301
|
Seo JS, Wei J, Qin L, Kim Y, Yan Z, Greengard P. Cellular and molecular basis for stress-induced depression. Mol Psychiatry 2017; 22:1440-1447. [PMID: 27457815 PMCID: PMC5269558 DOI: 10.1038/mp.2016.118] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/20/2016] [Accepted: 04/27/2016] [Indexed: 12/17/2022]
Abstract
Chronic stress has a crucial role in the development of psychiatric diseases, such as anxiety and depression. Dysfunction of the medial prefrontal cortex (mPFC) has been linked to the cognitive and emotional deficits induced by stress. However, little is known about the molecular and cellular determinants in mPFC for stress-associated mental disorders. Here we show that chronic restraint stress induces the selective loss of p11 (also known as annexin II light chain, S100A10), a multifunctional protein binding to 5-HT receptors, in layer II/III neurons of the prelimbic cortex (PrL), as well as depression-like behaviors, both of which are reversed by selective serotonin reuptake inhibitors (SSRIs) and the tricyclic class of antidepressant (TCA) agents. In layer II/III of the PrL, p11 is highly concentrated in dopamine D2 receptor-expressing (D2+) glutamatergic neurons. Viral expression of p11 in D2+ PrL neurons alleviates the depression-like behaviors exhibited by genetically manipulated mice with D2+ neuron-specific or global deletion of p11. In stressed animals, overexpression of p11 in D2+ PrL neurons rescues depression-like behaviors by restoring glutamatergic transmission. Our results have identified p11 as a key molecule in a specific cell type that regulates stress-induced depression, which provides a framework for the development of new strategies to treat stress-associated mental illnesses.
Collapse
Affiliation(s)
- J-S Seo
- grid.134907.80000 0001 2166 1519Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY USA
| | - J Wei
- grid.273335.30000 0004 1936 9887Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY USA
| | - L Qin
- grid.273335.30000 0004 1936 9887Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY USA
| | - Y Kim
- grid.134907.80000 0001 2166 1519Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY USA
| | - Z Yan
- grid.273335.30000 0004 1936 9887Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY USA
| | - P Greengard
- grid.134907.80000 0001 2166 1519Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY USA
| |
Collapse
|
302
|
Vasilescu AN, Schweinfurth N, Borgwardt S, Gass P, Lang UE, Inta D, Eckart S. Modulation of the activity of N-methyl-d-aspartate receptors as a novel treatment option for depression: current clinical evidence and therapeutic potential of rapastinel (GLYX-13). Neuropsychiatr Dis Treat 2017; 13:973-980. [PMID: 28408831 PMCID: PMC5384686 DOI: 10.2147/ndt.s119004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Classical monoaminergic antidepressants show several disadvantages, such as protracted onset of therapeutic action. Conversely, the fast and sustained antidepressant effect of the N-methyl-d-aspartate receptor (NMDAR) antagonist ketamine raises vast interest in understanding the role of the glutamate system in mood disorders. Indeed, numerous data support the existence of glutamatergic dysfunction in major depressive disorder (MDD). Drawback to this short-latency therapy is its side effect profile, especially the psychotomimetic action, which seriously hampers the common and widespread clinical use of ketamine. Therefore, there is a substantial need for alternative glutamatergic antidepressants with milder side effects. In this article, we review evidence that implicates NMDARs in the prospective treatment of MDD with focus on rapastinel (formerly known as GLYX-13), a novel synthetic NMDAR modulator with fast antidepressant effect, which acts by enhancing NMDAR function as opposed to blocking it. We summarize and discuss current clinical and animal studies regarding the therapeutic potential of rapastinel not only in MDD but also in other psychiatric disorders, such as obsessive-compulsive disorder and posttraumatic stress disorder. Additionally, we discuss current data concerning the molecular mechanisms underlying the antidepressant effect of rapastinel, highlighting common aspects as well as differences to ketamine. In 2016, rapastinel received the Breakthrough Therapy designation for the treatment of MDD from the US Food and Drug Administration, representing one of the most promising alternative antidepressants under current investigation.
Collapse
Affiliation(s)
- Andrei-Nicolae Vasilescu
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Nina Schweinfurth
- Department of Psychiatry (Universitäre Psychiatrische Kliniken), University of Basel, Basel, Switzerland
| | - Stefan Borgwardt
- Department of Psychiatry (Universitäre Psychiatrische Kliniken), University of Basel, Basel, Switzerland
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Undine E Lang
- Department of Psychiatry (Universitäre Psychiatrische Kliniken), University of Basel, Basel, Switzerland
| | - Dragos Inta
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.,Department of Psychiatry (Universitäre Psychiatrische Kliniken), University of Basel, Basel, Switzerland
| | - Sarah Eckart
- Department of Psychiatry (Universitäre Psychiatrische Kliniken), University of Basel, Basel, Switzerland
| |
Collapse
|
303
|
Lip PZY, Demasi M, Bonatto D. The role of the ubiquitin proteasome system in the memory process. Neurochem Int 2016; 102:57-65. [PMID: 27916542 DOI: 10.1016/j.neuint.2016.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/27/2016] [Accepted: 11/29/2016] [Indexed: 01/20/2023]
Abstract
Quite intuitive is the notion that memory formation and consolidation is orchestrated by protein synthesis because of the synaptic plasticity necessary for those processes. Nevertheless, recent advances have begun accumulating evidences of a high requirement for protein degradation on the molecular mechanisms of the memory process in the mammalian brain. Because degradation determines protein half-life, degradation has been increasingly recognized as an important intracellular regulatory mechanism. The proteasome is the main player in the degradation of intracellular proteins. Proteasomal substrates are mainly degraded after a post-translational modification by a poly-ubiquitin chain. Latter process, namely poly-ubiquitination, is highly regulated at the step of the ubiquitin molecule transferring to the protein substrate mediated by a set of proteins whose genes represent almost 2% of the human genome. Understanding the role of polyubiquitin-mediated protein degradation has challenging researchers in many fields of investigation as a new source of targets for therapeutic intervention, e.g. E3 ligases that transfer ubiquitin moieties to the substrate. The goal of present work was to uncover mechanisms underlying memory processes regarding the role of the ubiquitin-proteasome system (UPS). For that purpose, preceded of a short review on UPS and memory processes a top-down systems biology approach was applied to establish central proteins involved in memory formation and consolidation highlighting their cross-talking with the UPS. According to that approach, the pattern of expression of several elements of the UPS were found overexpressed in regions of the brain involved in processing cortical inputs.
Collapse
Affiliation(s)
- Philomena Z Y Lip
- Laboratory of Biochemistry and Biophysics, Instituto Butantan, São Paulo, SP, Brazil; Medical Sciences Division, University of Oxford, Oxford, UK
| | - Marilene Demasi
- Medical Sciences Division, University of Oxford, Oxford, UK.
| | - Diego Bonatto
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
304
|
De Rossi P, Harde E, Dupuis JP, Martin L, Chounlamountri N, Bardin M, Watrin C, Benetollo C, Pernet-Gallay K, Luhmann HJ, Honnorat J, Malleret G, Groc L, Acker-Palmer A, Salin PA, Meissirel C. A critical role for VEGF and VEGFR2 in NMDA receptor synaptic function and fear-related behavior. Mol Psychiatry 2016; 21:1768-1780. [PMID: 26728568 PMCID: PMC5116482 DOI: 10.1038/mp.2015.195] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 10/07/2015] [Accepted: 10/22/2015] [Indexed: 01/17/2023]
Abstract
Vascular endothelial growth factor (VEGF) is known to be required for the action of antidepressant therapies but its impact on brain synaptic function is poorly characterized. Using a combination of electrophysiological, single-molecule imaging and conditional transgenic approaches, we identified the molecular basis of the VEGF effect on synaptic transmission and plasticity. VEGF increases the postsynaptic responses mediated by the N-methyl-D-aspartate type of glutamate receptors (GluNRs) in hippocampal neurons. This is concurrent with the formation of new synapses and with the synaptic recruitment of GluNR expressing the GluN2B subunit (GluNR-2B). VEGF induces a rapid redistribution of GluNR-2B at synaptic sites by increasing the surface dynamics of these receptors within the membrane. Consistently, silencing the expression of the VEGF receptor 2 (VEGFR2) in neural cells impairs hippocampal-dependent synaptic plasticity and consolidation of emotional memory. These findings demonstrated the direct implication of VEGF signaling in neurons via VEGFR2 in proper synaptic function. They highlight the potential of VEGF as a key regulator of GluNR synaptic function and suggest a role for VEGF in new therapeutic approaches targeting GluNR in depression.
Collapse
Affiliation(s)
- P De Rossi
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Neurooncology and Neuroinflammation, Lyon Neuroscience Research Center, Lyon, France
| | - E Harde
- Institute of Cell Biology and Neuroscience and BMLS, Goethe University Frankfurt, Frankfurt, Germany,Max Planck Institute for Brain Research, Frankfurt, Germany,Focus Program Translational Neurosciences, University of Mainz, Mainz, Germany
| | - J P Dupuis
- Interdisciplinary Institute for Neuroscience, Unité Mixte de Recherche 5297, Université de Bordeaux, Bordeaux, France,Interdisciplinary Institute for Neuroscience, UMR 5297, Centre National de la Recherche Scientifique, Bordeaux, France
| | - L Martin
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Neurooncology and Neuroinflammation, Lyon Neuroscience Research Center, Lyon, France
| | - N Chounlamountri
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Neurooncology and Neuroinflammation, Lyon Neuroscience Research Center, Lyon, France
| | - M Bardin
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Neurooncology and Neuroinflammation, Lyon Neuroscience Research Center, Lyon, France
| | - C Watrin
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Neurooncology and Neuroinflammation, Lyon Neuroscience Research Center, Lyon, France
| | - C Benetollo
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Functional Neurogenomics and Optogenetics, Lyon Neuroscience Research Center, Lyon, France
| | - K Pernet-Gallay
- Grenoble Institute of Neurosciences, Grenoble, France,INSERM U836, Microscopy and Electron Microscopy Platform, Grenoble, France
| | - H J Luhmann
- Institute of Physiology, University Medical Center, University of Mainz, Mainz, Germany
| | - J Honnorat
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Neuro-Oncology Department, Hospices Civils de Lyon, Hôpital Neurologique, Lyon, France
| | - G Malleret
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Forgetting and Cortical Dynamics, Lyon Neuroscience Research Center, Lyon, France
| | - L Groc
- Interdisciplinary Institute for Neuroscience, Unité Mixte de Recherche 5297, Université de Bordeaux, Bordeaux, France,Interdisciplinary Institute for Neuroscience, UMR 5297, Centre National de la Recherche Scientifique, Bordeaux, France
| | - A Acker-Palmer
- Institute of Cell Biology and Neuroscience and BMLS, Goethe University Frankfurt, Frankfurt, Germany,Max Planck Institute for Brain Research, Frankfurt, Germany,Focus Program Translational Neurosciences, University of Mainz, Mainz, Germany
| | - P A Salin
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Forgetting and Cortical Dynamics, Lyon Neuroscience Research Center, Lyon, France
| | - C Meissirel
- Institut National de la Santé et de la Recherche Médicale, Unité 1028, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5292, Lyon, France,Claude Bernard University Lyon 1, Lyon, France,Neurooncology and Neuroinflammation, Lyon Neuroscience Research Center, Lyon, France,Equipe Neurooncologie et Neuroinflammation, Centre de Recherche en Neurosciences de Lyon, Institut National de la Santé et de la Recherche Médicale, Unité 1028, Faculté de Médecine Laennec, Lyon cedex O8, 69372 Lyon, France. E-mail:
| |
Collapse
|
305
|
Rafalo-Ulinska A, Piotrowska J, Kryczyk A, Opoka W, Sowa-Kucma M, Misztak P, Rajkowska G, Stockmeier CA, Datka W, Nowak G, Szewczyk B. Zinc transporters protein level in postmortem brain of depressed subjects and suicide victims. J Psychiatr Res 2016; 83:220-229. [PMID: 27661418 PMCID: PMC5107146 DOI: 10.1016/j.jpsychires.2016.09.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/31/2016] [Accepted: 09/08/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a serious psychiatric illness, associated with an increasing rate of suicide. The pathogenesis of depression may be associated with the disruption of zinc (Zn) homeostasis. In the brain, several proteins that regulate Zn homeostasis are present, including Zn transporters (ZnTs) which remove Zn from the cytosol. The present study was designed to investigate whether depression and suicide are associated with alterations in the expression of the ZnTs protein. METHODS Protein levels of ZnT1, ZnT3, ZnT4, ZnT5 and ZnT6 were measured in postmortem brain tissue from two different cohorts. Cohort A contained 10 subjects diagnosed with MDD (7 were suicide victims) and 10 psychiatrically-normal control subjects and cohort B contained 11 non-diagnosed suicide victims and 8 sudden-death control subjects. Moreover, in cohort A we measured protein level of NMDA (GluN2A subunit), AMPA (GluA1 subunit) and 5-HT1A receptors and PSD-95. Proteins were measured in the prefrontal cortex (PFC) using Western blotting. In addition, Zn concentration was measured using a voltammetric method. RESULTS There was a significant increase in protein levels of ZnT1, ZnT4, ZnT5 in the PFC in MDD, relative to control subjects, while ZnT3 protein level was decreased in MDD. There was no significant difference in the Zn concentration in the PFC between control and MDD subjects. Similarly, in the PFC of suicide victims (non-diagnosed), an increase in protein levels of ZnT1, ZnT4, ZnT5 and ZnT6 was observed. Conversely, protein levels of ZnT3 were decreased in both suicide victims and subjects with MDD, in comparison with control subjects. There was also a significant decrease in the protein level of GluA1, GluN2A, PSD-95 and 5-HT1A in MDD. CONCLUSIONS Our studies suggest that alterations in Zn transport proteins are associated with the pathophysiology of MDD and suicide.
