and neuroglia. Glial cells represent a highly heterogeneous population of cells of neural (astroglia and oligodendroglia) and non-neural (microglia) origin populating the central nervous system. The variety of glia reflects the innumerable functions that glial cells perform to support functions of the nervous system. Aberrant execution of glial functions contributes to the development of neuropsychiatric pathologies. Arguably, all types of glial cells are implicated in the neuropathology; however, astrocytes have received particular attention in recent years because of their pleiotropic functions that make them decisive in maintaining cerebral homeostasis. This chapter describes the multiple roles of astrocytes in the healthy central nervous system and discusses the diversity of astroglial responses in neuropsychiatric disorders suggesting that targeting astrocytes may represent an effective therapeutic strategy.
Collapse
Affiliation(s)
- Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy.
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
| |
Collapse
|
22
|
Song R, Shi Y, Li X, Zhu J, Zhang H, Li K, Wang B, Zhang H, Yang Y, Gao L, Zhao Y, Zhang Z. Potential of Antithrombin III as a Biomarker of Antidepressive Effect in Major Depressive Disorder. Front Psychiatry 2021; 12:678384. [PMID: 34777034 PMCID: PMC8580946 DOI: 10.3389/fpsyt.2021.678384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 09/20/2021] [Indexed: 12/20/2022] Open
Abstract
Background: The evaluation of treatment response to antidepressant therapy commonly depends on neuropsychologic assessments, as there are currently no suitable biomarkers. Previous research has identified a panel of increased proteins in patients with major depressive disorder (MDD), including antithrombin III (ATIII), as potential biomarkers of depression. Methods: A total of 90 MDD patients were recruited. Of these, 74 patients received occipital repetitive transcranial magnetic stimulation (rTMS) as individualized, standard, or sham treatment for 5 days, and underwent the complete procedure, including clinical assessments, blood collection, and protein measurement. Results: After treatment, ATIII was significantly decreased in both the individualized and standard groups (both p < 0.001) relative to the sham group. In the individualized group, reduction in ATIII was associated with improvements in several neuropsychological assessments. Furthermore, ATIII at baseline in the standard group and after individualized rTMS showed good performance for evaluating or predicting the response to five-day treatment (AUC = 0.771, 95% CI, 0.571-0.971; AUC = 0.875, 95% CI, 0.714-1.000, respectively) and remission at follow-up (AUC = 0.736, 95% CI, 0.529-0.943; AUC = 0.828, 95% CI, 0.656-1.000, respectively). Lastly, both baseline ATIII and change in ATIII showed good predictive value for the 24-item Hamilton Depression Rating Scale at follow-up (p = 0.024 and 0.023, respectively). Conclusion: Our study revealed a reduction in ATIII after occipital rTMS in MDD patients and a relationship between change in ATIII and therapeutic response. Taken together, these findings provide evidence for the potential of ATIII as a biomarker for the evaluation and prediction of antidepressive effects.
Collapse
Affiliation(s)
- Ruize Song
- Department of Neurology, School of Medicine, Affiliated ZhongDa Hospital, Southeast University, Nanjing, China
| | - Yachen Shi
- Department of Neurology, School of Medicine, Affiliated ZhongDa Hospital, Southeast University, Nanjing, China
| | - Xianrui Li
- Department of Psychology, Xinxiang Medical University, Xinxiang, China
| | - Jianli Zhu
- Department of Psychology, Xinxiang Medical University, Xinxiang, China
| | - Hongxing Zhang
- Department of Psychology, Xinxiang Medical University, Xinxiang, China.,Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Kun Li
- Department of Psychology, Xinxiang Medical University, Xinxiang, China
| | - Bi Wang
- Department of Psychology, Xinxiang Medical University, Xinxiang, China
| | - Haisan Zhang
- Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yongfeng Yang
- Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Lijuan Gao
- Department of Neurology, School of Medicine, Affiliated ZhongDa Hospital, Southeast University, Nanjing, China
| | - Yang Zhao
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhijun Zhang
- Department of Neurology, School of Medicine, Affiliated ZhongDa Hospital, Southeast University, Nanjing, China.,Department of Psychology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
23
|
Alshammari TK. The Ketamine Antidepressant Story: New Insights. Molecules 2020; 25:molecules25235777. [PMID: 33297563 PMCID: PMC7730956 DOI: 10.3390/molecules25235777] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 12/12/2022] Open
Abstract
Ketamine is a versatile agent primarily utilized as a dissociative anesthetic, which acts by blocking the excitatory receptor N-methyl-d-aspartate receptor (NMDA). It functions to inhibit the current of both Na+ and K+ voltage-gated channels, thus preventing serotonin and dopamine reuptake. Studies have indicated that administering a single subanesthetic dose of ketamine relieves depression rapidly and that the effect is sustained. For decades antidepressant agents were based on the monoamine theory. Although ketamine may not be the golden antidepressant, it has opened new avenues toward mechanisms involved in the pathology of treatment-resistant depression and achieving rapid antidepressant effects. Thus, preclinical studies focusing on deciphering the molecular mechanisms involved in the antidepressant action of ketamine will assist in the development of a new antidepressant. This review was conducted to elucidate the emerging pathways that can explain the complex dose-dependent mechanisms achieved by administering ketamine to treat major depressive disorders. Special attention was paid to reviewing the literature on hydroxynorketamines, which are ketamine metabolites that have recently attracted attention in the context of depression.
