1
|
Velasco ER, Nabás JF, Torrents-Rodas D, Arias B, Torrubia R, Fullana MA, Andero R. The PAC1 receptor risk genotype does not influence fear acquisition, extinction, or generalization in women with no trauma/low trauma. Biol Psychol 2025; 194:108981. [PMID: 39733787 DOI: 10.1016/j.biopsycho.2024.108981] [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] [Received: 09/03/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
Women are known to have twice as much lifetime prevalence of post-traumatic stress disorder (PTSD) as men do. It has been reported that the risk genotype (CC) of a single nucleotide polymorphism (SNP) (rs2267735) in the pituitary adenylate cyclase-activating polypeptide (PACAP-PAC1R) system is associated with PTSD risk and altered fear conditioning and fear extinction in women. Surprisingly, no previous work has studied the effect of this SNP on fear conditioning, extinction, or generalization in non-traumatized/low trauma load women. Here, two separate groups of women underwent either a two-day fear conditioning and fear extinction paradigm, or a one-day fear conditioning and fear generalization paradigm. Results showed no significant differences between genotypes in conditioned stimulus discrimination, during fear acquisition, extinction, or generalization. These findings suggest that the previously reported fear processing impairments in traumatized CC women are not a consequence of this genotype alone, but likely dependent on the interaction between this genetic risk and the exposure to traumatic stressors.
Collapse
Affiliation(s)
- Eric R Velasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Jaime F Nabás
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - David Torrents-Rodas
- Clinical Psychology and Psychotherapy, Institute of Psychology, Faculty of Psychology and Movement Sciences, Universität Hamburg, Germany
| | - Bárbara Arias
- Secció de Zoologia i Antropologia Biològica (Dpt. Biologia Evolutiva, Ecologia i Ciències Ambientals), Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica En Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Torrubia
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain; Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Miquel A Fullana
- Adult Psychiatry and Psychology Department, Institute of Neurosciences, Hospital Clinic, Barcelona, Spain; Imaging of Mood, and Anxiety-Related Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERSAM, University of Barcelona, Barcelona, Spain
| | - Raül Andero
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain; Centro de Investigación Biomédica En Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; ICREA, Barcelona, Spain.
| |
Collapse
|
2
|
Duggins P, Eliasmith C. A scalable spiking amygdala model that explains fear conditioning, extinction, renewal and generalization. Eur J Neurosci 2024; 59:3093-3116. [PMID: 38616566 DOI: 10.1111/ejn.16338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/03/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024]
Abstract
The amygdala (AMY) is widely implicated in fear learning and fear behaviour, but it remains unclear how the many biological components present within AMY interact to achieve these abilities. Building on previous work, we hypothesize that individual AMY nuclei represent different quantities and that fear conditioning arises from error-driven learning on the synapses between AMY nuclei. We present a computational model of AMY that (a) recreates the divisions and connections between AMY nuclei and their constituent pyramidal and inhibitory neurons; (b) accommodates scalable high-dimensional representations of external stimuli; (c) learns to associate complex stimuli with the presence (or absence) of an aversive stimulus; (d) preserves feature information when mapping inputs to salience estimates, such that these estimates generalize to similar stimuli; and (e) induces a diverse profile of neural responses within each nucleus. Our model predicts (1) defensive responses and neural activities in several experimental conditions, (2) the consequence of artificially ablating particular nuclei and (3) the tendency to generalize defensive responses to novel stimuli. We test these predictions by comparing model outputs to neural and behavioural data from animals and humans. Despite the relative simplicity of our model, we find significant overlap between simulated and empirical data, which supports our claim that the model captures many of the neural mechanisms that support fear conditioning. We conclude by comparing our model to other computational models and by characterizing the theoretical relationship between pattern separation and fear generalization in healthy versus anxious individuals.
Collapse
Affiliation(s)
- Peter Duggins
- Centre for Theoretical Neuroscience, University of Waterloo, Waterloo, Ontario, Canada
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Chris Eliasmith
- Centre for Theoretical Neuroscience, University of Waterloo, Waterloo, Ontario, Canada
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
- Department of Philosophy, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
3
|
Camera F, Colantoni E, Garcia-Sanchez T, Benassi B, Consales C, Muscat A, Vallet L, Mir LM, Andre F, Merla C. In Vitro Imaging and Molecular Characterization of Ca 2+ Flux Modulation by Nanosecond Pulsed Electric Fields. Int J Mol Sci 2023; 24:15616. [PMID: 37958601 PMCID: PMC10647260 DOI: 10.3390/ijms242115616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
In recent years, the application of pulsed electric fields with very short durations (nanoseconds) and extremely high amplitudes (MV/m) has been investigated for novel medical purposes. Various electric protocols have been explored for different objectives, including the utilization of fractionated pulse doses to enhance cell electrosensitization to the uptake of different markers or an increase in apoptosis. This study focused on the use of fluorescence imaging to examine molecular calcium fluxes induced by different fractionated protocols of short electric pulses in neuroblastoma (SH-SY5Y) and mesenchymal stem cells (HaMSCs) that were electroporated using nanosecond pulsed electric fields. In our experimental setup, we did not observe cell electrosensitization in terms of an increase in calcium flux following the administration of fractionated doses of nanosecond pulsed electric fields with respect to the non-fractionated dose. However, we observed the targeted activation of calcium-dependent genes (c-FOS, c-JUN, EGR1, NURR-1, β3-TUBULIN) based on the duration of calcium flux, independent of the instantaneous levels achieved but solely dependent on the final plateau reached. This level of control may have potential applications in various medical and biological treatments that rely on calcium and the delivery of nanosecond pulsed electric fields.
Collapse
Affiliation(s)
- Francesca Camera
- Division of Health Protection Technologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (F.C.); (E.C.); (B.B.); (C.C.)
| | - Eleonora Colantoni
- Division of Health Protection Technologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (F.C.); (E.C.); (B.B.); (C.C.)
| | - Tomas Garcia-Sanchez
- Department of Information and Communication Technologies, Pompeu Fabra University, 08002 Barcelona, Spain;
| | - Barbara Benassi
- Division of Health Protection Technologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (F.C.); (E.C.); (B.B.); (C.C.)
| | - Claudia Consales
- Division of Health Protection Technologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (F.C.); (E.C.); (B.B.); (C.C.)
| | - Adeline Muscat
- CNRS, Metabolic and Systemic Aspects of the Oncogenesis, (METSY), Institute Gustave Roussy, Paris-Saclay University, 94805 Villejuif, France; (A.M.); (L.V.); (L.M.M.); (F.A.)
| | - Leslie Vallet
- CNRS, Metabolic and Systemic Aspects of the Oncogenesis, (METSY), Institute Gustave Roussy, Paris-Saclay University, 94805 Villejuif, France; (A.M.); (L.V.); (L.M.M.); (F.A.)
| | - Luis M. Mir
- CNRS, Metabolic and Systemic Aspects of the Oncogenesis, (METSY), Institute Gustave Roussy, Paris-Saclay University, 94805 Villejuif, France; (A.M.); (L.V.); (L.M.M.); (F.A.)
| | - Franck Andre
- CNRS, Metabolic and Systemic Aspects of the Oncogenesis, (METSY), Institute Gustave Roussy, Paris-Saclay University, 94805 Villejuif, France; (A.M.); (L.V.); (L.M.M.); (F.A.)
| | - Caterina Merla
- Division of Health Protection Technologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (F.C.); (E.C.); (B.B.); (C.C.)
| |
Collapse
|
4
|
Székiová E, Michalová Z, Blaško J, Mucha R, Slovinská L, Kello M, Vanický I. Characterisation of mesenchymal stem cells conditioned media obtained at different conditioning times: their effect on glial cells in in vitro scratch model. Growth Factors 2023; 41:57-70. [PMID: 36825505 DOI: 10.1080/08977194.2023.2182145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023]
Abstract
In this study, the bone marrow mesenchymal stem cells conditioned media (BMMSC-CM) obtained by conditioning for 24(CM24), 48(CM48) and 72(CM72) hours was characterised. In vitro, the impact of BMMSC-CM on the astrocyte migratory response and oligodendrocyte density was evaluated using the scratch model. The proteomic profiles of individual secretomes were analysed by mass spectrometry and the concentrations of four selected neurotrophins (BDNF, NGF, GDNF and VEGF) were determined by ELISA. Our results revealed an increased number of proteins at CM72, many of which are involved in neuroregenerative processes. ELISA documented a gradual increase in the concentration of two neurotrophins (NGF, VEGF), peaking at CM72. In vitro, the different effect of individual BMMSC-CM on astrocyte migration response and oligodendrocyte density was observed, most pronounced with CM72. The outcomes demonstrate that the prolonged conditioning results in increased release of detectable proteins, neurotrophic factors concentration and stronger effect on reparative processes in neural cell cultures.
