1
|
Palchaudhuri S, Osypenko D, Schneggenburger R. Fear Learning: An Evolving Picture for Plasticity at Synaptic Afferents to the Amygdala. Neuroscientist 2024; 30:87-104. [PMID: 35822657 DOI: 10.1177/10738584221108083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Unraveling the neuronal mechanisms of fear learning might allow neuroscientists to make links between a learned behavior and the underlying plasticity at specific synaptic connections. In fear learning, an innocuous sensory event such as a tone (called the conditioned stimulus, CS) acquires an emotional value when paired with an aversive outcome (unconditioned stimulus, US). Here, we review earlier studies that have shown that synaptic plasticity at thalamic and cortical afferents to the lateral amygdala (LA) is critical for the formation of auditory-cued fear memories. Despite the early progress, it has remained unclear whether there are separate synaptic inputs that carry US information to the LA to act as a teaching signal for plasticity at CS-coding synapses. Recent findings have begun to fill this gap by showing, first, that thalamic and cortical auditory afferents can also carry US information; second, that the release of neuromodulators contributes to US-driven teaching signals; and third, that synaptic plasticity additionally happens at connections up- and downstream of the LA. Together, a picture emerges in which coordinated synaptic plasticity in serial and parallel circuits enables the formation of a finely regulated fear memory.
Collapse
Affiliation(s)
- Shriya Palchaudhuri
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Denys Osypenko
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Ralf Schneggenburger
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Eltokhi A, Bertocchi I, Rozov A, Jensen V, Borchardt T, Taylor A, Proenca CC, Rawlins JNP, Bannerman DM, Sprengel R. Distinct effects of AMPAR subunit depletion on spatial memory. iScience 2023; 26:108116. [PMID: 37876813 PMCID: PMC10590979 DOI: 10.1016/j.isci.2023.108116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 07/01/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023] Open
Abstract
Pharmacological studies established a role for AMPARs in the mammalian forebrain in spatial memory performance. Here we generated global GluA1/3 double knockout mice (Gria1/3-/-) and conditional knockouts lacking GluA1 and GluA3 AMPAR subunits specifically from principal cells across the forebrain (Gria1/3ΔFb). In both models, loss of GluA1 and GluA3 resulted in reduced hippocampal GluA2 and increased levels of the NMDAR subunit GluN2A. Electrically-evoked AMPAR-mediated EPSPs were greatly diminished, and there was an absence of tetanus-induced LTP. Gria1/3-/- mice showed premature mortality. Gria1/3ΔFb mice were viable, and their memory performance could be analyzed. In the Morris water maze (MWM), Gria1/3ΔFb mice showed profound long-term memory deficits, in marked contrast to the normal MWM learning previously seen in single Gria1-/- and Gria3-/- knockout mice. Our results suggest a redundancy of function within the pool of available ionotropic glutamate receptors for long-term spatial memory performance.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Pharmacolog, University of Washington, Seattle, WA, USA
| | - Ilaria Bertocchi
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy
- Neuroscience Institute - Cavalieri-Ottolenghi Foundation (NICO), Laboratory of Neuropsychopharmacology, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Andrei Rozov
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
- Institute of Neuroscience, Lobachevsky State University of Nizhniy, 603022 Novgorod, Russia
- Federal Center of Brain Research and Neurotechnology, 117997 Moscow, Russia
| | - Vidar Jensen
- Department of Molecular Medicine, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway
| | - Thilo Borchardt
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Amy Taylor
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Catia C. Proenca
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | | | | | - Rolf Sprengel
- Departments of Molecular Neurobiology and Physiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| |
Collapse
|
3
|
Gugustea R, Jia Z. Genetic manipulations of AMPA glutamate receptors in hippocampal synaptic plasticity. Neuropharmacology 2021; 194:108630. [PMID: 34089730 DOI: 10.1016/j.neuropharm.2021.108630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/06/2021] [Accepted: 05/18/2021] [Indexed: 01/17/2023]
Abstract
Alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are the principal mediators of fast excitatory synaptic transmission and they are required for various forms of synaptic plasticity, including long-term potentiation (LTP) and depression (LTD), which are key mechanisms of learning and memory. AMPARs are tetrameric complexes assembled from four subunits (GluA1-4), however, the lack of subunit-specific pharmacological tools has made the assessment of individual subunits difficult. The application of genetic techniques, particularly gene targeting, allows for precise manipulation and dissection of each subunit in the regulation of neuronal function and behaviour. In this review, we summarize studies using various mouse models with genetically altered AMPARs and focus on their roles in basal synaptic transmission, LTP, and LTD at the hippocampal CA1 synapse. These studies provide strong evidence that there are multiple forms of LTP and LTD at this synapse which can be induced by various induction protocols, and they are differentially regulated by different AMPAR subunits and domains. We conclude that it is necessary to delineate the mechanism of each of these forms of plasticity and their contribution to memory and brain disorders.
Collapse
Affiliation(s)
- Radu Gugustea
- The Hospital for Sick Children, Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Zhengping Jia
- The Hospital for Sick Children, Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Tucker LB, Fu AH, McCabe JT. Hippocampal-Dependent Cognitive Dysfunction following Repeated Diffuse Rotational Brain Injury in Male and Female Mice. J Neurotrauma 2021; 38:1585-1606. [PMID: 33622092 PMCID: PMC8126427 DOI: 10.1089/neu.2021.0025] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cognitive dysfunction is a common, often long-term complaint following acquired traumatic brain injury (TBI). Cognitive deficits suggest dysfunction in hippocampal circuits. The goal of the studies described here is to phenotype in both male and female mice the hippocampal-dependent learning and memory deficits resulting from TBI sustained by the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA) device—a model that delivers both a contact–concussion injury as well as unrestrained rotational head movement. Mice sustained either sham procedures or four injuries (0.7 J, 24-h intervals). Spatial learning and memory skills assessed in the Morris water maze (MWM) approximately 3 weeks following injuries were significantly impaired by brain injuries; however, slower swimming speeds and poor performance on visible platform trials suggest that measurement of cognitive impairment with this test is confounded by injury-induced motor and/or visual impairments. A separate experiment confirmed hippocampal-dependent cognitive deficits with trace fear conditioning (TFC), a behavioral test less dependent on motor and visual function. Male mice had greater injury-induced deficits on both the MWM and TFC tests than female mice. Pathologically, the injury was characterized by white matter damage as observed by silver staining and glial fibrillary acidic protein (astrogliosis) in the optic tracts, with milder damage seen in the corpus callosum, and fimbria and brainstem (cerebral peduncles) of some animals. No changes in the density of GABAergic parvalbumin-expressing cells in the hippocampus, amygdala, or parietal cortex were found. This experiment confirmed significant sexually dimorphic cognitive impairments following a repeated, diffuse brain injury.