Collapse
Affiliation(s)
- Tahani K Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2475, Riyadh 11451, Saudi Arabia
| |
Collapse
|
24
|
Antontseva E, Bondar N, Reshetnikov V, Merkulova T. The Effects of Chronic Stress on Brain Myelination in Humans and in Various Rodent Models. Neuroscience 2020; 441:226-238. [DOI: 10.1016/j.neuroscience.2020.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/23/2022]
|
25
|
Adell A. Brain NMDA Receptors in Schizophrenia and Depression. Biomolecules 2020; 10:biom10060947. [PMID: 32585886 PMCID: PMC7355879 DOI: 10.3390/biom10060947] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/21/2022] Open
Abstract
N-methyl-D-aspartate (NMDA) receptor antagonists such as phencyclidine (PCP), dizocilpine (MK-801) and ketamine have long been considered a model of schizophrenia, both in animals and humans. However, ketamine has been recently approved for treatment-resistant depression, although with severe restrictions. Interestingly, the dosage in both conditions is similar, and positive symptoms of schizophrenia appear before antidepressant effects emerge. Here, we describe the temporal mechanisms implicated in schizophrenia-like and antidepressant-like effects of NMDA blockade in rats, and postulate that such effects may indicate that NMDA receptor antagonists induce similar mechanistic effects, and only the basal pre-drug state of the organism delimitates the overall outcome. Hence, blockade of NMDA receptors in depressive-like status can lead to amelioration or remission of symptoms, whereas healthy individuals develop psychotic symptoms and schizophrenia patients show an exacerbation of these symptoms after the administration of NMDA receptor antagonists.
Collapse
Affiliation(s)
- Albert Adell
- Institute of Biomedicine and Biotechnology of Cantabria, IBBTEC (CSIC-University of Cantabria), Calle Albert Einstein 22 (PCTCAN), 39011 Santander, Spain; or
- Biomedical Research Networking Center for Mental Health (CIBERSAM), 39011 Santander, Spain
| |
Collapse
|
26
|
Sawamura D, Narita H, Hashimoto N, Nakagawa S, Hamaguchi H, Fujima N, Kudo K, Shirato H, Tha KK. Microstructural Alterations in Bipolar and Major Depressive Disorders: A Diffusion Kurtosis Imaging Study. J Magn Reson Imaging 2020; 52:1187-1196. [PMID: 32329208 DOI: 10.1002/jmri.27174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Identifying structural and functional abnormalities in bipolar (BD) and major depressive disorders (MDD) is important for understanding biological processes. HYPOTHESIS Diffusion kurtosis imaging (DKI) may be able to detect the brain's microstructural alterations in BD and MDD and any differences between the two. STUDY TYPE Prospective. SUBJECTS In all, 16 BD patients, 19 MDD patients, and 20 age- and gender-matched healthy volunteers. FIELD STRENGTH/SEQUENCE DKI at 3.0T. ASSESSMENT The major DKI indices of the brain were compared voxel-by-voxel among the three groups. Significantly different voxels were tested for correlation with clinical variables (ie, Young Mania Rating Scale [YMRS], 17-item Hamilton Depression Rating Scale [17-HDRS], Montgomery-Åsberg Depression Rating Scale, total disease duration, duration of current episode, and the number of past manic/depressive episodes). The performance of the DKI indices in identifying microstructural alterations was estimated. STATISTICAL TESTS One-way analysis of variance (ANOVA) was used for group comparison of DKI indices. The performance of these indices in detecting microstructural alterations was determined by receiver operating characteristic (ROC) analysis. Pearson's product-moment correlation analyses were used to test the correlations of these indices with clinical variables. RESULTS DKI revealed widespread microstructural alterations across the brain in each disorder (P < 0.05). Some were significantly different between the two disorders. Mean kurtosis (MK) in the gray matter of the right inferior parietal lobe was able to distinguish BD and MDD with an accuracy of 0.906. A strong correlation was revealed between MK in that region and YMRS in BD patients (r = -0.641, corrected P = 0.042) or 17-HDRS in MDD patients (r = -0.613, corrected P = 0.030). There were also strong correlations between a few other DKI indices and disease duration (r = -0.676 or 0.626, corrected P < 0.05). DATA CONCLUSION DKI detected microstructural brain alterations in BD and MDD. Its indices may be useful to distinguish the two disorders or to reflect disease severity and duration. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY STAGE: 3 J. Magn. Reson. Imaging 2020;52:1187-1196.
Collapse
Affiliation(s)
- Daisuke Sawamura
- Department of Biomarker Imaging Science, Graduate School of Medical Science and Engineering, Hokkaido University, Sapporo, Japan.,Department of Functioning and Disability, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Hisashi Narita
- Department of Psychiatry, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoki Hashimoto
- Department of Psychiatry, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shin Nakagawa
- Department of Psychiatry, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Hamaguchi
- Department of Biomarker Imaging Science, Graduate School of Medical Science and Engineering, Hokkaido University, Sapporo, Japan.,Department of Radiological Technology, Hokkaido University Hospital, Sapporo, Japan
| | - Noriyuki Fujima
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan.,Department of Radiology, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kohsuke Kudo
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan.,Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan
| | - Hiroki Shirato
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan.,Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Khin K Tha
- Department of Biomarker Imaging Science, Graduate School of Medical Science and Engineering, Hokkaido University, Sapporo, Japan.,Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan.,Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan.,Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
27
|
Khan AR, Geiger L, Wiborg O, Czéh B. Stress-Induced Morphological, Cellular and Molecular Changes in the Brain-Lessons Learned from the Chronic Mild Stress Model of Depression. Cells 2020; 9:cells9041026. [PMID: 32326205 PMCID: PMC7226496 DOI: 10.3390/cells9041026] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/14/2020] [Accepted: 04/19/2020] [Indexed: 02/07/2023] Open
Abstract
Major depressive disorder (MDD) is a severe illness imposing an increasing social and economic burden worldwide. Numerous rodent models have been developed to investigate the pathophysiology of MDD. One of the best characterized and most widely used models is the chronic mild stress (CMS) model which was developed more than 30 years ago by Paul Willner. More than 2000 published studies used this model, mainly to assess novel compounds with potential antidepressant efficacy. Most of these studies examined the behavioral consequences of stress and concomitant drug intervention. Much fewer studies focused on the CMS-induced neurobiological changes. However, the stress-induced cellular and molecular changes are important as they may serve as potential translational biomarkers and increase our understanding of the pathophysiology of MDD. Here, we summarize current knowledge on the structural and molecular alterations in the brain that have been described using the CMS model. We discuss the latest neuroimaging and postmortem histopathological data as well as molecular changes including recent findings on microRNA levels. Different chronic stress paradigms occasionally deliver dissimilar findings, but the available experimental data provide convincing evidence that the CMS model has a high translational value. Future studies examining the neurobiological changes in the CMS model in combination with clinically effective antidepressant drug intervention will likely deliver further valuable information on the pathophysiology of MDD.