Collapse
Affiliation(s)
- Eva Székiová
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovakia
| | - Zuzana Michalová
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovakia
| | - Juraj Blaško
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovakia
| | - Rastislav Mucha
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovakia
| | - Lucia Slovinská
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovakia
- Associated Tissue Bank, P. J. Šafárik University and L. Pasteur University Hospital, Košice, Slovakia
| | - Martin Kello
- Department of Pharmacology, P. J. Šafárik University, Košice, Slovakia
| | - Ivo Vanický
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovakia
| |
Collapse
|
5
|
Poli A, Viglione A, Mazziotti R, Totaro V, Morea S, Melani R, Silingardi D, Putignano E, Berardi N, Pizzorusso T. Selective Disruption of Perineuronal Nets in Mice Lacking Crtl1 is Sufficient to Make Fear Memories Susceptible to Erasure. Mol Neurobiol 2023; 60:4105-4119. [PMID: 37022587 DOI: 10.1007/s12035-023-03314-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/09/2023] [Indexed: 04/07/2023]
Abstract
The ability to store, retrieve, and extinguish memories of adverse experiences is an essential skill for animals' survival. The cellular and molecular factors that underlie such processes are only partially known. Using chondroitinase ABC treatment targeting chondroitin sulfate proteoglycans (CSPGs), previous studies showed that the maturation of the extracellular matrix makes fear memory resistant to deletion. Mice lacking the cartilage link protein Crtl1 (Crtl1-KO mice) display normal CSPG levels but impaired CSPG condensation in perineuronal nets (PNNs). Thus, we asked whether the presence of PNNs in the adult brain is responsible for the appearance of persistent fear memories by investigating fear extinction in Crtl1-KO mice. We found that mutant mice displayed fear memory erasure after an extinction protocol as revealed by analysis of freezing and pupil dynamics. Fear memory erasure did not depend on passive loss of retention; moreover, we demonstrated that, after extinction training, conditioned Crtl1-KO mice display no neural activation in the amygdala (Zif268 staining) in comparison to control animals. Taken together, our findings suggest that the aggregation of CSPGs into PNNs regulates the boundaries of the critical period for fear extinction.
Collapse
Affiliation(s)
- Andrea Poli
- BIO@SNS Lab, Scuola Normale Superiore Via G, Moruzzi 1, 56124, Pisa, Italy
| | - Aurelia Viglione
- BIO@SNS Lab, Scuola Normale Superiore Via G, Moruzzi 1, 56124, Pisa, Italy
| | - Raffaele Mazziotti
- Institute of Neuroscience, National Research Council, Via Moruzzi, 1, 56124, Pisa, Italy
| | - Valentino Totaro
- BIO@SNS Lab, Scuola Normale Superiore Via G, Moruzzi 1, 56124, Pisa, Italy
| | - Silvia Morea
- Institute of Neuroscience, National Research Council, Via Moruzzi, 1, 56124, Pisa, Italy
| | - Riccardo Melani
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Davide Silingardi
- Department of Neuroscience, Psychology, Drug Research, and Child Health NEUROFARBA, University of Florence, 50134, Florence, Italy
| | - Elena Putignano
- Institute of Neuroscience, National Research Council, Via Moruzzi, 1, 56124, Pisa, Italy
| | - Nicoletta Berardi
- Institute of Neuroscience, National Research Council, Via Moruzzi, 1, 56124, Pisa, Italy
- Department of Neuroscience, Psychology, Drug Research, and Child Health NEUROFARBA, University of Florence, 50134, Florence, Italy
| | - Tommaso Pizzorusso
- BIO@SNS Lab, Scuola Normale Superiore Via G, Moruzzi 1, 56124, Pisa, Italy.
- Institute of Neuroscience, National Research Council, Via Moruzzi, 1, 56124, Pisa, Italy.
| |
Collapse
|
6
|
Lewis V, Laberge F, Heyland A. Transcriptomic signature of extinction learning in the brain of the fire-bellied toad, Bombina orientalis. Neurobiol Learn Mem 2021; 184:107502. [PMID: 34391934 DOI: 10.1016/j.nlm.2021.107502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/21/2021] [Accepted: 08/08/2021] [Indexed: 11/26/2022]
Abstract
Insight into the molecular and cellular mechanisms of learning and memory from a diverse array of taxa contributes to our understanding of the evolution of these processes. The fire-bellied toad, Bombina orientalis, is a basal anuran amphibian model species who could help us describe shared and divergent characteristics of learning and memory mechanisms between amphibians and other vertebrates, and hence answer questions about the evolution of learning. Utilizing next generation sequencing techniques, we profiled gene expression patterns associated with the extinction of prey-catching conditioning in the brain of the fire-bellied toad. For this purpose, gene expression was at first compared between toads sacrificed after acquisition and extinction of the conditioned response. A second comparison was done between toads submitted to extinction following either short or long acquisition training, which results in toads displaying response extinction or resistance to extinction, respectively. We analyzed brain tissue transcription profiles common to both acquisition and extinction learning, or unique to extinction learning and resistance to extinction, and found significant overlap in gene expression related to molecular pathways involving neuronal plasticity (e.g. structural modification, transcription). However, extinction learning induced a unique GABAergic transcriptomic signal, which may be responsible for suppression of the original response memory. Further, when comparing extinction learning in short- and long-trained groups, short training engaged many pathways related to neuronal plasticity, as expected, but long training engaged molecular pathways related to the suppression of learning through epigenetic mediated transcriptional suppression and inhibitory neurotransmission. Overall, gene expression patterns associated with extinction learning in the fire-bellied toad were similar to those found in mammals submitted to extinction, although some divergent profiles highlighted potential differences in the mechanisms of learning and memory among tetrapods.
Collapse
Affiliation(s)
- Vern Lewis
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Frédéric Laberge
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Andreas Heyland
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
7
|
Gilmartin MR, Ferrara NC. Pituitary Adenylate Cyclase-Activating Polypeptide in Learning and Memory. Front Cell Neurosci 2021; 15:663418. [PMID: 34239418 PMCID: PMC8258392 DOI: 10.3389/fncel.2021.663418] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/02/2021] [Indexed: 02/01/2023] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a highly conserved neuropeptide that regulates neuronal physiology and transcription through Gs/Gq-coupled receptors. Its actions within hypothalamic, limbic, and mnemonic systems underlie its roles in stress regulation, affective processing, neuroprotection, and cognition. Recently, elevated PACAP levels and genetic disruption of PAC1 receptor signaling in humans has been linked to maladaptive threat learning and pathological stress and fear in post-traumatic stress disorder (PTSD). PACAP is positioned to integrate stress and memory in PTSD for which memory of the traumatic experience is central to the disorder. However, PACAP's role in memory has received comparatively less attention than its role in stress. In this review, we consider the evidence for PACAP-PAC1 receptor signaling in learning and plasticity, discuss emerging data on sex differences in PACAP signaling, and raise key questions for further study toward elucidating the contribution of PACAP to adaptive and maladaptive fear learning.
Collapse
Affiliation(s)
| | - Nicole C Ferrara
- Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
8
|
Ressler KJ. Translating Across Circuits and Genetics Toward Progress in Fear- and Anxiety-Related Disorders. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2021; 19:247-255. [PMID: 34690590 PMCID: PMC8475910 DOI: 10.1176/appi.focus.19205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/15/2020] [Indexed: 06/13/2023]
Abstract
(Reprinted with permission from Am J Psychiatry 2020; 177:214-222).
Collapse
|
9
|
Ruiz-López CX, Medina AC, Bello-Medina PC, Quirarte GL, Prado-Alcalá RA. Recruitment of neurons in basolateral amygdala after intense training produces a stronger memory trace. Neurobiol Learn Mem 2021; 181:107428. [PMID: 33798697 DOI: 10.1016/j.nlm.2021.107428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 03/18/2021] [Accepted: 03/28/2021] [Indexed: 11/17/2022]
Abstract
Typical amnestic treatments are ineffective when administered to subjects trained in aversively-motivated tasks using relatively high foot-shock intensities. This effect has been found when treatments that disrupt neuronal activity are administered to different regions of the brain, including the amygdala. However, the molecular mechanisms induced by this intense training are unknown. We made a detailed mapping of c-Fos-expressing neurons in four regions of the amygdala after moderate and intense one-trial inhibitory avoidance training. Rats were sacrificed 90 min after training or after appropriate control procedures, and their brains were prepared for immunohistochemical c-Fos protein detection in the central, lateral, and in the anterior and posterior parts of the basolateral amygdaloid nucleus. We found a high percentage of neurons expressing c-Fos in the anterior part of the basolateral nucleus after moderate training, and this percentage increased further after intense training. Moderate and intense training did not induce changes in c-Fos expression in the other explored amygdaloid regions. These results show that inhibitory avoidance training produces a localized expression of c-Fos in the basolateral anterior nucleus of the amygdala, which is dependent upon the intensity of training, and indicate that synaptic plastic changes in this region may be required for the formation of memory of moderate and intense aversive learning.