Collapse
Affiliation(s)
- Laura B Tucker
- Center for Neuroscience and Regenerative Medicine, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Amanda H Fu
- Center for Neuroscience and Regenerative Medicine, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Joseph T McCabe
- Center for Neuroscience and Regenerative Medicine, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Nonaka M, Taylor WW, Bukalo O, Tucker LB, Fu AH, Kim Y, McCabe JT, Holmes A. Behavioral and Myelin-Related Abnormalities after Blast-Induced Mild Traumatic Brain Injury in Mice. J Neurotrauma 2021; 38:1551-1571. [PMID: 33605175 DOI: 10.1089/neu.2020.7254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In civilian and military settings, mild traumatic brain injury (mTBI) is a common consequence of impacts to the head, sudden blows to the body, and exposure to high-energy atmospheric shockwaves from blast. In some cases, mTBI from blast exposure results in long-term emotional and cognitive deficits and an elevated risk for certain neuropsychiatric diseases. Here, we tested the effects of mTBI on various forms of auditory-cued fear learning and other measures of cognition in male C57BL/6J mice after single or repeated blast exposure (blast TBI; bTBI). bTBI produced an abnormality in the temporal organization of cue-induced freezing behavior in a conditioned trace fear test. Spatial working memory, evaluated by the Y-maze task performance, was also deleteriously affected by bTBI. Reverse-transcription quantitative real-time polymerase chain reaction (RT-qPCR) analysis for glial markers indicated an alteration in the expression of myelin-related genes in the hippocampus and corpus callosum 1-8 weeks after bTBI. Immunohistochemical and ultrastructural analyses detected bTBI-related myelin and axonal damage in the hippocampus and corpus callosum. Together, these data suggest a possible link between blast-induced mTBI, myelin/axonal injury, and cognitive dysfunction.
Collapse
Affiliation(s)
- Mio Nonaka
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIH), Rockville, Maryland, USA
| | - William W Taylor
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIH), Rockville, Maryland, USA
| | - Olena Bukalo
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIH), Rockville, Maryland, USA
| | - Laura B Tucker
- Department of Anatomy, Physiology and Genetics, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Preclinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Amanda H Fu
- Department of Anatomy, Physiology and Genetics, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Preclinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Yeonho Kim
- Department of Anatomy, Physiology and Genetics, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Preclinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Joseph T McCabe
- Department of Anatomy, Physiology and Genetics, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Preclinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIH), Rockville, Maryland, USA
| |
Collapse
|
6
|
Aitta-Aho T, Maksimovic M, Dahl K, Sprengel R, Korpi ER. Attenuation of Novelty-Induced Hyperactivity of Gria1-/- Mice by Cannabidiol and Hippocampal Inhibitory Chemogenetics. Front Pharmacol 2019; 10:309. [PMID: 30984001 PMCID: PMC6449460 DOI: 10.3389/fphar.2019.00309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/13/2019] [Indexed: 11/13/2022] Open
Abstract
Gene-targeted mice with deficient AMPA receptor GluA1 subunits (Gria1-/- mice) show robust hyperlocomotion in a novel environment, suggesting them to constitute a model for hyperactivity disorders such as mania, schizophrenia and attention deficit hyperactivity disorder. This behavioral alteration has been associated with increased neuronal activation in the hippocampus, and it can be attenuated by chronic treatment with antimanic drugs, such as lithium, valproic acid, and lamotrigine. Now we found that systemic cannabidiol strongly blunted the hyperactivity and the hippocampal c-Fos expression of the Gria1-/- mice, while not affecting the wild-type littermate controls. Acute bilateral intra-dorsal hippocampal infusion of cannabidiol partially blocked the hyperactivity of the Gria1-/- mice, but had no effect on wild-types. The activation of the inhibitory DREADD receptor hM4Gi in the dorsal hippocampus by clozapine-N-oxide robustly inhibited the hyperactivity of the Gria1-/- mice, but had no effect on the locomotion of wild-type mice. Our results show that enhanced neuronal excitability in the hippocampus is associated with pronounced novelty-induced hyperactivity of GluA1 subunit-deficient mice. When this enhanced response of hippocampal neurons to novel stimuli is specifically reduced in the hippocampus by pharmacological treatment or by chemogenetic inhibition, Gria1-/- mice recover from behavioral hyperactivity, suggesting a hippocampal dysfunction in hyperactive behaviors that can be treated with cannabidiol.
