1
|
Smith JR, Hopkins CE, Xiong J, Luccarelli J, Shultz E, Vandekar S. Use of ECT in Autism Spectrum Disorder and/or Intellectual Disability: A Single Site Retrospective Analysis. J Autism Dev Disord 2024; 54:963-982. [PMID: 36528758 PMCID: PMC10276173 DOI: 10.1007/s10803-022-05868-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Autism spectrum disorder (ASD) and intellectual disability (ID) are heterogenous and prevalent conditions which may occur in isolation or as a co-morbidity. Psychiatric co-morbidity is common with limited treatment options. Preliminary research into electroconvulsive therapy (ECT) for these conditions has been encouraging. Thus, further research in this patient population is warranted. We conducted a 10-year retrospective review of the electronic medical record and identified intellectually capable individuals with ASD (IC-ASD), and those with ASD+ID or ID who received at least three ECT treatments. 32 patients were identified of which 30 (94%) experienced positive clinical response, defined as a clinical global impression-improvement (CGI-I) score of 3 or less. The average retrospective CGI-I score across all groups was 1.97, and results of a t-test performed on CGI-I scores indicated improvement across all groups [t = - 16.54, df = 31, p < 0.001, 95% CI = (1.72, 2.22)]. No significant adverse events were identified based on clinical documentation. Our findings further support previous ECT research in this patient population.
Collapse
Affiliation(s)
- Joshua R Smith
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center at Village of Vanderbilt, 1500 21st Avenue South, Suite 2200, Nashville, TN, 37212, USA.
- Vanderbilt Kennedy Center, Vanderbilt University, 110 Magnolia Circle, Nashville, TN, 37203, USA.
| | - Corey E Hopkins
- Division of General Psychiatry, Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Ave South, Nashville, TN, 37212, USA
| | - Jiangmei Xiong
- Department of Biostatistics, Vanderbilt University, 2424 West End Avenue, Suite 1100, Nashville, TN, 37203, USA
| | - James Luccarelli
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
- Department of Psychiatry, Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
| | - Elizabeth Shultz
- Division of General Psychiatry, Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Ave South, Nashville, TN, 37212, USA
| | - Simon Vandekar
- Department of Biostatistics, Vanderbilt University, 2424 West End Avenue, Suite 1100, Nashville, TN, 37203, USA
| |
Collapse
|
2
|
Yu X, Chen K, Ma Y, Bai T, Zhu S, Cai D, Zhang X, Wang K, Tian Y, Wang J. Molecular basis underlying changes of brain entropy and functional connectivity in major depressive disorders after electroconvulsive therapy. CNS Neurosci Ther 2024; 30:e14690. [PMID: 38529527 PMCID: PMC10964037 DOI: 10.1111/cns.14690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/03/2024] [Accepted: 02/23/2024] [Indexed: 03/27/2024] Open
Abstract
INTRODUCTION Electroconvulsive therapy (ECT) is widely used for treatment-resistant depression. However, it is unclear whether/how ECT can be targeted to affect brain regions and circuits in the brain to dynamically regulate mood and cognition. METHODS This study used brain entropy (BEN) to measure the irregular levels of brain systems in 46 major depressive disorder (MDD) patients before and after ECT treatment. Functional connectivity (FC) was further adopted to reveal changes of functional couplings. Moreover, transcriptomic and neurotransmitter receptor data were used to reveal genetic and molecular basis of the changes of BEN and functional connectivities. RESULTS Compared to pretreatment, the BEN in the posterior cerebellar lobe (PCL) significantly decreased and FC between the PCL and the right temporal pole (TP) significantly increased in MDD patients after treatment. Moreover, we found that these changes of BEN and FC were closely associated with genes' expression profiles involved in MAPK signaling pathway, GABAergic synapse, and dopaminergic synapse and were significantly correlated with the receptor/transporter density of 5-HT, norepinephrine, glutamate, etc. CONCLUSION: These findings suggest that loops in the cerebellum and TP are crucial for ECT regulation of mood and cognition, which provides new evidence for the antidepressant effects of ECT and the potential molecular mechanism leading to cognitive impairment.
Collapse
Affiliation(s)
- Xiaohui Yu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Kexuan Chen
- Medical SchoolKunming University of Science and TechnologyKunmingChina
| | - Yingzi Ma
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Tongjian Bai
- Department of NeurologyThe First Hospital of Anhui Medical UniversityHefeiChina
| | - Shunli Zhu
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Defang Cai
- The Second People's Hospital of YuxiThe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Xing Zhang
- The Second People's Hospital of YuxiThe Affiliated Hospital of Kunming University of Science and TechnologyYuxiChina
| | - Kai Wang
- Department of NeurologyThe First Hospital of Anhui Medical UniversityHefeiChina
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric DisordersHefeiChina
- School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental HealthHefeiChina
- Anhui Province Clinical Research Center for Neurological DiseaseHefeiChina
| | - Yanghua Tian
- Department of NeurologyThe First Hospital of Anhui Medical UniversityHefeiChina
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric DisordersHefeiChina
- School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental HealthHefeiChina
- Anhui Province Clinical Research Center for Neurological DiseaseHefeiChina
- Institute of Artificial IntelligenceHefei Comprehensive National Science CenterHefeiChina
| | - Jiaojian Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| |
Collapse
|
3
|
Yi C, Verkhratsky A, Niu J. Pathological potential of oligodendrocyte precursor cells: terra incognita. Trends Neurosci 2023:S0166-2236(23)00103-0. [PMID: 37183154 DOI: 10.1016/j.tins.2023.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/12/2023] [Accepted: 04/13/2023] [Indexed: 05/16/2023]
Abstract
Adult oligodendrocyte precursor cells (aOPCs), transformed from fetal OPCs, are idiosyncratic neuroglia of the central nervous system (CNS) that are distinct in many ways from other glial cells. OPCs have been classically studied in the context of their remyelinating capacity. Recent studies, however, revealed that aOPCs not only contribute to post-lesional remyelination but also play diverse crucial roles in multiple neurological diseases. In this review we briefly present the physiology of aOPCs and summarize current knowledge of the beneficial and detrimental roles of aOPCs in different CNS diseases. We discuss unique features of aOPC death, reactivity, and changes during senescence, as well as aOPC interactions with other glial cells and pathological remodeling during disease. Finally, we outline future perspectives for the study of aOPCs in brain pathologies which may instigate the development of aOPC-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Department of Pathology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China; Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen 518107, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China.
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PL, UK; Achucarro Centre for Neuroscience, Basque Foundation for Science (IKERBASQUE), Bilbao 48011, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
| | - Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
4
|
Kritzer MD, Peterchev AV, Camprodon JA. Electroconvulsive Therapy: Mechanisms of Action, Clinical Considerations, and Future Directions. Harv Rev Psychiatry 2023; 31:101-113. [PMID: 37171471 PMCID: PMC10198476 DOI: 10.1097/hrp.0000000000000365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
LEARNING OBJECTIVES • Outline and discuss the fundamental physiologic, cellular, and molecular mechanisms of ECT to devise strategies to optimize therapeutic outcomes• Summarize the overview of ECT, its efficacy in treating depression, the known effects on cognition, evidence of mechanisms, and future directions. ABSTRACT Electroconvulsive therapy (ECT) is the most effective treatment for a variety of psychiatric illnesses, including treatment-resistant depression, bipolar depression, mania, catatonia, and clozapine-resistant schizophrenia. ECT is a medical and psychiatric procedure whereby electrical current is delivered to the brain under general anesthesia to induce a generalized seizure. ECT has evolved a great deal since the 1930s. Though it has been optimized for safety and to reduce adverse effects on cognition, issues persist. There is a need to understand fundamental physiologic, cellular, and molecular mechanisms of ECT to devise strategies to optimize therapeutic outcomes. Clinical trials that set out to adjust parameters, electrode placement, adjunctive medications, and patient selection are critical steps towards the goal of improving outcomes with ECT. This narrative review provides an overview of ECT, its efficacy in treating depression, its known effects on cognition, evidence of its mechanisms, and future directions.
Collapse
Affiliation(s)
- Michael D Kritzer
- From the Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA (Drs. Kritzer, Camprodon); Department of Psychiatry and Behavioral Sciences, Department of Biomedical Engineering, Department of Electrical and Computer Engineering, Department of Neurosurgery, Duke University, Durham, NC (Dr. Peterchev)
| | | | | |
Collapse
|
5
|
Issler O, van der Zee YY, Ramakrishnan A, Xia S, Zinsmaier AK, Tan C, Li W, Browne CJ, Walker DM, Salery M, Torres-Berrío A, Futamura R, Duffy JE, Labonte B, Girgenti MJ, Tamminga CA, Dupree JL, Dong Y, Murrough JW, Shen L, Nestler EJ. The long noncoding RNA FEDORA is a cell type- and sex-specific regulator of depression. SCIENCE ADVANCES 2022; 8:eabn9494. [PMID: 36449610 PMCID: PMC9710883 DOI: 10.1126/sciadv.abn9494] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 10/12/2022] [Indexed: 05/31/2023]
Abstract
Women suffer from depression at twice the rate of men, but the underlying molecular mechanisms are poorly understood. Here, we identify marked baseline sex differences in the expression of long noncoding RNAs (lncRNAs), a class of regulatory transcripts, in human postmortem brain tissue that are profoundly lost in depression. One such human lncRNA, RP11-298D21.1 (which we termed FEDORA), is enriched in oligodendrocytes and neurons and up-regulated in the prefrontal cortex (PFC) of depressed females only. We found that virally expressing FEDORA selectively either in neurons or in oligodendrocytes of PFC promoted depression-like behavioral abnormalities in female mice only, changes associated with cell type-specific regulation of synaptic properties, myelin thickness, and gene expression. We also found that blood FEDORA levels have diagnostic implications for depressed women and are associated with clinical response to ketamine. These findings demonstrate the important role played by lncRNAs, and FEDORA in particular, in shaping the sex-specific landscape of the brain and contributing to sex differences in depression.
Collapse
Affiliation(s)
- Orna Issler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yentl Y. van der Zee
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sunhui Xia
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Chunfeng Tan
- Department of Psychiatry, UT Southwestern, Dallas, TX, USA
| | - Wei Li
- Department of Psychiatry, UT Southwestern, Dallas, TX, USA
| | - Caleb J. Browne
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deena M. Walker
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marine Salery
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Angélica Torres-Berrío
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rita Futamura
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia E. Duffy
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benoit Labonte
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew J. Girgenti
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Jeffrey L. Dupree
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - James W. Murrough
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
6
|
Jones KL, Zhou M, Jhaveri DJ. Dissecting the role of adult hippocampal neurogenesis towards resilience versus susceptibility to stress-related mood disorders. NPJ SCIENCE OF LEARNING 2022; 7:16. [PMID: 35842419 PMCID: PMC9288448 DOI: 10.1038/s41539-022-00133-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/01/2022] [Indexed: 05/13/2023]
Abstract
Adult hippocampal neurogenesis in the developmental process of generating and integrating new neurons in the hippocampus during adulthood and is a unique form of structural plasticity with enormous potential to modulate neural circuit function and behaviour. Dysregulation of this process is strongly linked to stress-related neuropsychiatric conditions such as anxiety and depression, and efforts have focused on unravelling the contribution of adult-born neurons in regulating stress response and recovery. Chronic stress has been shown to impair this process, whereas treatment with clinical antidepressants was found to enhance the production of new neurons in the hippocampus. However, the precise role of adult hippocampal neurogenesis in mediating the behavioural response to chronic stress is not clear and whether these adult-born neurons buffer or increase susceptibility to stress-induced mood-related maladaptation remains one of the controversial issues. In this review, we appraise evidence probing the causal role of adult hippocampal neurogenesis in the regulation of emotional behaviour in rodents. We find that the relationship between adult-born hippocampal neurons and stress-related mood disorders is not linear, and that simple subtraction or addition of these neurons alone is not sufficient to lead to anxiety/depression or have antidepressant-like effects. We propose that future studies examining how stress affects unique properties of adult-born neurons, such as the excitability and the pattern of connectivity during their critical period of maturation will provide a deeper understanding of the mechanisms by which these neurons contribute to functional outcomes in stress-related mood disorders.
Collapse
Affiliation(s)
- Katherine L Jones
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Mei Zhou
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia.