Collapse
Affiliation(s)
- C X Ruiz-López
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - A C Medina
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - P C Bello-Medina
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Qro. 76230, Mexico; División de Ciencias de Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Lerma, Estado de México 52005, Mexico
| | - G L Quirarte
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - R A Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico.
| |
Collapse
|
10
|
Hwang H, Szucs MJ, Ding LJ, Allen A, Ren X, Haensgen H, Gao F, Rhim H, Andrade A, Pan JQ, Carr SA, Ahmad R, Xu W. Neurogranin, Encoded by the Schizophrenia Risk Gene NRGN, Bidirectionally Modulates Synaptic Plasticity via Calmodulin-Dependent Regulation of the Neuronal Phosphoproteome. Biol Psychiatry 2021; 89:256-269. [PMID: 33032807 PMCID: PMC9258036 DOI: 10.1016/j.biopsych.2020.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Neurogranin (Ng), encoded by the schizophrenia risk gene NRGN, is a calmodulin-binding protein enriched in the postsynaptic compartments, and its expression is reduced in the postmortem brains of patients with schizophrenia. Experience-dependent translation of Ng is critical for encoding contextual memory, and Ng regulates developmental plasticity in the primary visual cortex during the critical period. However, the overall impact of Ng on the neuronal signaling that regulates synaptic plasticity is unknown. METHODS Altered Ng expression was achieved via virus-mediated gene manipulation in mice. The effect on long-term potentiation (LTP) was accessed using spike timing-dependent plasticity protocols. Quantitative phosphoproteomics analyses led to discoveries in significant phosphorylated targets. An identified candidate was examined with high-throughput planar patch clamp and was validated with pharmacological manipulation. RESULTS Ng bidirectionally modulated LTP in the hippocampus. Decreasing Ng levels significantly affected the phosphorylation pattern of postsynaptic density proteins, including glutamate receptors, GTPases, kinases, RNA binding proteins, selective ion channels, and ionic transporters, some of which highlighted clusters of schizophrenia- and autism-related genes. Hypophosphorylation of NMDA receptor subunit Grin2A, one significant phosphorylated target, resulted in accelerated decay of NMDA receptor currents. Blocking protein phosphatase PP2B activity rescued the accelerated NMDA receptor current decay and the impairment of LTP mediated by Ng knockdown, implicating the requirement of synaptic PP2B activity for the deficits. CONCLUSIONS Altered Ng levels affect the phosphorylation landscape of neuronal proteins. PP2B activity is required for mediating the deficit in synaptic plasticity caused by decreasing Ng levels, revealing a novel mechanistic link of a schizophrenia risk gene to cognitive deficits.
Collapse
Affiliation(s)
- Hongik Hwang
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts; Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.
| | | | - Lei J. Ding
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andrew Allen
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Xiaobai Ren
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Henny Haensgen
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Fan Gao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hyewhon Rhim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Arturo Andrade
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Jen Q. Pan
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Steven A. Carr
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Rushdy Ahmad
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Weifeng Xu
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
11
|
Abstract
Anxiety and fear-related disorders are common and disabling, and they significantly increase risk for suicide and other causes of morbidity and mortality. However, there is tremendous potential for translational neuroscience to advance our understanding of these disorders, leading to novel and powerful interventions and even to preventing their initial development. This overview examines the general circuits and processes thought to underlie fear and anxiety, along with the promise of translational research. It then examines some of the data-driven "next-generation" approaches that are needed for discovery and understanding but that do not always fit neatly into established models. From one perspective, these disorders offer among the most tractable problems in psychiatry, with a great deal of accumulated understanding, across species, of neurocircuit, behavioral, and, increasingly, genetic mechanisms, of how dysregulation of fear and threat processes contributes to anxiety-related disorders. One example is the progressively sophisticated understanding of how extinction underlies the exposure therapy component of cognitive-behavioral therapy approaches, which are ubiquitously used across anxiety and fear-related disorders. However, it is also critical to examine gaps in our understanding between reasonably well-replicated examples of successful translation, areas of significant deficits in knowledge, and the role of large-scale data-driven approaches in future progress and discovery. Although a tremendous amount of progress is still needed, translational approaches to understanding, treating, and even preventing anxiety and fear-related disorders offer great opportunities for successfully bridging neuroscience discovery to clinical practice.
Collapse
|
12
|
Epigenetic regulation of immediate-early gene Nr4a2/Nurr1 in the medial habenula during reinstatement of cocaine-associated behavior. Neuropharmacology 2019; 153:13-19. [PMID: 30998946 DOI: 10.1016/j.neuropharm.2019.04.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/23/2019] [Accepted: 04/13/2019] [Indexed: 01/17/2023]
Abstract
Propensity to relapse following long periods of abstinence is a key feature of substance use disorder. Drugs of abuse, such as cocaine, cause long-term changes in the neural circuitry regulating reward, motivation, and memory processes through dysregulation of various molecular mechanisms, including epigenetic regulation of activity-dependent gene expression. Underlying drug-induced changes to neural circuit function are the molecular mechanisms regulating activity-dependent gene expression. Of note, histone acetyltransferases and histone deacetylases (HDACs), powerful epigenetic regulators of gene expression, are dysregulated following both acute and chronic cocaine exposure and are linked to cocaine-induced changes in neural circuit function. To better understand the effect of drug-induced changes on epigenetic function and behavior, we investigated HDAC3-mediated regulation of Nr4a2/Nurr1 in the medial habenula, an understudied pathway in cocaine-associated behaviors. Nr4a2, a transcription factor critical in cocaine-associated behaviors and necessary for MHb development, is enriched in the cholinergic cell-population of the MHb; yet, the role of NR4A2 within the MHb in the adult brain remains elusive. Here, we evaluated whether epigenetic regulation of Nr4a2 in the MHb has a role in reinstatement of cocaine-associated behaviors. We found that HDAC3 disengages from Nr4a2 in the MHb in response to cocaine-primed reinstatement. Whereas enhancing HDAC3 function in the MHb had no effect on reinstatement, we found, using a dominant-negative splice variant (NURR2C), that loss of NR4A2 function in the MHb blocked reinstatement behaviors. These results show for the first time that regulation of NR4A2 function in the MHb is critical in relapse-like behaviors.
Collapse
|
13
|
López AJ, Siciliano CA, Calipari ES. Activity-Dependent Epigenetic Remodeling in Cocaine Use Disorder. Handb Exp Pharmacol 2019; 258:231-263. [PMID: 31628597 DOI: 10.1007/164_2019_257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Substance use disorder (SUD) is a behavioral disorder characterized by cycles of abstinence, drug seeking, and relapse. SUD is characterized by aberrant learning processes which develop after repeated exposure to drugs of abuse. At the core of this phenotype is the persistence of symptoms, such as craving and relapse to drug seeking, long after the cessation of drug use. The neural basis of these behavioral changes has been linked to dysfunction in neural circuits across the brain; however, the molecular drivers that allow for these changes to persist beyond the lifespan of any individual protein remain opaque. Epigenetic adaptations - where DNA is modified to increase or decrease the probability of gene expression at key genes - have been identified as a mechanism underlying the long-lasting nature of drug-seeking behavior. Thus, to understand SUD, it is critical to define the interplay between neuronal activation and longer-term changes in transcription and epigenetic remodeling and define their role in addictive behaviors. In this review, we discuss the current understanding of drug-induced changes to circuit function, recent discoveries in epigenetic mechanisms that mediate these changes, and, ultimately, how these adaptations drive the persistent nature of relapse, with emphasis on adaptations in models of cocaine use disorder. Understanding the complex interplay between epigenetic gene regulation and circuit activity will be critical in elucidating the neural mechanisms underlying SUD. This, with the advent of novel genetic-based techniques, will allow for the generation of novel therapeutic avenues to improve treatment outcomes in SUD.
Collapse
Affiliation(s)
- Alberto J López
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Department of Molecular Physiology and Biophysics, Vanderbilt Institute for Infection, Immunology, and Infection, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Department of Psychiatry and Behavioral Sciences, Vanderbilt Institute for Infection, Immunology, and Infection, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
14
|
Chaaya N, Battle AR, Johnson LR. An update on contextual fear memory mechanisms: Transition between Amygdala and Hippocampus. Neurosci Biobehav Rev 2018; 92:43-54. [DOI: 10.1016/j.neubiorev.2018.05.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 03/02/2018] [Accepted: 05/08/2018] [Indexed: 12/27/2022]
|
15
|
Lucas EK, Clem RL. GABAergic interneurons: The orchestra or the conductor in fear learning and memory? Brain Res Bull 2018; 141:13-19. [PMID: 29197563 PMCID: PMC6178932 DOI: 10.1016/j.brainresbull.2017.11.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/15/2017] [Accepted: 11/28/2017] [Indexed: 10/18/2022]
Abstract
Fear conditioning is a form of associative learning that is fundamental to survival and involves potentiation of activity in excitatory projection neurons (PNs). Current models stipulate that the mechanisms underlying this process involve plasticity of PN synapses, which exhibit strengthening in response to fear conditioning. However, excitatory PNs are extensively modulated by a diverse array of GABAergic interneurons whose contributions to acquisition, storage, and expression of fear memory remain poorly understood. Here we review emerging evidence that genetically-defined interneurons play important subtype-specific roles in processing of fear-related stimuli and that these dynamics shape PN firing through both inhibition and disinhibition. Furthermore, interneurons exhibit structural, molecular, and electrophysiological evidence of fear learning-induced synaptic plasticity. These studies warrant discarding the notion of interneurons as passive bystanders in long-term memory.