Collapse
Affiliation(s)
- Teemu Aitta-Aho
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Milica Maksimovic
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristiina Dahl
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Rolf Sprengel
- Research Group of the Max Planck Institute for Medical Research at the Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
AnkG hemizygous mice present cognitive impairment and elevated anxiety/depressive-like traits associated with decreased expression of GABA receptors and postsynaptic density protein. Exp Brain Res 2017; 235:3375-3390. [PMID: 28821923 DOI: 10.1007/s00221-017-5056-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 08/03/2017] [Indexed: 10/19/2022]
Abstract
Recent genome-wide association studies (GWAS) of patient populations and genetic linkage assessments have demonstrated that the ankyrin-G (AnkG) gene is involved in neuropsychiatric disorders, including bipolar disorder, schizophrenia, and Alzheimer's disease, but it remains unclear how the genetic variants of AnkG contribute to neuropsychiatric disorders. Here, we generated AnkG hemizygous mice using the gene trapping approach. Homozygous AnkG was embryonically lethal. Western blotting and real-time polymerase chain reaction (qPCR) assessments of wild type (WT) and AnkG +/- mutant mice demonstrated a 50% reduction of ANKG levels, at the gene and protein levels, in AnkG hemizygous mice. In behavioral tests, AnkG hemizygous mice exhibited elevated anxiety- and depression-like traits, as well as cognitive impairment. Moreover, the expression levels of cognitive-related proteins (including metabotropic glutamate receptor subtype-1, brain-derived neurotrophic factor, postsynaptic density-95, GABA-B receptor, and GABA-A receptor alpha-1) were significantly decreased (P < 0.05), suggesting a possible role for AnkG in cognition. It is possible that the loss of AnkG in the brain disrupts the excitation/inhibition balance of neurotransmitters, hindering the synaptic plasticity of neurons, and consequently leading to abnormal behavioral symptoms. Therefore, AnkG possibly contributes to neuroprotection and normal brain function, and may constitute a new target for treating neuropsychiatric diseases, especially cognitive dysfunction.
Collapse
|
8
|
Gutierrez-Castellanos N, Da Silva-Matos CM, Zhou K, Canto CB, Renner MC, Koene LMC, Ozyildirim O, Sprengel R, Kessels HW, De Zeeuw CI. Motor Learning Requires Purkinje Cell Synaptic Potentiation through Activation of AMPA-Receptor Subunit GluA3. Neuron 2017; 93:409-424. [PMID: 28103481 PMCID: PMC5263704 DOI: 10.1016/j.neuron.2016.11.046] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 09/28/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022]
Abstract
Accumulating evidence indicates that cerebellar long-term potentiation (LTP) is necessary for procedural learning. However, little is known about its underlying molecular mechanisms. Whereas AMPA receptor (AMPAR) subunit rules for synaptic plasticity have been extensively studied in relation to declarative learning, it is unclear whether these rules apply to cerebellum-dependent motor learning. Here we show that LTP at the parallel-fiber-to-Purkinje-cell synapse and adaptation of the vestibulo-ocular reflex depend not on GluA1- but on GluA3-containing AMPARs. In contrast to the classic form of LTP implicated in declarative memory formation, this form of LTP does not require GluA1-AMPAR trafficking but rather requires changes in open-channel probability of GluA3-AMPARs mediated by cAMP signaling and activation of the protein directly activated by cAMP (Epac). We conclude that vestibulo-cerebellar motor learning is the first form of memory acquisition shown to depend on GluA3-dependent synaptic potentiation by increasing single-channel conductance. Cerebellar learning depends on expression of GluA3, but not GluA1, in Purkinje cells GluA3 is required to induce LTP, but not LTD, at PF-PC synapses GluA3-dependent potentiation involves a cAMP-driven change in channel conductance GluA3-mediated LTP and learning are induced via cAMP-mediated Epac activation
Collapse
Affiliation(s)
- Nicolas Gutierrez-Castellanos
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands; Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands; Department of Neuroscience, Erasmus MC Rotterdam, 3015 GE Rotterdam, the Netherlands
| | - Carla M Da Silva-Matos
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands; Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Kuikui Zhou
- Department of Neuroscience, Erasmus MC Rotterdam, 3015 GE Rotterdam, the Netherlands
| | - Cathrin B Canto
- Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Maria C Renner
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Linda M C Koene
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Ozgecan Ozyildirim
- Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Rolf Sprengel
- Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Helmut W Kessels
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands.
| | - Chris I De Zeeuw
- Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands; Department of Neuroscience, Erasmus MC Rotterdam, 3015 GE Rotterdam, the Netherlands
| |
Collapse
|
9
|
Zelena D, Mikics É, Balázsfi D, Varga J, Klausz B, Urbán E, Sipos E, Biró L, Miskolczi C, Kovács K, Ferenczi S, Haller J. Enduring abolishment of remote but not recent expression of conditioned fear by the blockade of calcium-permeable AMPA receptors before extinction training. Psychopharmacology (Berl) 2016; 233:2065-2076. [PMID: 27020785 DOI: 10.1007/s00213-016-4255-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 02/21/2016] [Indexed: 11/29/2022]
Abstract
RATIONALE Calcium-permeable (GluA2 subunit-free) AMPA receptors (CP-AMPAR) play prominent roles in fear extinction; however, no blockers of these receptors were studied in tests relevant to extinction learning so far. METHODS The CP-AMPAR antagonist IEM-1460 was administered once before extinction trainings, which were started either 1 or 28 days after fear conditioning (FC). We used a mild extinction protocol that durably decreased but did not abolish conditioned fear. The messenger RNA (mRNA) expression of GluA1 and GluA2 subunits were investigated at both time points in the ventromedial prefrontal cortex (vmPFC) and amygdala. RESULTS IEM-1460 transiently facilitated extinction 1 day after conditioning, but learned fear spontaneously recovered 4 weeks later. When the extinction protocol was applied 28 days after training, IEM-1460 enhanced extinction memory, moreover abolished conditioned fear for at least a month. The expression of GluA1 and GluA2 mRNAs was increased at both time points in the vmPFC. In the basolateral and central amygdala, the GluA1/GluA2 mRNA ratio increased, suggesting a shift towards the preponderance of GluA1 over GluA2 expression. CONCLUSIONS AMPAR blockade lastingly enhanced the extinction of remote but not recent fear memories. Time-dependent changes in AMPA receptor subunit mRNA expression may explain the differential effects of CP-AMPAR blockade on recent and remote conditioned fear, further supporting the notion that the mechanisms maintaining learned fear change over time. Our findings suggest clinical implications for CP-AMPAR blockers, particularly for acquired anxieties (e.g., post-traumatic stress disorder) which have a slow onset and are durable.