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
7
|
An X, Wang Y. Electroconvulsive shock increases neurotrophy and neurogenesis: Time course and treatment session effects. Psychiatry Res 2022; 309:114390. [PMID: 35063747 DOI: 10.1016/j.psychres.2022.114390] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 01/03/2022] [Accepted: 01/08/2022] [Indexed: 10/19/2022]
Abstract
Increasing evidence suggests that hippocampal neurotrophy may be related to the development of major depressive disorders. Neurogenesis, which can be regulated by neurotrophic factors, is also involved in antidepressant efficacy. This paper reviewed literature on neurotrophic signaling and cell proliferation after electroconvulsive shock (ECS) treatment. All articles were from PubMed, Web of Science, and Scopus databases between 2000 and 2020. The keywords used in the literature search are: "ECS," "ECT," "electroconvulsive seizure," "electroconvulsive shock," "electroconvulsive therapy," "neurotrophic factor," "nerve growth factor," "neurotrophins," "neurogenesis," and "cell proliferation." Eighty-two articles were included in the final analysis. It was shown that compared with acute ECS, repeated ECS increased neurotrophin expression in more brain regions at higher levels and was maintained for a longer time. Similarly, ECS increased cell proliferation in a dose- and time-dependent manner. The increase in cell proliferation was positively correlated with the amount of ECS administered and the newly born cells survived for a long time. The effects of ECS in inducing increases in neurotrophin levels and neurogenesis may contribute to brain function changes and antidepressant effects. Future research may focus on optimal sessions of ECT treatment to obtain the best therapeutic effect.
Collapse
Affiliation(s)
- Xianli An
- School of Educational Science, Yangzhou University, Yangzhou, JiangSu Province, China.
| | - Yaqing Wang
- School of Educational Science, Yangzhou University, Yangzhou, JiangSu Province, China
| |
Collapse
|
8
|
Machado-Santos AR, Loureiro-Campos E, Patrício P, Araújo B, Alves ND, Mateus-Pinheiro A, Correia JS, Morais M, Bessa JM, Sousa N, Rodrigues AJ, Oliveira JF, Pinto L. Beyond New Neurons in the Adult Hippocampus: Imipramine Acts as a Pro-Astrogliogenic Factor and Rescues Cognitive Impairments Induced by Stress Exposure. Cells 2022; 11:cells11030390. [PMID: 35159199 PMCID: PMC8834148 DOI: 10.3390/cells11030390] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Depression is a prevalent, socially burdensome disease. Different studies have demonstrated the important role of astrocytes in the pathophysiology of depression as modulators of neurotransmission and neurovascular coupling. This is evidenced by astrocyte impairments observed in brains of depressed patients and the appearance of depressive-like behaviors upon astrocytic dysfunctions in animal models. However, little is known about the importance of de novo generated astrocytes in the mammalian brain and in particular its possible involvement in the precipitation of depression and in the therapeutic actions of current antidepressants (ADs). Therefore, we studied the modulation of astrocytes and adult astrogliogenesis in the hippocampal dentate gyrus (DG) of rats exposed to an unpredictable chronic mild stress (uCMS) protocol, untreated and treated for two weeks with antidepressants—fluoxetine and imipramine. Our results show that adult astrogliogenesis in the DG is modulated by stress and imipramine. This study reveals that distinct classes of ADs impact differently in the astrogliogenic process, showing different cellular mechanisms relevant to the recovery from behavioral deficits induced by chronic stress exposure. As such, in addition to those resident, the newborn astrocytes in the hippocampal DG might also be promising therapeutic targets for future therapies in the neuropsychiatric field.
Collapse
Affiliation(s)
- Ana R Machado-Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Eduardo Loureiro-Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Patrício
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bruna Araújo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Dinis Alves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António Mateus-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Sofia Correia
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mónica Morais
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João M Bessa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João Filipe Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- IPCA-EST-2Ai, Polytechnic Institute of Cávado and Ave, Applied Artificial Intelligence Laboratory, Campus of IPCA, 4750-810 Barcelos, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
9
|
Koizumi S. Glial Purinergic Signals and Psychiatric Disorders. Front Cell Neurosci 2022; 15:822614. [PMID: 35069121 PMCID: PMC8766327 DOI: 10.3389/fncel.2021.822614] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 12/08/2021] [Indexed: 12/30/2022] Open
Abstract
Emotion-related neural networks are regulated in part by the activity of glial cells, and glial dysfunction can be directly related to emotional diseases such as depression. Here, we discuss three different therapeutic strategies involving astrocytes that are effective for treating depression. First, the antidepressant, fluoxetine, acts on astrocytes and increases exocytosis of ATP. This has therapeutic effects via brain-derived neurotrophic factor-dependent mechanisms. Second, electroconvulsive therapy is a well-known treatment for drug-resistant depression. Electroconvulsive therapy releases ATP from astrocytes to induce leukemia inhibitory factors and fibroblast growth factor 2, which leads to antidepressive actions. Finally, sleep deprivation therapy is well-known to cause antidepressive effects. Sleep deprivation also increases release of ATP, whose metabolite, adenosine, has antidepressive effects. These independent treatments share the same mechanism, i.e., ATP release from astrocytes, indicating an essential role of glial purinergic signals in the pathogenesis of depression.
Collapse
Affiliation(s)
- Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- GLIA Center, University of Yamanashi, Yamanashi, Japan
- *Correspondence: Schuichi Koizumi
| |
Collapse
|
10
|
Martins-Macedo J, Salgado AJ, Gomes ED, Pinto L. Adult brain cytogenesis in the context of mood disorders: From neurogenesis to the emergent role of gliogenesis. Neurosci Biobehav Rev 2021; 131:411-428. [PMID: 34555383 DOI: 10.1016/j.neubiorev.2021.09.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/06/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022]
Abstract
Psychiatric disorders severely impact patients' lives. Motivational, cognitive and emotional deficits are the most common symptoms observed in these patients and no effective treatment is still available, either due to the adverse side effects or the low rate of efficacy of currently available drugs. Neurogenesis recovery has been one important focus in the treatment of psychiatric disorders, which undeniably contributes to the therapeutic action of antidepressants. However, glial plasticity is emerging as a new strategy to explore the deficits observed in mood disorders and the efficacy of therapeutic interventions. Thus, it is crucial to understand the mechanisms behind glio- and neurogenesis to better define treatments and preventive therapies, once adult cytogenesis is of pivotal importance to cognitive and emotional components of behavior, both in healthy and pathological contexts, including in psychiatric disorders. Here, we review the concepts and history of neuro- and gliogenesis, providing as well a reflection on the functional importance of cytogenesis in the context of disease.
Collapse
Affiliation(s)
- Joana Martins-Macedo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
11
|
Goldfarb S, Fainstein N, Ganz T, Vershkov D, Lachish M, Ben-Hur T. Electric neurostimulation regulates microglial activation via retinoic acid receptor α signaling. Brain Behav Immun 2021; 96:40-53. [PMID: 33989746 DOI: 10.1016/j.bbi.2021.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/20/2021] [Accepted: 05/09/2021] [Indexed: 12/14/2022] Open
Abstract
Brain stimulation by electroconvulsive therapy is effective in neuropsychiatric disorders by unknown mechanisms. Microglial toxicity plays key role in neuropsychiatric, neuroinflammatory and degenerative diseases. We examined the mechanism by which electroconvulsive seizures (ECS) regulates microglial phenotype and response to stimuli. Microglial responses were examined by morphological analysis, Iba1 and cytokine expression. ECS did not affect resting microglial phenotype or morphology but regulated their activation by Lipopolysaccharide stimulation. Microglia were isolated after ECS or sham sessions in naïve mice for transcriptome analysis. RNA sequencing identified 141 differentially expressed genes. ECS modulated multiple immune-associated gene families and attenuated neurotoxicity-associated gene expression. Blood brain barrier was examined by injecting Biocytin-TMR tracer. There was no breakdown of the BBB, nor increase in gene-signature of peripheral monocytes, suggesting that ECS effect is mainly on resident microglia. Unbiased analysis of regulatory sequences identified the induction of microglial retinoic acid receptor α (RARα) gene expression and a putative common RARα-binding motif in multiple ECS-upregulated genes. The effects of AM580, a selective RARα agonist on microglial response to LPS was examined in vitro. AM580 prevented LPS-induced cytokine expression and reactive oxygen species production. Chronic murine experimental autoimmune encephalomyelitis (EAE) was utilized to confirm the role RARα signaling as mediator of ECS-induced transcriptional pathway in regulating microglial toxicity. Continuous intracerebroventricular delivery of AM580 attenuated effectively EAE severity. In conclusion, ECS regulates CNS innate immune system responses by activating microglial retinoic acid receptor α pathway, signifying a novel therapeutic approach for chronic neuroinflammatory, neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Smadar Goldfarb
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Dan Vershkov
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel; The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
12
|
Early life adversity targets the transcriptional signature of hippocampal NG2+ glia and affects voltage gated sodium (Na v) channels properties. Neurobiol Stress 2021; 15:100338. [PMID: 34095364 PMCID: PMC8164094 DOI: 10.1016/j.ynstr.2021.100338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/27/2022] Open
Abstract
The precise mechanisms underlying the detrimental effects of early life adversity (ELA) on adult mental health remain still elusive. To date, most studies have exclusively targeted neuronal populations and not considered neuron-glia crosstalk as a crucially important element for the integrity of stress-related brain function. Here, we have investigated the impact of ELA, in the form of a limited bedding and nesting material (LBN) paradigm, on a glial subpopulation with unique properties in brain homeostasis, the NG2+ cells. First, we have established a link between maternal behavior, activation of the offspring's stress response and heterogeneity in the outcome to LBN manipulation. We further showed that LBN targets the hippocampal NG2+ transcriptome with glucocorticoids being an important mediator of the LBN-induced molecular changes. LBN altered the NG2+ transcriptome and these transcriptional effects were correlated with glucocorticoids levels. The functional relevance of one LBN-induced candidate gene, Scn7a, could be confirmed by an increase in the density of voltage-gated sodium (Nav) channel activated currents in hippocampal NG2+ cells. Scn7a remained upregulated until adulthood in LBN animals, which displayed impaired cognitive performance. Considering that Nav channels are important for NG2+ cell-to-neuron communication, our findings provide novel insights into the disruption of this process in LBN mice.
Collapse
|
13
|
Breton JM, Barraza M, Hu KY, Frias SJ, Long KL, Kaufer D. Juvenile exposure to acute traumatic stress leads to long-lasting alterations in grey matter myelination in adult female but not male rats. Neurobiol Stress 2021; 14:100319. [PMID: 33937444 PMCID: PMC8079662 DOI: 10.1016/j.ynstr.2021.100319] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/21/2021] [Accepted: 03/15/2021] [Indexed: 12/02/2022] Open
Abstract
Stress early in life can have a major impact on brain development, and there is increasing evidence that childhood stress confers vulnerability for later developing psychiatric disorders. In particular, during peri-adolescence, brain regions crucial for emotional regulation, such as the prefrontal cortex (PFC), amygdala (AMY) and hippocampus (HPC), are still developing and are highly sensitive to stress. Changes in myelin levels have been implicated in mental illnesses and stress effects on myelin and oligodendrocytes (OLs) are beginning to be explored as a novel and underappreciated mechanism underlying psychopathologies. Yet there is little research on the effects of acute stress on myelin during peri-adolescence, and even less work exploring sex-differences. Here, we used a rodent model to test the hypothesis that exposure to acute traumatic stress as a juvenile would induce changes in OLs and myelin content across limbic brain regions. Male and female juvenile rats underwent 3 h of restraint stress with exposure to a predator odor on postnatal day (p) 28. Acute stress induced a physiological response, increasing corticosterone release and reducing weight gain in stress-exposed animals. Brain sections containing the PFC, AMY and HPC were taken either in adolescence (p40), or in adulthood (p95) and stained for markers of OLs and myelin. We found that acute stress induced sex-specific changes in grey matter (GM) myelination and OLs in both the short- and long-term. Exposure to a single stressor as a juvenile increased GM myelin content in the AMY and HPC in p40 males, compared to the respective control group. At p40, corticosterone release during stress exposure was also positively correlated with GM myelin content in the AMY of male rats. Single exposure to juvenile stress also led to long-term effects exclusively in female rats. Compared to controls, stress-exposed females showed reduced GM myelin content in all three brain regions. Acute stress exposure decreased PFC and HPC OL density in p40 females, perhaps contributing towards this observed long-term decrease in myelin content. Overall, our findings suggest that the juvenile brain is vulnerable to exposure to a brief severe stressor. Exposure to a single short traumatic event during peri-adolescence produces long-lasting changes in GM myelin content in the adult brain of female, but not male, rats. These findings highlight myelin plasticity as a potential contributor to sex-specific sensitivity to perturbation during a critical window of development.