Collapse
Affiliation(s)
- Elizabeth K Lucas
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Roger L Clem
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States.
| |
Collapse
|
16
|
Abstract
De novo protein synthesis is critical for memory formation. We found that protein synthesis during acquisition is transiently required for contextual memory formation. We identified one candidate gene, Nrgn (encoding protein neurogranin, Ng) with enhanced translation upon novel-context exposure, and found that experience-dependent translation of Ng in the hippocampus is required for contextual memory formation. Furthermore, fragile-X mental retardation protein interacts with the 3′UTR of the Nrgn mRNA, which is required for activity-dependent translation of Ng in the synaptic compartment and contextual memory formation. Together, these results indicate that experience-dependent and acute translation of Ng in the hippocampus during memory acquisition enables durable context memory encoding. Experience induces de novo protein synthesis in the brain and protein synthesis is required for long-term memory. It is important to define the critical temporal window of protein synthesis and identify newly synthesized proteins required for memory formation. Using a behavioral paradigm that temporally separates the contextual exposure from the association with fear, we found that protein synthesis during the transient window of context exposure is required for contextual memory formation. Among an array of putative activity-dependent translational neuronal targets tested, we identified one candidate, a schizophrenia-associated candidate mRNA, neurogranin (Ng, encoded by the Nrgn gene) responding to novel-context exposure. The Ng mRNA was recruited to the actively translating mRNA pool upon novel-context exposure, and its protein levels were rapidly increased in the hippocampus. By specifically blocking activity-dependent translation of Ng using virus-mediated molecular perturbation, we show that experience-dependent translation of Ng in the hippocampus is required for contextual memory formation. We further interrogated the molecular mechanism underlying the experience-dependent translation of Ng, and found that fragile-X mental retardation protein (FMRP) interacts with the 3′UTR of the Nrgn mRNA and is required for activity-dependent translation of Ng in the synaptic compartment and contextual memory formation. Our results reveal that FMRP-mediated, experience-dependent, rapid enhancement of Ng translation in the hippocampus during the memory acquisition enables durable context memory encoding.
Collapse
|
17
|
Shvetsov AV, Lopatina NG, Vaido AI. Histone H3 methylation at lysine 4 is involved in long-term memory formation in the honeybee Apis mellifera L. J EVOL BIOCHEM PHYS+ 2018. [DOI: 10.1134/s0022093017060072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Lori A, Maddox SA, Sharma S, Andero R, Ressler KJ, Smith AK. Dynamic Patterns of Threat-Associated Gene Expression in the Amygdala and Blood. Front Psychiatry 2018; 9:778. [PMID: 30705647 PMCID: PMC6344436 DOI: 10.3389/fpsyt.2018.00778] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/24/2018] [Indexed: 12/16/2022] Open
Abstract
Stress and trauma profoundly influence psychiatric biobehavioral outcomes. The identification of treatment and biomarker targets would be accelerated by a broad understanding of the biological responses to these events. The goal of this study was to determine genes responsive to auditory fear conditioning (FC), a well-characterized amygdala-dependent rodent model of threat-exposure, in the presence or absence of prior stress history, providing insight into the physiological processes underlying response to trauma. RNA-sequencing was performed in blood and amygdala from mice that underwent fear conditioning with (Immo+FC) and without (FC) prior immobilization stress, a paradigm that induces HPA axis, and behavioral stress sensitization. In the amygdala, 607 genes were regulated by FC vs. home-cage (HC) controls, and 516 genes differed in stress-sensitized mice (Immo+FC vs. FC). In the former, we observed an enhancement of specific biological processes involved in learning and synaptic transmission, and in the latter processes associated with cell proliferation and the cellular response to drugs. In the blood of stress-sensitized animals, 468 genes were dynamically regulated when compared to FC, and were enriched for the biological pathways of inflammation and cytokine signaling. This study identified genes and pathways that respond to threat in the amygdala and blood of mice with and without a prior stress history and reveals the impact of stress history on subsequent inflammation. Future studies will be needed to examine the role of these dynamically regulated genes may play in human clinical stress and trauma-related disorders.
Collapse
Affiliation(s)
- Adriana Lori
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Stephanie A Maddox
- Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Sumeet Sharma
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States.,Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, United States
| | - Raül Andero
- Institut de Neurociènces, Universitat Autònoma de Barcelona, Bellaterra, Spain.,CIBERSAM, Corporació Sanitaria Parc Taulí, Sabadell, Spain.,Department of Psychobiology and Methodology in Health Sciences, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Kerry J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States.,Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Alicia K Smith
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States.,Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
19
|
Ganglberger F, Kaczanowska J, Penninger JM, Hess A, Bühler K, Haubensak W. Predicting functional neuroanatomical maps from fusing brain networks with genetic information. Neuroimage 2017; 170:113-120. [PMID: 28877513 DOI: 10.1016/j.neuroimage.2017.08.070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 02/07/2023] Open
Abstract
Functional neuroanatomical maps provide a mesoscale reference framework for studies from molecular to systems neuroscience and psychiatry. The underlying structure-function relationships are typically derived from functional manipulations or imaging approaches. Although highly informative, these are experimentally costly. The increasing amount of publicly available brain and genetic data offers a rich source that could be mined to address this problem computationally. Here, we developed an algorithm that fuses gene expression and connectivity data with functional genetic meta data and exploits cumulative effects to derive neuroanatomical maps related to multi-genic functions. We validated the approach by using public available mouse and human data. The generated neuroanatomical maps recapture known functional anatomical annotations from literature and functional MRI data. When applied to multi-genic meta data from mouse quantitative trait loci (QTL) studies and human neuropsychiatric databases, this method predicted known functional maps underlying behavioral or psychiatric traits. Taken together, genetically weighted connectivity analysis (GWCA) allows for high throughput functional exploration of brain anatomy in silico. It maps functional genetic associations onto brain circuitry for refining functional neuroanatomy, or identifying trait-associated brain circuitry, from genetic data.
Collapse
Affiliation(s)
| | - Joanna Kaczanowska
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), 1030, Vienna, Austria
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University Erlangen-Nuremberg, Fahrstrasse 17, 91054, Erlangen, Germany
| | - Katja Bühler
- VRVis Research Center, Donau-City Strasse 11, 1220, Vienna, Austria.
| | - Wulf Haubensak
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria.
| |
Collapse
|
20
|
de Solis CA, Hosek MP, Holehonnur R, Ho A, Banerjee A, Luong JA, Jones LE, Chaturvedi D, Ploski JE. Adeno-associated viral serotypes differentially transduce inhibitory neurons within the rat amygdala. Brain Res 2017; 1672:148-162. [PMID: 28764932 DOI: 10.1016/j.brainres.2017.07.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/21/2017] [Accepted: 07/26/2017] [Indexed: 11/18/2022]
Abstract
Recombinant adeno-associated viruses (AAV) are frequently used to make localized genetic manipulations within the rodent brain. It is accepted that the different viral serotypes possess differing affinities for particular cell types, but it is not clear how these properties affect their ability to transduce specific neuronal cell sub-types. Here, we examined ten AAV serotypes for their ability to transduce neurons within the rat basal and lateral nuclei of the amygdala (BLA) and the central nucleus of the amygdala (CeA). AAV2 based viral genomes designed to express either green fluorescent protein (GFP) from a glutamate decarboxylase (GAD65) promoter or the far-red fluorescent protein (E2-Crimson) from a phosphate-activated glutaminase (PAG) promoter were created and pseudotyped as AAV2/1, AAV2/4, AAV2/5, AAV2/6, AAV2/7, AAV 2/8, AAV2/9, AAV2/rh10, AAV2/DJ and AAV2/DJ8. These viruses were infused into the BLA and CeA at equal titers and twenty-one days later tissue within the amygdala was examined for viral transduction efficiency. These serotypes transduced neurons with similar efficiency, except for AAV4 and AAV5, which exhibited significantly less efficient neuronal transduction. Notably, AAV4 and AAV5 possess the most divergent capsid protein sequences compared to the other commonly available serotypes. We found that the Gad65-GFP virus did not exclusively express GFP within inhibitory neurons, as assessed by fluorescent in situ hybridization (FISH), but when this virus was used to transduce CeA neurons, the majority of the neurons that expressed GFP were in fact inhibitory neurons and this was likely due to the fact that this nucleus contains a very high percentage of inhibitory neurons.
Collapse
Affiliation(s)
- C A de Solis
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, The University of Texas at Dallas, United States
| | - M P Hosek
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, The University of Texas at Dallas, United States
| | - R Holehonnur
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, The University of Texas at Dallas, United States
| | - A Ho
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, The University of Texas at Dallas, United States
| | - A Banerjee
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, The University of Texas at Dallas, United States
| | - J A Luong
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, The University of Texas at Dallas, United States
| | - L E Jones
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, The University of Texas at Dallas, United States
| | - D Chaturvedi
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, The University of Texas at Dallas, United States
| | - J E Ploski
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, The University of Texas at Dallas, United States.
| |
Collapse
|
21
|
Circadian Rhythms in Fear Conditioning: An Overview of Behavioral, Brain System, and Molecular Interactions. Neural Plast 2017; 2017:3750307. [PMID: 28698810 PMCID: PMC5494081 DOI: 10.1155/2017/3750307] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/28/2017] [Accepted: 05/14/2017] [Indexed: 12/17/2022] Open
Abstract
The formation of fear memories is a powerful and highly evolutionary conserved mechanism that serves the behavioral adaptation to environmental threats. Accordingly, classical fear conditioning paradigms have been employed to investigate fundamental molecular processes of memory formation. Evidence suggests that a circadian regulation mechanism allows for a timestamping of such fear memories and controlling memory salience during both their acquisition and their modification after retrieval. These mechanisms include an expression of molecular clocks in neurons of the amygdala, hippocampus, and medial prefrontal cortex and their tight interaction with the intracellular signaling pathways that mediate neural plasticity and information storage. The cellular activities are coordinated across different brain regions and neural circuits through the release of glucocorticoids and neuromodulators such as acetylcholine, which integrate circadian and memory-related activation. Disturbance of this interplay by circadian phase shifts or traumatic experience appears to be an important factor in the development of stress-related psychopathology, considering these circadian components are of critical importance for optimizing therapeutic approaches to these disorders.