Collapse
Affiliation(s)
- Ahmad Raza Khan
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute (SGPGI) Campus, Lucknow-226017, U.P, India;
| | - Lili Geiger
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary;
- Department of Laboratory Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Ove Wiborg
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark;
| | - Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary;
- Department of Laboratory Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
- Correspondence:
| |
Collapse
|
28
|
Obuchowicz E, Bielecka-Wajdman A, Zieliński M, Machnik G, Gołyszny M, Ludyga T. Imipramine and Venlafaxine Differentially Affect Primary Glial Cultures of Prenatally Stressed Rats. Front Pharmacol 2020; 10:1687. [PMID: 32076407 PMCID: PMC7006619 DOI: 10.3389/fphar.2019.01687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/24/2019] [Indexed: 12/25/2022] Open
Abstract
Here, we examine the effects of prenatal administration of two antidepressants—imipramine (IMI) and venlafaxine (VEN)—on morphology and activity of a primary glial culture. Microglia are targeted by antidepressants used for antenatal depression and are important regulators of central nervous system development. In this study, female Wistar rats were assigned to one of four groups: a control group that received water ad libitum (1), and groups that received additionally once daily either water (2), IMI (10 mg/kg) (3), or VEN (20 mg/kg) (4) by oral gavage from gestation day 7 to 22. Oral gavage administration induced prenatal stress. Cell cultures were obtained from the brains of 1-day-old pups. Prenatal stress caused a disturbance of sensorimotor function in pups. Prenatal stress also produced alterations in the glial cultures, specifically, an increased percentage of microglia in the mixed glial cultures and an increased percentage of dead cells. Moreover, increased levels of IL1-β, TNF-α, NO, and an increased expression of CX3CR1 mRNA were found in microglia. However, the ratio of Bax/Bcl2 mRNA was reduced. Prenatal stress increased the vulnerability of microglia to lipopolysaccharide (LPS). The mixed glial culture derived from pups exposed to IMI showed greater morphological changes and the highest percentage of microglia. Microglia were characterized by the largest increase in the production of pro-inflammatory cytokines and NO, and the greatest reduction in the expression of CX3CR1 mRNA. Exposure to IMI reduced the effects of LPS on IL-1β production and Bax/Bcl2 mRNA, and exacerbated the effects of LPS on CX3CR1 mRNA expression. Prenatal administration of VEN induced protective effects on microglia, as measured by all studied parameters. Taken together, our data suggest that, by disturbing microglia function, exposure to even mild forms of chronic prenatal stress may predispose individuals to psychiatric or neurodevelopmental disorders. These data also indicate that chronic mild stress sensitizes microglia to immune challenges, which may lead to enhanced neuronal damage in the embryonic brain. The observed detrimental effects of IMI on microglial activity under conditions of prenatal stress may help to explain the teratogenic effects of IMI reported in the literature.
Collapse
Affiliation(s)
- Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Anna Bielecka-Wajdman
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Michał Zieliński
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Grzegorz Machnik
- Department of Internal Medicine and Clinical Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Tomasz Ludyga
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
29
|
Microglial activation contributes to depressive-like behavior in dopamine D3 receptor knockout mice. Brain Behav Immun 2020; 83:226-238. [PMID: 31626970 DOI: 10.1016/j.bbi.2019.10.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/14/2019] [Accepted: 10/14/2019] [Indexed: 12/29/2022] Open
Abstract
We previously demonstrated that the dopamine D3 receptor (D3R) inhibitor, NGB2904, increases susceptibility to depressive-like symptoms, elevates pro-inflammatory cytokine expression, and alters brain-derived neurotrophic factor (BDNF) levels in mesolimbic dopaminergic regions, including the medial prefrontal cortex (mPFC), nucleus accumbens (NAc), and ventral tegmental area (VTA) in mice. The mechanisms by which D3R inhibition affects neuroinflammation and onset of depression remain unclear. Here, using D3R-knockout (D3RKO) and congenic wild-type C56BL/6 (WT) mice, we demonstrated that D3RKO mice displayed depressive-like behaviors, increased tumornecrosisfactor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 levels, and altered BDNF expression in selected mesolimbic dopaminergic regions. D3R expression was localized to astrocytes or microglia in the mPFC, NAc, and VTA in WT mice. D3RKO mice exhibited a large number of Iba1-labelled microglia in the absence of glial fibrillary acidic protein (GFAP)-labelled astrocytes in mesolimbic dopaminergic brain areas. Inhibition or ablation of microglia by minocycline (25 mg/kg and 50 mg/kg) or PLX3397 (40 mg/kg) treatment ameliorated depressive-like symptoms, alterations in pro-inflammatory cytokine levels, and BDNF expression in the indicated brain regions in D3RKO mice. Minocycline therapy alleviated the increase in synaptic density in the NAc in D3RKO mice. These findings suggest that microglial activation in selected mesolimbic reward regions affects depressive-like behaviors induced by D3R deficiency.