Collapse
Affiliation(s)
- Jocelyn M. Breton
- University of California, Berkeley, Helen Wills Neuroscience Institute, United States
| | - Matthew Barraza
- University of California, Berkeley, Molecular and Cellular Biology, United States
| | - Kelsey Y. Hu
- University of California, Berkeley, Molecular and Cellular Biology, United States
| | - Samantha Joy Frias
- University of California, Berkeley, Molecular and Cellular Biology, United States
| | - Kimberly L.P. Long
- University of California, Berkeley, Helen Wills Neuroscience Institute, United States
| | - Daniela Kaufer
- University of California, Berkeley, Helen Wills Neuroscience Institute, United States
- University of California, Berkeley, Integrative Biology, United States
- Canadian Institute for Advanced Research, Toronto, ON, M5G1M1, Canada
| |
Collapse
|
14
|
The effect of electroconvulsive therapy on neuroinflammation, behavior and amyloid plaques in the 5xFAD mouse model of Alzheimer's disease. Sci Rep 2021; 11:4910. [PMID: 33649346 PMCID: PMC7921388 DOI: 10.1038/s41598-021-83998-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
Microglial cells are affected in Alzheimer’s disease (AD) and interact with amyloid-beta (Aβ) plaques. Apart from memory loss, depression is common in patients with AD. Electroconvulsive therapy (ECT) is an anti-depressive treatment that may stimulate microglia, induce neuroinflammation and alter the levels of soluble Aβ, but the effects of ECT on microglia and Aβ aggregation in AD are not known. We investigated the short- and long-term effects of ECT on neuroinflammation and Aβ accumulation. 5xFAD mice received either electroconvulsive stimulation (ECS n = 26) or sham treatment (n = 25) for 3 weeks. Microglia and Aβ were analyzed in samples collected 24 h, 5 weeks, or 9 weeks after the last treatment. Aβ plaques and microglia were quantified using immunohistochemistry. The concentration of soluble Aβ and cytokines was quantified using ELISA and levels of Aβ aggregates were measured with Western Blot. Microglial phagocytosis of Aβ in the hippocampus was evaluated by flow cytometry in Methoxy-X04 injected mice 24 h following the last ECS treatment. Y-maze and Elevated plus maze were performed to study behavior after 5 weeks. We could not detect any significant short- or long-term effects of ECS on Aβ pathology or neuroinflammation, but ECS reduced abnormal behavior in the Elevated Plus maze.
Collapse
|
15
|
Long KLP, Breton JM, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis I: Effects across the Lifespan. Biomolecules 2021; 11:biom11020283. [PMID: 33672939 PMCID: PMC7918364 DOI: 10.3390/biom11020283] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
The brain’s capacity to respond to changing environments via hormonal signaling is critical to fine-tuned function. An emerging body of literature highlights a role for myelin plasticity as a prominent type of experience-dependent plasticity in the adult brain. Myelin plasticity is driven by oligodendrocytes (OLs) and their precursor cells (OPCs). OPC differentiation regulates the trajectory of myelin production throughout development, and importantly, OPCs maintain the ability to proliferate and generate new OLs throughout adulthood. The process of oligodendrogenesis, the creation of new OLs, can be dramatically influenced during early development and in adulthood by internal and environmental conditions such as hormones. Here, we review the current literature describing hormonal regulation of oligodendrogenesis within physiological conditions, focusing on several classes of hormones: steroid, peptide, and thyroid hormones. We discuss hormonal regulation at each stage of oligodendrogenesis and describe mechanisms of action, where known. Overall, the majority of hormones enhance oligodendrogenesis, increasing OPC differentiation and inducing maturation and myelin production in OLs. The mechanisms underlying these processes vary for each hormone but may ultimately converge upon common signaling pathways, mediated by specific receptors expressed across the OL lineage. However, not all of the mechanisms have been fully elucidated, and here, we note the remaining gaps in the literature, including the complex interactions between hormonal systems and with the immune system. In the companion manuscript in this issue, we discuss the implications of hormonal regulation of oligodendrogenesis for neurological and psychiatric disorders characterized by white matter loss. Ultimately, a better understanding of the fundamental mechanisms of hormonal regulation of oligodendrogenesis across the entire lifespan, especially in vivo, will progress both basic and translational research.
Collapse
Affiliation(s)
- Kimberly L. P. Long
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
- Correspondence:
| | - Jocelyn M. Breton
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
| | - Matthew K. Barraza
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 94720, USA;
| | - Olga S. Perloff
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA 94143, USA;
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
16
|
Goldfarb S, Fainstein N, Ben-Hur T. Electroconvulsive stimulation attenuates chronic neuroinflammation. JCI Insight 2020; 5:137028. [PMID: 32780728 PMCID: PMC7526446 DOI: 10.1172/jci.insight.137028] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
Electroconvulsive therapy is highly effective in resistant depression by unknown mechanisms. Microglial toxicity was suggested to mediate depression and plays key roles in neuroinflammatory and degenerative diseases, where there is critical shortage in therapies. We examined the effects of electroconvulsive seizures (ECS) on chronic neuroinflammation and microglial neurotoxicity. Electric brain stimulation inducing full tonic-clonic seizures during chronic relapsing-progressive experimental autoimmune encephalomyelitis (EAE) reduced spinal immune cell infiltration, reduced myelin and axonal loss, and prevented clinical deterioration. Using the transfer EAE model, we examined the effect of ECS on systemic immune response in donor mice versus ECS effect on CNS innate immune activity in recipient mice. ECS did not affect encephalitogenicity of systemic T cells, but it targeted the CNS directly to inhibit T cell-induced neuroinflammation. In vivo and ex vivo assays indicated that ECS suppressed microglial neurotoxicity by reducing inducible NOS expression, nitric oxide, and reactive oxygen species (ROS) production, and by reducing CNS oxidative stress. Microglia from ECS-treated EAE mice expressed less T cell stimulatory and chemoattractant factors. Our findings indicate that electroconvulsive therapy targets the CNS innate immune system to reduce neuroinflammation by attenuating microglial neurotoxicity. These findings signify a potentially novel therapeutic approach for chronic neuroinflammatory, neuropsychiatric, and neurodegenerative diseases.
Collapse
|
17
|
Quantitative characterization of proliferative cells subpopulations in the hilus of the hippocampus of adult Wistar rats: an integrative study. J Mol Histol 2020; 51:437-453. [PMID: 32653982 DOI: 10.1007/s10735-020-09895-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023]
Abstract
The hilus plays an important role modulating the excitability of the hippocampal dentate gyrus (DG). It also harbors proliferative cells whose proliferation rate is modified during pathological events. However, the characterization of these cells, in terms of cellular identity, lineage, and fate, as well as the morphology and proportion of each cell subpopulation has been poorly studied. Therefore, a deeper investigation of hilar proliferative cells might expand the knowledge not only in the physiology, but in the pathophysiological processes related to the hippocampus too. The aim of this work was to perform an integrative study characterizing the identity of proliferative cells populations harbored in the hilus, along with morphology and proportion. In addition, this study provides comparative evidence of the subgranular zone (SGZ) of the DG. Quantified cells included proliferative, neural precursor, Type 1, oligodendrocyte progenitor (OPCs), neural progenitor (NPCs), and proliferative mature astrocytes in the hilus and SGZ of Wistar adult rats. Our results showed that 84% of the hilar proliferative cells correspond to neural precursor cells, OPCs and NPCs being the most abundant at 54 and 45%, respectively, unlike the SGZ, where OPCs represent only 11%. Proliferative mature astrocytes and Type 1-like cells were rarely observed in the hilus. Together, our results lay the basis for future studies focused on the lineage and fate of hilar proliferative cells and suggest that the hilus could be relevant to the formation of new cells that modulate multiple physiological processes governed by the hippocampus.
Collapse
|
18
|
Vasogenic edema versus neuroplasticity as neural correlates of hippocampal volume increase following electroconvulsive therapy. Brain Stimul 2020; 13:1080-1086. [DOI: 10.1016/j.brs.2020.04.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023] Open
|
19
|
Volume increase in the dentate gyrus after electroconvulsive therapy in depressed patients as measured with 7T. Mol Psychiatry 2020; 25:1559-1568. [PMID: 30867562 DOI: 10.1038/s41380-019-0392-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 01/03/2023]
Abstract
Electroconvulsive therapy (ECT) is the most effective treatment for depression, yet its working mechanism remains unclear. In the animal analog of ECT, neurogenesis in the dentate gyrus (DG) of the hippocampus is observed. In humans, volume increase of the hippocampus has been reported, but accurately measuring the volume of subfields is limited with common MRI protocols. If the volume increase of the hippocampus in humans is attributable to neurogenesis, it is expected to be exclusively present in the DG, whereas other processes (angiogenesis, synaptogenesis) also affect other subfields. Therefore, we acquired an optimized MRI scan at 7-tesla field strength allowing sensitive investigation of hippocampal subfields. A further increase in sensitivity of the within-subjects measurements is gained by automatic placement of the field of view. Patients receive two MRI scans: at baseline and after ten bilateral ECT sessions (corresponding to a 5-week interval). Matched controls are also scanned twice, with a similar 5-week interval. A total of 31 participants (23 patients, 8 controls) completed the study. A large and significant increase in DG volume was observed after ECT (M = 75.44 mm3, std error = 9.65, p < 0.001), while other hippocampal subfields were unaffected. We note that possible type II errors may be present due to the small sample size. In controls no changes in volume were found. Furthermore, an increase in DG volume was related to a decrease in depression scores, and baseline DG volume predicted clinical response. These findings suggest that the volume change of the DG is related to the antidepressant properties of ECT, and may reflect neurogenesis.
Collapse
|
20
|
The Bidirectional Relationship of Depression and Inflammation: Double Trouble. Neuron 2020; 107:234-256. [PMID: 32553197 DOI: 10.1016/j.neuron.2020.06.002] [Citation(s) in RCA: 994] [Impact Index Per Article: 198.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/21/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
Depression represents the number one cause of disability worldwide and is often fatal. Inflammatory processes have been implicated in the pathophysiology of depression. It is now well established that dysregulation of both the innate and adaptive immune systems occur in depressed patients and hinder favorable prognosis, including antidepressant responses. In this review, we describe how the immune system regulates mood and the potential causes of the dysregulated inflammatory responses in depressed patients. However, the proportion of never-treated major depressive disorder (MDD) patients who exhibit inflammation remains to be clarified, as the heterogeneity in inflammation findings may stem in part from examining MDD patients with varied interventions. Inflammation is likely a critical disease modifier, promoting susceptibility to depression. Controlling inflammation might provide an overall therapeutic benefit, regardless of whether it is secondary to early life trauma, a more acute stress response, microbiome alterations, a genetic diathesis, or a combination of these and other factors.
Collapse
|
21
|
Repple J, Meinert S, Bollettini I, Grotegerd D, Redlich R, Zaremba D, Bürger C, Förster K, Dohm K, Stahl F, Opel N, Hahn T, Enneking V, Leehr EJ, Böhnlein J, Leenings R, Kaehler C, Emden D, Winter NR, Heindel W, Kugel H, Bauer J, Arolt V, Benedetti F, Dannlowski U. Influence of electroconvulsive therapy on white matter structure in a diffusion tensor imaging study. Psychol Med 2020; 50:849-856. [PMID: 31010441 DOI: 10.1017/s0033291719000758] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is a fast-acting intervention for major depressive disorder. Previous studies indicated neurotrophic effects following ECT that might contribute to changes in white matter brain structure. We investigated the influence of ECT in a non-randomized prospective study focusing on white matter changes over time. METHODS Twenty-nine severely depressed patients receiving ECT in addition to inpatient treatment, 69 severely depressed patients with inpatient treatment (NON-ECT) and 52 healthy controls (HC) took part in a non-randomized prospective study. Participants were scanned twice, approximately 6 weeks apart, using diffusion tensor imaging, applying tract-based spatial statistics. Additional correlational analyses were conducted in the ECT subsample to investigate the effects of seizure duration and therapeutic response. RESULTS Mean diffusivity (MD) increased after ECT in the right hemisphere, which was an ECT-group-specific effect. Seizure duration was associated with decreased fractional anisotropy (FA) following ECT. Longitudinal changes in ECT were not associated with therapy response. However, within the ECT group only, baseline FA was positively and MD negatively associated with post-ECT symptomatology. CONCLUSION Our data suggest that ECT changes white matter integrity, possibly reflecting increased permeability of the blood-brain barrier, resulting in disturbed communication of fibers. Further, baseline diffusion metrics were associated with therapy response. Coherent fiber structure could be a prerequisite for a generalized seizure and inhibitory brain signaling necessary to successfully inhibit increased seizure activity.