Collapse
|
22
|
Olson I, Suryanarayana SM, Robertson B, Grillner S. Griseum centrale, a homologue of the periaqueductal gray in the lamprey. IBRO Rep 2017; 2:24-30. [PMID: 30135930 PMCID: PMC6084820 DOI: 10.1016/j.ibror.2017.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/19/2017] [Accepted: 01/21/2017] [Indexed: 11/27/2022] Open
Abstract
Fear, a response to threatening stimuli and important for survival, is a behavior found throughout the animal kingdom. One critical structure involved in the expression of fear-related behavior is the periaqueductal gray (PAG) in mammals, and in the zebrafish, the griseum centrale. Here, we show in the lamprey, belonging to the oldest now living group of vertebrates, that a bilateral periventricular nucleus in the ventral mesencephalon has a similar location to that of the PAG and griseum centrale. It targets the pretectum and the substantia nigra pars compacta (SNc), expresses the dopamine D1 and D2 receptors and receives input from the pallium (cortex in mammals), hypothalamus, the raphe area and SNc. These are all hallmarks of the mammalian PAG. In addition, like in the zebrafish, there is an input from the interpeduncular nucleus. Our results thus suggest that a structure homologous to the PAG/griseum centrale was present very early in vertebrate evolution. A homologue of the mammalian PAG is present in the lamprey. As in the zebrafish, this structure is named griseum centrale. The neuronal circuitry for fear-related behavior is evolutionarily conserved.
Collapse
Affiliation(s)
- Ian Olson
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Shreyas M Suryanarayana
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Brita Robertson
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Sten Grillner
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| |
Collapse
|
23
|
Cortés R, Vilaró MT, Mengod G. Visualization of 5-HT Receptors Using Radioligand-Binding Autoradiography. CURRENT PROTOCOLS IN PHARMACOLOGY 2016; 75:8.3.1-8.3.20. [PMID: 27960027 DOI: 10.1002/cpph.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Described in this unit are techniques to visualize the majority of serotonin (5-hydroxytryptamine, 5-HT) receptor subtypes in sections of frozen brain tissue using receptor autoradiography. Protocols for brain extraction and sectioning, radioligand exposure, autoradiogram generation, and data quantification are provided, as are the optimal incubation conditions for the autoradiographic visualization of receptors using agonist and antagonist radioligands. © 2016 by John Wiley & Sons, Inc.
Collapse
|
24
|
Taguchi YH. Principal component analysis based unsupervised feature extraction applied to publicly available gene expression profiles provides new insights into the mechanisms of action of histone deacetylase inhibitors. NEUROEPIGENETICS 2016; 8:1-18. [DOI: 10.1016/j.nepig.2016.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
25
|
Cho JH, Huang BS, Gray JM. RNA sequencing from neural ensembles activated during fear conditioning in the mouse temporal association cortex. Sci Rep 2016; 6:31753. [PMID: 27557751 PMCID: PMC4997356 DOI: 10.1038/srep31753] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 07/25/2016] [Indexed: 12/13/2022] Open
Abstract
The stable formation of remote fear memories is thought to require neuronal gene induction in cortical ensembles that are activated during learning. However, the set of genes expressed specifically in these activated ensembles is not known; knowledge of such transcriptional profiles may offer insights into the molecular program underlying stable memory formation. Here we use RNA-Seq to identify genes whose expression is enriched in activated cortical ensembles labeled during associative fear learning. We first establish that mouse temporal association cortex (TeA) is required for remote recall of auditory fear memories. We then perform RNA-Seq in TeA neurons that are labeled by the activity reporter Arc-dVenus during learning. We identify 944 genes with enriched expression in Arc-dVenus+ neurons. These genes include markers of L2/3, L5b, and L6 excitatory neurons but not glial or inhibitory markers, confirming Arc-dVenus to be an excitatory neuron-specific but non-layer-specific activity reporter. Cross comparisons to other transcriptional profiles show that 125 of the enriched genes are also activity-regulated in vitro or induced by visual stimulus in the visual cortex, suggesting that they may be induced generally in the cortex in an experience-dependent fashion. Prominent among the enriched genes are those encoding potassium channels that down-regulate neuronal activity, suggesting the possibility that part of the molecular program induced by fear conditioning may initiate homeostatic plasticity.
Collapse
Affiliation(s)
- Jin-Hyung Cho
- Harvard Medical School, Genetics Department, 77 Ave Louis Pasteur NRB Room 356, Boston, Massachusetts 02115, USA
| | - Ben S Huang
- Harvard Medical School, Genetics Department, 77 Ave Louis Pasteur NRB Room 356, Boston, Massachusetts 02115, USA.,University of California at Los Angeles, David Geffen School of Medicine, Department of Neurology, 710 Westwood Plaza, Los Angeles, California 90095, USA
| | - Jesse M Gray
- Harvard Medical School, Genetics Department, 77 Ave Louis Pasteur NRB Room 356, Boston, Massachusetts 02115, USA
| |
Collapse
|
26
|
Jasnow AM, Lynch JF, Gilman TL, Riccio DC. Perspectives on fear generalization and its implications for emotional disorders. J Neurosci Res 2016; 95:821-835. [PMID: 27448175 DOI: 10.1002/jnr.23837] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/28/2022]
Abstract
Although generalization to conditioned stimuli is not a new phenomenon, renewed interest in understanding its biological underpinning has stemmed from its association with a number of anxiety disorders. Generalization as it relates to fear processing is a temporally dynamic process in which animals, including humans, display fear in response to similar yet distinct cues or contexts as the time between training and testing increases. This Review surveys the literature on contextual fear generalization and its relation to several views of memory, including systems consolidation, forgetting, and transformation hypothesis, which differentially implicate roles of the hippocampus and neocortex in memory consolidation and retrieval. We discuss recent evidence on the neurobiological mechanisms contributing to the increase in fear generalization over time and how generalized responding may be modulated by acquisition, consolidation, and retrieval mechanisms. Whereas clinical perspectives of generalization emphasize a lack of fear inhibition to CS- cues or fear toward intermediate CS cues, the time-dependent nature of generalization and its relation to traditional views on memory consolidation and retrieval are often overlooked. Understanding the time-dependent increase in fear generalization has important implications not only for understanding how generalization contributes to anxiety disorders but also for understanding basic long-term memory function. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aaron M Jasnow
- Department of Psychological Sciences, Kent State University, Kent, Ohio
| | - Joseph F Lynch
- Department of Psychological Sciences, Kent State University, Kent, Ohio
| | - T Lee Gilman
- Department of Psychological Sciences, Kent State University, Kent, Ohio
| | - David C Riccio
- Department of Psychological Sciences, Kent State University, Kent, Ohio
| |
Collapse
|
27
|
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) has recently emerged as a possible molecular mediator of activity-dependent synaptic plasticity underlying learning and memory. Long-term potentiation (LTP) within the hippocampus and hippocampally dependent behaviors has been the primary model for examining the role of BDNF in learning and memory. However, these studies are limited by an incomplete understanding of the complex behavioral function of hippocampal circuitry, making it difficult to unravel the molecular machinery responsible for the formation and storage of these memories. In contrast, the amygdala and its role in Pavlovian fear conditioning promise to provide us with new insights into the mechanisms of BDNF-mediated synaptic plasticity during the learning and memory process. This article reviews the different levels of research on BDNF in learning and memory. The focus is primarily on the use of Pavlovian fear conditioning as a learning model that allows for the examination of the role of BDNF in the amygdala, following a single learning session and within a well-understood neural circuit.
Collapse
Affiliation(s)
- Lisa M Rattiner
- Emory University School of Medicine, Department of Psychiatry, Center for Behavioral Neuroscience, Atlanta, Georgia 30329, USA
| | | | | |
Collapse
|
28
|
Kim S, Kim T, Lee HR, Kong YY, Kaang BK. Mind Bomb-2 Regulates Hippocampus-dependent Memory Formation and Synaptic Plasticity. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:515-22. [PMID: 26557018 PMCID: PMC4637354 DOI: 10.4196/kjpp.2015.19.6.515] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/03/2015] [Accepted: 07/15/2015] [Indexed: 01/06/2023]
Abstract
Notch signaling is a key regulator of neuronal fate during embryonic development, but its function in the adult brain is still largely unknown. Mind bomb-2 (Mib2) is an essential positive regulator of the Notch pathway, which acts in the Notch signal-sending cells. Therefore, genetic deletion of Mib2 in the mouse brain might help understand Notch signaling-mediated cell-cell interactions between neurons and their physiological function. Here we show that deletion of Mib2 in the mouse brain results in impaired hippocampal spatial memory and contextual fear memory. Accordingly, we found impaired hippocampal synaptic plasticity in Mib2 knock-out (KO) mice; however, basal synaptic transmission did not change at the Schaffer collateral-CA1 synapses. Using western blot analysis, we found that the level of cleaved Notch1 was lower in Mib2 KO mice than in wild type (WT) littermates after mild foot shock. Taken together, these data suggest that Mib2 plays a critical role in synaptic plasticity and spatial memory through the Notch signaling pathway.