Collapse
Affiliation(s)
- Anna Rafalo-Ulinska
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland,Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - Joanna Piotrowska
- Department of Inorganic and Analytical Chemistry, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Agata Kryczyk
- Department of Inorganic and Analytical Chemistry, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Włodzimierz Opoka
- Department of Inorganic and Analytical Chemistry, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Magdalena Sowa-Kucma
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland
| | - Paulina Misztak
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland,Department of Pharmacobiology, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA,Department of Psychiatry, Case Western Reserve University, 10524 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wojciech Datka
- Department of Affective Disorders, Jagiellonian University Medical College, Kopernika 21a, 31-501 Kraków, Poland
| | - Gabriel Nowak
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland,Department of Pharmacobiology, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Bernadeta Szewczyk
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland.
| |
Collapse
|
306
|
Wohleb ES. Neuron-Microglia Interactions in Mental Health Disorders: "For Better, and For Worse". Front Immunol 2016; 7:544. [PMID: 27965671 PMCID: PMC5126117 DOI: 10.3389/fimmu.2016.00544] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/16/2016] [Indexed: 12/13/2022] Open
Abstract
Persistent cognitive and behavioral symptoms that characterize many mental health disorders arise from impaired neuroplasticity in several key corticolimbic brain regions. Recent evidence suggests that reciprocal neuron–microglia interactions shape neuroplasticity during physiological conditions, implicating microglia in the neurobiology of mental health disorders. Neuron–microglia interactions are modulated by several molecular and cellular pathways, and dysregulation of these pathways often have neurobiological consequences, including aberrant neuronal responses and microglia activation. Impaired neuron-microglia interactions are implicated in mental health disorders because rodent stress models lead to concomitant neuronal dystrophy and alterations in microglia morphology and function. In this context, functional changes in microglia may be indicative of an immune state termed parainflammation in which tissue-resident macrophages (i.e., microglia) respond to malfunctioning cells by initiating modest inflammation in an attempt to restore homeostasis. Thus, aberrant neuronal activity and release of damage-associated signals during repeated stress exposure may contribute to functional changes in microglia and resultant parainflammation. Furthermore, accumulating evidence shows that uncoupling neuron–microglia interactions may contribute to altered neuroplasticity and associated anxiety- or depressive-like behaviors. Additional work shows that microglia have varied phenotypes in specific brain regions, which may underlie divergent neuroplasticity observed in corticolimbic structures following stress exposure. These findings indicate that neuron–microglia interactions are critical mediators of the interface between adaptive, homeostatic neuronal function and the neurobiology of mental health disorders.
Collapse
Affiliation(s)
- Eric S Wohleb
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
307
|
McKlveen JM, Morano RL, Fitzgerald M, Zoubovsky S, Cassella SN, Scheimann JR, Ghosal S, Mahbod P, Packard BA, Myers B, Baccei ML, Herman JP. Chronic Stress Increases Prefrontal Inhibition: A Mechanism for Stress-Induced Prefrontal Dysfunction. Biol Psychiatry 2016; 80:754-764. [PMID: 27241140 PMCID: PMC5629635 DOI: 10.1016/j.biopsych.2016.03.2101] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 03/06/2016] [Accepted: 03/10/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Multiple neuropsychiatric disorders, e.g., depression, are linked to imbalances in excitatory and inhibitory neurotransmission and prefrontal cortical dysfunction, and are concomitant with chronic stress. METHODS We used electrophysiologic (n = 5-6 animals, 21-25 cells/group), neuroanatomic (n = 6-8/group), and behavioral (n = 12/group) techniques to test the hypothesis that chronic stress increases inhibition of medial prefrontal cortex (mPFC) glutamatergic output neurons. RESULTS Using patch clamp recordings from infralimbic mPFC pyramidal neurons, we found that chronic stress selectively increases the frequency of miniature inhibitory postsynaptic currents with no effect on amplitude, which suggests that chronic stress increases presynaptic gamma-aminobutyric acid release. Elevated gamma-aminobutyric acid release under chronic stress is accompanied by increased inhibitory appositions and terminals onto glutamatergic cells, as assessed by both immunohistochemistry and electron microscopy. Furthermore, chronic stress decreases glucocorticoid receptor immunoreactivity specifically in a subset of inhibitory neurons, which suggests that increased inhibitory tone in the mPFC after chronic stress may be caused by loss of a glucocorticoid receptor-mediated brake on interneuron activity. These neuroanatomic and functional changes are associated with impairment of a prefrontal-mediated behavior. During chronic stress, rats initially make significantly more errors in the delayed spatial win-shift task, an mPFC-mediated behavior, which suggests a diminished impact of the mPFC on decision making. CONCLUSIONS Taken together, the data suggest that chronic stress increases synaptic inhibition onto prefrontal glutamatergic output neurons, limiting the influence of the prefrontal cortex in control of stress reactivity and behavior. Thus, these data provide a mechanistic link among chronic stress, prefrontal cortical hypofunction, and behavioral dysfunction.
Collapse
|
308
|
Tada H, Miyazaki T, Takemoto K, Takase K, Jitsuki S, Nakajima W, Koide M, Yamamoto N, Komiya K, Suyama K, Sano A, Taguchi A, Takahashi T. Neonatal isolation augments social dominance by altering actin dynamics in the medial prefrontal cortex. Proc Natl Acad Sci U S A 2016; 113:E7097-E7105. [PMID: 27791080 PMCID: PMC5111648 DOI: 10.1073/pnas.1606351113] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Social separation early in life can lead to the development of impaired interpersonal relationships and profound social disorders. However, the underlying cellular and molecular mechanisms involved are largely unknown. Here, we found that isolation of neonatal rats induced glucocorticoid-dependent social dominance over nonisolated control rats in juveniles from the same litter. Furthermore, neonatal isolation inactivated the actin-depolymerizing factor (ADF)/cofilin in the juvenile medial prefrontal cortex (mPFC). Isolation-induced inactivation of ADF/cofilin increased stable actin fractions at dendritic spines in the juvenile mPFC, decreasing glutamate synaptic AMPA receptors. Expression of constitutively active ADF/cofilin in the mPFC rescued the effect of isolation on social dominance. Thus, neonatal isolation affects spines in the mPFC by reducing actin dynamics, leading to altered social behavior later in life.
Collapse
Affiliation(s)
- Hirobumi Tada
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Tomoyuki Miyazaki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kiwamu Takemoto
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Kenkichi Takase
- Laboratory of Psychology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Susumu Jitsuki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Waki Nakajima
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Mayu Koide
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Naoko Yamamoto
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kasane Komiya
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kumiko Suyama
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Akane Sano
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Akiko Taguchi
- Department of Integrative Aging Neuroscience, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan;
| |
Collapse
|
309
|
Qin L, Liu W, Ma K, Wei J, Zhong P, Cho K, Yan Z. The ADHD-linked human dopamine D4 receptor variant D4.7 induces over-suppression of NMDA receptor function in prefrontal cortex. Neurobiol Dis 2016; 95:194-203. [PMID: 27475724 PMCID: PMC5391260 DOI: 10.1016/j.nbd.2016.07.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 06/16/2016] [Accepted: 07/25/2016] [Indexed: 12/15/2022] Open
Abstract
The human dopamine D4 receptor (hD4R) variants with long tandem repeats in the third intracellular loop have been strongly associated with attention deficit hyperactivity disorder (ADHD) and risk taking behaviors. To understand the potential molecular mechanism underlying the connection, we have investigated the synaptic function of human D4R polymorphism by virally expressing the ADHD-linked 7-repeat allele, hD4.7, or its normal counterpart, hD4.4, in the prefrontal cortex (PFC) of D4R knockout mice. We found that hD4R bound to the SH3 domain of PSD-95 in a state-dependent manner. Activation of hD4.7 caused more reduction of NR1/PSD-95 binding and NR1 surface expression than hD4.4 in PFC slices. Moreover, the NMDAR-mediated excitatory postsynaptic currents (NMDAR-EPSC) in PFC pyramidal neurons were suppressed to a larger extent by hD4.7 than hD4.4 activation. Direct stimulation of NMDARs with the partial agonist d-cycloserine prevented the NMDAR hypofunction induced by hD4.7 activation. Moreover, hD4.7-expressing mice exhibited the increased exploratory and novelty seeking behaviors, mimicking the phenotypic hallmark of human ADHD. d-cycloserine administration ameliorated the ADHD-like behaviors in hD4.7-expressing mice. Our results suggest that over-suppression of NMDAR function may underlie the role of hD4.7 in ADHD, and enhancing NMDAR signaling may be a viable therapeutic strategy to ADHD humans carrying the D4.7 allele.
Collapse
Affiliation(s)
- Luye Qin
- Dept. of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Wenhua Liu
- Dept. of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA; School of Life Science, Zhaoqing University, Zhaoqing 526061, China
| | - Kaijie Ma
- Dept. of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Jing Wei
- Dept. of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Ping Zhong
- Dept. of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Kei Cho
- The MRC Centre for Synaptic Plasticity, University of Bristol Whitson, Bristol, UK
| | - Zhen Yan
- Dept. of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA.
| |
Collapse
|
310
|
Smith M, Piehler T, Benjamin R, Farizatto KL, Pait MC, Almeida MF, Ghukasyan VV, Bahr BA. Blast waves from detonated military explosive reduce GluR1 and synaptophysin levels in hippocampal slice cultures. Exp Neurol 2016; 286:107-115. [PMID: 27720798 DOI: 10.1016/j.expneurol.2016.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 09/27/2016] [Accepted: 10/04/2016] [Indexed: 12/15/2022]
Abstract
Explosives create shockwaves that cause blast-induced neurotrauma, one of the most common types of traumatic brain injury (TBI) linked to military service. Blast-induced TBIs are often associated with reduced cognitive and behavioral functions due to a variety of factors. To study the direct effects of military explosive blasts on brain tissue, we removed systemic factors by utilizing rat hippocampal slice cultures. The long-term slice cultures were briefly sealed air-tight in serum-free medium, lowered into a 37°C water-filled tank, and small 1.7-gram assemblies of cyclotrimethylene trinitramine (RDX) were detonated 15cm outside the tank, creating a distinct shockwave recorded at the culture plate position. Compared to control mock-treated groups of slices that received equal submerge time, 1-3 blast impacts caused a dose-dependent reduction in the AMPA receptor subunit GluR1. While only a small reduction was found in hippocampal slices exposed to a single RDX blast and harvested 1-2days later, slices that received two consecutive RDX blasts 4min apart exhibited a 26-40% reduction in GluR1, and the receptor subunit was further reduced by 64-72% after three consecutive blasts. Such loss correlated with increased levels of HDAC2, a histone deacetylase implicated in stress-induced reduction of glutamatergic transmission. No evidence of synaptic marker recovery was found at 72h post-blast. The presynaptic marker synaptophysin was found to have similar susceptibility as GluR1 to the multiple explosive detonations. In contrast to the synaptic protein reductions, actin levels were unchanged, spectrin breakdown was not detected, and Fluoro-Jade B staining found no indication of degenerating neurons in slices exposed to three RDX blasts, suggesting that small, sub-lethal explosives are capable of producing selective alterations to synaptic integrity. Together, these results indicate that blast waves from military explosive cause signs of synaptic compromise without producing severe neurodegeneration, perhaps explaining the cognitive and behavioral changes in those blast-induced TBI sufferers that have no detectable neuropathology.