Collapse
|
30
|
|
31
|
Binzer S, McKay KA, Brenner P, Hillert J, Manouchehrinia A. Disability worsening among persons with multiple sclerosis and depression: A Swedish cohort study. Neurology 2019; 93:e2216-e2223. [PMID: 31704791 PMCID: PMC6937491 DOI: 10.1212/wnl.0000000000008617] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/20/2019] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Depression is common in multiple sclerosis (MS), but its impact on disability worsening has not yet been determined. We explored the risk of disability worsening associated with depression in a nationwide longitudinal cohort. METHODS This retrospective cohort study used linked data from 3 Swedish nationwide registries: the MS Register, National Patient Register, and Prescribed Drug Register. Two incident cohorts were developed: cohort 1 included all registered cases of MS in the MS Registry (2001-2014) with depression defined as ≥1 ICD-10 code for depression; and cohort 2 comprised all cases of MS in the MS Registry (2005-2014) with depression defined as ≥1 prescription filled for an antidepressant. Cox regression models were used to compare the risk of reaching sustained disability milestone scores of 3.0, 4.0, and 6.0 on the Expanded Disability Status Scale (EDSS) between persons with MS with and without depression. RESULTS Cohort 1 included 5,875 cases; 502 (8.5%) had depression. Cohort 2 had 3,817 cases; 1,289 (33.8%) were prescribed an antidepressant. Persons with depression were at a significantly higher risk of reaching sustained EDSS scores of 3.0, 4.0, and 6.0, with hazard ratios of 1.50 (95% confidence interval [CI] 1.20-1.87), 1.79 (95% CI 1.40-2.29), and 1.89 (95% CI 1.38-2.57), respectively. A similar increased risk among persons exposed to antidepressants was observed, with hazard ratios of 1.37 (95% CI 1.18-1.60), 1.93 (95% CI 1.61-2.31), and 1.86 (95% CI 1.45-2.40) for sustained EDSS scores of 3.0, 4.0, and 6.0, respectively. CONCLUSION Persons with MS and comorbid depression had a significantly increased risk of disability worsening. This finding highlights the need for early recognition and appropriate treatment of depression in persons with MS.
Collapse
Affiliation(s)
- Stefanie Binzer
- From the Department of Clinical Neuroscience (S.B., K.A.M., J.H., A.M.), Department of Medicine Solna (P.B.), and Karolinska Neuroimmunology & Multiple Sclerosis Centre and Centre for Molecular Medicine (A.M.) Karolinska Institutet, Stockholm, Sweden, Odense University Hospital (S.B.), Department of Neurology, Denmark; and Karolinska University Hospital (J.H.), Stockholm, Sweden.
| | - Kyla A McKay
- From the Department of Clinical Neuroscience (S.B., K.A.M., J.H., A.M.), Department of Medicine Solna (P.B.), and Karolinska Neuroimmunology & Multiple Sclerosis Centre and Centre for Molecular Medicine (A.M.) Karolinska Institutet, Stockholm, Sweden, Odense University Hospital (S.B.), Department of Neurology, Denmark; and Karolinska University Hospital (J.H.), Stockholm, Sweden
| | - Philip Brenner
- From the Department of Clinical Neuroscience (S.B., K.A.M., J.H., A.M.), Department of Medicine Solna (P.B.), and Karolinska Neuroimmunology & Multiple Sclerosis Centre and Centre for Molecular Medicine (A.M.) Karolinska Institutet, Stockholm, Sweden, Odense University Hospital (S.B.), Department of Neurology, Denmark; and Karolinska University Hospital (J.H.), Stockholm, Sweden
| | - Jan Hillert
- From the Department of Clinical Neuroscience (S.B., K.A.M., J.H., A.M.), Department of Medicine Solna (P.B.), and Karolinska Neuroimmunology & Multiple Sclerosis Centre and Centre for Molecular Medicine (A.M.) Karolinska Institutet, Stockholm, Sweden, Odense University Hospital (S.B.), Department of Neurology, Denmark; and Karolinska University Hospital (J.H.), Stockholm, Sweden
| | - Ali Manouchehrinia
- From the Department of Clinical Neuroscience (S.B., K.A.M., J.H., A.M.), Department of Medicine Solna (P.B.), and Karolinska Neuroimmunology & Multiple Sclerosis Centre and Centre for Molecular Medicine (A.M.) Karolinska Institutet, Stockholm, Sweden, Odense University Hospital (S.B.), Department of Neurology, Denmark; and Karolinska University Hospital (J.H.), Stockholm, Sweden
| |
Collapse
|
32
|
Al-Haddad BJS, Oler E, Armistead B, Elsayed NA, Weinberger DR, Bernier R, Burd I, Kapur R, Jacobsson B, Wang C, Mysorekar I, Rajagopal L, Adams Waldorf KM. The fetal origins of mental illness. Am J Obstet Gynecol 2019; 221:549-562. [PMID: 31207234 PMCID: PMC6889013 DOI: 10.1016/j.ajog.2019.06.013] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022]
Abstract