Collapse
Affiliation(s)
- Somi Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - TaeHyun Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Hye-Ryeon Lee
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Young-Yun Kong
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Bong-Kiun Kaang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
29
|
Baker KD, Richardson R. Forming competing fear learning and extinction memories in adolescence makes fear difficult to inhibit. ACTA ACUST UNITED AC 2015; 22:537-43. [PMID: 26472643 PMCID: PMC4749725 DOI: 10.1101/lm.039487.114] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/21/2015] [Indexed: 01/06/2023]
Abstract
Fear inhibition is markedly impaired in adolescent rodents and humans. The present experiments investigated whether this impairment is critically determined by the animal's age at the time of fear learning or their age at fear extinction. Male rats (n = 170) were tested for extinction retention after conditioning and extinction at different ages. We examined neural correlates of impaired extinction retention by detection of phosphorylated mitogen-activated protein kinase immunoreactivity (pMAPK-IR) in several brain regions. Unexpectedly, adolescent rats exhibited good extinction retention if fear was acquired before adolescence. Further, fear acquired in adolescence could be successfully extinguished in adulthood but not within adolescence. Adolescent rats did not show extinction-induced increases in pMAPK-IR in the medial prefrontal cortex or the basolateral amygdala, or a pattern of reduced caudal central amygdala pMAPK-IR, as was observed in juveniles. This dampened prefrontal and basolateral amygdala MAPK activation following extinction in adolescence occurred even when there was no impairment in extinction retention. In contrast, only adolescent animals that exhibited impaired extinction retention showed elevated pMAPK-IR in the posterior paraventricular thalamus. These data suggest that neither the animal's age at the time of fear acquisition or extinction determines whether impaired extinction retention is exhibited. Rather, it appears that forming competing fear conditioning and extinction memories in adolescence renders this a vulnerable developmental period in which fear is difficult to inhibit. Furthermore, even under conditions that promote good extinction, the neural correlates of extinction in adolescence are different than those recruited in animals of other ages.
Collapse
Affiliation(s)
- Kathryn D Baker
- School of Psychology, UNSW Australia, Sydney, New South Wales 2052, Australia
| | - Rick Richardson
- School of Psychology, UNSW Australia, Sydney, New South Wales 2052, Australia
| |
Collapse
|
30
|
Activity of the anterior cingulate cortex and ventral hippocampus underlie increases in contextual fear generalization. Neurobiol Learn Mem 2015; 124:19-27. [DOI: 10.1016/j.nlm.2015.07.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/30/2015] [Accepted: 07/02/2015] [Indexed: 11/21/2022]
|
31
|
Donev R, Alawam K. Alterations in Gene Expression in Depression: Prospects for Personalize Patient Treatment. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 101:97-124. [PMID: 26572977 DOI: 10.1016/bs.apcsb.2015.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The number of people around the world suffering from depression has dramatically increased in last few decades. It has been predicted that by 2020 depression will become the second most common cause of disability. Furthermore, depression is often misdiagnosed and confused with other psychiatric disorders showing similar symptoms, i.e., anxiety and bipolar disorder, due to the fact that diagnosing is often carried out by medical workers who are not psychiatrically trained. These facts prompt us to prepare this review which focuses on alterations in gene expression in depression. We believe that an in-depth knowledge of molecular bases of behavior in depression and other mood disorders would be of a great benefit for the correct diagnosing of these disorders, as well as for prescribing a treatment that best suits each individual depending on expression alterations in depression-related genes. Therefore, the main aim of this review is to promote further translational research on the biochemistry of mood disorders and take the results further for the design of new targeted therapeutics that can be used for personalized treatment with minimal adverse effects.
Collapse
Affiliation(s)
| | - Khaled Alawam
- Forensic Medicine Department, Ministry of Interior, Kuwait City, Kuwait
| |
Collapse
|
32
|
Voxel-based morphometry predicts shifts in dendritic spine density and morphology with auditory fear conditioning. Nat Commun 2015; 6:7582. [PMID: 26151911 PMCID: PMC4506522 DOI: 10.1038/ncomms8582] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/21/2015] [Indexed: 01/27/2023] Open
Abstract
Neuroimaging has provided compelling data about the brain. Yet the underlying mechanisms of many neuroimaging techniques have not been elucidated. Here we report a voxel-based morphometry (VBM) study of Thy1-YFP mice following auditory fear conditioning complemented by confocal microscopy analysis of cortical thickness, neuronal morphometric features and nuclei size/density. Significant VBM results included the nuclei of the amygdala, the insula and the auditory cortex. There were no significant VBM changes in a control brain area. Focusing on the auditory cortex, confocal analysis showed that fear conditioning led to a significantly increased density of shorter and wider dendritic spines, while there were no spine differences in the control area. Of all the morphology metrics studied, the spine density was the only one to show significant correlation with the VBM signal. These data demonstrate that learning-induced structural changes detected by VBM may be partially explained by increases in dendritic spine density. Voxel-based morphometry (VBM) involves comparisons of high resolution structural images of the brain between groups, but what causes changes in the VBM signal is unclear. Here the authors perform a VBM study of Thy1-YFP mice following auditory fear conditioning and propose that the signal changes can be partially explained by increases in dendritic spine density.
Collapse
|
33
|
Deletion of Rapgef6, a candidate schizophrenia susceptibility gene, disrupts amygdala function in mice. Transl Psychiatry 2015; 5:e577. [PMID: 26057047 PMCID: PMC4490285 DOI: 10.1038/tp.2015.75] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/22/2015] [Accepted: 04/23/2015] [Indexed: 02/01/2023] Open
Abstract
In human genetic studies of schizophrenia, we uncovered copy-number variants in RAPGEF6 and RAPGEF2 genes. To discern the effects of RAPGEF6 deletion in humans, we investigated the behavior and neural functions of a mouse lacking Rapgef6. Rapgef6 deletion resulted in impaired amygdala function measured as reduced fear conditioning and anxiolysis. Hippocampal-dependent spatial memory and prefrontal cortex-dependent working memory tasks were intact. Neural activation measured by cFOS phosphorylation demonstrated a reduction in hippocampal and amygdala activation after fear conditioning, while neural morphology assessment uncovered reduced spine density and primary dendrite number in pyramidal neurons of the CA3 hippocampal region of knockout mice. Electrophysiological analysis showed enhanced long-term potentiation at cortico-amygdala synapses. Rapgef6 deletion mice were most impaired in hippocampal and amygdalar function, brain regions implicated in schizophrenia pathophysiology. The results provide a deeper understanding of the role of the amygdala in schizophrenia and suggest that RAPGEF6 may be a novel therapeutic target in schizophrenia.
Collapse
|
34
|
Cummings ME. The mate choice mind: studying mate preference, aversion and social cognition in the female poeciliid brain. Anim Behav 2015. [DOI: 10.1016/j.anbehav.2015.02.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Bowers ME, Xia B, Carreiro S, Ressler KJ. The Class I HDAC inhibitor RGFP963 enhances consolidation of cued fear extinction. ACTA ACUST UNITED AC 2015; 22:225-31. [PMID: 25776040 PMCID: PMC4371170 DOI: 10.1101/lm.036699.114] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Evidence indicates that broad, nonspecific histone deacetylase (HDAC) inhibition enhances learning and memory, however, the contribution of the various HDACs to specific forms of learning is incompletely understood. Here, we show that the Class I HDAC inhibitor, RGFP963, enhances consolidation of cued fear extinction. However, RGFP966, a strong inhibitor of HDAC3, does not significantly enhance consolidation of cued fear extinction. These data extend previous evidence that demonstrate the Class I HDACs play a role in the consolidation of long-term memory, suggesting that HDAC1 and/or HDAC2, but less likely HDAC3, may function as negative regulators of extinction retention. The development of specific HDAC inhibitors, such as RGFP963, will further illuminate the role of specific HDACs in various types of learning and memory. Moreover, HDAC inhibitors that enhance cued fear extinction may show translational promise for the treatment of fear-related disorders, including post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Mallory E Bowers
- Behavioral Neuroscience and Psychiatric Disorders, Emory University, Atlanta, Georgia 30329, USA
| | - Bing Xia
- RepliGen Waltham, Massachusetts 02453, USA
| | | | - Kerry J Ressler
- Behavioral Neuroscience and Psychiatric Disorders, Emory University, Atlanta, Georgia 30329, USA Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
36
|
Affiliation(s)
- James L. McGaugh
- Center for the Neurobiology of Learning and Memory and Department of Neurobiology and Behavior, University of California, Irvine, California 92697-3800;
| |
Collapse
|
37
|
Grinkevich LN, Vorobiova OV. Role of modulatory mediator serotonin in induction of epigenetic processes during long-term memory formation in Helix. ACTA ACUST UNITED AC 2014. [DOI: 10.1134/s2079059714060094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Petrini EM, Barberis A. Diffusion dynamics of synaptic molecules during inhibitory postsynaptic plasticity. Front Cell Neurosci 2014; 8:300. [PMID: 25294987 PMCID: PMC4171989 DOI: 10.3389/fncel.2014.00300] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/05/2014] [Indexed: 12/24/2022] Open
Abstract
The plasticity of inhibitory transmission is expected to play a key role in the modulation of neuronal excitability and network function. Over the last two decades, the investigation of the determinants of inhibitory synaptic plasticity has allowed distinguishing presynaptic and postsynaptic mechanisms. While there has been a remarkable progress in the characterization of presynaptically-expressed plasticity of inhibition, the postsynaptic mechanisms of inhibitory long-term synaptic plasticity only begin to be unraveled. At postsynaptic level, the expression of inhibitory synaptic plasticity involves the rearrangement of the postsynaptic molecular components of the GABAergic synapse, including GABAA receptors, scaffold proteins and structural molecules. This implies a dynamic modulation of receptor intracellular trafficking and receptor surface lateral diffusion, along with regulation of the availability and distribution of scaffold proteins. This Review will focus on the mechanisms of the multifaceted molecular reorganization of the inhibitory synapse during postsynaptic plasticity, with special emphasis on the key role of protein dynamics to ensure prompt and reliable activity-dependent adjustments of synaptic strength.