Collapse
Affiliation(s)
- Marquitta Smith
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Thuvan Piehler
- U.S. Army Research Laboratory, Aberdeen Proving Ground, MD 21005, USA
| | - Richard Benjamin
- U.S. Army Research Laboratory, Aberdeen Proving Ground, MD 21005, USA
| | - Karen L Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Morgan C Pait
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Vladimir V Ghukasyan
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA.
| |
Collapse
|
311
|
Lentinan produces a robust antidepressant-like effect via enhancing the prefrontal Dectin-1/AMPA receptor signaling pathway. Behav Brain Res 2016; 317:263-271. [PMID: 27693847 DOI: 10.1016/j.bbr.2016.09.062] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/24/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022]
Abstract
Lentinan (LNT) is an immune regulator and its potential and mechanism for the treatment of mood disorder is of our interest. Dectin-1 is a β-glucan (including LNT) receptor that regulates immune functions in many immune cell types. Cumulative evidence has suggested that the glutamatergic system seems to play an important role in the treatment of depression. Here, we studied the antidepressant-like effects of LNT and its therapeutical target in regulating the functions of AMPA receptors. We found that 60min treatment with LNT leads to a significant antidepressant-like effect in the tail suspension test (TST) and the forced swim test (FST) in mice. The antidepressant-like effects of LNT in TST and FST remained after 1day or 5days of injections. Additionally, LNT did not show a hyperactive effect in the open field test. Dectin-1 receptor levels were increased after LNT treatment for 5days and the specific Dectin-1 inhibitor laminarin was able to block the antidepressant-like effects of LNT. After 5days of treatment, LNT enhanced p-GluR1 (S845) in the prefrontal cortex (PFC); however, the total GluR1, GluR2, and GluR3 expression levels remained unchanged. We also found that the AMPA-specific blocker GYKI 52466 was able to block the antidepressant-like effects of LNT. This study identified LNT as a novel antidepressant with clinical potential and a new antidepressant mechanism for regulating prefrontal Dectin-1/AMPA receptor signaling.
Collapse
|
312
|
Wang W, Guo H, Zhang SX, Li J, Cheng K, Bai SJ, Yang DY, Wang HY, Liang ZH, Liao L, Sun L, Xie P. Targeted Metabolomic Pathway Analysis and Validation Revealed Glutamatergic Disorder in the Prefrontal Cortex among the Chronic Social Defeat Stress Mice Model of Depression. J Proteome Res 2016; 15:3784-3792. [PMID: 27599184 DOI: 10.1021/acs.jproteome.6b00577] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Major depressive disorder (MDD) is a severe psychiatric disease that has critically affected life quality for millions of people. Chronic stress is gradually recognized as a primary pathogenesis risk factor of MDD. Despite the remarkable progress in mechanism research, the pathogenesis mechanism of MDD is still not well understood. Therefore, we conducted a liquid chromatography-tandem mass spectrometry (LC-MS/MS) detection of 25 major metabolites of tryptophanic, GABAergic, and catecholaminergic pathways in the prefontal cortex (PFC) of mice in chronic social defeat stress (CSDS). The depressed mice exhibit significant reduction of glutamate in the GABAergic pathway and an increase of L-DOPA and vanillylmandelic acid in catecholaminergic pathways. The data of real-time-quantitative polymerase chain reaction (RT-qPCR) and Western blotting analysis revealed an altered level of glutamatergic circuitry. The metabolomic and molecular data reveal that the glutamatergic disorder in mice shed lights to reveal a mechanism on depression-like and stress resilient phenotype.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University , Chongqing 402460, China.,Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China
| | - Hua Guo
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University , Chongqing 402460, China.,Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China
| | - Shu-Xiao Zhang
- Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China
| | - Juan Li
- Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China.,Department of Neurology, the First Affiliated Hospital of Chongqing Medical University , Chongqing 400016, China
| | - Ke Cheng
- Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China.,Department of Neurology, the First Affiliated Hospital of Chongqing Medical University , Chongqing 400016, China
| | - Shun-Jie Bai
- Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China
| | - De-Yu Yang
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University , Chongqing 402460, China
| | - Hai-Yang Wang
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University , Chongqing 402460, China.,Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China
| | - Zi-Hong Liang
- Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China.,Department of Neurology, the First Affiliated Hospital of Chongqing Medical University , Chongqing 400016, China
| | - Li Liao
- Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China
| | - Lin Sun
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University , Chongqing 400016, China
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University , Chongqing 402460, China.,Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing 400016, China.,Chongqing Key Laboratory of Neurobiology , Chongqing 400016, China.,Department of Neurology, the First Affiliated Hospital of Chongqing Medical University , Chongqing 400016, China
| |
Collapse
|
313
|
Ren Z, Pribiag H, Jefferson SJ, Shorey M, Fuchs T, Stellwagen D, Luscher B. Bidirectional Homeostatic Regulation of a Depression-Related Brain State by Gamma-Aminobutyric Acidergic Deficits and Ketamine Treatment. Biol Psychiatry 2016; 80:457-468. [PMID: 27062563 PMCID: PMC4983262 DOI: 10.1016/j.biopsych.2016.02.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/06/2016] [Accepted: 02/08/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Major depressive disorder is increasingly recognized to involve functional deficits in both gamma-aminobutyric acid (GABA)ergic and glutamatergic synaptic transmission. To elucidate the relationship between these phenotypes, we used GABAA receptor γ2 subunit heterozygous (γ2(+/-)) mice, which we previously characterized as a model animal with construct, face, and predictive validity for major depressive disorder. METHODS To assess possible consequences of GABAergic deficits on glutamatergic transmission, we quantitated the cell surface expression of N-methyl-D-aspartate (NMDA)-type and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)-type glutamate receptors and the function of synapses in the hippocampus and medial prefrontal cortex of γ2(+/-) mice. We also analyzed the effects of an acute dose of the experimental antidepressant ketamine on all these parameters in γ2(+/-) versus wild-type mice. RESULTS Modest defects in GABAergic synaptic transmission of γ2(+/-) mice resulted in a strikingly prominent homeostatic-like reduction in the cell surface expression of NMDA-type and AMPA-type glutamate receptors, along with prominent functional impairment of glutamatergic synapses in the hippocampus and medial prefrontal cortex. A single subanesthetic dose of ketamine normalized glutamate receptor expression and synaptic function of γ2(+/-) mice to wild-type levels for a prolonged period, along with antidepressant-like behavioral consequences selectively in γ2(+/-) mice. The GABAergic synapses of γ2(+/-) mice were potentiated by ketamine in parallel but only in the medial prefrontal cortex. CONCLUSIONS Depressive-like brain states that are caused by GABAergic deficits involve a homeostatic-like reduction of glutamatergic transmission that is reversible by an acute, subanesthetic dose of ketamine, along with regionally selective potentiation of GABAergic synapses. The data merge the GABAergic and glutamatergic deficit hypotheses of major depressive disorder.
Collapse
Affiliation(s)
- Zhen Ren
- Department of Biology, Pennsylvania State University, University Park, PA 16802
| | - Horia Pribiag
- Center for Research in Neuroscience, McGill University, Montreal General Hospital, L7-132, 1650 Cedar Av, Montreal, QC H3G 1A4, Canada
| | - Sarah J. Jefferson
- Department of Biology, Pennsylvania State University, University Park, PA 16802
| | - Matthew Shorey
- Department of Biology, Pennsylvania State University, University Park, PA 16802
| | - Thomas Fuchs
- Department of Biology, Pennsylvania State University, University Park, PA 16802
| | - David Stellwagen
- Center for Research in Neuroscience, McGill University, Montreal General Hospital, L7-132, 1650 Cedar Av, Montreal, QC H3G 1A4, Canada
| | - Bernhard Luscher
- Departments of Biology, Pennsylvania State University, University Park, Pennsylvania; Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania; Center for Molecular Investigation of Neurological Disorders, Pennsylvania State University, University Park, Pennsylvania.
| |
Collapse
|
314
|
EphB2 in the Medial Prefrontal Cortex Regulates Vulnerability to Stress. Neuropsychopharmacology 2016; 41:2541-56. [PMID: 27103064 PMCID: PMC4987853 DOI: 10.1038/npp.2016.58] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 03/28/2016] [Accepted: 04/12/2016] [Indexed: 01/23/2023]
Abstract
The ephrin B2 (EphB2) receptor is a tyrosine kinase receptor that is associated with synaptic development and maturation. It has recently been implicated in cognitive deficits and anxiety. However, still unknown is the involvement of EphB2 in the vulnerability to stress. In the present study, we observed decreases in EphB2 levels and their downstream molecules in the medial prefrontal cortex (mPFC) but not in the orbitofrontal cortex (OFC) in mice that were susceptible to chronic social defeat stress. The activation of EphB2 receptors with EphrinB1-Fc in the mPFC produced stress-resistant and antidepressant-like behavioral effects in susceptible mice that lasted for at least 10 days. EphB2 receptor knockdown by short-hairpin RNA in the mPFC increased the susceptibility to stress and induced depressive-like behaviors in a subthreshold chronic social defeat stress paradigm. These behavioral effects were associated with changes in the phosphorylation of cofilin and membrane α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking and the expression of some synaptic proteins in the mPFC. We also found that EphB2 regulated stress-induced spine remodeling in the mPFC. Altogether, these results indicate that EphB2 is a critical regulator of stress vulnerability and might be a potential target for the treatment of depression.
Collapse
|
315
|
Abdallah CG, Adams TG, Kelmendi B, Esterlis I, Sanacora G, Krystal JH. KETAMINE'S MECHANISM OF ACTION: A PATH TO RAPID-ACTING ANTIDEPRESSANTS. Depress Anxiety 2016; 33:689-97. [PMID: 27062302 PMCID: PMC4961540 DOI: 10.1002/da.22501] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/16/2016] [Accepted: 03/01/2016] [Indexed: 12/11/2022] Open
Abstract
Major depressive disorder (MDD) is a common and debilitating psychiatric disorder. Traditional antidepressants are of limited efficacy and take weeks to months to yield full therapeutic effects. Thus, there is a clear need for effective rapid-acting antidepressant medications. The N-methyl-d-aspartate receptor (NMDA-R) antagonist, ketamine, has received a great deal of attention over the last 20 years due to the discovery that a single subanesthetic dose leads to a rapid antidepressant effect in individuals with treatment-resistant depression. Animal and human research suggest that ketamine's antidepressant effects are mediated by a glutamate surge that leads to a cascade of events that result in synaptogenesis and reversal of the negative effects of chronic stress and depression, particularly within the prefrontal cortex (PFC). Preclinical and clinical data have provided compelling insights into the mechanisms underlying the rapid-acting antidepressant effects of ketamine. This review discusses stress-related neurobiology of depression and the safety, tolerability, and efficacy of ketamine for MDD, along with a review of ketamine's mechanism of action and prospective predictors of treatment response. Research limitations and future clinical prospects are also discussed.