Collapse
Affiliation(s)
- Dóra Zelena
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary.
| | - Éva Mikics
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - Diána Balázsfi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary.,János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - János Varga
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - Barbara Klausz
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - Eszter Urbán
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - Eszter Sipos
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - László Biró
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary.,János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Christina Miskolczi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - Krisztina Kovács
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - Szilamér Ferenczi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - József Haller
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| |
Collapse
|
10
|
Robertson JM, Prince MA, Achua JK, Carpenter RE, Arendt DH, Smith JP, Summers TL, Summers TR, Summers CH. Nuance and behavioral cogency: How the Visible Burrow System inspired the Stress-Alternatives Model and conceptualization of the continuum of anxiety. Physiol Behav 2016; 146:86-97. [PMID: 26066728 DOI: 10.1016/j.physbeh.2015.03.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 03/24/2015] [Accepted: 03/25/2015] [Indexed: 12/20/2022]
Abstract
By creating the Visible Burrow System (VBS) Bob Blanchard found a way to study the interaction of genetics, physiology, environment, and adaptive significance in a model with broad validity. The VBS changed the way we think about anxiety and affective disorders by allowing the mechanisms which control them to be observed in a dynamic setting. Critically, Blanchard used the VBS and other models to show how behavioral systems like defense are dependent upon context and behavioral elements unique to the individual. Inspired by the VBS, we developed a Stress Alternatives Model (SAM) to further explore the multifaceted dynamics of the stress response with a dichotomous choice condition. Like the VBS, the SAM is a naturalistic model built upon risk assessment and defensive behavior, but with a choice of response: escape or submission to a large conspecific aggressor. The anxiety of novelty during the first escape must be weighed against fear of the aggressor, and a decision must be made. Both outcomes are adaptively significant, evidenced by a 50/50 split in outcome across several study systems. By manipulating the variables of the SAM, we show that a gradient of anxiety exists that spans the contextual settings of escaping an open field, escaping from aggression, and submitting to aggression. These findings correspond with increasing levels of corticosterone and increasing levels of NPS and BDNF in the central amygdala as the context changes.Whereas some anxiolytics were able to reduce the latency to escape for some animals, only with the potent anxiolytic drug antalarmin (CRF1R-blocker) and the anxiogenic drug yohimbine (α2 antagonist) were we able to reverse the outcome for a substantial proportion of individuals. Our findings promote a novel method for modeling anxiety, offering a distinction between low-and-high levels, and accounting for individual variability. The translational value of the VBS is immeasurable, and it guided us and many other researchers to seek potential clinical solutions through a deeper understanding of regional neurochemistry and gene expression in concert with an ecological behavioral model.
Collapse
|
11
|
Radke AK, Nakazawa K, Holmes A. Cortical GluN2B deletion attenuates punished suppression of food reward-seeking. Psychopharmacology (Berl) 2015; 232. [PMID: 26223494 PMCID: PMC6065108 DOI: 10.1007/s00213-015-4033-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE Compulsive behavior, which is a hallmark of psychiatric disorders such as addiction and obsessive-compulsive disorder, engages corticostriatal circuits. Previous studies indicate a role for corticostriatal N-methyl-D-aspartate receptors (NMDARs) in mediating compulsive-like responding for drugs of abuse, but the specific receptor subunits controlling reward-seeking in the face of punishment remain unclear. OBJECTIVES The current study assessed the involvement of corticostriatal GluN2B-containing NMDARs in measures of persistent and punished food reward-seeking. METHODS Mice with genetic deletion of GluN2B in one of three distinct neuronal populations, cortical principal neurons, forebrain interneurons, or striatal medium spiny neurons, were tested for (1) sustained food reward-seeking when reward was absent, (2) reward-seeking under a progressive ratio schedule of reinforcement, and (3) persistent reward-seeking after a footshock punishment. RESULTS Mutant mice with genetic deletion of GluN2B in cortical principal neurons demonstrated attenuated suppression of reward-seeking during punishment. These mice performed normally on other behavioral measures, including an assay for pain sensitivity. Mutants with interneuronal or striatal GluN2B deletions were normal on all behavioral assays. CONCLUSIONS Current findings offer novel evidence that loss of GluN2B-containing NMDARs expressed on principal neurons in the cortex results in reduced punished food reward-seeking. These data support the involvement of GluN2B subunit in cortical circuits regulating cognitive flexibility in a variety of settings, with implications for understanding the basis of inflexible behavior in neuropsychiatric disorders including obsessive-compulsive disorders (OCD) and addictions.
Collapse
Affiliation(s)
- Anna K. Radke
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA,5625 Fishers Lane Room 2N09, Rockville, MD, USA
| | - Kazu Nakazawa
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| |
Collapse
|
12
|
Fitzgerald PJ, Pinard CR, Camp MC, Feyder M, Sah A, Bergstrom H, Graybeal C, Liu Y, Schlüter O, Grant SG, Singewald N, Xu W, Holmes A. Durable fear memories require PSD-95. Mol Psychiatry 2015; 20:901-12. [PMID: 25510511 PMCID: PMC4469631 DOI: 10.1038/mp.2014.161] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 07/28/2014] [Accepted: 10/15/2014] [Indexed: 12/13/2022]
Abstract
Traumatic fear memories are highly durable but also dynamic, undergoing repeated reactivation and rehearsal over time. Although overly persistent fear memories underlie anxiety disorders, such as posttraumatic stress disorder, the key neural and molecular mechanisms underlying fear memory durability remain unclear. Postsynaptic density 95 (PSD-95) is a synaptic protein regulating glutamate receptor anchoring, synaptic stability and certain types of memory. Using a loss-of-function mutant mouse lacking the guanylate kinase domain of PSD-95 (PSD-95(GK)), we analyzed the contribution of PSD-95 to fear memory formation and retrieval, and sought to identify the neural basis of PSD-95-mediated memory maintenance using ex vivo immediate-early gene mapping, in vivo neuronal recordings and viral-mediated knockdown (KD) approaches. We show that PSD-95 is dispensable for the formation and expression of recent fear memories, but essential for the formation of precise and flexible fear memories and for the maintenance of memories at remote time points. The failure of PSD-95(GK) mice to retrieve remote cued fear memory was associated with hypoactivation of the infralimbic (IL) cortex (but not the anterior cingulate cortex (ACC) or prelimbic cortex), reduced IL single-unit firing and bursting, and attenuated IL gamma and theta oscillations. Adeno-associated virus-mediated PSD-95 KD in the IL, but not the ACC, was sufficient to impair recent fear extinction and remote fear memory, and remodel IL dendritic spines. Collectively, these data identify PSD-95 in the IL as a critical mechanism supporting the durability of fear memories over time. These preclinical findings have implications for developing novel approaches to treating trauma-based anxiety disorders that target the weakening of overly persistent fear memories.