Collapse
Affiliation(s)
- Enrica Maria Petrini
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genoa, Italy
| | - Andrea Barberis
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genoa, Italy
| |
Collapse
|
39
|
Caroni P, Chowdhury A, Lahr M. Synapse rearrangements upon learning: from divergent-sparse connectivity to dedicated sub-circuits. Trends Neurosci 2014; 37:604-14. [PMID: 25257207 DOI: 10.1016/j.tins.2014.08.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 01/24/2023]
Abstract
Learning can involve formation of new synapses and loss of synapses, providing memory traces of learned skills. Recent findings suggest that these synapse rearrangements reflect assembly of task-related sub-circuits from initially broadly distributed and sparse connectivity in the brain. These local circuit remodeling processes involve rapid emergence of synapses upon learning, followed by protracted validation involving strengthening of some new synapses, and selective elimination of others. The timing of these consolidation processes can vary. Here, we review these findings, focusing on how molecular/cellular mechanisms of synapse assembly, strengthening, and elimination might interface with circuit/system mechanisms of learning and memory consolidation. An integrated understanding of these learning-related processes should provide a better basis to elucidate how experience, genetic background, and disease influence brain function.
Collapse
Affiliation(s)
- Pico Caroni
- Friedrich Miescher Institut, Basel, Switzerland.
| | | | - Maria Lahr
- Friedrich Miescher Institut, Basel, Switzerland
| |
Collapse
|
40
|
Andero R, Dias BG, Ressler KJ. A role for Tac2, NkB, and Nk3 receptor in normal and dysregulated fear memory consolidation. Neuron 2014; 83:444-454. [PMID: 24976214 DOI: 10.1016/j.neuron.2014.05.028] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2014] [Indexed: 01/06/2023]
Abstract
The centromedial amygdala (CeM), a subdivision of the central amygdala (CeA), is believed to be the main output station of the amygdala for fear expression. We provide evidence that the Tac2 gene, expressed by neurons specifically within the CeM, is required for modulating fear memories. Tac2 is colocalized with GAD65 and CaMKIIα but not with PKCd and Enk neurons in the CeM. Moreover, the Tac2 product, NkB, and its specific receptor, Nk3R, are also involved in the consolidation of fear memories. Increased Tac2 expression, through a stress-induced PTSD-like model, or following lentiviral CeA overexpression, are sufficient to enhance fear consolidation. This effect is blocked by the Nk3R antagonist osanetant. Concordantly, silencing of Tac2-expressing neurons in CeA with DREADDs impairs fear consolidation. Together, these studies further our understanding of the role of the Tac2 gene and CeM in fear processing and may provide approaches to intervention for fear-related disorders.
Collapse
Affiliation(s)
- Raül Andero
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, USA; Center for Behavioral Neuroscience, Yerkes National Primate Research Center, Atlanta, GA 30329, USA.
| | - Brian G Dias
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, USA; Center for Behavioral Neuroscience, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Kerry J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, USA; Center for Behavioral Neuroscience, Yerkes National Primate Research Center, Atlanta, GA 30329, USA; Howard Hughes Medical Institute, Bethesda, MD 20814, USA
| |
Collapse
|
41
|
The neuronal activity-driven transcriptome. Mol Neurobiol 2014; 51:1071-88. [PMID: 24935719 DOI: 10.1007/s12035-014-8772-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/01/2014] [Indexed: 10/25/2022]
Abstract
Activity-driven transcription is a key event associated with long-lasting forms of neuronal plasticity. Despite the efforts to investigate the regulatory mechanisms that control this complex process and the important advances in the knowledge of the function of many activity-induced genes in neurons, as well as the specific contribution of activity-regulated transcription factors, our understanding of how activity-driven transcription operates at the systems biology level is still very limited. This review focuses on the research of neuronal activity-driven transcription from an "omics" perspective. We will discuss the different high-throughput approaches undertaken to characterize the gene programs downstream of specific activity-regulated transcription factors, including CREB, SRF, MeCP2, Fos, Npas4, and others, and the interplay between epigenetic and transcriptional mechanisms underlying neuronal plasticity changes. Although basic questions remain unanswered and important challenges still lie ahead, the refinement of genome-wide techniques for investigating the neuronal transcriptome and epigenome promises great advances.
Collapse
|
42
|
Xu N, Zhou WJ, Wang Y, Huang SH, Li X, Chen ZY. Hippocampal Wnt3a is Necessary and Sufficient for Contextual Fear Memory Acquisition and Consolidation. Cereb Cortex 2014; 25:4062-75. [PMID: 24904070 DOI: 10.1093/cercor/bhu121] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Wnt signaling pathway plays critical roles in development. However, to date, the role of Wnts in learning and memory in adults is still not well understood. Here, we aimed to investigate the roles and mechanisms of Wnts in hippocampal-dependent contextual fear conditioning (CFC) memory formation in adult mice. CFC training induced the secretion and expression of Wnt3a and the activation of its downstream Wnt/Ca(2+) and Wnt/β-catenin signaling pathways in the dorsal hippocampus (DH). Intrahippocampal infusion of Wnt3a antibody impaired CFC acquisition and consolidation, but not expression. Using the Wnt antagonist sFRP1 or the canonical Wnt inhibitor Dkk1, we found that Wnt/Ca(2+) and Wnt/β-catenin signaling pathways were involved in acquisition and consolidation, respectively. Moreover, we found Wnt3a signaling is not only necessary but also sufficient for CFC memory. Intrahippocampal infusion of exogenous Wnt3a could enhance acquisition and consolidation of CFC. Overexpression of constitutively active β-catenin in the DH could rescue the deficit in CFC memory consolidation, but not acquisition induced by Wnt3a antibody injection, which suggests β-catenin signaling pathway acts downstream of Wnt3a to mediate CFC memory consolidation. Our study may help further the understanding of the precise regulation of Wnt3a in differential memory phases depending on divergent signaling pathways.
Collapse
Affiliation(s)
- Ning Xu
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wen-Juan Zhou
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yue Wang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shu-Hong Huang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xian Li
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhe-Yu Chen
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
43
|
Grinkevich LN. Studies of Histone H3 Methylation during the Formation of Long-Term Memory. ACTA ACUST UNITED AC 2014. [DOI: 10.1007/s11055-014-9953-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
44
|
Holehonnur R, Luong JA, Chaturvedi D, Ho A, Lella SK, Hosek MP, Ploski JE. Adeno-associated viral serotypes produce differing titers and differentially transduce neurons within the rat basal and lateral amygdala. BMC Neurosci 2014; 15:28. [PMID: 24533621 PMCID: PMC3937004 DOI: 10.1186/1471-2202-15-28] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 02/11/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In recent years, there has been an increased interest in using recombinant adeno-associated viruses (AAV) to make localized genetic manipulations within the rodent brain. Differing serotypes of AAV possess divergent capsid protein sequences and these variations greatly influence each serotype's ability to transduce particular cell types and brain regions. We therefore aimed to determine the AAV serotype that is optimal for targeting neurons within the Basal and Lateral Amygdala (BLA) since the transduction efficiency of AAV has not been previously examined within the BLA. This region is desirable to genetically manipulate due to its role in emotion, learning & memory, and numerous psychiatric disorders. We accomplished this by screening 9 different AAV serotypes (AAV2/1, AAV2/2, AAV2/5, AAV2/7, AAV2/8, AAV2/9, AAV2/rh10, AAV2/DJ and AAV2/DJ8) designed to express red fluorescent protein (RFP) under the regulation of an alpha Ca2+/calmodulin-dependent protein kinase II promoter (αCaMKII). RESULTS We determined that these serotypes produce differing amounts of virus under standard laboratory production. Notably AAV2/2 consistently produced the lowest titers compared to the other serotypes examined. These nine serotypes were bilaterally infused into the rat BLA at the highest titers achieved for each serotype and at a normalized titer of 7.8E + 11 GC/ml. Twenty one days following viral infusion the degree of transduction was quantitated throughout the amygdala. These viruses exhibited differential transduction of neurons within the BLA. AAV2/7 exhibited a trend toward having the highest efficiency of transduction and AAV2/5 exhibited significantly lower transduction efficiency as compared to the serotypes examined. AAV2/5's decreased ability to transduce BLA neurons correlates with its significantly different capsid protein sequences as compared to the other serotypes examined. CONCLUSIONS For laboratories producing their own recombinant adeno-associated viruses, the use of AAV2/2 is likely less desirable since AAV2/2 produces significantly lower titers than many other serotypes of AAV. Numerous AAV serotypes appear to efficiently transduce BLA neurons, with the exception of AAV2/5. Taking into consideration the ability of certain serotypes to achieve high titers and transduce BLA neurons well, in our hands AAV2/DJ8 and AAV2/9 appear to be ideal serotypes to use when targeting neurons within the BLA.