Collapse
Affiliation(s)
- Chadi G. Abdallah
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
- Clinical Neuroscience Division, Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT
| | - Thomas G. Adams
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
- Clinical Neuroscience Division, Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT
| | - Benjamin Kelmendi
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
- Clinical Neuroscience Division, Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT
| | - Irina Esterlis
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
- Clinical Neuroscience Division, Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT
| | - Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
- Clinical Neuroscience Division, Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT
- Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT
| | - John H. Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
- Clinical Neuroscience Division, Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT
- Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT
| |
Collapse
|
316
|
Stelly CE, Pomrenze MB, Cook JB, Morikawa H. Repeated social defeat stress enhances glutamatergic synaptic plasticity in the VTA and cocaine place conditioning. eLife 2016; 5. [PMID: 27374604 PMCID: PMC4931908 DOI: 10.7554/elife.15448] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/07/2016] [Indexed: 11/13/2022] Open
Abstract
Enduring memories of sensory cues associated with drug intake drive addiction. It is well known that stressful experiences increase addiction vulnerability. However, it is not clear how repeated stress promotes learning of cue-drug associations, as repeated stress generally impairs learning and memory processes unrelated to stressful experiences. Here, we show that repeated social defeat stress in rats causes persistent enhancement of long-term potentiation (LTP) of NMDA receptor-mediated glutamatergic transmission in the ventral tegmental area (VTA). Protein kinase A-dependent increase in the potency of inositol 1,4,5-triphosphate-induced Ca2+ signaling underlies LTP facilitation. Notably, defeated rats display enhanced learning of contextual cues paired with cocaine experience assessed using a conditioned place preference (CPP) paradigm. Enhancement of LTP in the VTA and cocaine CPP in behaving rats both require glucocorticoid receptor activation during defeat episodes. These findings suggest that enhanced glutamatergic plasticity in the VTA may contribute, at least partially, to increased addiction vulnerability following repeated stressful experiences. DOI:http://dx.doi.org/10.7554/eLife.15448.001 Daily stress increases the likelihood that people who take drugs will become addicted. A very early step in the development of addiction is learning that certain people, places, or paraphernalia are associated with obtaining drugs. These ‘cues’ – drug dealers, bars, cigarette advertisements, etc. – become powerful motivators to seek out drugs and can trigger relapse in recovering addicts. It is thought that learning happens when synapses (the connections between neurons in the brain) that relay information about particular cues become stronger. However, it is not clear how stress promotes the learning of cue-drug associations. Stelly et al. investigated whether repeated episodes of stress make it easier to strengthen synapses on dopamine neurons, which are involved in processing rewards and addiction. For the experiments, rats were repeatedly exposed to a stressful situation – an encounter with an unfamiliar aggressive rat – every day for five days. Stelly et al. found that these stressed rats formed stronger associations between the drug cocaine and the place where they were given the drug (the cue). Furthermore, a mechanism that strengthens synapses was more sensitive in the stressed rats than in unstressed rats. These changes persisted for 10-30 days after the stressful situation, suggesting that stress might begin a period of time during which the individual is more vulnerable to addiction. The experiments also show that a hormone called corticosterone – which is released during stressful experiences – is necessary for stress to trigger the changes in the synapses and behavior of the rats. However, corticosterone must work with other factors because giving this hormone to unstressed rats was not sufficient to trigger the changes seen in the stressed rats. Future experiments will investigate what these other stress factors are and how they work together with corticosterone. DOI:http://dx.doi.org/10.7554/eLife.15448.002
Collapse
Affiliation(s)
- Claire E Stelly
- Department of Neuroscience, University of Texas, Austin, United States.,Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, United States
| | - Matthew B Pomrenze
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, United States.,Division of Pharmacology and Toxicology, University of Texas, Austin, United States
| | - Jason B Cook
- Department of Neuroscience, University of Texas, Austin, United States.,Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, United States
| | - Hitoshi Morikawa
- Department of Neuroscience, University of Texas, Austin, United States.,Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, United States
| |
Collapse
|
317
|
Milior G, Lecours C, Samson L, Bisht K, Poggini S, Pagani F, Deflorio C, Lauro C, Alboni S, Limatola C, Branchi I, Tremblay ME, Maggi L. Fractalkine receptor deficiency impairs microglial and neuronal responsiveness to chronic stress. Brain Behav Immun 2016; 55:114-125. [PMID: 26231972 DOI: 10.1016/j.bbi.2015.07.024] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/25/2015] [Accepted: 07/26/2015] [Indexed: 12/25/2022] Open
Abstract
Chronic stress is one of the most relevant triggering factors for major depression. Microglial cells are highly sensitive to stress and, more generally, to environmental challenges. However, the role of these brain immune cells in mediating the effects of stress is still unclear. Fractalkine signaling - which comprises the chemokine CX3CL1, mainly expressed by neurons, and its receptor CX3CR1, almost exclusively present on microglia in the healthy brain - has been reported to critically regulate microglial activity. Here, we investigated whether interfering with microglial function by deleting the Cx3cr1 gene affects the brain's response to chronic stress. To this purpose, we housed Cx3cr1 knockout and wild-type adult mice in either control or stressful environments for 2weeks, and investigated the consequences on microglial phenotype and interactions with synapses, synaptic transmission, behavioral response and corticosterone levels. Our results show that hampering neuron-microglia communication via the CX3CR1-CX3CL1 pathway prevents the effects of chronic unpredictable stress on microglial function, short- and long-term neuronal plasticity and depressive-like behavior. Overall, the present findings suggest that microglia-regulated mechanisms may underlie the differential susceptibility to stress and consequently the vulnerability to diseases triggered by the experience of stressful events, such as major depression.
Collapse
Affiliation(s)
- Giampaolo Milior
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| | - Cynthia Lecours
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada
| | - Louis Samson
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada
| | - Kanchan Bisht
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada
| | - Silvia Poggini
- Section of Behavioural Neurosciences, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Pagani
- Center for Life Nanoscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | - Cristina Deflorio
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy; Département de Neuroscience, Institut Pasteur, Unité Neurobiologie Intégrative des Systèmes Cholinergiques, Paris Cedex 15, Paris, France
| | - Clotilde Lauro
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy
| | - Igor Branchi
- Section of Behavioural Neurosciences, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada.
| | - Laura Maggi
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| |
Collapse
|
318
|
Histone Modification of Nedd4 Ubiquitin Ligase Controls the Loss of AMPA Receptors and Cognitive Impairment Induced by Repeated Stress. J Neurosci 2016; 36:2119-30. [PMID: 26888924 DOI: 10.1523/jneurosci.3056-15.2016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
UNLABELLED Stress and the major stress hormone corticosterone induce profound influences in the brain. Altered histone modification and transcriptional dysfunction have been implicated in stress-related mental disorders. We previously found that repeated stress caused an impairment of prefrontal cortex (PFC)-mediated cognitive functions by increasing the ubiquitination and degradation of AMPA-type glutamate receptors via a mechanism depending on the E3 ubiquitin ligase Nedd4. Here, we demonstrated that in PFC of repeatedly stressed rats, active glucocorticoid receptor had the increased binding to the glucocorticoid response element of histone deacetylase 2 (HDAC2) promoter, resulting in the upregulation of HDAC2. Inhibition or knock-down of HDAC2 blocked the stress-induced impairment of synaptic transmission, AMPAR expression, and recognition memory. Furthermore, we found that, in stressed animals, the HDAC2-dependent downregulation of histone methyltransferase Ehmt2 (G9a) led to the loss of repressive histone methylation at the Nedd4-1 promoter and the transcriptional activation of Nedd4. These results have provided an epigenetic mechanism and a potential treatment strategy for the detrimental effects of chronic stress. SIGNIFICANCE STATEMENT Prolonged stress exposure can induce altered histone modification and transcriptional dysfunction, which may underlie the profound influence of stress in regulating brain functions. We report an important finding about the epigenetic mechanism controlling the detrimental effects of repeated stress on synaptic transmission and cognitive function. First, it has revealed the stress-induced alteration of key epigenetic regulators HDAC2 and Ehmt2, which determines the synaptic and behavioral effects of repeated stress. Second, it has uncovered the stress-induced histone modification of the target gene Nedd4, an E3 ligase that is critically involved in the ubiquitination and degradation of AMPA receptors and cognition. Third, it has provided the epigenetic approach, HDAC2 inhibition or knock-down, to rescue synaptic and cognitive functions in stressed animals.
Collapse
|
319
|
Hultman R, Mague SD, Li Q, Katz BM, Michel N, Lin L, Wang J, David LK, Blount C, Chandy R, Carlson D, Ulrich K, Carin L, Dunson D, Kumar S, Deisseroth K, Moore SD, Dzirasa K. Dysregulation of Prefrontal Cortex-Mediated Slow-Evolving Limbic Dynamics Drives Stress-Induced Emotional Pathology. Neuron 2016; 91:439-52. [PMID: 27346529 DOI: 10.1016/j.neuron.2016.05.038] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 04/21/2016] [Accepted: 05/25/2016] [Indexed: 11/28/2022]
Abstract
Circuits distributed across cortico-limbic brain regions compose the networks that mediate emotional behavior. The prefrontal cortex (PFC) regulates ultraslow (<1 Hz) dynamics across these networks, and PFC dysfunction is implicated in stress-related illnesses including major depressive disorder (MDD). To uncover the mechanism whereby stress-induced changes in PFC circuitry alter emotional networks to yield pathology, we used a multi-disciplinary approach including in vivo recordings in mice and chronic social defeat stress. Our network model, inferred using machine learning, linked stress-induced behavioral pathology to the capacity of PFC to synchronize amygdala and VTA activity. Direct stimulation of PFC-amygdala circuitry with DREADDs normalized PFC-dependent limbic synchrony in stress-susceptible animals and restored normal behavior. In addition to providing insights into MDD mechanisms, our findings demonstrate an interdisciplinary approach that can be used to identify the large-scale network changes that underlie complex emotional pathologies and the specific network nodes that can be used to develop targeted interventions.
Collapse
Affiliation(s)
- Rainbo Hultman
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Stephen D Mague
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Qiang Li
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Brittany M Katz
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Nadine Michel
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Lizhen Lin
- Department of Statistics and Data Science, University of Texas at Austin, Austin, TX 78712, USA
| | - Joyce Wang
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Lisa K David
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Cameron Blount
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Rithi Chandy
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - David Carlson
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 22208, USA
| | - Kyle Ulrich
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 22208, USA
| | - Lawrence Carin
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 22208, USA
| | - David Dunson
- Department of Statistical Sciences, Duke University, Durham, NC 22208, USA
| | - Sunil Kumar
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Karl Deisseroth
- Departments of Bioengineering and Psychiatry and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Scott D Moore
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Kafui Dzirasa
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA; Center for Neuroengineering, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences, Duke University Medical Center, Durham, NC 27710, USA; Departments of Biomedical Engineering and Neurobiology, Duke University, Durham, NC 22208, USA.
| |
Collapse
|
320
|
Bao H, Ran P, Zhu M, Sun L, Li B, Hou Y, Nie J, Shan L, Li H, Zheng S, Xu X, Xiao C, Du J. The Prefrontal Dectin-1/AMPA Receptor Signaling Pathway Mediates The Robust and Prolonged Antidepressant Effect of Proteo-β-Glucan from Maitake. Sci Rep 2016; 6:28395. [PMID: 27329257 PMCID: PMC4916609 DOI: 10.1038/srep28395] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/03/2016] [Indexed: 01/13/2023] Open
Abstract
Proteo-β-glucan from Maitake (PGM) is a strong immune regulator, and its receptor is called Dectin-1. Cumulative evidence suggests that AMPA receptors are important for the treatment of depression. Here, we report that PGM treatment leads to a significant antidepressant effect in the tail suspension test and forced swim test after sixty minutes of treatment in mice. After five consecutive days of PGM treatment, this antidepressant effect remained. PGM treatment did not show a hyperactive effect in the open field test. PGM significantly enhanced the expression of its receptor Dectin-1, as well as p-GluA1(S845) and GluA1, but not GluA2 or GluA3 in the prefrontal cortex (PFC) after five days of treatment. The Dectin-1 inhibitor Laminarin was able to block the antidepressant effect of PGM. At the synapses of PFC, PGM treatment significantly up-regulated the p-GluA1(S845), GluA1, GluA2, and GluA3 levels. Moreover, PGM’s antidepressant effects and the increase of p-GluA1(S845)/GluA1 lasted for 3 days after stopping treatment. The AMPA-specific antagonist GYKI 52466 was able to block the antidepressant effect of PGM. This study identified PGM as a novel antidepressant with clinical potential and a new antidepressant mechanism for regulating prefrontal Dectin-1/AMPA receptor signalling.
Collapse
Affiliation(s)
- Hongkun Bao
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Pengzhan Ran
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Ming Zhu
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Lijuan Sun
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Bai Li
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Yangyang Hou
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Jun Nie
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Liping Shan
- Beijing Gragen Biotechnology Co. Ltd., Beijing, P. R. China
| | - Hongliang Li
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Shangyong Zheng
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Xiufeng Xu
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Chunjie Xiao
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| | - Jing Du
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, Yunnan, P. R. China.,School of Medicine, Yunnan University, Kunming, Yunnan, P. R. China
| |
Collapse
|
321
|
Goff DC, Romero K, Paul J, Mercedes Perez-Rodriguez M, Crandall D, Potkin SG. Biomarkers for drug development in early psychosis: Current issues and promising directions. Eur Neuropsychopharmacol 2016; 26:923-37. [PMID: 27005595 DOI: 10.1016/j.euroneuro.2016.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/20/2016] [Accepted: 01/23/2016] [Indexed: 12/14/2022]
Abstract
A major goal of current research in schizophrenia is to understand the biology underlying onset and early progression and to develop interventions that modify these processes. Biomarkers can play a critical role in identifying disease state, factors contributing to underlying progression, as well as predicting and monitoring response to treatment. Once biomarker-based therapeutics are established, biomarkers can guide treatment selection. It is increasingly clear that a wide range of potential biomarkers should be examined in schizophrenia, given the large number of genetic and environmental factors that have been identified as risk factors. New models for analysis of biomarkers are needed that represent the central nervous system as a highly complex, dynamic, and interactive system. Many tools are available with which to study relevant brain chemistry, but most are indirect measures and represent only a small fraction of the potential etiologic factors contributing to the molecular, structural and functional components of schizophrenia. This review represents the work of the International Society for CNS Clinical Trials and Methodology (ISCTM) Biomarkers Working Group. It discusses advantages and disadvantages of different categories of biomarkers and provides a summary of evidence that biomarkers representing inflammation, oxidative stress, endocannabinoids, glucocorticoid, and biogenic amines systems are dysregulated and potentially interactive in early phase schizophrenia. As has been recently demonstrated in several neurodevelopmental and neurodegenerative disorders, a multi-modal, longitudinal strategy involving a diverse array of biomarkers and new approaches to statistical modeling are needed to improve early interventions based on the fuller understanding.