Collapse
Affiliation(s)
- Paul J. Fitzgerald
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, NIH
| | - Courtney R. Pinard
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, NIH
| | - Marguerite C. Camp
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, NIH
| | - Michael Feyder
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, NIH
| | - Anupam Sah
- Department of Pharmacology & Toxicology, University of Innsbruck, Innsbruck, Austria
| | - Hadley Bergstrom
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, NIH
| | - Carolyn Graybeal
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, NIH
| | - Yan Liu
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Seth G.N. Grant
- Centre for Clinical Brain Sciences and Centre for Neuroregeneration, The University of Edinburgh, Edinburgh, UK
| | - Nicolas Singewald
- Department of Pharmacology & Toxicology, University of Innsbruck, Innsbruck, Austria
| | - Weifeng Xu
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, NIH
| |
Collapse
|
13
|
Udagawa T, Fujioka Y, Tanaka M, Honda D, Yokoi S, Riku Y, Ibi D, Nagai T, Yamada K, Watanabe H, Katsuno M, Inada T, Ohno K, Sokabe M, Okado H, Ishigaki S, Sobue G. FUS regulates AMPA receptor function and FTLD/ALS-associated behaviour via GluA1 mRNA stabilization. Nat Commun 2015; 6:7098. [PMID: 25968143 PMCID: PMC4479014 DOI: 10.1038/ncomms8098] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/02/2015] [Indexed: 12/12/2022] Open
Abstract
FUS is an RNA/DNA-binding protein involved in multiple steps of gene expression and is associated with amyotrophic lateral sclerosis (ALS) and fronto-temporal lobar degeneration (FTLD). However, the specific disease-causing and/or modifying mechanism mediated by FUS is largely unknown. Here we evaluate intrinsic roles of FUS on synaptic functions and animal behaviours. We find that FUS depletion downregulates GluA1, a subunit of AMPA receptor. FUS binds GluA1 mRNA in the vicinity of the 3′ terminus and controls poly (A) tail maintenance, thus regulating stability. GluA1 reduction upon FUS knockdown reduces miniature EPSC amplitude both in cultured neurons and in vivo. FUS knockdown in hippocampus attenuates dendritic spine maturation and causes behavioural aberrations including hyperactivity, disinhibition and social interaction defects, which are partly ameliorated by GluA1 reintroduction. These results highlight the pivotal role of FUS in regulating GluA1 mRNA stability, post-synaptic function and FTLD-like animal behaviours. FUS is an RNA/DNA-binding protein involved in gene expression regulation and associated with amyotrophic lateral sclerosis and frontotemporal dementia (FTLD) but the disease-causing mechanisms are unclear. Here the authors show that FUS regulates the stability of GluA1 mRNA and dendritic maturation and plays a role in FTLD-associated behaviours.
Collapse
Affiliation(s)
- Tsuyoshi Udagawa
- 1] Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan [2] Graduate School of pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Yusuke Fujioka
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Motoki Tanaka
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Daiyu Honda
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Satoshi Yokoi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yuichi Riku
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Daisuke Ibi
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Toshifumi Inada
- Graduate School of pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Haruo Okado
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Shinsuke Ishigaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
14
|
Kiselycznyk C, Jury NJ, Halladay LR, Nakazawa K, Mishina M, Sprengel R, Grant SGN, Svenningsson P, Holmes A. NMDA receptor subunits and associated signaling molecules mediating antidepressant-related effects of NMDA-GluN2B antagonism. Behav Brain Res 2015; 287:89-95. [PMID: 25800971 PMCID: PMC4425283 DOI: 10.1016/j.bbr.2015.03.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/07/2015] [Accepted: 03/12/2015] [Indexed: 12/11/2022]
Abstract
Drugs targeting the glutamate N-methyl-d-aspartate receptor (NMDAR) may be efficacious for treating mood disorders, as exemplified by the rapid antidepressant effects produced by single administration of the NMDAR antagonist ketamine. Though the precise mechanisms underlying the antidepressant-related effects of NMDAR antagonism remain unclear, recent studies implicate specific NMDAR subunits, including GluN2A and GluN2B, as well as the alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR) subunit glutamate receptor interacting molecule, PSD-95. Here, integrating mutant and pharmacological in mice, we investigated the contribution of these subunits and molecules to antidepressant-related behaviors and the antidepressant-related effects of the GluN2B blocker, Ro 25-6981. We found that global deletion of GluA1 or PSD-95 reduced forced swim test (FST) immobility, mimicking the antidepressant-related effect produced by systemically administered Ro 25-6981 in C57BL/6J mice. Moreover, the FST antidepressant-like effects of systemic Ro 25-6981 were intact in mutants with global GluA1 deletion or GluN1 deletion in forebrain interneurons, but were absent in mutants constitutively lacking GluN2A or PSD-95. Next, we found that microinfusing Ro 25-6981 into the medial prefrontal cortex (mPFC), but not basolateral amygdala, of C57BL/6J mice was sufficient to produce an antidepressant-like effect. Together, these findings extend and refine current understanding of the mechanisms mediating antidepressant-like effects produced by NMDAR-GluN2B antagonists, and may inform the development of a novel class of medications for treating depression that target the GluN2B subtype of NMDAR.