Collapse
Affiliation(s)
| | | | - Irene Bollettini
- Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | | | - Ronny Redlich
- Department of Psychiatry, University of Muenster, Germany
| | - Dario Zaremba
- Department of Psychiatry, University of Muenster, Germany
| | | | | | - Katharina Dohm
- Department of Psychiatry, University of Muenster, Germany
| | - Felix Stahl
- Department of Psychiatry, University of Muenster, Germany
| | - Nils Opel
- Department of Psychiatry, University of Muenster, Germany
| | - Tim Hahn
- Department of Psychiatry, University of Muenster, Germany
| | | | | | | | | | - Claas Kaehler
- Department of Psychiatry, University of Muenster, Germany
- Institute of Pattern Recognition and Image Analysis, University of Muenster, Germany
| | - Daniel Emden
- Department of Psychiatry, University of Muenster, Germany
| | - Nils R Winter
- Department of Psychiatry, University of Muenster, Germany
| | - Walter Heindel
- Department of Clinical Radiology, University of Muenster, Germany
| | - Harald Kugel
- Department of Clinical Radiology, University of Muenster, Germany
| | - Jochen Bauer
- Department of Clinical Radiology, University of Muenster, Germany
| | - Volker Arolt
- Department of Psychiatry, University of Muenster, Germany
| | - Francesco Benedetti
- Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
- University Vita-Salute San Raffaele, Italy
| | - Udo Dannlowski
- Department of Psychiatry, University of Muenster, Germany
| |
Collapse
|
22
|
Ousdal OT, Argyelan M, Narr KL, Abbott C, Wade B, Vandenbulcke M, Urretavizcaya M, Tendolkar I, Takamiya A, Stek ML, Soriano-Mas C, Redlich R, Paulson OB, Oudega ML, Opel N, Nordanskog P, Kishimoto T, Kampe R, Jorgensen A, Hanson LG, Hamilton JP, Espinoza R, Emsell L, van Eijndhoven P, Dols A, Dannlowski U, Cardoner N, Bouckaert F, Anand A, Bartsch H, Kessler U, Oedegaard KJ, Dale AM, Oltedal L. Brain Changes Induced by Electroconvulsive Therapy Are Broadly Distributed. Biol Psychiatry 2020; 87:451-461. [PMID: 31561859 DOI: 10.1016/j.biopsych.2019.07.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/14/2019] [Accepted: 07/15/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is associated with volumetric enlargements of corticolimbic brain regions. However, the pattern of whole-brain structural alterations following ECT remains unresolved. Here, we examined the longitudinal effects of ECT on global and local variations in gray matter, white matter, and ventricle volumes in patients with major depressive disorder as well as predictors of ECT-related clinical response. METHODS Longitudinal magnetic resonance imaging and clinical data from the Global ECT-MRI Research Collaboration (GEMRIC) were used to investigate changes in white matter, gray matter, and ventricle volumes before and after ECT in 328 patients experiencing a major depressive episode. In addition, 95 nondepressed control subjects were scanned twice. We performed a mega-analysis of single subject data from 14 independent GEMRIC sites. RESULTS Volumetric increases occurred in 79 of 84 gray matter regions of interest. In total, the cortical volume increased by mean ± SD of 1.04 ± 1.03% (Cohen's d = 1.01, p < .001) and the subcortical gray matter volume increased by 1.47 ± 1.05% (d = 1.40, p < .001) in patients. The subcortical gray matter increase was negatively associated with total ventricle volume (Spearman's rank correlation ρ = -.44, p < .001), while total white matter volume remained unchanged (d = -0.05, p = .41). The changes were modulated by number of ECTs and mode of electrode placements. However, the gray matter volumetric enlargements were not associated with clinical outcome. CONCLUSIONS The findings suggest that ECT induces gray matter volumetric increases that are broadly distributed. However, gross volumetric increases of specific anatomically defined regions may not serve as feasible biomarkers of clinical response.
Collapse
Affiliation(s)
| | - Miklos Argyelan
- Center for Psychiatric Neuroscience at the Feinstein Institute for Medical Research, New York, New York
| | - Katherine L Narr
- Departments of Neurology, Psychiatry, and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles
| | - Christopher Abbott
- Department of Psychiatry, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Benjamin Wade
- Departments of Neurology, Psychiatry, and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles
| | - Mathieu Vandenbulcke
- Department of Geriatric Psychiatry, University Psychiatric Center Katholieke Universiteit Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Mikel Urretavizcaya
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Biomedical Research Institute; Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain
| | - Indira Tendolkar
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands; Donders Institute for Brain Cognition and Behavior, Centre for Cognitive Neuroimaging, Nijmegen, The Netherlands; Faculty of Medicine and Landschaftsverband Rheinland Clinic for Psychiatry and Psychotherapy, University of Duisburg-Essen, Duisburg-Essen, Germany
| | - Akihiro Takamiya
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; Center for Psychiatry and Behavioral Science, Komagino Hospital, Tokyo, Japan
| | - Max L Stek
- Geestelijke GezondheidsZorg inGeest Specialized Mental Health Care, Amsterdam, The Netherlands; Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Psychiatry, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Carles Soriano-Mas
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Biomedical Research Institute; Department of Psychobiology and Methodology in Health Sciences, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain
| | - Ronny Redlich
- Department of Psychiatry and Psychotherapy, University of Muenster, Muenster, Germany
| | - Olaf B Paulson
- Neurobiology Research Unit, Department of Neurology, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mardien L Oudega
- Geestelijke GezondheidsZorg inGeest Specialized Mental Health Care, Amsterdam, The Netherlands; Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Psychiatry, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Nils Opel
- Department of Psychiatry and Psychotherapy, University of Muenster, Muenster, Germany; Interdisciplinary Centre for Clinical Research (IZKF), University of Muenster, Muenster, Germany
| | - Pia Nordanskog
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Taishiro Kishimoto
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Robin Kampe
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Anders Jorgensen
- Psychiatric Center Copenhagen (Rigshospitalet), Mental Health Services of the Capital Region of Denmark, Copenhagen, Denmark
| | - Lars G Hanson
- Center for Magnetic Resonance, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark; Danish Research Centre for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark
| | - J Paul Hamilton
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Randall Espinoza
- Departments of Neurology, Psychiatry, and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles
| | - Louise Emsell
- Department of Geriatric Psychiatry, University Psychiatric Center Katholieke Universiteit Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Philip van Eijndhoven
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands; Donders Institute for Brain Cognition and Behavior, Centre for Cognitive Neuroimaging, Nijmegen, The Netherlands
| | - Annemieke Dols
- Geestelijke GezondheidsZorg inGeest Specialized Mental Health Care, Amsterdam, The Netherlands; Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Psychiatry, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Udo Dannlowski
- Department of Psychiatry and Psychotherapy, University of Muenster, Muenster, Germany
| | - Narcis Cardoner
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain; Department of Mental Health, University Hospital Parc Taulí-I3PT, Sabadell, Spain
| | - Filip Bouckaert
- Department of Geriatric Psychiatry, University Psychiatric Center Katholieke Universiteit Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Amit Anand
- Cleveland Clinic, Center for Behavioral Health, Cleveland, Ohio
| | - Hauke Bartsch
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, California; Department of Radiology, University of California, San Diego, La Jolla, California
| | - Ute Kessler
- Norwegian Centre for Mental Disorders Research, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Ketil J Oedegaard
- Norwegian Centre for Mental Disorders Research, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, California; Department of Radiology, University of California, San Diego, La Jolla, California; Department of Neurosciences, University of California, San Diego, La Jolla, California
| | - Leif Oltedal
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | |
Collapse
|
23
|
Boda E. Myelin and oligodendrocyte lineage cell dysfunctions: New players in the etiology and treatment of depression and stress‐related disorders. Eur J Neurosci 2019; 53:281-297. [DOI: 10.1111/ejn.14621] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/06/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi‐Montalcini University of Turin Turin Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO) University of Turin Turin Italy
| |
Collapse
|
24
|
Running exercise protects oligodendrocytes in the medial prefrontal cortex in chronic unpredictable stress rat model. Transl Psychiatry 2019; 9:322. [PMID: 31780641 PMCID: PMC6882819 DOI: 10.1038/s41398-019-0662-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/05/2019] [Accepted: 11/08/2019] [Indexed: 12/28/2022] Open
Abstract
Previous postmortem and animal studies have shown decreases in the prefrontal cortex (PFC) volume and the number of glial cells in the PFC of depression. Running exercise has been shown to alleviate depressive symptoms. However, the effects of running exercise on the medial prefrontal cortex (mPFC) volume and oligodendrocytes in the mPFC of depressed patients and animals have not been investigated. To address these issues, adult male rats were subjected to chronic unpredictable stress (CUS) for 5 weeks, followed by treadmill running for 6 weeks. Then, the mPFC volume and the mPFC oligodendrocytes were investigated using stereology, immunohistochemistry, immunofluorescence and western blotting. Using a CUS paradigm that allowed for the analysis of anhedonia, we found that running exercise alleviated the deficits in sucrose preference, as well as the decrease in the mPFC volume. Meanwhile, we found that running exercise significantly increased the number of CNPase+ oligodendrocytes and Olig2+ oligodendrocytes, reduced the ratio between Olig2+/NG2+ oligodendrocytes and Olig2+ oligodendrocytes and increased myelin basic protein (MBP), CNPase and Olig2 protein expression in the mPFC of the CUS rat model. However, running exercise did not change NG2+ oligodendrocyte number in the mPFC in these rats. These results indicated that running exercise promoted the differentiation of oligodendrocytes and myelin-forming ability in the mPFC in the context of depression. These findings suggest that the beneficial effects of running exercise on mPFC volume and oligodendrocytes in mPFC might be an important structural basis for the antidepressant effects of running exercise.
Collapse
|
25
|
Acute and long-term effects of electroconvulsive therapy on human dentate gyrus. Neuropsychopharmacology 2019; 44:1805-1811. [PMID: 30622299 PMCID: PMC6785137 DOI: 10.1038/s41386-019-0312-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/27/2018] [Accepted: 12/30/2018] [Indexed: 12/29/2022]
Abstract
Electroconvulsive therapy (ECT) is the most effective treatment for severe depression, although the underlying mechanisms remain unclear. Animal studies have consistently shown that electroconvulsive stimulation induces neuroplastic changes in the dentate gyrus. To date, few studies have investigated the effect of ECT on human hippocampal subfields. In the current study, structural magnetic resonance imaging (MRI) was conducted in 25 patients with major depressive episodes at 3 time points: before ECT (TP1), after 1 week of the last ECT (TP2) and after 3 months of the last ECT (TP3). Twenty healthy controls were scanned twice with an interval similar to patients between TP1 and TP2. Volumetric analyses of the cornu ammonis (CA)4/dentate gyrus (DG) were performed using the MAGeT-Brain (Multiple Automatically Generated Templates) algorithm. Clinically remitted patients after ECT showed larger volume increases in the right CA4/DG than non-remitted patients. Volume increases in the right CA4/DG were negatively associated with age. Increased CA4/DG volumes after ECT returned to baseline levels after 3 months irrespective of clinical state. ECT-induced volume increase in the CA4/DG was associated with age and clinical remission. These findings are consistent with the neurotrophic processes seen in preclinical studies. Neuroplastic change in the CA4/DG might mediate some of the short-term antidepressant effects of ECT.
Collapse
|
26
|
Variations in Hippocampal White Matter Diffusivity Differentiate Response to Electroconvulsive Therapy in Major Depression. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 4:300-309. [PMID: 30658916 DOI: 10.1016/j.bpsc.2018.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/03/2018] [Accepted: 11/08/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is an effective treatment for severe depression and is shown to increase hippocampal volume and modulate hippocampal functional connectivity. Whether variations in hippocampal structural connectivity occur with ECT and relate to clinical response is unknown. METHODS Patients with major depression (n = 36, 20 women, age 41.49 ± 13.57 years) underwent diffusion magnetic resonance imaging at baseline and after ECT. Control subjects (n = 32, 17 women, age 39.34 ± 12.27 years) underwent scanning twice. Functionally defined seeds in the left and right anterior hippocampus and probabilistic tractography were used to extract tract volume and diffusion metrics (fractional anisotropy and axial, radial, and mean diffusivity). Statistical analyses determined effects of ECT and time-by-response group interactions (>50% change in symptoms before and after ECT defined response). Differences between baseline measures across diagnostic groups and in association with treatment outcome were also examined. RESULTS Significant effects of ECT (all p < .01) and time-by-response group interactions (all p < .04) were observed for axial, radial, and mean diffusivity for right, but not left, hippocampal pathways. Follow-up analyses showed that ECT-related changes occurred in responders only (all p < .01) as well as in relation to change in mood examined continuously (all p < .004). Baseline measures did not relate to symptom change or differ between patients and control subjects. All measures remained stable across time in control subjects. No significant effects were observed for fractional anisotropy and volume. CONCLUSIONS Structural connectivity of hippocampal neural circuits changed with ECT and distinguished treatment responders. The findings suggested neurotrophic, glial, or inflammatory response mechanisms affecting axonal integrity.