Collapse
Affiliation(s)
| | | | - Jeffrey Paul
- Astellas Pharma Global Development, Northbrook, IL, USA
| | | | | | | |
Collapse
|
322
|
Yuen EY, Wei J, Yan Z. Estrogen in prefrontal cortex blocks stress-induced cognitive impairments in female rats. J Steroid Biochem Mol Biol 2016; 160:221-6. [PMID: 26321384 PMCID: PMC4769981 DOI: 10.1016/j.jsbmb.2015.08.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/20/2015] [Accepted: 08/24/2015] [Indexed: 11/15/2022]
Abstract
Animal and human studies have found that males and females show distinct stress responses. Recent studies suggest the contribution of estrogen in the brain to this sexual dimorphism. Repeated stress has been found to impair cognitive behaviors via suppressing glutamatergic transmission and glutamate receptor surface expression in pyramidal neurons of prefrontal cortex (PFC) in male rats. On the contrary, female rats exposed to the same stress paradigms show normal synaptic function and PFC-mediated cognition. The level of aromatase, the enzyme for the biosynthesis of estrogen, is significantly higher in the PFC of females than males. The stress-induced glutamatergic deficits and memory impairment are unmasked by blocking estrogen receptors or aromatase in females, suggesting a protective role of estrogen against the detrimental effects of repeated stress.
Collapse
Affiliation(s)
- Eunice Y Yuen
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Jing Wei
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA.
| |
Collapse
|
323
|
South M, Stephenson KG, Nielson CA, Maisel M, Top DN, Kirwan CB. Overactive Pattern Separation Memory Associated with Negative Emotionality in Adults Diagnosed with Autism Spectrum Disorder. J Autism Dev Disord 2016; 45:3458-67. [PMID: 26231206 DOI: 10.1007/s10803-015-2547-x] [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] [Indexed: 10/23/2022]
Abstract
Bowler et al. (Journal of Autism and Developmental Disorders 44(9):2355-2362. doi:10.1007/s10803-014-2105-y, 2014) have suggested that a specific memory impairment in autism spectrum disorders (ASD) arises from hippocampal failure to consolidate multiple related pieces of information. Twenty-four adults diagnosed with ASD and matched healthy controls completed a pattern separation memory task that is known to critically depend on hippocampal involvement. They additionally completed questionnaires regarding anxiety, depression, and behavioral motivation. Specific deficits in pattern separation were significantly correlated with negative emotionality; the best predictor of memory deficit was from a measure of achievement motivation that has also been associated with hyperactivity and impulsivity. In the context of impaired emotion regulation in ASD, there is a need for integrated cognitive, affective, and neural systems approaches to build targeted interventions.
Collapse
Affiliation(s)
- M South
- Department of Psychology, Brigham Young University, 245 TLRB, Provo, UT, 84602, USA. .,Neuroscience Center, Brigham Young University, Provo, UT, USA.
| | - K G Stephenson
- Department of Psychology, Brigham Young University, 245 TLRB, Provo, UT, 84602, USA
| | - C A Nielson
- Neuroscience Center, Brigham Young University, Provo, UT, USA
| | - M Maisel
- Department of Psychology, Brigham Young University, 245 TLRB, Provo, UT, 84602, USA
| | - D N Top
- Department of Psychology, Brigham Young University, 245 TLRB, Provo, UT, 84602, USA
| | - C B Kirwan
- Department of Psychology, Brigham Young University, 245 TLRB, Provo, UT, 84602, USA.,Neuroscience Center, Brigham Young University, Provo, UT, USA
| |
Collapse
|
324
|
Xu A, Cui S, Wang JH. Incoordination among Subcellular Compartments Is Associated with Depression-Like Behavior Induced by Chronic Mild Stress. Int J Neuropsychopharmacol 2016; 19:pyv122. [PMID: 26506857 PMCID: PMC4886664 DOI: 10.1093/ijnp/pyv122] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/16/2015] [Accepted: 10/23/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Major depressive disorder is characterized as persistent low mood. A chronically stressful life in genetically susceptible individuals is presumably the major etiology that leads to dysfunctions of monoamine and hypothalamus-pituitary-adrenal axis. These pathogenic factors cause neuron atrophy in the limbic system for major depressive disorder. Cell-specific pathophysiology is unclear, so we investigated prelimbic cortical GABAergic neurons and their interaction with glutamatergic neurons in depression-like mice. METHODS Mice were treated with chronic unpredictable mild stress for 3 weeks until they expressed depression-like behaviors confirmed by sucrose preference, Y-maze, and forced swimming tests. The structures and functions of GABAergic and glutamatergic units in prelimbic cortices were studied by cell imaging and electrophysiology in chronic unpredictable mild stress-induced depression mice vs controls. RESULTS In depression-like mice, prelimbic cortical GABAergic neurons show incoordination among the subcellular compartments, such as decreased excitability and synaptic outputs as well as increased reception from excitatory inputs. GABAergic synapses on glutamatergic cells demonstrate decreased presynaptic innervation and increased postsynaptic responsiveness. CONCLUSIONS Chronic unpredictable mild stress-induced incoordination in prelimbic cortical GABAergic and glutamatergic neurons dysregulates their target neurons, which may be the pathological basis for depressive mood. The rebalance of compatibility among subcellular compartments would be an ideal strategy to treat neural disorders.
Collapse
MESH Headings
- Animals
- Behavior, Animal
- Chronic Disease
- Depressive Disorder, Major/etiology
- Depressive Disorder, Major/metabolism
- Depressive Disorder, Major/physiopathology
- Depressive Disorder, Major/psychology
- Dietary Sucrose/administration & dosage
- Disease Models, Animal
- Excitatory Postsynaptic Potentials
- Food Preferences
- GABAergic Neurons/metabolism
- Glutamic Acid/metabolism
- In Vitro Techniques
- Inhibitory Postsynaptic Potentials
- Male
- Maze Learning
- Mice, Transgenic
- Motor Activity
- Neural Inhibition
- Neural Pathways/metabolism
- Neural Pathways/physiopathology
- Prefrontal Cortex/metabolism
- Prefrontal Cortex/physiopathology
- Stress, Psychological/complications
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
- Stress, Psychological/psychology
- Swimming
- Time Factors
Collapse
Affiliation(s)
- Aiping Xu
- College of Life Science, University of Science and Technology of China, Hefei Anhui, China (Ms Xu and Dr Wang); State Key Lab of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (Ms Xu, Ms Cui, and Dr Wang); University of Chinese Academy of Sciences, Beijing, China (Dr Wang)
| | - Shan Cui
- College of Life Science, University of Science and Technology of China, Hefei Anhui, China (Ms Xu and Dr Wang); State Key Lab of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (Ms Xu, Ms Cui, and Dr Wang); University of Chinese Academy of Sciences, Beijing, China (Dr Wang)
| | - Jin-Hui Wang
- College of Life Science, University of Science and Technology of China, Hefei Anhui, China (Ms Xu and Dr Wang); State Key Lab of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (Ms Xu, Ms Cui, and Dr Wang); University of Chinese Academy of Sciences, Beijing, China (Dr Wang).
| |
Collapse
|
325
|
Antidepressant-like effect of cannabidiol injection into the ventral medial prefrontal cortex—Possible involvement of 5-HT1A and CB1 receptors. Behav Brain Res 2016; 303:218-27. [DOI: 10.1016/j.bbr.2016.01.033] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 01/12/2016] [Accepted: 01/15/2016] [Indexed: 01/24/2023]
|
326
|
Kos A, Olde Loohuis N, Meinhardt J, van Bokhoven H, Kaplan BB, Martens GJ, Aschrafi A. MicroRNA-181 promotes synaptogenesis and attenuates axonal outgrowth in cortical neurons. Cell Mol Life Sci 2016; 73:3555-67. [PMID: 27017280 DOI: 10.1007/s00018-016-2179-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 02/13/2016] [Accepted: 03/10/2016] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRs) are non-coding gene transcripts abundantly expressed in both the developing and adult mammalian brain. They act as important modulators of complex gene regulatory networks during neuronal development and plasticity. miR-181c is highly abundant in cerebellar cortex and its expression is increased in autism patients as well as in an animal model of autism. To systematically identify putative targets of miR-181c, we repressed this miR in growing cortical neurons and found over 70 differentially expressed target genes using transcriptome profiling. Pathway analysis showed that the miR-181c-modulated genes converge on signaling cascades relevant to neurite and synapse developmental processes. To experimentally examine the significance of these data, we inhibited miR-181c during rat cortical neuronal maturation in vitro; this loss-of miR-181c function resulted in enhanced neurite sprouting and reduced synaptogenesis. Collectively, our findings suggest that miR-181c is a modulator of gene networks associated with cortical neuronal maturation.
Collapse
Affiliation(s)
- Aron Kos
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Nikkie Olde Loohuis
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Julia Meinhardt
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Barry B Kaplan
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Gerard J Martens
- Department of Molecular Animal Physiology, Radboud University, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Armaz Aschrafi
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands.
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
327
|
Soares JM, Marques P, Magalhães R, Santos NC, Sousa N. The association between stress and mood across the adult lifespan on default mode network. Brain Struct Funct 2016; 222:101-112. [PMID: 26971253 PMCID: PMC5225218 DOI: 10.1007/s00429-016-1203-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/19/2016] [Indexed: 01/20/2023]
Abstract
Aging of brain structure and function is a complex process characterized by high inter- and intra-individual variability. Such variability may arise from the interaction of multiple factors, including exposure to stressful experience and mood variation, across the lifespan. Using a multimodal neuroimaging and neurocognitive approach, we investigated the association of stress, mood and their interaction, in the structure and function of the default mode network (DMN), both during rest and task-induced deactivation, throughout the adult lifespan. Data confirmed a decreased functional connectivity (FC) and task-induced deactivation of the DMN during the aging process and in subjects with lower mood; on the contrary, an increased FC was observed in subjects with higher perceived stress. Surprisingly, the association of aging with DMN was altered by stress and mood in specific regions. An increased difficulty to deactivate the DMN was noted in older participants with lower mood, contrasting with an increased deactivation in individuals presenting high stress, independently of their mood levels, with aging. Interestingly, this constant interaction across aging was globally most significant in the combination of high stress levels with a more depressed mood state, both during resting state and task-induced deactivations. The present results contribute to characterize the spectrum of FC and deactivation patterns of the DMN, highlighting the crucial association of stress and mood levels, during the adult aging process. These combinatorial approaches may help to understand the heterogeneity of the aging process in brain structure and function and several states that may lead to neuropsychiatric disorders.
Collapse
Affiliation(s)
- José Miguel Soares
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal.,Clinical Academic Center - Braga, Braga, Portugal
| | - Paulo Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal.,Clinical Academic Center - Braga, Braga, Portugal
| | - Ricardo Magalhães
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal.,Clinical Academic Center - Braga, Braga, Portugal
| | - Nadine Correia Santos
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal.,Clinical Academic Center - Braga, Braga, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal. .,Clinical Academic Center - Braga, Braga, Portugal.
| |
Collapse
|
328
|
Yang Y, Zhang Y, Luo F, Li B. Chronic stress regulates NG2+ cell maturation and myelination in the prefrontal cortex through induction of death receptor 6. Exp Neurol 2016; 277:202-214. [DOI: 10.1016/j.expneurol.2016.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 12/24/2015] [Accepted: 01/05/2016] [Indexed: 10/22/2022]
|
329
|
Sousa N. The dynamics of the stress neuromatrix. Mol Psychiatry 2016; 21:302-12. [PMID: 26754952 PMCID: PMC4759204 DOI: 10.1038/mp.2015.196] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 10/04/2015] [Accepted: 10/21/2015] [Indexed: 01/08/2023]
Abstract
Stressful stimuli in healthy subjects trigger activation of a consistent and reproducible set of brain regions; yet, the notion that there is a single and constant stress neuromatrix is not sustainable. Indeed, after chronic stress exposure there is activation of many brain regions outside that network. This suggests that there is a distinction between the acute and the chronic stress neuromatrix. Herein, a new working model is proposed to understand the shift between these networks. The understanding of the factors that modulate these networks and their interplay will allow for a more comprehensive and holistic perspective of how the brain shifts 'back and forth' from a healthy to a stressed pattern and, ultimately, how the latter can be a trigger for several neurological and psychiatric conditions.