Collapse
Affiliation(s)
- Carly Kiselycznyk
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Nicholas J Jury
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| | - Lindsay R Halladay
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Kazu Nakazawa
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Masayoshi Mishina
- Brain Science Laboratory, The Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Rolf Sprengel
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Seth G N Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Per Svenningsson
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| |
Collapse
|
15
|
Freudenberg F, Celikel T, Reif A. The role of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in depression: central mediators of pathophysiology and antidepressant activity? Neurosci Biobehav Rev 2015; 52:193-206. [PMID: 25783220 DOI: 10.1016/j.neubiorev.2015.03.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/23/2015] [Accepted: 03/06/2015] [Indexed: 12/27/2022]
Abstract
Depression is a major psychiatric disorder affecting more than 120 million people worldwide every year. Changes in monoaminergic transmitter release are suggested to take part in the pathophysiology of depression. However, more recent experimental evidence suggests that glutamatergic mechanisms might play a more central role in the development of this disorder. The importance of the glutamatergic system in depression was particularly highlighted by the discovery that N-methyl-D-aspartate (NMDA) receptor antagonists (particularly ketamine) exert relatively long-lasting antidepressant like effects with rapid onset. Importantly, the antidepressant-like effects of NMDA receptor antagonists, but also other antidepressants (both classical and novel), require activation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Additionally, expression of AMPA receptors is altered in patients with depression. Moreover, preclinical evidence supports an important involvement of AMPA receptor-dependent signaling and plasticity in the pathophysiology and treatment of depression. Here we summarize work published on the involvement of AMPA receptors in depression and discuss a possible central role for AMPA receptors in the pathophysiology, course and treatment of depression.
Collapse
Affiliation(s)
- Florian Freudenberg
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Frankfurt, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany.
| | - Tansu Celikel
- Department of Neurophysiology, Donders Center for Neuroscience, Radboud University Nijmegen, 6500 AA Nijmegen, The Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Frankfurt, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| |
Collapse
|
16
|
Freudenberg F, Marx V, Seeburg PH, Sprengel R, Celikel T. Circuit mechanisms of GluA1-dependent spatial working memory. Hippocampus 2013; 23:1359-66. [DOI: 10.1002/hipo.22184] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Florian Freudenberg
- Laboratory of Neural Circuits and Plasticity; University of Southern California, 3641 Watt Way; Los Angeles California
- Department of Molecular Neurobiology; Max Planck Institute for Medical Research, Jahnstrasse 29; 69120 Heidelberg Germany
| | - Verena Marx
- Laboratory of Neural Circuits and Plasticity; University of Southern California, 3641 Watt Way; Los Angeles California
- Department of Molecular Neurobiology; Max Planck Institute for Medical Research, Jahnstrasse 29; 69120 Heidelberg Germany
| | - Peter H. Seeburg
- Department of Molecular Neurobiology; Max Planck Institute for Medical Research, Jahnstrasse 29; 69120 Heidelberg Germany
| | - Rolf Sprengel
- Department of Molecular Neurobiology; Max Planck Institute for Medical Research, Jahnstrasse 29; 69120 Heidelberg Germany
| | - Tansu Celikel
- Laboratory of Neural Circuits and Plasticity; University of Southern California, 3641 Watt Way; Los Angeles California
- Department of Neurophysiology; Donders Center for Neuroscience, Radboud University Nijmegen; 6500 AA Nijmegen The Netherlands
| |
Collapse
|
17
|
Meis S, Endres T, Lessmann V. Postsynaptic BDNF signalling regulates long-term potentiation at thalamo-amygdala afferents. J Physiol 2011; 590:193-208. [PMID: 22083603 DOI: 10.1113/jphysiol.2011.220434] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) is known to regulate synaptic plasticity and memory formation in the hippocampus and the neocortex of the mammalian brain. In contrast, a role of BDNF in mediating synaptic plasticity and fear learning in the amygdala is just beginning to evolve. Using patch clamp recordings from projection neurons of the dorsal lateral amygdala (LA) in acute slices of mice, we now investigated the cellular mechanism of BDNF-mediated long-term potentiation (LTP) of excitatory postsynaptic currents (EPSCs) in the amygdala. LTP was elicited in cortical and thalamic synaptic inputs by pairing postsynaptic depolarisation with presynaptic stimulation. LTP in the cortico-amygdala pathway was not changed in heterozygous BDNF-knockout (BDNF(+/-)) mice. In contrast, pairing induced LTP in the thalamic input was abolished in BDNF(+/-) mice (BDNF(+/-): 104.0 ± 5.7% of initial EPSC values; WT: 132.5 ± 7.3%). Likewise, inhibition of BDNF/TrkB signalling with TrkB-IgGs as scavenger molecules for endogenous BDNF blocked LTP in wild-type mice in this pathway (TrkB-IgG: 102.7 ± 6.9% of initial EPSC values; control: 132.5 ± 8.7%). Inclusion of the tyrosine kinase inhibitor K252a in the pipette solution also prevented the induction of LTP in the thalamic pathway, indicating a postsynaptic site of action of BDNF in regulating LTP. Reduced BDNF levels in BDNF(+/-) mice did not affect intrinsic membrane properties of LA projection neurons. Likewise, presynaptic glutamate release, and postsynaptic membrane properties also remained unaffected in BDNF(+/-) mice. These data suggest a postsynaptic site of action of BDNF in mediating LTP selectively in the thalamic fear conditioning pathway.