The impact of infections and inflammation during pregnancy on the developing fetal brain remains incompletely defined, with important clinical and research gaps. Although the classic infectious TORCH pathogens (ie, Toxoplasma gondii, rubella virus, cytomegalovirus [CMV], herpes simplex virus) are known to be directly teratogenic, emerging evidence suggests that these infections represent the most extreme end of a much larger spectrum of injury. We present the accumulating evidence that prenatal exposure to a wide variety of viral and bacterial infections-or simply inflammation-may subtly alter fetal brain development, leading to neuropsychiatric consequences for the child later in life. The link between influenza infections in pregnant women and an increased risk for development of schizophrenia in their children was first described more than 30 years ago. Since then, evidence suggests that a range of infections during pregnancy may also increase risk for autism spectrum disorder and depression in the child. Subsequent studies in animal models demonstrated that both pregnancy infections and inflammation can result in direct injury to neurons and neural progenitor cells or indirect injury through activation of microglia and astrocytes, which can trigger cytokine production and oxidative stress. Infectious exposures can also alter placental serotonin production, which can perturb neurotransmitter signaling in the developing brain. Clinically, detection of these subtle injuries to the fetal brain is difficult. As the neuropsychiatric impact of perinatal infections or inflammation may not be known for decades after birth, our construct for defining teratogenic infections in pregnancy (eg, TORCH) based on congenital anomalies is insufficient to capture the full adverse impact on the child. We discuss the clinical implications of this body of evidence and how we might place greater emphasis on prevention of prenatal infections. For example, increasing uptake of the seasonal influenza vaccine is a key strategy to reduce perinatal infections and the risk for fetal brain injury. An important research gap exists in understanding how antibiotic therapy during pregnancy affects the fetal inflammatory load and how to avoid inflammation-mediated injury to the fetal brain. In summary, we discuss the current evidence and mechanisms linking infections and inflammation with the increased lifelong risk of neuropsychiatric disorders in the child, and how we might improve prenatal care to protect the fetal brain.
Collapse
Affiliation(s)
| | - Elizabeth Oler
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA
| | - Blair Armistead
- Department of Global Health, University of Washington Seattle, WA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Nada A Elsayed
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Departments of Psychiatry, Neurology, Neuroscience, and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD
| | - Raphael Bernier
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Raj Kapur
- Department of Pediatrics, University of Washington, Seattle Children's Hospital, Seattle, WA
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Genetics and Bioinformatics, Domain of Health Data and Digitalization, Institute of Public Health, Oslo, Norway
| | - Caihong Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO
| | - Indira Mysorekar
- Departments of Obstetrics and Gynecology and Pathology and Immunology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO
| | - Lakshmi Rajagopal
- Center for Innate Immunity and Immune Disease, Department of Pediatrics, University of Washington, Seattle, WA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Kristina M Adams Waldorf
- Department of Obstetrics & Gynecology and Global Health, Center for Innate Immunity and Immune Disease, Center for Emerging and Reemerging Infectious Diseases, University of Washington, Seattle, WA; Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
33
|
Westfall S, Pasinetti GM. The Gut Microbiota Links Dietary Polyphenols With Management of Psychiatric Mood Disorders. Front Neurosci 2019; 13:1196. [PMID: 31749681 PMCID: PMC6848798 DOI: 10.3389/fnins.2019.01196] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022] Open
Abstract
The pathophysiology of depression is multifactorial yet generally aggravated by stress and its associated physiological consequences. To effectively treat these diverse risk factors, a broad acting strategy is required and is has been suggested that gut-brain-axis signaling may play a pinnacle role in promoting resilience to several of these stress-induced changes including pathogenic load, inflammation, HPA-axis activation, oxidative stress and neurotransmitter imbalances. The gut microbiota also manages the bioaccessibility of phenolic metabolites from dietary polyphenols whose multiple beneficial properties have known therapeutic efficacy against depression. Although several potential therapeutic mechanisms of dietary polyphenols toward establishing cognitive resilience to neuropsychiatric disorders have been established, only a handful of studies have systematically identified how the interaction of the gut microbiota with dietary polyphenols can synergistically alleviate the biological signatures of depression. The current review investigates several of these potential mechanisms and how synbiotics, that combine probiotics with dietary polyphenols, may provide a novel therapeutic strategy for depression. In particular, synbiotics have the potential to alleviate neuroinflammation by modulating microglial and inflammasome activation, reduce oxidative stress and balance serotonin metabolism therefore simultaneously targeting several of the major pathological risk factors of depression. Overall, synbiotics may act as a novel therapeutic paradigm for neuropsychiatric disorders and further understanding the fundamental mechanisms of gut-brain-axis signaling will allow full utilization of the gut microbiota's as a therapeutic tool.