Collapse
Affiliation(s)
- N Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga, Portugal,ICVS/3B's–PT Government Associate Laboratory, Braga/Guimarães, Portugal,Clinical Academic Center–Braga, Braga, Portugal,Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal. E-mail:
| |
Collapse
|
330
|
Acute Footshock Stress Induces Time-Dependent Modifications of AMPA/NMDA Protein Expression and AMPA Phosphorylation. Neural Plast 2016; 2016:7267865. [PMID: 26966584 PMCID: PMC4757710 DOI: 10.1155/2016/7267865] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/21/2015] [Accepted: 01/10/2016] [Indexed: 12/04/2022] Open
Abstract
Clinical studies on patients with stress-related neuropsychiatric disorders reported functional and morphological changes in brain areas where glutamatergic transmission is predominant, including frontal and prefrontal areas. In line with this evidence, several preclinical works suggest that glutamate receptors are targets of both rapid and long-lasting effects of stress. Here we found that acute footshock- (FS-) stress, although inducing no transcriptional and RNA editing alterations of ionotropic AMPA and NMDA glutamate receptor subunits, rapidly and transiently modulates their protein expression, phosphorylation, and localization at postsynaptic spines in prefrontal and frontal cortex. In total extract, FS-stress increased the phosphorylation levels of GluA1 AMPA subunit at Ser845 immediately after stress and of GluA2 Ser880 2 h after start of stress. At postsynaptic spines, stress induced a rapid decrease of GluA2 expression, together with an increase of its phosphorylation at Ser880, suggesting internalization of GluA2 AMPA containing receptors. GluN1 and GluN2A NMDA receptor subunits were found markedly upregulated in postsynaptic spines, 2 h after start of stress. These results suggest selected time-dependent changes in glutamatergic receptor subunits induced by acute stress, which may suggest early and transient enhancement of AMPA-mediated currents, followed by a transient activation of NMDA receptors.
Collapse
|
331
|
NMDA antagonist treatment of depression. Curr Opin Neurobiol 2016; 36:112-7. [DOI: 10.1016/j.conb.2015.11.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/15/2015] [Accepted: 11/20/2015] [Indexed: 12/16/2022]
|
332
|
Negrón-Oyarzo I, Aboitiz F, Fuentealba P. Impaired Functional Connectivity in the Prefrontal Cortex: A Mechanism for Chronic Stress-Induced Neuropsychiatric Disorders. Neural Plast 2016; 2016:7539065. [PMID: 26904302 PMCID: PMC4745936 DOI: 10.1155/2016/7539065] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/25/2015] [Accepted: 12/16/2015] [Indexed: 12/20/2022] Open
Abstract
Chronic stress-related psychiatric diseases, such as major depression, posttraumatic stress disorder, and schizophrenia, are characterized by a maladaptive organization of behavioral responses that strongly affect the well-being of patients. Current evidence suggests that a functional impairment of the prefrontal cortex (PFC) is implicated in the pathophysiology of these diseases. Therefore, chronic stress may impair PFC functions required for the adaptive orchestration of behavioral responses. In the present review, we integrate evidence obtained from cognitive neuroscience with neurophysiological research with animal models, to put forward a hypothesis that addresses stress-induced behavioral dysfunctions observed in stress-related neuropsychiatric disorders. We propose that chronic stress impairs mechanisms involved in neuronal functional connectivity in the PFC that are required for the formation of adaptive representations for the execution of adaptive behavioral responses. These considerations could be particularly relevant for understanding the pathophysiology of chronic stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ignacio Negrón-Oyarzo
- Departamento de Psiquiatría, Facultad de Medicina, Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Avenida Marcoleta No. 391, 8320000 Santiago, Chile
| | - Francisco Aboitiz
- Departamento de Psiquiatría, Facultad de Medicina, Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Avenida Marcoleta No. 391, 8320000 Santiago, Chile
| | - Pablo Fuentealba
- Departamento de Psiquiatría, Facultad de Medicina, Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Avenida Marcoleta No. 391, 8320000 Santiago, Chile
| |
Collapse
|
333
|
Dendritic Spines in Depression: What We Learned from Animal Models. Neural Plast 2016; 2016:8056370. [PMID: 26881133 PMCID: PMC4736982 DOI: 10.1155/2016/8056370] [Citation(s) in RCA: 286] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/26/2015] [Indexed: 02/07/2023] Open
Abstract
Depression, a severe psychiatric disorder, has been studied for decades, but the underlying mechanisms still remain largely unknown. Depression is closely associated with alterations in dendritic spine morphology and spine density. Therefore, understanding dendritic spines is vital for uncovering the mechanisms underlying depression. Several chronic stress models, including chronic restraint stress (CRS), chronic unpredictable mild stress (CUMS), and chronic social defeat stress (CSDS), have been used to recapitulate depression-like behaviors in rodents and study the underlying mechanisms. In comparison with CRS, CUMS overcomes the stress habituation and has been widely used to model depression-like behaviors. CSDS is one of the most frequently used models for depression, but it is limited to the study of male mice. Generally, chronic stress causes dendritic atrophy and spine loss in the neurons of the hippocampus and prefrontal cortex. Meanwhile, neurons of the amygdala and nucleus accumbens exhibit an increase in spine density. These alterations induced by chronic stress are often accompanied by depression-like behaviors. However, the underlying mechanisms are poorly understood. This review summarizes our current understanding of the chronic stress-induced remodeling of dendritic spines in the hippocampus, prefrontal cortex, orbitofrontal cortex, amygdala, and nucleus accumbens and also discusses the putative underlying mechanisms.
Collapse
|
334
|
McEwen BS, Nasca C, Gray JD. Stress Effects on Neuronal Structure: Hippocampus, Amygdala, and Prefrontal Cortex. Neuropsychopharmacology 2016; 41:3-23. [PMID: 26076834 PMCID: PMC4677120 DOI: 10.1038/npp.2015.171] [Citation(s) in RCA: 887] [Impact Index Per Article: 98.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/05/2015] [Accepted: 06/08/2015] [Indexed: 12/18/2022]
Abstract
The hippocampus provided the gateway into much of what we have learned about stress and brain structural and functional plasticity, and this initial focus has expanded to other interconnected brain regions, such as the amygdala and prefrontal cortex. Starting with the discovery of adrenal steroid, and later, estrogen receptors in the hippocampal formation, and subsequent discovery of dendritic and spine synapse remodeling and neurogenesis in the dentate gyrus, mechanistic studies have revealed both genomic and rapid non-genomic actions of circulating steroid hormones in the brain. Many of these actions occur epigenetically and result in ever-changing patterns of gene expression, in which there are important sex differences that need further exploration. Moreover, glucocorticoid and estrogen actions occur synergistically with an increasing number of cellular mediators that help determine the qualitative nature of the response. The hippocampus has also been a gateway to understanding lasting epigenetic effects of early-life experiences. These findings in animal models have resulted in translation to the human brain and have helped change thinking about the nature of brain malfunction in psychiatric disorders and during aging, as well as the mechanisms of the effects of early-life adversity on the brain and the body.
Collapse
Affiliation(s)
- Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, USA,Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, 1230 York Avenue, New York, NY 10065, USA. Tel: +1 212 327 8624, Fax: +1 212 327 8634, E-mail: or http://www.rockefeller.edu/labheads/mcewen/mcewen-lab.php
| | - Carla Nasca
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, USA
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, USA
| |
Collapse
|
335
|
Kula J, Blasiak A, Czerw A, Tylko G, Sowa J, Hess G. Short-term repeated corticosterone administration enhances glutamatergic but not GABAergic transmission in the rat motor cortex. Pflugers Arch 2015; 468:679-91. [PMID: 26696244 PMCID: PMC4792354 DOI: 10.1007/s00424-015-1773-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 11/22/2015] [Accepted: 12/13/2015] [Indexed: 01/26/2023]
Abstract
It has been demonstrated that stress impairs performance of skilled reaching and walking tasks in rats due to the action of glucocorticoids involved in the stress response. Skilled reaching and walking are controlled by the primary motor cortex (M1); however, it is not known whether stress-related impairments in skilled motor tasks are related to functional and/or structural alterations within the M1. We studied the effects of single and repeated injections of corticosterone (twice daily for 7 days) on spontaneous excitatory and inhibitory postsynaptic currents (sEPSCs and sIPSCs) recorded from layer II/III pyramidal neurons in ex vivo slices of the M1, prepared 2 days after the last administration of the hormone. We also measured the density of dendritic spines on pyramidal cells and the protein levels of selected subunits of AMPA, NMDA, and GABAA receptors after repeated corticosterone administration. Repeatedly administered corticosterone induced an increase in the frequency but not in the amplitude of sEPSCs, while a single administration had no effect on the recorded excitatory currents. The frequency and amplitude of sIPSCs as well as the excitability of pyramidal cells were changed neither after single nor after repeated corticosterone administration. Treatment with corticosterone for 7 days did not modify the density of dendritic spines on pyramidal neurons. Corticosterone influenced neither the protein levels of GluA1, GluA2, GluN1, GluN2A, and GluN2B subunits of glutamate receptors nor those of α1, β2, and γ2 subunits of the GABAA receptor. The increase in sEPSCs frequency induced by repeated corticosterone administration faded out within 7 days. These data indicate that prolonged administration of exogenous corticosterone selectively and reversibly enhances glutamatergic, but not GABAergic transmission in the rat motor cortex. Our results suggest that corticosterone treatment results in an enhancement of spontaneous glutamate release from presynaptic terminals in the M1 and thereby uncovers a potential mechanism underlying stress-induced motor functions impairment.
Collapse
Affiliation(s)
- Joanna Kula
- Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Anna Blasiak
- Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Anna Czerw
- Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Grzegorz Tylko
- Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Joanna Sowa
- Department of Physiology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343, Krakow, Poland
| | - Grzegorz Hess
- Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland. .,Department of Physiology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343, Krakow, Poland.
| |
Collapse
|
336
|
Long Noncoding RNA-Directed Epigenetic Regulation of Gene Expression Is Associated With Anxiety-like Behavior in Mice. Biol Psychiatry 2015; 78:848-59. [PMID: 25792222 PMCID: PMC4532653 DOI: 10.1016/j.biopsych.2015.02.004] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 01/29/2023]
Abstract
BACKGROUND RNA-directed regulation of epigenetic processes has recently emerged as an important feature of mammalian differentiation and development. Perturbation of this regulatory system in the brain may contribute to the development of neuropsychiatric disorders. METHODS RNA sequencing was used to identify changes in the experience-dependent expression of long noncoding RNAs (lncRNAs) within the medial prefrontal cortex of adult mice. Transcripts were validated by real-time quantitative polymerase chain reaction and a candidate lncRNA, Gomafu, was selected for further investigation. The functional role of this schizophrenia-related lncRNA was explored in vivo by antisense oligonucleotide-mediated gene knockdown in the medial prefrontal cortex, followed by behavioral training and assessment of fear-related anxiety. Long noncoding RNA-directed epigenetic regulation of gene expression was investigated by chromatin and RNA immunoprecipitation assays. RESULTS RNA sequencing analysis revealed changes in the expression of a significant number of genes related to neural plasticity and stress, as well as the dynamic regulation of lncRNAs. In particular, we detected a significant downregulation of Gomafu lncRNA. Our results revealed that Gomafu plays a role in mediating anxiety-like behavior and suggest that this may occur through an interaction with a key member of the polycomb repressive complex 1, BMI1, which regulates the expression of the schizophrenia-related gene beta crystallin (Crybb1). We also demonstrated a novel role for Crybb1 in mediating fear-induced anxiety-like behavior. CONCLUSIONS Experience-dependent expression of lncRNAs plays an important role in the epigenetic regulation of adaptive behavior, and the perturbation of Gomafu may be related to anxiety and the development of neuropsychiatric disorders.