Collapse
Affiliation(s)
- S Meis
- Institut für Physiologie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany.
| | | | | |
Collapse
|
18
|
Dachtler J, Fox KD, Good MA. Gender specific requirement of GluR1 receptors in contextual conditioning but not spatial learning. Neurobiol Learn Mem 2011; 96:461-7. [PMID: 21810476 DOI: 10.1016/j.nlm.2011.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/12/2011] [Accepted: 07/15/2011] [Indexed: 12/13/2022]
Abstract
The GluR1 subunit of the AMPA receptor is required for hippocampal-dependent memory formation, emotional learning and synaptic plasticity. Recent work has shown that GluR1-independent synaptic plasticity is mediated by nitric oxide. Nitric oxide activity is influenced by estrogen. It is unknown whether this gender-dependent effect conveys a gender dimorphic requirement of GluR1 for learning. This hypothesis was tested in two behavioral paradigms. In Experiment 1, the retention of contextual fear conditioning was impaired in male but not female GluR1 knockout mice. In Experiment 2, GluR1 knockout mice made significantly more arm entry errors during acquisition of a radial-arm watermaze task. This deficit was independent of gender. These results indicate that some forms of learning are gender dimorphic in GluR1 knockout mice. The results are discussed with reference to task and gender-specific interactions between GluR1 receptor intracellular signalling pathways.
Collapse
Affiliation(s)
- James Dachtler
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF103AX, UK.
| | | | | |
Collapse
|
19
|
Sartori SB, Landgraf R, Singewald N. The clinical implications of mouse models of enhanced anxiety. FUTURE NEUROLOGY 2011; 6:531-571. [PMID: 21901080 PMCID: PMC3166843 DOI: 10.2217/fnl.11.34] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Mice are increasingly overtaking the rat model organism in important aspects of anxiety research, including drug development. However, translating the results obtained in mouse studies into information that can be applied in clinics remains challenging. One reason may be that most of the studies so far have used animals displaying 'normal' anxiety rather than 'psychopathological' animal models with abnormal (elevated) anxiety, which more closely reflect core features and sensitivities to therapeutic interventions of human anxiety disorders, and which would, thus, narrow the translational gap. Here, we discuss manipulations aimed at persistently enhancing anxiety-related behavior in the laboratory mouse using phenotypic selection, genetic techniques and/or environmental manipulations. It is hoped that such models with enhanced construct validity will provide improved ways of studying the neurobiology and treatment of pathological anxiety. Examples of findings from mouse models of enhanced anxiety-related behavior will be discussed, as well as their relation to findings in anxiety disorder patients regarding neuroanatomy, neurobiology, genetic involvement and epigenetic modifications. Finally, we highlight novel targets for potential anxiolytic pharmacotherapeutics that have been established with the help of research involving mice. Since the use of psychopathological mouse models is only just beginning to increase, it is still unclear as to the extent to which such approaches will enhance the success rate of drug development in translating identified therapeutic targets into clinical trials and, thus, helping to introduce the next anxiolytic class of drugs.
Collapse
Affiliation(s)
- Simone B Sartori
- Department of Pharmacology & Toxicology, Institute of Pharmacy & Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Peter-Mayr-Street 1, A-6020, Innsbruck, Austria
| | - Rainer Landgraf
- Max Planck Institute of Psychiatry, Department of Behavioral Neuroendocrinology, Munich, Germany
| | - Nicolas Singewald
- Department of Pharmacology & Toxicology, Institute of Pharmacy & Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Peter-Mayr-Street 1, A-6020, Innsbruck, Austria
| |
Collapse
|
20
|
Do GluA1 knockout mice exhibit behavioral abnormalities relevant to the negative or cognitive symptoms of schizophrenia and schizoaffective disorder? Neuropharmacology 2011; 62:1263-72. [PMID: 21693126 DOI: 10.1016/j.neuropharm.2011.06.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 03/28/2011] [Accepted: 06/07/2011] [Indexed: 12/27/2022]
Abstract
The glutamate system has been strongly implicated in the pathophysiology of psychotic illnesses, including schizophrenia and schizoaffective disorder. We recently found that knockout (KO) mice lacking the AMPA GluA1 subunit displayed behavioral abnormalities relevant to some of the positive symptoms of these disorders. Here we phenotyped GluA1 KO mice for behavioral phenotypes pertinent to negative and cognitive/executive symptoms. GluA1 KO mice were tested for conspecific social interactions, the acquisition and extinction of an operant response for food-reward, operant-based pairwise visual discrimination and reversal learning, and impulsive choice in a delay-based cost/benefit decision-making T-maze task. Results showed that GluA1 KO mice engaged in less social interaction than wildtype (WT) controls when tested in a non-habituated, novel environment, but, conversely, displayed more social interaction in a well habituated, familiar environment. GluA1 KO mice were faster to acquire an operant stimulus-response for food reward than WT and were subsequently slower to extinguish the response. Genotypes showed similar pairwise discrimination learning and reversal, although GluA1 KO mice made fewer errors during early reversal. GluA1 KO mice also displayed increased impulsive choice, being less inclined to choose a delayed, larger reward when given a choice between this and a smaller, immediate reward, compared to WT mice. Finally, sucrose preference did not differ between genotypes. Collectively, these data add to the growing evidence that GluA1 KO mice display at least some phenotypic abnormalities mimicking those found in schizophrenia/schizoaffective disorder. Although these mice, like any other single mutant line, are unlikely to model the entire disease, they may nevertheless provide a useful tool for studying the role of GluA1 in certain aspects of the pathophysiology of major psychotic illness.
Collapse
|
21
|
O'Tuathaigh CMP, Desbonnet L, Moran PM, Waddington JL. Susceptibility genes for schizophrenia: mutant models, endophenotypes and psychobiology. Curr Top Behav Neurosci 2011; 12:209-50. [PMID: 22367925 DOI: 10.1007/7854_2011_194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Schizophrenia is characterised by a multifactorial aetiology that involves genetic liability interacting with epigenetic and environmental factors to increase risk for developing the disorder. A consensus view is that the genetic component involves several common risk alleles of small effect and/or rare but penetrant copy number variations. Furthermore, there is increasing evidence for broader, overlapping genetic-phenotypic relationships in psychosis; for example, the same susceptibility genes also confer risk for bipolar disorder. Phenotypic characterisation of genetic models of candidate risk genes and/or putative pathophysiological processes implicated in schizophrenia, as well as examination of epidemiologically relevant gene × environment interactions in these models, can illuminate molecular and pathobiological mechanisms involved in schizophrenia. The present chapter outlines both the evidence from phenotypic studies in mutant mouse models related to schizophrenia and recently described mutant models addressing such gene × environment interactions. Emphasis is placed on evaluating the extent to which mutant phenotypes recapitulate the totality of the disease phenotype or model selective endophenotypes. We also discuss new developments and trends in relation to the functional genomics of psychosis which might help to inform on the construct validity of mutant models of schizophrenia and highlight methodological challenges in phenotypic evaluation that relate to such models.