Collapse
|
27
|
Child abuse associates with an imbalance of oligodendrocyte-lineage cells in ventromedial prefrontal white matter. Mol Psychiatry 2018; 23:2018-2028. [PMID: 29158585 DOI: 10.1038/mp.2017.231] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/14/2017] [Accepted: 09/06/2017] [Indexed: 12/19/2022]
Abstract
Child abuse (CA) is a major risk factor for depression, and strongly associates with suicidal behavior during adulthood. Neuroimaging studies have reported widespread changes in white matter integrity and brain connectivity in subjects with a history of CA. Although such observations could reflect changes in myelin and oligodendrocyte function, their cellular underpinnings have never been addressed. Using postmortem brain samples from depressed suicides with or without history of CA and matched controls (18 per group), we aimed to characterize the effects of CA on oligodendrocyte-lineage (OL) cells in the ventromedial prefrontal white matter. Using immunoblotting, double-labeling immunofluorescence and stereological estimates of stage-specific markers, we found that CA is associated with increased numbers of mature myelinating oligodendrocytes, accompanied by decreased numbers of more immature OL cells. This was paralleled by an increased expression of transcription factor MASH1, which is involved in the terminal differentiation of the OL, suggesting that CA may trigger an increased maturation, or bias the populations of OL cells toward a more mature phenotype. Some of these effects, which were absent in the brain of depressed suicides with no history of CA, were also found to recover with age, suggesting that changes in the balance of the OL may reflect a transient adaptive mechanism triggered by early-life adversity. In conclusion, our results indicate that CA in depressed suicides is associated with an imbalance of the OL in the ventromedial prefrontal white matter, an effect that could lead to myelin remodeling and long-term connectivity changes within the limbic network.
Collapse
|
28
|
Kranaster L, Hoyer C, Aksay SS, Bumb JM, Müller N, Zill P, Schwarz MJ, Sartorius A. Antidepressant efficacy of electroconvulsive therapy is associated with a reduction of the innate cellular immune activity in the cerebrospinal fluid in patients with depression. World J Biol Psychiatry 2018; 19:379-389. [PMID: 28714751 DOI: 10.1080/15622975.2017.1355473] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES A bidirectional link between the antidepressant effects of electroconvulsive therapy (ECT) and the modulation of the immune system has been proposed. To elucidate the interplay between antidepressant treatment and macrophage/microglia activation in humans, we performed a study on the effects of the antidepressant treatment by ECT on markers of macrophage/microglia activation in patients with depression. METHODS We measured six different markers (IL-6, neopterin, sCD14, sCD163 MIF and MCP1) of macrophage/microglia activation in the cerebrospinal fluid (CSF) and blood of 12 patients with a severe, treatment-resistant depressive episode before and after a course of ECT. RESULTS Some markers in the CSF of remitters were reduced after the ECT course and differed from non-remitters, but no differences were found before and after ECT independently from the antidepressant efficacy. CSF baseline levels of some markers could predict the reduction of depressive psychopathology during ECT. Higher CSF levels indicating increased macrophage/microglia activation at baseline predicted a better treatment response to ECT. CONCLUSIONS Although the sample size was small, our data suggest that macrophages/microglia are involved in the pathophysiology of major depression and that antidepressant efficacy by ECT might be partly explained by the modulation of the innate immune system within the brain.
Collapse
Affiliation(s)
- Laura Kranaster
- a Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim , Heidelberg University , Mannheim , Germany
| | - Carolin Hoyer
- b Department of Neurology , University Medical Centre Mannheim , Mannheim , Germany
| | - Suna S Aksay
- a Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim , Heidelberg University , Mannheim , Germany
| | - Jan Malte Bumb
- c Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim , Heidelberg University , Mannheim , Germany
| | - Norbert Müller
- d Department of Psychiatry and Psychotherapy , Ludwig Maximilian University Munich , Munich , Germany
| | - Peter Zill
- d Department of Psychiatry and Psychotherapy , Ludwig Maximilian University Munich , Munich , Germany
| | - Markus J Schwarz
- e Department of Laboratory Medicine , Ludwig Maximilian University Munich , Munich , Germany
| | - Alexander Sartorius
- a Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim , Heidelberg University , Mannheim , Germany
| |
Collapse
|
29
|
Dario MFR, Sara T, Estela CO, Margarita PM, Guillermo ET, Fernando RDF, Javier SL, Carmen P. Stress, Depression, Resilience and Ageing: A Role for the LPA-LPA1 Pathway. Curr Neuropharmacol 2018; 16:271-283. [PMID: 28699486 PMCID: PMC5843979 DOI: 10.2174/1570159x15666170710200352] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/26/2017] [Accepted: 06/30/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Chronic stress affects health and the quality of life, with its effects being particularly relevant in ageing due to the psychobiological characteristics of this population. However, while some people develop psychiatric disorders, especially depression, others seem very capable of dealing with adversity. There is no doubt that along with the identification of neurobiological mechanisms involved in developing depression, discovering which factors are involved in positive adaptation under circumstances of extreme difficulty will be crucial for promoting resilience. METHODS Here, we review recent work in our laboratory, using an animal model lacking the LPA1 receptor, together with pharmacological studies and clinical evidence for the possible participation of the LPA1 receptor in mood and resilience to stress. RESULTS Substantial evidence has shown that the LPA1 receptor is involved in emotional regulation and in coping responses to chronic stress, which, if dysfunctional, may induce vulnerability to stress and predisposition to the development of depression. Given that there is commonality of mechanisms between those involved in negative consequences of stress and in ageing, this is not surprising, considering that the LPA1 receptor may be involved in coping with adversity during ageing. CONCLUSION Alterations in this receptor may be a susceptibility factor for the presence of depression and cognitive deficits in the elderly population. However, because this is only a promising hypothesis based on previous data, future studies should focus on the involvement of the LPA-LPA1 pathway in coping with stress and resilience in ageing.
Collapse
Affiliation(s)
- Moreno-Fernández Román Dario
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| | - Tabbai Sara
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| | - Castilla-Ortega Estela
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga; Málaga 29010, Spain
| | - Pérez-Martín Margarita
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de
Málaga; Málaga 29071, Spain
| | - Estivill-Torrús Guillermo
- Unidad de Gestión Clínica de Neurociencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitarios de Málaga, Málaga, Spain
| | - Rodríguez de Fonseca Fernando
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga; Málaga 29010, Spain
| | - Santin Luis Javier
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| | - Pedraza Carmen
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| |
Collapse
|
30
|
Czéh B, Nagy SA. Clinical Findings Documenting Cellular and Molecular Abnormalities of Glia in Depressive Disorders. Front Mol Neurosci 2018. [PMID: 29535607 PMCID: PMC5835102 DOI: 10.3389/fnmol.2018.00056] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Depressive disorders are complex, multifactorial mental disorders with unknown neurobiology. Numerous theories aim to explain the pathophysiology. According to the “gliocentric theory”, glial abnormalities are responsible for the development of the disease. The aim of this review article is to summarize the rapidly growing number of cellular and molecular evidences indicating disturbed glial functioning in depressive disorders. We focus here exclusively on the clinical studies and present the in vivo neuroimaging findings together with the postmortem molecular and histopathological data. Postmortem studies demonstrate glial cell loss while the in vivo imaging data reveal disturbed glial functioning and altered white matter microstructure. Molecular studies report on altered gene expression of glial specific genes. In sum, the clinical findings provide ample evidences on glial pathology and demonstrate that all major glial cell types are affected. However, we still lack convincing theories explaining how the glial abnormalities develop and how exactly contribute to the emotional and cognitive disturbances. Abnormal astrocytic functioning may lead to disturbed metabolism affecting ion homeostasis and glutamate clearance, which in turn, affect synaptic communication. Abnormal oligodendrocyte functioning may disrupt the connectivity of neuronal networks, while microglial activation indicates neuroinflammatory processes. These cellular changes may relate to each other or they may indicate different endophenotypes. A theory has been put forward that the stress-induced inflammation—mediated by microglial activation—triggers a cascade of events leading to damaged astrocytes and oligodendroglia and consequently to their dysfunctions. The clinical data support the “gliocentric” theory, but future research should clarify whether these glial changes are truly the cause or simply the consequences of this devastating disorder.
Collapse
Affiliation(s)
- Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary.,Department of Laboratory Medicine, University of Pécs, Medical School, Pécs, Hungary
| | - Szilvia A Nagy
- Neurobiology of Stress Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary.,Department of Neurosurgery, University of Pécs, Medical School, Pécs, Hungary.,MTA-PTE, Clinical Neuroscience MR Research Group, Pécs, Hungary.,Pécs Diagnostic Centre, Pécs, Hungary
| |
Collapse
|
31
|
Takamiya A, Chung JK, Liang KC, Graff-Guerrero A, Mimura M, Kishimoto T. Effect of electroconvulsive therapy on hippocampal and amygdala volumes: systematic review and meta-analysis. Br J Psychiatry 2018; 212:19-26. [PMID: 29433612 DOI: 10.1192/bjp.2017.11] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is one of the most effective treatments for depression, although the underlying mechanisms remain unclear. Animal studies have shown that electroconvulsive shock induced neuroplastic changes in the hippocampus. Aims To summarise volumetric magnetic resonance imaging studies investigating the effects of ECT on limbic brain structures. METHOD A systematic review and meta-analysis was conducted to assess volumetric changes of each side of the hippocampus and amygdala before and after ECT. Standardised mean difference (SMD) was calculated. RESULTS A total of 8 studies (n = 193) were selected for our analyses. Both right and left hippocampal and amygdala volumes increased after ECT. Meta-regression analyses revealed that age, percentage of those responding and percentage of those in remission were negatively associated with volume increases in the left hippocampus. CONCLUSIONS ECT increased brain volume in the limbic structures. The clinical relevance of volume increase needs further investigation. Declaration of interest None.
Collapse
Affiliation(s)
- Akihiro Takamiya
- Department of Neuropsychiatry,Keio University School of Medicine and Komagino Hospital,Tokyo,Japan
| | - Jun Ku Chung
- Institute of Medical Science,Faculty of Medicine,University of Toronto, and Multimodal Imaging Group Research Imaging Centre, Centre for Addiction and Mental Health,Toronto,Canada
| | - Kuo-Ching Liang
- Department of Neuropsychiatry,Keio University School of Medicine,Tokyo,Japan
| | - Ariel Graff-Guerrero
- Institute of Medical Science,Faculty of Medicine,University of Toronto, Multimodal Imaging Group Research Imaging Centre, Centre for Addiction and Mental Health,Toronto,Department of Psychiatry,University of Toronto,and Geriatric Mental Health Division,Centre for Addiction and Mental Health,Toronto,Canada
| | - Masaru Mimura
- Department of Neuropsychiatry,Keio University School of Medicine,Tokyo,Japan
| | - Taishiro Kishimoto
- Department of Neuropsychiatry,Keio University School of Medicine,Tokyo, Japan, andHofstra Northwell School of Medicine, Hempstead, New York,USA
| |
Collapse
|
32
|
Schoenfeld TJ, McCausland HC, Morris HD, Padmanaban V, Cameron HA. Stress and Loss of Adult Neurogenesis Differentially Reduce Hippocampal Volume. Biol Psychiatry 2017; 82:914-923. [PMID: 28629541 PMCID: PMC5683934 DOI: 10.1016/j.biopsych.2017.05.013] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/18/2017] [Accepted: 05/05/2017] [Indexed: 12/09/2022]
Abstract
BACKGROUND Hippocampal volume loss is a hallmark of clinical depression. Chronic stress produces volume loss in the hippocampus in humans and atrophy of CA3 pyramidal cells and suppression of adult neurogenesis in rodents. METHODS To investigate the relationship between decreased adult neurogenesis and stress-induced changes in hippocampal structure and volume, we compared the effects of chronic unpredictable restraint stress and inhibition of neurogenesis in a rat pharmacogenetic model. RESULTS Chronic unpredictable restraint stress over 4 weeks decreased total hippocampal volume, reflecting loss of volume in all hippocampal subfields and in both dorsal and ventral hippocampus. In contrast, complete inhibition of adult neurogenesis for 4 weeks led to volume reduction only in the dentate gyrus. With prolonged inhibition of neurogenesis for 8 or 16 weeks, volume loss spread to the CA3 region, but not CA1. Combining stress and inhibition of adult neurogenesis did not have additive effects on the magnitude of volume loss but did produce a volume reduction throughout the hippocampus. One month of chronic unpredictable restraint stress and inhibition of adult neurogenesis led to atrophy of pyramidal cell apical dendrites in dorsal CA3 and to neuronal reorganization in ventral CA3. Stress also significantly affected granule cell dendrites. CONCLUSIONS The findings suggest that adult neurogenesis is required to maintain hippocampal volume but is not responsible for stress-induced volume loss.