Collapse
Affiliation(s)
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
34
|
Evidence of decreased gap junction coupling between astrocytes and oligodendrocytes in the anterior cingulate cortex of depressed suicides. Neuropsychopharmacology 2019; 44:2099-2111. [PMID: 31374562 PMCID: PMC6897926 DOI: 10.1038/s41386-019-0471-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
Glial dysfunction is a major pathophysiological feature of mood disorders. While altered astrocyte (AS) and oligodendrocyte-lineage (OL) functions have been associated with depression, the crosstalk between these glial cell types has never been assessed in that context. AS are potent regulators of myelination, in part through gap junction (GJ) channels formed by the heterotypic coupling of AS-specific (Cx30 and Cx43) and OL-specific (Cx32 and Cx47) connexins. This study therefore aimed at addressing the integrity of AS/OL coupling in the anterior cingulate cortex (ACC) of depressed suicides. Using immunofluorescence and confocal imaging, we characterized the distribution of Cx30 and mapped its expression onto OL somas, myelinated axons, and brain vasculature in postmortem brain samples from depressed suicides (N = 48) and matched controls (N = 23). Differential gene expression of key components of the GJ nexus was also screened through RNA-sequencing previously generated by our group, and validated by quantitative real-time PCR. We show that Cx30 expression localized onto OL cells and myelinated fibers is decreased in deep cortical layers of the ACC in male-depressed suicides. This effect was associated with decreased expression of OL-specific connexins, as well as the downregulation of major connexin-interacting proteins essential for the scaffolding, trafficking, and function of GJs. These results provide a first evidence of impaired AS/OL GJ-mediated communication in the ACC of individuals with mood disorders. These changes in glial coupling are likely to have significant impact on brain function, and may contribute to the altered OL function previously reported in this brain region.
Collapse
|
35
|
Gultyaeva VV, Zinchenko MI, Uryumtsev DY, Krivoschekov SG, Aftanas LI. [Exercise for depression treatment. Physiological mechanisms]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:112-119. [PMID: 31464298 DOI: 10.17116/jnevro2019119071112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This literature review considers meta-analyzes, systematic reviews and original research over the last decade addressing a comprehensive analysis of the antidepressant effect of targeted physical exercise and physical activity in general. Exercise is a promising non-pharmacological treatment for depression, showing effects that are comparable or may even exceed other first-line treatments of depression. The article introduces modern ideas about the mechanisms of depression and mechanisms of exercise effects on depression manifestations. The structures of the central nervous system, changing with the effective exercise-based treatment of depression, are indicated. Physical activity stimulates the secretion of growth factors, maintenance of angio-, synapto-, and neurogenesis. The regulation of antioxidant protection of neuronal mitochondria, a decrease in pro-inflammatory reactions and stress reactivity are also observed in response to regular exercise. Physical activity has a multimodal effect that stimulates biochemical pathways and restores neuronal structures disturbed in depression.
Collapse
Affiliation(s)
- V V Gultyaeva
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - M I Zinchenko
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - D Y Uryumtsev
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - S G Krivoschekov
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - L I Aftanas
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| |
Collapse
|
36
|
Enache D, Pariante CM, Mondelli V. Markers of central inflammation in major depressive disorder: A systematic review and meta-analysis of studies examining cerebrospinal fluid, positron emission tomography and post-mortem brain tissue. Brain Behav Immun 2019; 81:24-40. [PMID: 31195092 DOI: 10.1016/j.bbi.2019.06.015] [Citation(s) in RCA: 372] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/04/2019] [Accepted: 06/09/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Increased peripheral inflammation has been consistently reported in patients with major depressive disorder (MDD). However, only few studies have explored markers of central (brain) inflammation in patients with MDD. The aim of this study is to systematically review in vivo and post-mortem markers of central inflammation, including studies examining cerebrospinal fluid (CSF), positron emission tomography, and post-mortem brain tissues in subjects suffering with MDD compared with controls. METHODS PubMed and Medline databases were searched up to December 2018. We included studies measuring cerebrospinal fluid (CSF) cytokines and chemokines, positron emission tomography (PET) studies; and post-mortem studies measuring cytokines, chemokines and cell-specific markers of microglia and astrocytes, all in MDD. A meta-analysis was performed only for CSF and PET studies, as studies on post-mortem markers of inflammation had different cell-specific markers and analysed different brain regions. RESULTS A total of 69 studies met the inclusion criteria. CSF levels of IL-6 and TNF-α were higher in patients with MDD compared with controls (standardised mean difference SMD 0.37, 95%CI: 0.17-0.57 and SMD 0.58, 95%CI 0.26-0.90, respectively). CSF levels of IL-6 were increased in suicide attempters regardless of their psychiatric diagnosis. Translocator protein, a PET marker of central inflammation, was elevated in the anterior cingulate cortex and temporal cortex of patients with MDD compared with controls (SMD 0.78, 95%CI: 0.41-1.16 and SMD 0.52, 95%CI: 0.19-0.85 respectively). Abnormalities in CSF and PET inflammatory markers were not correlated with those in peripheral blood. In post-mortem studies, two studies found increased markers of microglia in MDD brains, while four studies found no MDD related changes. Of the studies investigating expression of cell-specific marker for astrocytes, thirteen studies reported a decreased expression of astrocytes specific markers, two studies reported increased expression of astrocytes specific markers, and eleven studies did not detect any difference. Four out of six studies reported decreased markers of oligodendrocytes in the prefrontal cortex. Post-mortem brain levels of tumor necrosis alpha (TNF-α) were also found increased in MDD. CONCLUSIONS Our review suggests the presence of an increase in IL-6 and TNF-alpha levels in CSF and brain parenchyma, in the context of a possible increased microglia activity and reduction of astrocytes and oligodendrocytes markers in MDD. The reduced number of astrocytes may lead to compromised integrity of blood brain barrier with increased monocyte recruitment and infiltration, which is partly supported by post-mortem studies and by PET studies showing an increased TSPO expression in MDD.