Collapse
|
337
|
de Sousa CNS, Meneses LN, Vasconcelos GS, Silva MCC, da Silva JC, Macêdo D, de Lucena DF, Vasconcelos SMM. Reversal of corticosterone-induced BDNF alterations by the natural antioxidant alpha-lipoic acid alone and combined with desvenlafaxine: Emphasis on the neurotrophic hypothesis of depression. Psychiatry Res 2015; 230:211-9. [PMID: 26350703 DOI: 10.1016/j.psychres.2015.08.042] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 08/11/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022]
Abstract
Brain derived neurotrophic factor (BDNF) is linked to the pathophysiology of depression. We hypothesized that BDNF is one of the neurobiological pathways related to the augmentation effect of alpha-lipoic acid (ALA) when associated with antidepressants. Female mice were administered vehicle or CORT 20mg/kg during 14 days. From the 15th to 21st days the animals were divided in groups that were further administered: vehicle, desvenlafaxine (DVS) 10 or 20mg/kg, ALA 100 or 200mg/kg or the combinations of DVS10+ALA100, DVS20+ALA100, DVS10+ALA200 or DVS20+ALA200. ALA or DVS alone or in combination reversed CORT-induced increase in immobility time in the forced swimming test and decrease in sucrose preference, presenting, thus, an antidepressant-like effect. DVS10 alone reversed CORT-induced decrease in BDNF in the prefrontal cortex (PFC), hippocampus (HC) and striatum (ST). The same was observed in the HC and ST of ALA200 treated animals. The combination of DVS and ALA200 reversed CORT-induced alterations in BDNF and even, in some cases, increased the levels of this neurotrophin when compared to vehicle-treated animals in HC and ST. Taken together, these results suggest that the combination of the DVS+ALA may be valuable for treating conditions in which BDNF levels are decreased, such as depression.
Collapse
Affiliation(s)
- Caren Nádia Soares de Sousa
- Neuropsychopharmacology Laboratory, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| | - Lucas Nascimento Meneses
- Neuropsychopharmacology Laboratory, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| | - Germana Silva Vasconcelos
- Neuropsychopharmacology Laboratory, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| | - Márcia Calheiros Chaves Silva
- Neuropsychopharmacology Laboratory, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| | | | - Danielle Macêdo
- Neuropsychopharmacology Laboratory, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| | - David Freitas de Lucena
- Neuropsychopharmacology Laboratory, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| | - Silvânia Maria Mendes Vasconcelos
- Neuropsychopharmacology Laboratory, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| |
Collapse
|
338
|
Zhong P, Liu W, Yan Z. Aberrant regulation of synchronous network activity by the attention-deficit/hyperactivity disorder-associated human dopamine D4 receptor variant D4.7 in the prefrontal cortex. J Physiol 2015; 594:135-47. [PMID: 26541360 DOI: 10.1113/jp271317] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/29/2015] [Indexed: 01/28/2023] Open
Abstract
KEY POINTS The hD4.7 variant has been linked to attention-deficit/hyperactivity disorder (ADHD); however, the underlying mechanism is unknown. We found that activation of hD4.7 induced over-suppression of glutamatergic excitatory network bursts and under-suppression of GABAergic inhibitory network bursts in the prefrontal cortex (PFC) circuitry. Methylphenidate, a psychostimulant drug used to treat ADHD, normalized the effects of hD4.7 on synchronous network bursts in PFC pyramidal neurons. The findings of the present study suggest that the aberrant regulation of PFC synchronous network activity by hD4.7 may underlie its involvement in ADHD. A unique feature of the human D4 receptor (hD4 R) gene is the existence of a large number of polymorphisms in exon 3 coding for the third intracellular loop, which consists of a variable number of tandem repeats. The hD4 R variants with long repeats have been linked to attention-deficit/hyperactivity disorder (ADHD); however, the underlying mechanism is unknown. Emerging evidence suggests that selective attention is controlled by the rhythmic synchronization in the prefrontal cortex (PFC) and its connected networks. In the present study, we examined the role of hD4 R variants in regulating PFC synchronous network activity. D4 R knockout mice with viral infection of hD4.4 or hD4.7 in the medial PFC were used. Whole-cell patch-clamp recordings were performed to examine the effects of activating hD4.x on the spontaneous large scale correlated activity in PFC pyramidal neurons. We found that, compared to the normal four-repeat variant hD4.4, the ADHD-linked variant hD4.7 induces more suppression of glutamatergic excitatory network bursts and less suppression of GABAergic inhibitory network bursts in the PFC circuitry. Methylphenidate, a psychostimulant drug used to treat ADHD, normalized the effects of hD4.7 on synchronous network bursts in PFC pyramidal neurons. These results reveal the differential effects of hD4 R variants on the integrated excitability of PFC circuits. It is suggested that the aberrant regulation of PFC network activity by hD4.7 may underlie its involvement in ADHD. The methylphenidate-induced normalization of synaptic circuitry regulation may contribute to its effectiveness in ADHD treatment.
Collapse
Affiliation(s)
- Ping Zhong
- Department of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.,VA Western New York Healthcare System, Buffalo, NY, USA
| | - Wenhua Liu
- Department of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.,School of Lifescience, Zhaoqing University, Zhaoqing, China
| | - Zhen Yan
- Department of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.,VA Western New York Healthcare System, Buffalo, NY, USA
| |
Collapse
|
339
|
Birey F, Kloc M, Chavali M, Hussein I, Wilson M, Christoffel DJ, Chen T, Frohman MA, Robinson JK, Russo SJ, Maffei A, Aguirre A. Genetic and Stress-Induced Loss of NG2 Glia Triggers Emergence of Depressive-like Behaviors through Reduced Secretion of FGF2. Neuron 2015; 88:941-956. [PMID: 26606998 PMCID: PMC5354631 DOI: 10.1016/j.neuron.2015.10.046] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 09/29/2015] [Accepted: 10/28/2015] [Indexed: 01/25/2023]
Abstract
NG2-expressing glia (NG2 glia) are a uniformly distributed and mitotically active pool of cells in the central nervous system (CNS). In addition to serving as progenitors of myelinating oligodendrocytes, NG2 glia might also fulfill physiological roles in CNS homeostasis, although the mechanistic nature of such roles remains unclear. Here, we report that ablation of NG2 glia in the prefrontal cortex (PFC) of the adult brain causes deficits in excitatory glutamatergic neurotransmission and astrocytic extracellular glutamate uptake and induces depressive-like behaviors in mice. We show in parallel that chronic social stress causes NG2 glia density to decrease in areas critical to Major Depressive Disorder (MDD) pathophysiology at the time of symptom emergence in stress-susceptible mice. Finally, we demonstrate that loss of NG2 glial secretion of fibroblast growth factor 2 (FGF2) suffices to induce the same behavioral deficits. Our findings outline a pathway and role for NG2 glia in CNS homeostasis and mood disorders.
Collapse
Affiliation(s)
- Fikri Birey
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA; Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Michelle Kloc
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Manideep Chavali
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA; Department of Materials Science and Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Israa Hussein
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Michael Wilson
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Daniel J Christoffel
- Fishberg Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Tony Chen
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Michael A Frohman
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - John K Robinson
- Department of Psychology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Arianna Maffei
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Adan Aguirre
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
340
|
Chaudhury D, Liu H, Han MH. Neuronal correlates of depression. Cell Mol Life Sci 2015; 72:4825-48. [PMID: 26542802 PMCID: PMC4709015 DOI: 10.1007/s00018-015-2044-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 08/27/2015] [Accepted: 09/10/2015] [Indexed: 12/14/2022]
Abstract
Major depressive disorder (MDD) is a common psychiatric disorder effecting approximately 121 million people worldwide and recent reports from the World Health Organization (WHO) suggest that it will be the leading contributor to the global burden of diseases. At present, the most commonly used treatment strategies are still based on the monoamine hypothesis that has been the predominant theory in the last 60 years. Clinical observations show that only a subset of depressed patients exhibits full remission when treated with classical monoamine-based antidepressants together with the fact that patients exhibit multiple symptoms suggest that the pathophysiology leading to mood disorders may differ between patients. Accumulating evidence indicates that depression is a neural circuit disorder and that onset of depression may be located at different regions of the brain involving different transmitter systems and molecular mechanisms. This review synthesises findings from rodent studies from which emerges a role for different, yet interconnected, molecular systems and associated neural circuits to the aetiology of depression.
Collapse
Affiliation(s)
- Dipesh Chaudhury
- Division of Science, Experimental Research Building, Office 106, New York University Abu Dhabi (NYUAD), Saadiyat Island Campus, P.O. Box 129188, Abu Dhabi, United Arab Emirates.
| | - He Liu
- Division of Science, Experimental Research Building, Office 106, New York University Abu Dhabi (NYUAD), Saadiyat Island Campus, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Ming-Hu Han
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
341
|
Neurophysiological and Neurochemical Mechanisms Underlying Depression Disorders and Search for New Directions of Treatment. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9542-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
342
|
Negrón-Oyarzo I, Dagnino-Subiabre A, Muñoz Carvajal P. Synaptic Impairment in Layer 1 of the Prefrontal Cortex Induced by Repeated Stress During Adolescence is Reversed in Adulthood. Front Cell Neurosci 2015; 9:442. [PMID: 26617490 PMCID: PMC4641900 DOI: 10.3389/fncel.2015.00442] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/26/2015] [Indexed: 12/03/2022] Open
Abstract
Chronic stress is a risk factor for the development of psychiatric disorders, some of which involve dysfunction of the prefrontal cortex (PFC). There is a higher prevalence of these chronic stress-related psychiatric disorders during adolescence, when the PFC has not yet fully matured. In the present work we studied the effect of repeated stress during adolescence on synaptic function in the PFC in adolescence and adulthood. To this end, adolescent Sprague-Dawley rats were subjected to seven consecutive days of restraint stress. Afterward, both synaptic transmission and short- and long-term synaptic plasticity were evaluated in layer 1 of medial-PFC (mPFC) slices from adolescent and adult rats. We found that repeated stress significantly reduced the amplitude of evoked field excitatory post-synaptic potential (fEPSP) in the mPFC. Isolation of excitatory transmission reveled that lower-amplitude fEPSPs were associated with a reduction in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-mediated transmission. We also found that repeated stress significantly decreased long-term depression (LTD). Interestingly, AMPA/kainate receptor-mediated transmission and LTD were recovered in adult animals that experienced a three-week stress-free recovery period. The data indicates that the changes in synaptic transmission and plasticity in the mPFC induced by repeated stress during adolescence are reversed in adulthood after a stress-free period.
Collapse
Affiliation(s)
- Ignacio Negrón-Oyarzo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile ; Laboratorio de Neurobiología y Conducta, Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso Valparaíso, Chile ; Departamento de Psiquiatría, Facultad de Medicina, Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Alexies Dagnino-Subiabre
- Laboratorio de Neurobiología y Conducta, Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso Valparaíso, Chile
| | - Pablo Muñoz Carvajal
- Centro Interdisciplinario de Innovación en Salud, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso Valparaíso, Chile
| |
Collapse
|
343
|
Goo MS, Scudder SL, Patrick GN. Ubiquitin-dependent trafficking and turnover of ionotropic glutamate receptors. Front Mol Neurosci 2015; 8:60. [PMID: 26528125 PMCID: PMC4607782 DOI: 10.3389/fnmol.2015.00060] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/22/2015] [Indexed: 12/19/2022] Open
Abstract
Changes in synaptic strength underlie the basis of learning and memory and are controlled, in part, by the insertion or removal of AMPA-type glutamate receptors at the postsynaptic membrane of excitatory synapses. Once internalized, these receptors may be recycled back to the plasma membrane by subunit-specific interactions with other proteins or by post-translational modifications such as phosphorylation. Alternatively, these receptors may be targeted for destruction by multiple degradation pathways in the cell. Ubiquitination, another post-translational modification, has recently emerged as a key signal that regulates the recycling and trafficking of glutamate receptors. In this review, we will discuss recent findings on the role of ubiquitination in the trafficking and turnover of ionotropic glutamate receptors and plasticity of excitatory synapses.
Collapse
Affiliation(s)
- Marisa S Goo
- Section of Neurobiology, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Samantha L Scudder
- Section of Neurobiology, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Gentry N Patrick
- Section of Neurobiology, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
344
|
Lussier MP, Sanz-Clemente A, Roche KW. Dynamic Regulation of N-Methyl-d-aspartate (NMDA) and α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA) Receptors by Posttranslational Modifications. J Biol Chem 2015; 290:28596-603. [PMID: 26453298 DOI: 10.1074/jbc.r115.652750] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Many molecular mechanisms underlie the changes in synaptic glutamate receptor content that are required by neuronal networks to generate cellular correlates of learning and memory. During the last decade, posttranslational modifications have emerged as critical regulators of synaptic transmission and plasticity. Notably, phosphorylation, ubiquitination, and palmitoylation control the stability, trafficking, and synaptic expression of glutamate receptors in the central nervous system. In the current review, we will summarize some of the progress made by the neuroscience community regarding our understanding of phosphorylation, ubiquitination, and palmitoylation of the NMDA and AMPA subtypes of glutamate receptors.