Collapse
Affiliation(s)
- Timothy J Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland.
| | - Hayley C McCausland
- Section on Neuroplasticity, National Institute of Mental Health, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - H Douglas Morris
- Nuclear Magnetic Resonance Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Varun Padmanaban
- Section on Neuroplasticity, National Institute of Mental Health, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
33
|
Hayasaka S, Nakamura M, Noda Y, Izuno T, Saeki T, Iwanari H, Hirayasu Y. Lateralized hippocampal volume increase following high-frequency left prefrontal repetitive transcranial magnetic stimulation in patients with major depression. Psychiatry Clin Neurosci 2017. [PMID: 28631869 DOI: 10.1111/pcn.12547] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIM Repetitive transcranial magnetic stimulation (rTMS) has been applied as a treatment for patients with treatment-resistant depression in recent years, and a large body of evidence has demonstrated its therapeutic efficacy through stimulating neuronal plasticity. The aim of this study was to investigate structural alterations in the hippocampus (HIPP) and amygdala (AM) following conventional rTMS in patients with depression. METHODS Twenty-eight patients with depression underwent 10 daily 20-Hz left prefrontal rTMS over 2 weeks. The left dorsolateral prefrontal cortex (DLPFC) was identified using magnetic resonance imaging-guided neuronavigation prior to stimulation. Magnetic resonance imaging scans were obtained at baseline and after the completion of rTMS sessions. The therapeutic effects of rTMS were evaluated with the 17-item Hamilton Depression Rating Scale (HAM-D17 ), and the volumes of the HIPP and AM were measured by a manual tracing method. RESULTS Statistical analyses revealed a significant volume increase in the left HIPP (+3.4%) after rTMS but no significant volume change in the AM. No correlation was found between the left HIPP volume increase and clinical improvement, as measured by the HAM-D17 . CONCLUSION The present study demonstrated that conventional left prefrontal rTMS increases the HIPP volume in the stimulated side, indicating a remote neuroplastic effect through the cingulum bundle.
Collapse
Affiliation(s)
- Shunsuke Hayasaka
- Laboratory of Neuromodulation, Kanagawa Psychiatric Center, Yokohama, Japan.,Department of Psychiatry, Yokohama City University School of Medicine, Yokohama, Japan.,ATR Brain Information Communication Research Laboratory Group, Kyoto, Japan
| | - Motoaki Nakamura
- Laboratory of Neuromodulation, Kanagawa Psychiatric Center, Yokohama, Japan.,Department of Psychiatry, Yokohama City University School of Medicine, Yokohama, Japan.,ATR Brain Information Communication Research Laboratory Group, Kyoto, Japan
| | - Yoshihiro Noda
- Laboratory of Neuromodulation, Kanagawa Psychiatric Center, Yokohama, Japan.,Department of Neuropsychiatry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Takuji Izuno
- Laboratory of Neuromodulation, Kanagawa Psychiatric Center, Yokohama, Japan.,Department of Psychiatry, School of Medicine, Showa University, Tokyo, Japan
| | - Takashi Saeki
- Laboratory of Neuromodulation, Kanagawa Psychiatric Center, Yokohama, Japan.,Department of Psychiatry, Yokohama City University School of Medicine, Yokohama, Japan
| | - Hideo Iwanari
- Laboratory of Neuromodulation, Kanagawa Psychiatric Center, Yokohama, Japan
| | - Yoshio Hirayasu
- Department of Psychiatry, Yokohama City University School of Medicine, Yokohama, Japan
| |
Collapse
|
34
|
van Buel EM, Sigrist H, Seifritz E, Fikse L, Bosker FJ, Schoevers RA, Klein HC, Pryce CR, Eisel ULM. Mouse repeated electroconvulsive seizure (ECS) does not reverse social stress effects but does induce behavioral and hippocampal changes relevant to electroconvulsive therapy (ECT) side-effects in the treatment of depression. PLoS One 2017; 12:e0184603. [PMID: 28910337 PMCID: PMC5598988 DOI: 10.1371/journal.pone.0184603] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/28/2017] [Indexed: 01/21/2023] Open
Abstract
Electroconvulsive therapy (ECT) is an effective treatment for depression, but can have negative side effects including amnesia. The mechanisms of action underlying both the antidepressant and side effects of ECT are not well understood. An equivalent manipulation that is conducted in experimental animals is electroconvulsive seizure (ECS). Rodent studies have provided valuable insights into potential mechanisms underlying the antidepressant and side effects of ECT. However, relatively few studies have investigated the effects of ECS in animal models with a depression-relevant manipulation such as chronic stress. In the present study, mice were first exposed to chronic social stress (CSS) or a control procedure for 15 days followed by ECS or a sham procedure for 10 days. Behavioral effects were investigated using an auditory fear conditioning (learning) and expression (memory) test and a treadmill-running fatigue test. Thereafter, immunohistochemistry was conducted on brain material using the microglial marker Iba-1 and the cholinergic fibre marker ChAT. CSS did not increase fear learning and memory in the present experimental design; in both the control and CSS mice ECS reduced fear learning and fear memory expression. CSS induced the expected fatigue-like effect in the treadmill-running test; ECS induced increased fatigue in CSS and control mice. In CSS and control mice ECS induced inflammation in hippocampus in terms of increased expression of Iba-1 in radiatum of CA1 and CA3. CSS and ECS both reduced acetylcholine function in hippocampus as indicated by decreased expression of ChAT in several hippocampal sub-regions. Therefore, CSS increased fatigue and reduced hippocampal ChAT activity and, rather than reversing these effects, a repeated ECS regimen resulted in impaired fear learning-memory, increased fatigue, increased hippocampal Iba-1 expression, and decreased hippocampal ChAT expression. As such, the current model does not provide insights into the mechanism of ECT antidepressant function but does provide evidence for pathophysiological mechanisms that might contribute to important ECT side-effects.
Collapse
Affiliation(s)
- Erin M. van Buel
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Nuclear Medicine & Molecular Imaging, Groningen, Netherlands
- Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, Netherlands
| | - Hannes Sigrist
- Preclinical Laboratory for Translational Research into Affective Disorders (PLaTRAD), Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Erich Seifritz
- Preclinical Laboratory for Translational Research into Affective Disorders (PLaTRAD), Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Lianne Fikse
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
| | - Fokko J. Bosker
- University of Groningen, University Medical Centre Groningen, Dept of Nuclear Medicine & Molecular Imaging, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Psychiatry, Groningen, Netherlands
| | - Robert A. Schoevers
- Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Psychiatry, Groningen, Netherlands
| | - Hans C. Klein
- University of Groningen, University Medical Centre Groningen, Dept of Nuclear Medicine & Molecular Imaging, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Psychiatry, Groningen, Netherlands
| | - Christopher R. Pryce
- Preclinical Laboratory for Translational Research into Affective Disorders (PLaTRAD), Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Ulrich LM Eisel
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
- Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Psychiatry, Groningen, Netherlands
- * E-mail:
| |
Collapse
|
35
|
Singh A, Kar SK. How Electroconvulsive Therapy Works?: Understanding the Neurobiological Mechanisms. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2017; 15:210-221. [PMID: 28783929 PMCID: PMC5565084 DOI: 10.9758/cpn.2017.15.3.210] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/06/2016] [Accepted: 12/21/2016] [Indexed: 12/29/2022]
Abstract
Electroconvulsive therapy (ECT) is a time tested treatment modality for the management of various psychiatric disorders. There have been a lot of modifications in the techniques of delivering ECT over decades. Despite lots of criticisms encountered, ECT has still been used commonly in clinical practice due to its safety and efficacy. Research evidences found multiple neuro-biological mechanisms for the therapeutic effect of ECT. ECT brings about various neuro-physiological as well as neuro-chemical changes in the macro- and micro-environment of the brain. Diverse changes involving expression of genes, functional connectivity, neurochemicals, permeability of blood-brain-barrier, alteration in immune system has been suggested to be responsible for the therapeutic effects of ECT. This article reviews different neurobiological mechanisms responsible for the therapeutic efficacy of ECT.
Collapse
Affiliation(s)
- Amit Singh
- Department of Psychiatry, King George's Medical University, Lucknow, U.P, India
| | - Sujita Kumar Kar
- Department of Psychiatry, King George's Medical University, Lucknow, U.P, India
| |
Collapse
|
36
|
Toma VA, Farcas AD, Parvu M, Silaghi-Dumitrescu R, Roman I. CA3 hippocampal field: Cellular changes and its relation with blood nitro-oxidative stress reveal a balancing function of CA3 area in rats exposed to repetead restraint stress. Brain Res Bull 2016; 130:10-17. [PMID: 28013041 DOI: 10.1016/j.brainresbull.2016.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/29/2016] [Accepted: 12/20/2016] [Indexed: 12/23/2022]
Abstract
Rats exposed to repeated restraint stress exhibit structural and functional deficits in hippocampus that are similar to those observed in patients with depressive illnesses. Blood corticosterone concentrations are proportionally increased with catalase and glutathione-peroxidase activity and are inversely proportional with 3-nitrotyrosine concentrations.Cytochrome c oxidase, adenosin tryphosphatase and monoamine oxidase activities of CA3 hippocampal field mark a stress-time dependent decrease. Acridine-orange labeling of the CA3 field reveals an enhancing green fluorescence of glyocites in stress conditions. After three days of restraint stress, the secretory activity of CA3 neurons did not show significant decrease, and neurons appeared with normal shapes and distribution. CA3 neurons after seven days of restraint stress have marked a slight decrease of secretory activity. In contrast to a well-preserved histological appearance of the CA3 neurons, local and blood stress-related reactions are observed. CA3-glial activation and disturbance of blood oxidative homeostasis are tandem processes during three and seven days of RS. This study depicts the balancing role of CA3 area in time-varying stress conditions.
Collapse
Affiliation(s)
- Vlad Al Toma
- Babes-Bolyai University, Cluj-Napoca, Romania; Institute of Biological Research, Cluj-Napoca, Romania; NIRD of Isotopic and Molecular Technologies, Cluj-Napoca, Romania.
| | - Anca D Farcas
- Babes-Bolyai University, Cluj-Napoca, Romania; Institute of Biological Research, Cluj-Napoca, Romania; NIRD of Isotopic and Molecular Technologies, Cluj-Napoca, Romania
| | | | | | - Ioana Roman
- Institute of Biological Research, Cluj-Napoca, Romania
| |
Collapse
|
37
|
Electroconvulsive shock attenuated microgliosis and astrogliosis in the hippocampus and ameliorated schizophrenia-like behavior of Gunn rat. J Neuroinflammation 2016; 13:230. [PMID: 27590010 PMCID: PMC5009533 DOI: 10.1186/s12974-016-0688-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/18/2016] [Indexed: 11/23/2022] Open
Abstract
Background Although electroconvulsive therapy (ECT) is regarded as one of the efficient treatments for intractable psychiatric disorders, the mechanism of therapeutic action remains unclear. Recently, many studies indicate that ECT affects the immune-related cells, such as microglia, astrocytes, and lymphocytes. Moreover, microglial activation and astrocytic activation have been implicated in the postmortem brains of schizophrenia patients. We previously demonstrated that Gunn rats showed schizophrenia-like behavior and microglial activation in their brains. The present study examined the effects of electroconvulsive shock (ECS), an animal counterpart of ECT, on schizophrenia-like behavior, microgliosis, and astrogliosis in the brain of Gunn rats. Methods The rats were divided into four groups, i.e., Wistar sham, Wistar ECS, Gunn sham, and Gunn ECS. ECS groups received ECS once daily for six consecutive days. Subsequently, prepulse inhibition (PPI) test was performed, and immunohistochemistry analysis was carried out to determine the activation degree of microglia and astrocytes in the hippocampus by using anti-CD11b and anti-glial fibrillary acidic protein (GFAP) antibody, respectively. Results We found PPI deficit in Gunn rats compared to Wistar rats, and it was significantly improved by ECS. Immunohistochemistry analysis revealed that immunoreactivity of CD11b and GFAP was significantly increased in Gunn rats compared to Wistar rats. ECS significantly attenuated the immunoreactivity of both CD11b and GFAP in Gunn rats. Conclusions ECS ameliorated schizophrenia-like behavior of Gunn rats and attenuated microgliosis and astrogliosis in the hippocampus of Gunn rats. Accordingly, therapeutic effects of ECT may be exerted, at least in part, by inhibition of glial activation. These results may provide crucial information to elucidate the role of activated glia in the pathogenesis of schizophrenia and to determine whether future therapeutic interventions should attempt to up-regulate or down-regulate glial functions. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0688-2) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Variations in myo-inositol in fronto-limbic regions and clinical response to electroconvulsive therapy in major depression. J Psychiatr Res 2016; 80:45-51. [PMID: 27285661 PMCID: PMC4980182 DOI: 10.1016/j.jpsychires.2016.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 05/03/2016] [Accepted: 05/26/2016] [Indexed: 11/22/2022]
Abstract
Though electroconvulsive therapy (ECT) is an established treatment for severe depression, the neurobiological factors accounting for the clinical effects of ECT are largely unknown. Myo-inositol, a neurometabolite linked with glial activity, is reported as reduced in fronto-limbic regions in patients with depression. Whether changes in myo-inositol relate to the antidepressant effects of ECT is unknown. Using magnetic resonance spectroscopy ((1)H-MRS), we measured dorsomedial anterior cingulate cortex (dmACC) and left and right hippocampal myo-inositol in 50 ECT patients (mean age: 43.78, 14 SD) and 33 controls (mean age: 39.33, 12 SD) to determine cross sectional effects of diagnosis and longitudinal effects of ECT. Patients were scanned prior to treatment, after the second ECT and at completion of the ECT index series. Controls were scanned twice at intervals corresponding to patients' baseline and end of treatment scans. Myo-inositol increased over the course of ECT in the dmACC (p = 0.042). A significant hemisphere by clinical response effect was observed for the hippocampus (p = 0.003) where decreased myo-inositol related to symptom improvement in the left hippocampus. Cross-sectional differences between patients and controls at baseline were not detected. Changes in myo-inositol observed in the dmACC in association with ECT and in the hippocampus in association with ECT-related clinical response suggest the mechanisms of ECT could include gliogenesis or a reversal of gliosis that differentially affect dorsal and ventral limbic regions. Change in dmACC myo-inositol diverged from control values with ECT suggesting compensation, while hippocampal change suggested normalization.