Collapse
Affiliation(s)
- Daniela Enache
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, UK; Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden.
| | - Carmine M Pariante
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, UK; National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK.
| | - Valeria Mondelli
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, UK; National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK.
| |
Collapse
|
37
|
Verkhratsky A, Rodrigues JJ, Pivoriunas A, Zorec R, Semyanov A. Astroglial atrophy in Alzheimer’s disease. Pflugers Arch 2019; 471:1247-1261. [DOI: 10.1007/s00424-019-02310-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/23/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
|
38
|
Saffari R, Grotefeld K, Kravchenko M, Zhang M, Zhang W. Calretinin +-neurons-mediated GABAergic inhibition in mouse prefrontal cortex. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109658. [PMID: 31145926 DOI: 10.1016/j.pnpbp.2019.109658] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 10/26/2022]
Abstract
The prefrontal cortex (PFC) is a center for executive and cognitive functions. Although many studies have been carried out to elucidate the role of different subtypes of GABAergic neurons in other brain areas, their functional relevance in PFC is still not fully understood. Calretinin+-GABAergic neurons are heterogeneous in their morphology and intrinsic properties. Previous studies showed an involvement of CR+-GABAergic neurons in the disinhibition of the other GABAergic neurons in neocortex and hippocampus. Furthermore, the loss of CR+- and PV+-interneurons in human brain has been linked to the vulnerability of the interneurons and to the overall increase in the network excitability associated with mental diseases. In the present study, the intensity of CR+-neuropil was higher in layer II/III, whereas the intensity of PV+-neuropil was higher in deeper layers within the PFC. In addition, pronounced CR expression was detected in layer II and III of prelimbic and infralimbic cortex whereas they were less abundant in anterior cingulate cortex and motor cortex 2. Our results showed that bipolar CR+- neurons in layer V not only feedback inhibited multipolar CR+- and other interneurons in layer II/III, but the majority of bipolar CR+-neurons in layer II/III also provide long-range forward-inhibition to pyramidal neurons in deeper layers of PFC. Thus, given the importance of the neuronal network of PFC in central control of emotion and cognition and in the pathology of mental diseases, CR+-GABAergic neuron-mediated feed-forward and -backward modulation within PFC would differentially modulate the downstream limbic activity and subsequently shape the cognitive and emotional behavior.
Collapse
Affiliation(s)
- Roja Saffari
- Laboratory of molecular psychiatry, Department of psychiatry, University of Münster, Germany
| | - Kirsten Grotefeld
- Laboratory of molecular psychiatry, Department of psychiatry, University of Münster, Germany
| | - Mykola Kravchenko
- Laboratory of molecular psychiatry, Department of psychiatry, University of Münster, Germany
| | - Mingyue Zhang
- Laboratory of molecular psychiatry, Department of psychiatry, University of Münster, Germany
| | - Weiqi Zhang
- Laboratory of molecular psychiatry, Department of psychiatry, University of Münster, Germany.
| |
Collapse
|
39
|
Steardo L, de Filippis R, Carbone EA, Segura-Garcia C, Verkhratsky A, De Fazio P. Sleep Disturbance in Bipolar Disorder: Neuroglia and Circadian Rhythms. Front Psychiatry 2019; 10:501. [PMID: 31379620 PMCID: PMC6656854 DOI: 10.3389/fpsyt.2019.00501] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
The worldwide prevalence of sleep disorders is approximately 50%, with an even higher occurrence in a psychiatric population. Bipolar disorder (BD) is a severe mental illness characterized by shifts in mood and activity. The BD syndrome also involves heterogeneous symptomatology, including cognitive dysfunctions and impairments of the autonomic nervous system. Sleep abnormalities are frequently associated with BD and are often a good predictor of a mood swing. Preservation of stable sleep-wake cycles is therefore a key to the maintenance of stability in BD, indicating the crucial role of circadian rhythms in this syndrome. The symptom most widespread in BD is insomnia, followed by excessive daytime sleepiness, nightmares, difficulty falling asleep or maintaining sleep, poor sleep quality, sleep talking, sleep walking, and obstructive sleep apnea. Alterations in the structure or duration of sleep are reported in all phases of BD. Understanding the role of neuroglia in BD and in various aspects of sleep is in nascent state. Contributions of the different types of glial cells to BD and sleep abnormalities are discussed in this paper.
Collapse
Affiliation(s)
- Luca Steardo
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Renato de Filippis
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Elvira Anna Carbone
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Cristina Segura-Garcia
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Achucarro Center for Neuroscience, IKERBASQUE, Bilbao, Spain
| | - Pasquale De Fazio
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| |
Collapse
|
40
|
Arteaga-Henríquez G, Simon MS, Burger B, Weidinger E, Wijkhuijs A, Arolt V, Birkenhager TK, Musil R, Müller N, Drexhage HA. Low-Grade Inflammation as a Predictor of Antidepressant and Anti-Inflammatory Therapy Response in MDD Patients: A Systematic Review of the Literature in Combination With an Analysis of Experimental Data Collected in the EU-MOODINFLAME Consortium. Front Psychiatry 2019; 10:458. [PMID: 31354538 PMCID: PMC6630191 DOI: 10.3389/fpsyt.2019.00458] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 06/11/2019] [Indexed: 01/17/2023] Open
Abstract
Low-grade inflammation plays a role not only in the pathogenesis of major depressive disorder (MDD) but probably also in the poor responsiveness to regular antidepressants. There are also indications that anti-inflammatory agents improve the outcomes of antidepressants. Aim: To study whether the presence of low-grade inflammation predicts the outcome of antidepressants, anti-inflammatory agents, or combinations thereof. Methods: We carried out a systematic review of the literature on the prediction capability of the serum levels of inflammatory compounds and/or the inflammatory state of circulating leukocytes for the outcome of antidepressant/anti-inflammatory treatment in MDD. We compared outcomes of the review with original data (collected in two limited trials carried out in the EU project MOODINFLAME) on the prediction capability of the inflammatory state of monocytes (as measured by inflammatory gene expression) for the outcome of venlafaxine, imipramine, or sertraline treatment, the latter with and without celecoxib added. Results: Collectively, the literature and original data showed that: 1) raised serum levels of pro-inflammatory compounds (in particular of CRP/IL-6) characterize an inflammatory form of MDD with poor responsiveness to predominately serotonergic agents, but a better responsiveness to antidepressant regimens with a) (add-on) noradrenergic, dopaminergic, or glutamatergic action or b) (add-on) anti-inflammatory agents such as infliximab, minocycline, or eicosapentaenoic acid, showing-next to anti-inflammatory-dopaminergic or lipid corrective action; 2) these successful anti-inflammatory (add-on) agents, when used in patients with low serum levels of CRP/IL-6, decreased response rates in comparison to placebo. Add-on aspirin, in contrast, improved responsiveness in such "non-inflammatory" patients; 3) patients with increased inflammatory gene expression in circulating leukocytes had a poor responsiveness to serotonergic/noradrenergic agents. Conclusions: The presence of inflammation in patients with MDD heralds a poor outcome of first-line antidepressant therapies. Immediate step-ups to dopaminergic or glutamatergic regimens or to (add-on) anti-inflammatory agents are most likely indicated. However, at present, insufficient data exist to design protocols with reliable inflammation parameter cutoff points to guide such therapies, the more since detrimental outcomes are possible of anti-inflammatory agents in "non-inflamed" patients.