Collapse
Affiliation(s)
- Colm M P O'Tuathaigh
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland,
| | | | | | | |
Collapse
|
22
|
Does gene deletion of AMPA GluA1 phenocopy features of schizoaffective disorder? Neurobiol Dis 2010; 40:608-21. [PMID: 20699120 PMCID: PMC2955784 DOI: 10.1016/j.nbd.2010.08.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 07/06/2010] [Accepted: 08/03/2010] [Indexed: 12/19/2022] Open
Abstract
Glutamatergic dysfunction is strongly implicated in schizophrenia and
mood disorders. GluA1 knockout (KO) mice display schizophrenia- and
depression-related abnormalities. Here, we asked whether GluA1 KO show
mania-related abnormalities. KO were tested for behavior in approach/avoid
conflict tests, responses to repeated forced swim exposure, and locomotor
responses under stress and after psychostimulant treatment. The effects of rapid
dopamine depletion and treatment with lithium or GSK-3β inhibitor on KO
locomotor hyperactivity were tested. Results showed that KO exhibited novelty-
and stress-induced locomotor hyperactivity, reduced forced swim immobility and
alterations in approach/avoid conflict tests. Psychostimulant treatment and
dopamine depletion exacerbated KO locomotor hyperactivity. Lithium, but not
GSK-3β inhibitor, treatment normalized KO anxiety-related behavior and
partially reversed hyperlocomotor behavior, and also reversed elevated
prefrontal cortex levels of phospho-MARCKS and phospho-neuromodulin.
Collectively, these findings demonstrate mania-related abnormalities in GluA1 KO
and, combined with previous findings, suggest this mutant may provide a novel
model of features of schizoaffective disorder.
Collapse
|
23
|
Loss of GluN2B-containing NMDA receptors in CA1 hippocampus and cortex impairs long-term depression, reduces dendritic spine density, and disrupts learning. J Neurosci 2010; 30:4590-600. [PMID: 20357110 DOI: 10.1523/jneurosci.0640-10.2010] [Citation(s) in RCA: 261] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
NMDA receptors (NMDARs) are key mediators of certain forms of synaptic plasticity and learning. NMDAR complexes are heteromers composed of an obligatory GluN1 subunit and one or more GluN2 (GluN2A-GluN2D) subunits. Different subunits confer distinct physiological and molecular properties to NMDARs, but their contribution to synaptic plasticity and learning in the adult brain remains uncertain. Here, we generated mice lacking GluN2B in pyramidal neurons of cortex and CA1 subregion of hippocampus. We found that hippocampal principal neurons of adult GluN2B mutants had faster decaying NMDAR-mediated EPSCs than nonmutant controls and were insensitive to GluN2B but not NMDAR antagonism. A subsaturating form of hippocampal long-term potentiation (LTP) was impaired in the mutants, whereas a saturating form of LTP was intact. An NMDAR-dependent form of long-term depression (LTD) produced by low-frequency stimulation combined with glutamate transporter inhibition was abolished in the mutants. Additionally, mutants exhibited decreased dendritic spine density in CA1 hippocampal neurons compared with controls. On multiple assays for corticohippocampal-mediated learning and memory (hidden platform Morris water maze, T-maze spontaneous alternation, and pavlovian trace fear conditioning), mutants were impaired. These data further demonstrate the importance of GluN2B for synaptic plasticity in the adult hippocampus and suggest a particularly critical role in LTD, at least the form studied here. The finding that loss of GluN2B was sufficient to cause learning deficits illustrates the contribution of GluN2B-mediated forms of plasticity to memory formation, with implications for elucidating NMDAR-related dysfunction in disease-related cognitive impairment.
Collapse
|
24
|
Fear memory impairing effects of systemic treatment with the NMDA NR2B subunit antagonist, Ro 25-6981, in mice: attenuation with ageing. Pharmacol Biochem Behav 2008; 91:453-60. [PMID: 18809426 DOI: 10.1016/j.pbb.2008.08.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 08/21/2008] [Accepted: 08/26/2008] [Indexed: 12/22/2022]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are mediators of synaptic plasticity and learning and are implicated in the pathophysiology of neuropsychiatric disease and age-related cognitive dysfunction. NMDARs are heteromers, but the relative contribution of specific subunits to NMDAR-mediated learning is not fully understood. We characterized pre-conditioning systemic treatment of the NR2B subunit-selective antagonist Ro 25-6981 for effects on multi-trial, one-trial and low-shock Pavlovian fear conditioning in C57BL/6J mice. Ro 25-6981 was also profiled for effects on novel open field exploration, elevated plus-maze anxiety-like behavior, startle reactivity, prepulse inhibition of startle, and nociception. Three-month (adult) and 12-month old C57BL/6Tac mice were compared for Ro 25-6981 effects on multi-trial fear conditioning, and corticolimbic NR2B protein levels. Ro 25-6981 moderately impaired fear learning in the multi-trial and one-trial (but not low-shock) conditioning paradigms, but did not affect exploratory or anxiety-related behaviors or sensory functions. Memory impairing effects of Ro 25-6981 were absent in 12-month old mice, although NR2B protein levels were not significantly altered. Present data provide further evidence of the memory impairing effects of selective blockade of NR2B-containing NMDARs, and show loss of these effects with ageing. This work could ultimately have implications for elucidating the pathophysiology of learning dysfunction in neuropsychiatric disorders and ageing.
Collapse
|