Collapse
|
39
|
Wade BSC, Joshi SH, Njau S, Leaver AM, Vasavada M, Woods RP, Gutman BA, Thompson PM, Espinoza R, Narr KL. Effect of Electroconvulsive Therapy on Striatal Morphometry in Major Depressive Disorder. Neuropsychopharmacology 2016; 41:2481-91. [PMID: 27067127 PMCID: PMC4987846 DOI: 10.1038/npp.2016.48] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 03/01/2016] [Accepted: 03/25/2016] [Indexed: 12/17/2022]
Abstract
Patients with major depression show reductions in striatal and paleostriatal volumes. The functional integrity and connectivity of these regions are also shown to change with antidepressant response. Electroconvulsive therapy (ECT) is a robust and rapidly acting treatment for severe depression. However, whether morphological changes in the dorsal and ventral striatum/pallidum relate to or predict therapeutic response to ECT is unknown. Using structural MRI, we assessed cross-sectional effects of diagnosis and longitudinal effects of ECT for volume and surface-based shape metrics of the caudate, putamen, pallidum, and nucleus accumbens in 53 depressed patients (mean age: 44.1 years, 13.8 SD; 52% female) and 33 healthy controls (mean age: 39.3 years, 12.4 SD; 57% female). Patients were assessed before ECT, after their second ECT, and after completing an ECT treatment index. Controls were evaluated at two time points. Support vector machines determined whether morphometric measures at baseline predicted ECT-related clinical response. Patients showed smaller baseline accumbens and pallidal volumes than controls (P<0.05). Increases in left putamen volume (P<0.03) occurred with ECT. Global increases in accumbens volume and local changes in pallidum and caudate volume occurred in patients defined as treatment responders. Morphometric changes were absent across time in controls. Baseline volume and shape metrics predicted overall response to ECT with up to 89% accuracy. Results support that ECT elicits structural plasticity in the dorsal and ventral striatum/pallidum. The morphometry of these structures, forming key components of limbic-cortical-striatal-pallidal-thalamic circuitry involved in mood and emotional regulation, may determine patients likely to benefit from treatment.
Collapse
Affiliation(s)
- Benjamin S C Wade
- Imaging Genetics Center, University of Southern California, Los Angeles, CA, USA
| | - Shantanu H Joshi
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Stephanie Njau
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Amber M Leaver
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Megha Vasavada
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Roger P Woods
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
| | - Boris A Gutman
- Imaging Genetics Center, University of Southern California, Los Angeles, CA, USA
| | - Paul M Thompson
- Imaging Genetics Center, University of Southern California, Los Angeles, CA, USA
| | - Randall Espinoza
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
| | - Katherine L Narr
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
40
|
Lehmann ML, Cooper HA, Maric D, Herkenham M. Social defeat induces depressive-like states and microglial activation without involvement of peripheral macrophages. J Neuroinflammation 2016; 13:224. [PMID: 27581371 PMCID: PMC5007852 DOI: 10.1186/s12974-016-0672-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 08/17/2016] [Indexed: 01/27/2023] Open
Abstract
Background We are interested in the causal interactions between psychological stress and activity within different compartments of the immune system. Psychosocial stress has been reported to not only alter microglia morphology but also produce anxiety-like and depressive-like effects by triggering CNS infiltration of macrophages from the periphery. We sought to test these phenomena in a somewhat different but standardized model of chronic social defeat (SD) stress. Methods We used a paradigm of dyadic home pairing of dominant and subordinate mice that has been validated to induce powerful anxiety-like and depressive-like effects manifested by behavior assessed in social tasks. We administered the SD stress for 3 days (acute SD) or 14 days (chronic SD) and looked for monocyte entry into the brain by three independent means, including CD45 activation states assessed by flow cytometry and tracking fluorescently tagged peripheral cells from Ccr2wt/rfp and Ubcgfp/gfp reporter mice. We further characterized the effects of SD stress on microglia using quantitative morphometric analysis, ex vivo phagocytosis assays, flow cytometry, and immunochemistry. Results We saw no evidence of stress-induced macrophage entry after acute or chronic defeat stress. In comparison, brain infiltration of peripheral cells did occur after endotoxin administration. Furthermore, mutant mice lacking infiltrating macrophages due to CCR2 knockout developed the same degree of chronic SD-induced depressive behavior as wildtype mice. We therefore focused more closely on the intrinsic immune cells, the microglia. Using Cx3cr1wt/gpf microglial reporter mice, we saw by quantitative methods that microglial morphology was not altered by stress at either time point. However, chronic SD mice had elevated numbers of CD68hi microglia examined by flow cytometry. CD68 is a marker for phagocytic activity. Indeed, these cells ex vivo showed elevated phagocytosis, confirming the increased activation status of chronic SD microglia. Finally, acute SD but not chronic SD increased microglial proliferation, which occurred selectively in telencephalic stress-related brain areas. Conclusions In the SD paradigm, changes in CNS-resident microglia numbers and activation states might represent the main immunological component of the psychosocial stress-induced depressive state.
Collapse
Affiliation(s)
- Michael L Lehmann
- Section on Functional Neuroanatomy, Intramural Research Program, National Institute of Mental Health, NIH, Bldg. 35, Rm. 1C911, Bethesda, MD, 20892-3724, USA.
| | - Hannah A Cooper
- Section on Functional Neuroanatomy, Intramural Research Program, National Institute of Mental Health, NIH, Bldg. 35, Rm. 1C911, Bethesda, MD, 20892-3724, USA
| | - Dragan Maric
- NINDS Flow Cytometry Core Facility, NIH, Bethesda, MD, 20892, USA
| | - Miles Herkenham
- Section on Functional Neuroanatomy, Intramural Research Program, National Institute of Mental Health, NIH, Bldg. 35, Rm. 1C911, Bethesda, MD, 20892-3724, USA
| |
Collapse
|
41
|
Yirmiya R, Rimmerman N, Reshef R. Depression as a microglial disease. Trends Neurosci 2016; 38:637-658. [PMID: 26442697 DOI: 10.1016/j.tins.2015.08.001] [Citation(s) in RCA: 601] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/10/2015] [Accepted: 08/11/2015] [Indexed: 12/12/2022]
Abstract
Despite decades of intensive research, the biological mechanisms that causally underlie depression are still unclear, and therefore the development of novel effective antidepressant treatments is hindered. Recent studies indicate that impairment of the normal structure and function of microglia, caused by either intense inflammatory activation (e.g., following infections, trauma, stroke, short-term stress, autoimmune or neurodegenerative diseases) or by decline and senescence of these cells (e.g., during aging, Alzheimer's disease, or chronic unpredictable stress exposure), can lead to depression and associated impairments in neuroplasticity and neurogenesis. Accordingly, some forms of depression can be considered as a microglial disease (microgliopathy), which should be treated by a personalized medical approach using microglial inhibitors or stimulators depending on the microglial status of the depressed patient.
Collapse
Affiliation(s)
- Raz Yirmiya
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem 91905, Israel.
| | - Neta Rimmerman
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem 91905, Israel
| | - Ronen Reshef
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem 91905, Israel
| |
Collapse
|
42
|
Pirnia T, Joshi SH, Leaver AM, Vasavada M, Njau S, Woods RP, Espinoza R, Narr KL. Electroconvulsive therapy and structural neuroplasticity in neocortical, limbic and paralimbic cortex. Transl Psychiatry 2016; 6:e832. [PMID: 27271858 PMCID: PMC4931600 DOI: 10.1038/tp.2016.102] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 04/05/2016] [Accepted: 04/24/2016] [Indexed: 02/07/2023] Open
Abstract
Electroconvulsive therapy (ECT) is a highly effective and rapidly acting treatment for severe depression. To understand the biological bases of therapeutic response, we examined variations in cortical thickness from magnetic resonance imaging (MRI) data in 29 patients scanned at three time points during an ECT treatment index series and in 29 controls at two time points. Changes in thickness across time and with symptom improvement were evaluated at high spatial resolution across the cortex and within discrete cortical regions of interest. Patients showed increased thickness over the course of ECT in the bilateral anterior cingulate cortex (ACC), inferior and superior temporal, parahippocampal, entorhinal and fusiform cortex and in distributed prefrontal areas. No changes across time occurred in controls. In temporal and fusiform regions showing significant ECT effects, thickness differed between patients and controls at baseline and change in thickness related to therapeutic response in patients. In the ACC, these relationships occurred in treatment responders only, and thickness measured soon after treatment initiation predicted the overall ECT response. ECT leads to widespread neuroplasticity in neocortical, limbic and paralimbic regions and changes relate to the extent of antidepressant response. Variations in ACC thickness, which discriminate treatment responders and predict response early in the course of ECT, may represent a biomarker of overall clinical outcome. Because post-mortem studies show focal reductions in glial density and neuronal size in patients with severe depression, ECT-related increases in thickness may be attributable to neuroplastic processes affecting the size and/or density of neurons and glia and their connections.
Collapse
Affiliation(s)
- T Pirnia
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, University of California Los Angeles, Los Angeles, CA, USA
| | - S H Joshi
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, University of California Los Angeles, Los Angeles, CA, USA
| | - A M Leaver
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, University of California Los Angeles, Los Angeles, CA, USA
| | - M Vasavada
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, University of California Los Angeles, Los Angeles, CA, USA
| | - S Njau
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, University of California Los Angeles, Los Angeles, CA, USA
| | - R P Woods
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - R Espinoza
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - K L Narr
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
43
|
Shibasaki C, Takebayashi M, Itagaki K, Abe H, Kajitani N, Okada-Tsuchioka M, Yamawaki S. Altered Serum Levels of Matrix Metalloproteinase-2, -9 in Response to Electroconvulsive Therapy for Mood Disorders. Int J Neuropsychopharmacol 2016; 19:pyw019. [PMID: 26912606 PMCID: PMC5043640 DOI: 10.1093/ijnp/pyw019] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/18/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Inflammatory processes could underlie mood disorders. Matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMP) are inflammation-related molecules. The current study sought an association between mood disorders and systemic levels of MMPs and TIMPs. METHODS Serum was obtained from patients with mood disorders (n=21) and patients with schizophrenia (n=13) scheduled to undergo electroconvulsive therapy. Serum was also obtained from healthy controls (n=40). Clinical symptoms were assessed by the Hamilton Rating Score for Depression and the Brief Psychiatric Rating Scale. Serum levels of MMPs and TIMPs were quantified by ELISA. RESULTS The serum levels of MMP-2 in mood disorder patients, but not in schizophrenia patients, prior to the first electroconvulsive therapy session (baseline) was significantly lower than that of healthy controls. At baseline, levels of MMP-9 and TIMP-2, -1 were not different between patients with mood disorder and schizophrenia and healthy controls. After a course of electroconvulsive therapy, MMP-2 levels were significantly increased in mood disorder patients, but MMP-9 levels were significantly decreased in both mood disorder and schizophrenia patients. In mood disorder patients, there was a significant negative correlation between depressive symptoms and serum levels of MMP-2 and a positive correlation between depressive symptoms and MMP-9. In addition, alterations of serum levels of MMP-2 and MMP-9 were significantly correlated each other and were associated with certain depressive symptoms. CONCLUSION A change in inflammatory homeostasis, as indicated by MMP-2 and MMP-9, could be related to mood disorders, and these markers appear to be sensitive to electroconvulsive therapy.