Collapse
Affiliation(s)
- Gara Arteaga-Henríquez
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian-University, Munich, Germany.,Department of Immunology, Erasmus Medical Center, Rotterdam, Netherlands.,Psychiatry, Mental Health and Addictions Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Maria S Simon
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian-University, Munich, Germany
| | | | - Elif Weidinger
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian-University, Munich, Germany
| | | | - Volker Arolt
- Department of Psychiatry and Psychotherapy, University Hospital of Muenster, Muenster, Germany
| | - Tom K Birkenhager
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, Netherlands
| | - Richard Musil
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian-University, Munich, Germany
| | - Norbert Müller
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian-University, Munich, Germany.,Marion von Tessin Memory-Center, Munich, Germany
| | - Hemmo A Drexhage
- Department of Immunology, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
41
|
Verkhratsky A, Ho MS, Vardjan N, Zorec R, Parpura V. General Pathophysiology of Astroglia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1175:149-179. [PMID: 31583588 PMCID: PMC7188602 DOI: 10.1007/978-981-13-9913-8_7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Astroglial cells are involved in most if not in all pathologies of the brain. These cells can change the morpho-functional properties in response to pathology or innate changes of these cells can lead to pathologies. Overall pathological changes in astroglia are complex and diverse and often vary with different disease stages. We classify astrogliopathologies into reactive astrogliosis, astrodegeneration with astroglial atrophy and loss of function, and pathological remodelling of astrocytes. Such changes can occur in neurological, neurodevelopmental, metabolic and psychiatric disorders as well as in infection and toxic insults. Mutation in astrocyte-specific genes leads to specific pathologies, such as Alexander disease, which is a leukodystrophy. We discuss changes in astroglia in the pathological context and identify some molecular entities underlying pathology. These entities within astroglia may repent targets for novel therapeutic intervention in the management of brain pathologies.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.
| | - Margaret S Ho
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
- Celica BIOMEDICAL, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
- Celica BIOMEDICAL, Ljubljana, Slovenia
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
42
|
Zebrafish models of epigenetic regulation of CNS functions. Brain Res Bull 2018; 142:344-351. [DOI: 10.1016/j.brainresbull.2018.08.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/22/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022]
|
43
|
Pál B. Involvement of extrasynaptic glutamate in physiological and pathophysiological changes of neuronal excitability. Cell Mol Life Sci 2018; 75:2917-2949. [PMID: 29766217 PMCID: PMC11105518 DOI: 10.1007/s00018-018-2837-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Glutamate is the most abundant neurotransmitter of the central nervous system, as the majority of neurons use glutamate as neurotransmitter. It is also well known that this neurotransmitter is not restricted to synaptic clefts, but found in the extrasynaptic regions as ambient glutamate. Extrasynaptic glutamate originates from spillover of synaptic release, as well as from astrocytes and microglia. Its concentration is magnitudes lower than in the synaptic cleft, but receptors responding to it have higher affinity for it. Extrasynaptic glutamate receptors can be found in neuronal somatodendritic location, on astroglia, oligodendrocytes or microglia. Activation of them leads to changes of neuronal excitability with different amplitude and kinetics. Extrasynaptic glutamate is taken up by neurons and astrocytes mostly via EAAT transporters, and astrocytes, in turn metabolize it to glutamine. Extrasynaptic glutamate is involved in several physiological phenomena of the central nervous system. It regulates neuronal excitability and synaptic strength by involving astroglia; contributing to learning and memory formation, neurosecretory and neuromodulatory mechanisms, as well as sleep homeostasis.The extrasynaptic glutamatergic system is affected in several brain pathologies related to excitotoxicity, neurodegeneration or neuroinflammation. Being present in dementias, neurodegenerative and neuropsychiatric diseases or tumor invasion in a seemingly uniform way, the system possibly provides a common component of their pathogenesis. Although parts of the system are extensively discussed by several recent reviews, in this review I attempt to summarize physiological actions of the extrasynaptic glutamate on neuronal excitability and provide a brief insight to its pathology for basic understanding of the topic.
Collapse
Affiliation(s)
- Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Nagyerdei krt 98, Debrecen, 4012, Hungary.
| |
Collapse
|