Collapse
Affiliation(s)
- Marc P Lussier
- From the Département de Chimie, Université du Québec à Montréal, Montréal, Québec H3C 3P8, Canada
| | - Antonio Sanz-Clemente
- the Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, and
| | - Katherine W Roche
- the Receptor Biology Section, NINDS, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
345
|
Li B, Hou Y, Zhu M, Bao H, Nie J, Zhang GY, Shan L, Yao Y, Du K, Yang H, Li M, Zheng B, Xu X, Xiao C, Du J. 3'-Deoxyadenosine (Cordycepin) Produces a Rapid and Robust Antidepressant Effect via Enhancing Prefrontal AMPA Receptor Signaling Pathway. Int J Neuropsychopharmacol 2015; 19:pyv112. [PMID: 26443809 PMCID: PMC4851261 DOI: 10.1093/ijnp/pyv112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/29/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The development of rapid and safe antidepressants for the treatment of major depression is in urgent demand. Converging evidence suggests that glutamatergic signaling seems to play important roles in the pathophysiology of depression. METHODS We studied the antidepressant effects of 3(')-deoxyadenosine (3'-dA, Cordycepin) and the critical role of the α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor in male CD-1 mice via behavioral and biochemical experiments. After 3'-dA treatment, the phosphorylation and synaptic localization of the AMPA receptors GluR1 and GluR2 were determined in the prefrontal cortex (PFC) and hippocampus (HIP). The traditional antidepressant imipramine was applied as a positive control. RESULTS We found that an injection of 3'-dA led to a rapid and robust antidepressant effect, which was significantly faster and stronger than imipramine, after 45min in tail suspension and forced swim tests. This antidepressant effect remained after 5 days of treatment with 3'-dA. Unlike the psycho-stimulants, 3'-dA did not show a hyperactive effect in the open field test. After 45min or 5 days of treatment, 3'-dA enhanced GluR1 S845 phosphorylation in both the PFC and HIP. In addition, after 45min of treatment, 3'-dA significantly up-regulated GluR1 S845 phosphorylation and GluR1, but not GluR2 levels, at the synapses in the PFC. After 5 days of treatment, 3'-dA significantly enhanced GluR1 S845 phosphorylation and GluR1, but not GluR2, at the synapses in the PFC and HIP. Moreover, the AMPA-specific antagonist GYKI 52466 was able to block the rapid antidepressant effects of 3'-dA. CONCLUSION This study identified 3'-dA as a novel rapid antidepressant with clinical potential and multiple beneficial mechanisms, particularly in regulating the prefrontal AMPA receptor signaling pathway.
Collapse
Affiliation(s)
- Bai Li
- *These authors contributed equally to this work
| | | | - Ming Zhu
- *These authors contributed equally to this work
| | | | | | | | | | | | | | | | | | | | | | | | - Jing Du
- #These two authors are co-corresponding authors
| |
Collapse
|
346
|
Rapid Antidepressant Action and Restoration of Excitatory Synaptic Strength After Chronic Stress by Negative Modulators of Alpha5-Containing GABAA Receptors. Neuropsychopharmacology 2015; 40:2499-509. [PMID: 25900119 PMCID: PMC4569955 DOI: 10.1038/npp.2015.112] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 04/08/2015] [Accepted: 04/08/2015] [Indexed: 01/30/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the primary pharmacological treatment for depression, but SSRIs are effective in only half of the patients and typically take several weeks to relieve symptoms. The NMDA receptor antagonist ketamine exerts a rapid antidepressant action, but has troubling side effects. We hypothesized that negative allosteric modulators of GABAA receptors would exert similar effects on brain activity as ketamine, but would not exert as many side effects if targeted only to GABAA receptors containing α5 subunits, which are enriched in the hippocampus and prefrontal cortex. Here, we show that the α5-selective negative modulator L-655,708 reversed the alterations in hedonic behavior in the sucrose preference and social interaction tests produced by two different chronic stress paradigms in rats within 24 h of systemic administration. Similar effects were observed with another α5-selective negative modulator, MRK-016. L-655,708 had no effect on hedonic or open-field behavior in unstressed animals. Within 24 h, L-655,708 injection also restored the strength of pathologically weakened excitatory synaptic transmission at the stress-sensitive temporoammonic-CA1 synapse, measured electrophysiologically, and increased levels of the GluA1 subunit of the AMPA receptor, measured with western blotting. We suggest that the ability of L-655,708 to restore excitatory synaptic strength rapidly may underlie its ability to restore stress-induced behavioral alterations rapidly, supporting evidence that dysfunction of multiple excitatory synapses in cortico-mesolimbic reward pathways contributes, in part, to the genesis of depression. Negative allosteric modulators of α5 subunit-containing GABAA receptors represent a promising novel class of fast-acting and clinically viable antidepressant compounds.
Collapse
|
347
|
Michael Caudle W. This can't be stressed enough: The contribution of select environmental toxicants to disruption of the stress circuitry and response. Physiol Behav 2015; 166:65-75. [PMID: 26409212 DOI: 10.1016/j.physbeh.2015.09.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 02/06/2023]
Abstract
Integration of the hypothalamic-pituitary-adrenal (HPA) axis and the limbic system through glucocorticoid signaling is imperative in initiating and regulating a suitable stress response following real or perceived threats. Dysfunction of these circuits that results in a persistent or inhibited glucocorticoid secretion can severely affect processing of stressful experiences and lead to risk for developing further psychiatric pathology. Exposure to toxic chemicals found in our environment, including pesticides, metals, and industrial compounds, have been shown to have significant impact on neurological health and disease. Indeed, studies have begun to identify the HPA axis and limbic system as potential targets of many of these environmental chemicals, suggesting a possible environmental risk for damage to the stress circuit and response to stressful stimuli. This review will focus on our current understanding of the impact exposure to environmental toxicants, including bisphenol A and lead, has on the synaptic physiology of the HPA axis and limbic system and how this contributes to an alteration in behavior output. Further, this discussion will provide a starting point to continue to couple novel toxicological and neurological approaches to elaborate our understanding of the influence of environmental chemicals on the stress response and pathology.
Collapse
Affiliation(s)
- W Michael Caudle
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322-3090, USA; Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA 30322-3090, USA.
| |
Collapse
|
348
|
Briz V, Liu Y, Zhu G, Bi X, Baudry M. A novel form of synaptic plasticity in field CA3 of hippocampus requires GPER1 activation and BDNF release. J Cell Biol 2015; 210:1225-37. [PMID: 26391661 PMCID: PMC4586750 DOI: 10.1083/jcb.201504092] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/19/2015] [Indexed: 01/11/2023] Open
Abstract
Estrogen gates metabotropic glutamate receptor–dependent long-term depression at mossy fiber–CA3 synapses through a mechanism involving GPER1-mediated BDNF release, mTOR-dependent protein synthesis, and proteasome activity. Estrogen is an important modulator of hippocampal synaptic plasticity and memory consolidation through its rapid action on membrane-associated receptors. Here, we found that both estradiol and the G-protein–coupled estrogen receptor 1 (GPER1) specific agonist G1 rapidly induce brain-derived neurotrophic factor (BDNF) release, leading to transient stimulation of activity-regulated cytoskeleton-associated (Arc) protein translation and GluA1-containing AMPA receptor internalization in field CA3 of hippocampus. We also show that type-I metabotropic glutamate receptor (mGluR) activation does not induce Arc translation nor long-term depression (LTD) at the mossy fiber pathway, as opposed to its effects in CA1, and it only triggers LTD after GPER1 stimulation. Furthermore, this form of mGluR-dependent LTD is associated with ubiquitination and proteasome-mediated degradation of GluA1, and is prevented by proteasome inhibition. Overall, our study identifies a novel mechanism by which estrogen and BDNF regulate hippocampal synaptic plasticity in the adult brain.
Collapse
Affiliation(s)
- Victor Briz
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766 VIB Center for the Biology of Disease, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Yan Liu
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766 College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Guoqi Zhu
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766 Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei 230038, China
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766
| |
Collapse
|
349
|
Bredewold R, Schiavo JK, van der Hart M, Verreij M, Veenema AH. Dynamic changes in extracellular release of GABA and glutamate in the lateral septum during social play behavior in juvenile rats: Implications for sex-specific regulation of social play behavior. Neuroscience 2015; 307:117-27. [PMID: 26318330 DOI: 10.1016/j.neuroscience.2015.08.052] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/11/2015] [Accepted: 08/21/2015] [Indexed: 10/23/2022]
Abstract
Social play is a motivated and rewarding behavior that is displayed by nearly all mammals and peaks in the juvenile period. Moreover, social play is essential for the development of social skills and is impaired in social disorders like autism. We recently showed that the lateral septum (LS) is involved in the regulation of social play behavior in juvenile male and female rats. The LS is largely modulated by GABA and glutamate neurotransmission, but their role in social play behavior is unknown. Here, we determined whether social play behavior is associated with changes in the extracellular release of GABA and glutamate in the LS and to what extent such changes modulate social play behavior in male and female juvenile rats. Using intracerebral microdialysis in freely behaving rats, we found no sex difference in extracellular GABA concentrations, but extracellular glutamate concentrations are higher in males than in females under baseline conditions and during social play. This resulted in a higher glutamate/GABA concentration ratio in males vs. females and thus, an excitatory predominance in the LS of males. Furthermore, social play behavior in both sexes is associated with significant increases in extracellular release of GABA and glutamate in the LS. Pharmacological blockade of GABA-A receptors in the LS with bicuculline (100 ng/0.5 μl, 250 ng/0.5 μl) dose-dependently decreased the duration of social play behavior in both sexes. In contrast, pharmacological blockade of ionotropic glutamate receptors (NMDA and AMPA/kainate receptors) in the LS with AP-5+CNQX (2mM+0.4mM/0.5 μl, 30 mM+3mM/0.5 μl) dose-dependently decreased the duration of social play behavior in females, but did not alter social play behavior in males. Together, these data suggest a role for GABA neurotransmission in the LS in the regulation of juvenile social play behavior in both sexes, while glutamate neurotransmission in the LS is involved in the sex-specific regulation of juvenile social play behavior.
Collapse
Affiliation(s)
- R Bredewold
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA.
| | - J K Schiavo
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | | | - M Verreij
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - A H Veenema
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| |
Collapse
|
350
|
Arloth J, Bogdan R, Weber P, Frishman G, Menke A, Wagner KV, Balsevich G, Schmidt MV, Karbalai N, Czamara D, Altmann A, Trümbach D, Wurst W, Mehta D, Uhr M, Klengel T, Erhardt A, Carey CE, Conley ED, Ruepp A, Müller-Myhsok B, Hariri AR, Binder EB. Genetic Differences in the Immediate Transcriptome Response to Stress Predict Risk-Related Brain Function and Psychiatric Disorders. Neuron 2015; 86:1189-202. [PMID: 26050039 PMCID: PMC4490780 DOI: 10.1016/j.neuron.2015.05.034] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 03/26/2015] [Accepted: 05/13/2015] [Indexed: 12/27/2022]
Abstract
Depression risk is exacerbated by genetic factors and stress exposure; however, the biological mechanisms through which these factors interact to confer depression risk are poorly understood. One putative biological mechanism implicates variability in the ability of cortisol, released in response to stress, to trigger a cascade of adaptive genomic and non-genomic processes through glucocorticoid receptor (GR) activation. Here, we demonstrate that common genetic variants in long-range enhancer elements modulate the immediate transcriptional response to GR activation in human blood cells. These functional genetic variants increase risk for depression and co-heritable psychiatric disorders. Moreover, these risk variants are associated with inappropriate amygdala reactivity, a transdiagnostic psychiatric endophenotype and an important stress hormone response trigger. Network modeling and animal experiments suggest that these genetic differences in GR-induced transcriptional activation may mediate the risk for depression and other psychiatric disorders by altering a network of functionally related stress-sensitive genes in blood and brain.
Collapse
Affiliation(s)
- Janine Arloth
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Ryan Bogdan
- Department of Psychology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Peter Weber
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Goar Frishman
- Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Andreas Menke
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Klaus V Wagner
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Georgia Balsevich
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Mathias V Schmidt
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Nazanin Karbalai
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Andre Altmann
- Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Dietrich Trümbach
- Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Wolfgang Wurst
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany; Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany; Technische Universität München, c/o Helmholtz Zentrum München, German Research Centre for Environmental Health, Institute of Developmental Genetics, Neuherberg 85764, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Site Munich, Munich 80336, Germany
| | - Divya Mehta
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Manfred Uhr
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Torsten Klengel
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Angelika Erhardt
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Caitlin E Carey
- Department of Psychology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | | | | | - Andreas Ruepp
- Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Bertram Müller-Myhsok
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Ahmad R Hariri
- Department of Psychology and Neuroscience, Institute for Genome Sciences and Policy, Duke University, Durham, NC 27708, USA
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, USA.
| | | |
Collapse
|