Collapse
Affiliation(s)
| | - Minoru Takebayashi
- Division of Psychiatry and Neuroscience, Institute for Clinical Research (Drs Shibasaki, Takebayashi, Itagaki, Abe, Kajitani, and Okada-Tsuchioka), and Department of Psychiatry (Drs Takebayashi and Itagaki), National Hospital Organization, Kure Medical Center and Chugoku Cancer Center, Hiroshima, Japan; Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Programs for Biomedical Research, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan (Drs Shibasaki, Itagaki, and Yamawaki).
| | | | | | | | | | - Shigeto Yamawaki
- Division of Psychiatry and Neuroscience, Institute for Clinical Research (Drs Shibasaki, Takebayashi, Itagaki, Abe, Kajitani, and Okada-Tsuchioka), and Department of Psychiatry (Drs Takebayashi and Itagaki), National Hospital Organization, Kure Medical Center and Chugoku Cancer Center, Hiroshima, Japan; Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Programs for Biomedical Research, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan (Drs Shibasaki, Itagaki, and Yamawaki).
| |
Collapse
|
44
|
Elsayed M, Magistretti PJ. A New Outlook on Mental Illnesses: Glial Involvement Beyond the Glue. Front Cell Neurosci 2015; 9:468. [PMID: 26733803 PMCID: PMC4679853 DOI: 10.3389/fncel.2015.00468] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/16/2015] [Indexed: 12/27/2022] Open
Abstract
Mental illnesses have long been perceived as the exclusive consequence of abnormalities in neuronal functioning. Until recently, the role of glial cells in the pathophysiology of mental diseases has largely been overlooked. However recently, multiple lines of evidence suggest more diverse and significant functions of glia with behavior-altering effects. The newly ascribed roles of astrocytes, oligodendrocytes and microglia have led to their examination in brain pathology and mental illnesses. Indeed, abnormalities in glial function, structure and density have been observed in postmortem brain studies of subjects diagnosed with mental illnesses. In this review, we discuss the newly identified functions of glia and highlight the findings of glial abnormalities in psychiatric disorders. We discuss these preclinical and clinical findings implicating the involvement of glial cells in mental illnesses with the perspective that these cells may represent a new target for treatment.
Collapse
Affiliation(s)
- Maha Elsayed
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne Lausanne, Switzerland
| | - Pierre J Magistretti
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de LausanneLausanne, Switzerland; Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and TechnologyThuwal, Saudi Arabia; Department of Psychiatry, Center for Psychiatric Neurosciences, University of LausanneLausanne, Switzerland
| |
Collapse
|
45
|
Proliferating cells in the adolescent rat amygdala: Characterization and response to stress. Neuroscience 2015; 311:105-17. [PMID: 26476262 DOI: 10.1016/j.neuroscience.2015.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/24/2015] [Accepted: 10/02/2015] [Indexed: 12/27/2022]
Abstract
The amygdala is a heterogeneous group of nuclei that plays a role in emotional and social learning. As such, there has been increased interest in its development in adolescent animals, a period in which emotional/social learning increases dramatically. While many mechanisms of amygdala development have been studied, the role of cell proliferation during adolescence has received less attention. Using bromodeoxyuridine (BrdU) injections in adolescent and adult rats, we previously found an almost fivefold increase in BrdU-positive cells in the amygdala of adolescents compared to adults. Approximately one third of BrdU-labeled cells in the amygdala contained the putative neural marker doublecortin (DCX), suggesting a potential for neurogenesis. To further investigate this possibility in adolescents, we examined the proliferative dynamics of DCX/BrdU-labeled cells. Surprisingly, DCX/BrdU-positive cells were found to comprise a stable subpopulation of BrdU-containing cells across survivals up to 56 days, and there was no evidence of neural maturation by 28 days after BrdU injection. Additionally, we found that approximately 50% of BrdU+ cells within the adolescent amygdala contain neural-glial antigen (NG2) and are therefore presumptive oligodendrocyte precursors (OPCs). We next characterized the response to a short-lived stressor (3-day repeated variable stress, RVS). The total BrdU-labeled cell number decreased by ∼30% by 13 days following RVS (10 days post-BrdU injection) as assessed by stereologic counting methods, but the DCX/BrdU-labeled subpopulation was relatively resistant to RVS effects. In contrast, NG2/BrdU-labeled cells were strongly influenced by RVS. We conclude that typical neurogenesis is not a feature of the adolescent amygdala. These findings point to several possibilities, including the possibility that DCX/BrdU cells are late-developing neural precursors, or a unique subtype of NG2 cell that is relatively resistant to stress. In contrast, many proliferating OPCs are significantly impacted by a short-lived stressor, suggesting consequences for myelination in the developing amygdala.
Collapse
|
46
|
Tiwari SK, Seth B, Agarwal S, Yadav A, Karmakar M, Gupta SK, Choubey V, Sharma A, Chaturvedi RK. Ethosuximide Induces Hippocampal Neurogenesis and Reverses Cognitive Deficits in an Amyloid-β Toxin-induced Alzheimer Rat Model via the Phosphatidylinositol 3-Kinase (PI3K)/Akt/Wnt/β-Catenin Pathway. J Biol Chem 2015; 290:28540-28558. [PMID: 26420483 DOI: 10.1074/jbc.m115.652586] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Indexed: 01/20/2023] Open
Abstract
Neurogenesis involves generation of new neurons through finely tuned multistep processes, such as neural stem cell (NSC) proliferation, migration, differentiation, and integration into existing neuronal circuitry in the dentate gyrus of the hippocampus and subventricular zone. Adult hippocampal neurogenesis is involved in cognitive functions and altered in various neurodegenerative disorders, including Alzheimer disease (AD). Ethosuximide (ETH), an anticonvulsant drug is used for the treatment of epileptic seizures. However, the effects of ETH on adult hippocampal neurogenesis and the underlying cellular and molecular mechanism(s) are yet unexplored. Herein, we studied the effects of ETH on rat multipotent NSC proliferation and neuronal differentiation and adult hippocampal neurogenesis in an amyloid β (Aβ) toxin-induced rat model of AD-like phenotypes. ETH potently induced NSC proliferation and neuronal differentiation in the hippocampus-derived NSC in vitro. ETH enhanced NSC proliferation and neuronal differentiation and reduced Aβ toxin-mediated toxicity and neurodegeneration, leading to behavioral recovery in the rat AD model. ETH inhibited Aβ-mediated suppression of neurogenic and Akt/Wnt/β-catenin pathway gene expression in the hippocampus. ETH activated the PI3K·Akt and Wnt·β-catenin transduction pathways that are known to be involved in the regulation of neurogenesis. Inhibition of the PI3K·Akt and Wnt·β-catenin pathways effectively blocked the mitogenic and neurogenic effects of ETH. In silico molecular target prediction docking studies suggest that ETH interacts with Akt, Dkk-1, and GSK-3β. Our findings suggest that ETH stimulates NSC proliferation and differentiation in vitro and adult hippocampal neurogenesis via the PI3K·Akt and Wnt·β-catenin signaling.
Collapse
Affiliation(s)
- Shashi Kant Tiwari
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India; Academy of Scientific and Innovative Research, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India
| | - Brashket Seth
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India; Academy of Scientific and Innovative Research, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India
| | - Swati Agarwal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India; Academy of Scientific and Innovative Research, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India
| | - Anuradha Yadav
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India; Academy of Scientific and Innovative Research, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India
| | - Madhumita Karmakar
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India
| | - Shailendra Kumar Gupta
- Systems Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India
| | - Vinay Choubey
- Department of Pharmacology, Centre of Excellence for Translational Medicine; University of Tartu, Tartu 50411, Estonia
| | - Abhay Sharma
- CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, 110025 New Delhi, India.
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India; Academy of Scientific and Innovative Research, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India
| |
Collapse
|
47
|
van Buel EM, Patas K, Peters M, Bosker FJ, Eisel ULM, Klein HC. Immune and neurotrophin stimulation by electroconvulsive therapy: is some inflammation needed after all? Transl Psychiatry 2015; 5. [PMID: 26218851 PMCID: PMC5068722 DOI: 10.1038/tp.2015.100] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A low-grade inflammatory response is commonly seen in the peripheral blood of major depressive disorder (MDD) patients, especially those with refractory and chronic disease courses. However, electroconvulsive therapy (ECT), the most drastic intervention reserved for these patients, is closely associated with an enhanced haematogenous as well as neuroinflammatory immune response, as evidenced by both human and animal studies. A related line of experimental evidence further shows that inflammatory stimulation reinforces neurotrophin expression and may even mediate dramatic neurogenic and antidepressant-like effects following exposure to chronic stress. The current review therefore attempts a synthesis of our knowledge on the neurotrophic and immunological aspects of ECT and other electrically based treatments in psychiatry. Perhaps contrary to contemporary views, we conclude that targeted potentiation, rather than suppression, of inflammatory responses may be of therapeutic relevance to chronically depressed patients or a subgroup thereof.
Collapse
Affiliation(s)
- E M van Buel
- Department of Molecular Neurobiology, Center for Life Sciences, University of Groningen, Groningen, The Netherlands,Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands,Department of Molecular Neurobiology, Center for Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands. E-mail:
| | - K Patas
- Department of Molecular Neurobiology, Center for Life Sciences, University of Groningen, Groningen, The Netherlands,Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology, University Medical Center Eppendorf, Hamburg, Germany
| | - M Peters
- Department of Molecular Neurobiology, Center for Life Sciences, University of Groningen, Groningen, The Netherlands
| | - F J Bosker
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - U L M Eisel
- Department of Molecular Neurobiology, Center for Life Sciences, University of Groningen, Groningen, The Netherlands,Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - H C Klein
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands,Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
48
|
Stelzhammer V, Ozcan S, Gottschalk MG, Steeb H, Hodes GE, Guest PC, Rahmoune H, Wong EH, Russo SJ, Bahn S. Central and peripheral changes underlying susceptibility and resistance to social defeat stress – A proteomic profiling study. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.dineu.2015.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
49
|
Yang P, Gao Z, Zhang H, Fang Z, Wu C, Xu H, Huang QJ. Changes in proinflammatory cytokines and white matter in chronically stressed rats. Neuropsychiatr Dis Treat 2015; 11:597-607. [PMID: 25834438 PMCID: PMC4358419 DOI: 10.2147/ndt.s78131] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Although the pathogenesis of depression, an incapacitating psychiatric ailment, remains largely unknown, previous human and animal studies have suggested that both proinflammatory cytokines and altered oligodendrocytes play important roles in the condition. This study examined these two factors in the brains of rats following unpredictable chronic mild stress for 4 weeks, with the hypothesis that chronic stress may affect oligodendrocytes and elevate proinflammatory cytokines in the brain. After suffering unpredictable stressors for 4 weeks, the rats showed depression-like behaviors, including decreased locomotion in the open field, increased immobility time in the forced swim test, and decreased sucrose consumption and less sucrose preference when compared with controls. Immunohistochemical staining of brain sections showed higher immunoreactivity of proinflammatory cytokines in certain brain regions of stressed rats compared with controls; lower immunoreactivity of myelin basic protein and fewer mature oligodendrocytes were seen in the prefrontal cortex, but no demyelination was detected. These results are interpreted and discussed in the context of recent findings from human and animal studies.
Collapse
Affiliation(s)
- Ping Yang
- Mental Health Center, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Zhenyong Gao
- Mental Health Center, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Handi Zhang
- Mental Health Center, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Zeman Fang
- Mental Health Center, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Cairu Wu
- Mental Health Center, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Haiyun Xu
- Mental Health Center, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
- Department of Anatomy, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
- Correspondence: Haiyun Xu; Qing-Jun Huang, Mental Health Center, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041, People’s Republic of China, Tel +86 754 8890 0728, Email ;
| | - Qing-Jun Huang
- Mental Health Center, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
- Correspondence: Haiyun Xu; Qing-Jun Huang, Mental Health Center, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041, People’s Republic of China, Tel +86 754 8890 0728, Email ;
| |
Collapse
|
50
|
Somkuwar SS, Staples MC, Galinato MH, Fannon MJ, Mandyam CD. Role of NG2 expressing cells in addiction: a new approach for an old problem. Front Pharmacol 2014; 5:279. [PMID: 25566075 PMCID: PMC4271769 DOI: 10.3389/fphar.2014.00279] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/30/2014] [Indexed: 12/17/2022] Open
Abstract
Neuron-glial antigen 2 (NG2) is a proteoglycan expressed predominantly in oligodendrocyte progenitor cells (OPCs). NG2-expressing OPCs (NG2-OPCs) are self-renewing cells that are widely distributed in the gray and white matter areas of the central nervous system. NG2-OPCs can mature into premyelinating oligodendrocytes and myelinating oligodendroglia which serve as the primary source of myelin in the brain. This review characterizes NG2-OPCs in brain structure and function, conceptualizes the role of NG2-OPCs in brain regions associated with negative reinforcement and relapse to drug seeking and discusses how NG2-OPCs are regulated by neuromodulators linked to motivational withdrawal. We hope to provide the readers with an overview of the role of NG2-OPCs in brain structure and function in the context of negative affect state in substance abuse disorders and to integrate our current understanding of the physiological significance of the NG2-OPCs in the adult brain.
Collapse
Affiliation(s)
- Sucharita S Somkuwar
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| | - Miranda C Staples
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| | - Melissa H Galinato
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| | - McKenzie J Fannon
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| | - Chitra D Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| |
Collapse
|