Collapse
Affiliation(s)
- Roopashri Holehonnur
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, University of Texas at Dallas, 800 West Campbell RD, Richardson, TX 75080, USA
| | - Jonathan A Luong
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, University of Texas at Dallas, 800 West Campbell RD, Richardson, TX 75080, USA
| | - Dushyant Chaturvedi
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, University of Texas at Dallas, 800 West Campbell RD, Richardson, TX 75080, USA
| | - Anthony Ho
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, University of Texas at Dallas, 800 West Campbell RD, Richardson, TX 75080, USA
| | - Srihari K Lella
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, University of Texas at Dallas, 800 West Campbell RD, Richardson, TX 75080, USA
| | - Matthew P Hosek
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, University of Texas at Dallas, 800 West Campbell RD, Richardson, TX 75080, USA
| | - Jonathan E Ploski
- School of Behavioral and Brain Sciences and the Department of Molecular & Cell Biology, University of Texas at Dallas, 800 West Campbell RD, Richardson, TX 75080, USA
| |
Collapse
|
45
|
Andero R, Brothers SP, Jovanovic T, Chen YT, Salah-Uddin H, Cameron M, Bannister TD, Almli L, Stevens JS, Bradley B, Binder EB, Wahlestedt C, Ressler KJ. Amygdala-dependent fear is regulated by Oprl1 in mice and humans with PTSD. Sci Transl Med 2014; 5:188ra73. [PMID: 23740899 DOI: 10.1126/scitranslmed.3005656] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The amygdala-dependent molecular mechanisms driving the onset and persistence of posttraumatic stress disorder (PTSD) are poorly understood. Recent observational studies have suggested that opioid analgesia in the aftermath of trauma may decrease the development of PTSD. Using a mouse model of dysregulated fear, we found altered expression within the amygdala of the Oprl1 gene (opioid receptor-like 1), which encodes the amygdala nociceptin (NOP)/orphanin FQ receptor (NOP-R). Systemic and central amygdala infusion of SR-8993, a new highly selective NOP-R agonist, impaired fear memory consolidation. In humans, a single-nucleotide polymorphism (SNP) within OPRL1 is associated with a self-reported history of childhood trauma and PTSD symptoms (n = 1847) after a traumatic event. This SNP is also associated with physiological startle measures of fear discrimination and magnetic resonance imaging analysis of amygdala-insula functional connectivity. Together, these data suggest that Oprl1 is associated with amygdala function, fear processing, and PTSD symptoms. Further, our data suggest that activation of the Oprl1/NOP receptor may interfere with fear memory consolidation, with implications for prevention of PTSD after a traumatic event.
Collapse
Affiliation(s)
- Raül Andero
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mou L, Dias BG, Gosnell H, Ressler KJ. Gephyrin plays a key role in BDNF-dependent regulation of amygdala surface GABAARs. Neuroscience 2013; 255:33-44. [PMID: 24096136 DOI: 10.1016/j.neuroscience.2013.09.051] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 09/18/2013] [Accepted: 09/23/2013] [Indexed: 11/19/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is critically involved in synaptic plasticity and neurotransmission. Our lab has previously found that BDNF activation of neurotrophic tyrosine kinase, receptor, type 2 (TrkB) is required for fear memory formation and that GABAA receptor (GABAAR) subunits and the GABAA clustering protein gephyrin are dynamically regulated during fear memory consolidation. We hypothesize that TrkB-dependent internalization of GABAARs may partially underlie a transient period of amygdala hyperactivation during fear memory consolidation. We have previously reported that BDNF modulates GABAAR α1 subunit sequestration in cultured hippocampal and amygdala neurons by differential phosphorylation pathways. At present, no studies have investigated the regulation of gephyrin and GABAAR α1 subunits following BDNF activation in the amygdala. In this study, we confirm the association of GABAAR α1 and γ2 subunits with gephyrin on mouse amygdala neurons by coimmunoprecipitation and immunocytochemistry. We then demonstrate that rapid BDNF treatment, as well as suppression of gephyrin protein levels on amygdala neurons, induced sequestration of surface α1 subunits. Further, we find that rapid exposure of BDNF to primary amygdala cultures produced decreases in gephyrin levels, whereas longer exposure resulted in an eventual increase. While total α1 subunit levels remained unchanged, gephyrin was downregulated in whole cell homogenates, but enhanced in complexes with GABAARs. Our data with anisomycin suggest that BDNF may rapidly induce gephyrin protein degradation, with subsequent gephyrin synthesis occurring. Together, these findings suggest that gephyrin may be a key factor in BDNF-dependent GABAAR regulation in the amygdala. This work may inform future studies aimed at elucidating the pathways connecting BDNF, GABAA systems, gephyrin, and their role in underlying amygdala-dependent learning.
Collapse
Affiliation(s)
- L Mou
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | | | | |
Collapse
|
47
|
Olango W, Géranton S, Roche M, Hunt S, Finn D. Novel molecular correlates of endocannabinoid-mediated fear-conditioned analgesia in rats. Eur J Pain 2013; 18:182-91. [DOI: 10.1002/j.1532-2149.2013.00369.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2013] [Indexed: 11/12/2022]
Affiliation(s)
- W.M. Olango
- Pharmacology and Therapeutics; School of Medicine; National University of Ireland Galway; Ireland
- NCBES Neuroscience Cluster; National University of Ireland Galway; Ireland
- Centre for Pain Research; National University of Ireland Galway; Ireland
| | - S.M. Géranton
- Department of Cell and Developmental Biology; University College London; UK
| | - M. Roche
- Physiology; School of Medicine; National University of Ireland Galway; Ireland
- NCBES Neuroscience Cluster; National University of Ireland Galway; Ireland
- Centre for Pain Research; National University of Ireland Galway; Ireland
| | - S.P. Hunt
- Department of Cell and Developmental Biology; University College London; UK
| | - D.P. Finn
- Pharmacology and Therapeutics; School of Medicine; National University of Ireland Galway; Ireland
- NCBES Neuroscience Cluster; National University of Ireland Galway; Ireland
- Centre for Pain Research; National University of Ireland Galway; Ireland
| |
Collapse
|
48
|
Partin AC, Hosek MP, Luong JA, Lella SK, Sharma SAR, Ploski JE. Amygdala nuclei critical for emotional learning exhibit unique gene expression patterns. Neurobiol Learn Mem 2013; 104:110-21. [PMID: 23831498 DOI: 10.1016/j.nlm.2013.06.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 06/24/2013] [Accepted: 06/25/2013] [Indexed: 01/09/2023]
Abstract
The amygdala is a heterogeneous, medial temporal lobe structure that has been implicated in the formation, expression and extinction of emotional memories. This structure is composed of numerous nuclei that vary in cytoarchitectonics and neural connections. In particular the lateral nucleus of the amygdala (LA), central nucleus of the amygdala (CeA), and the basal (B) nucleus contribute an essential role to emotional learning. However, to date it is still unclear to what extent these nuclei differ at the molecular level. Therefore we have performed whole genome gene expression analysis on these nuclei to gain a better understanding of the molecular differences and similarities among these nuclei. Specifically the LA, CeA and B nuclei were laser microdissected from the rat brain, and total RNA was isolated from these nuclei and subjected to RNA amplification. Amplified RNA was analyzed by whole genome microarray analysis which revealed that 129 genes are differentially expressed among these nuclei. Notably gene expression patterns differed between the CeA nucleus and the LA and B nuclei. However gene expression differences were not considerably different between the LA and B nuclei. Secondary confirmation of numerous genes was performed by in situ hybridization to validate the microarray findings, which also revealed that for many genes, expression differences among these nuclei were consistent with the embryological origins of these nuclei. Knowing the stable gene expression differences among these nuclei will provide novel avenues of investigation into how these nuclei contribute to emotional arousal and emotional learning, and potentially offer new genetic targets to manipulate emotional learning and memory.
Collapse
Affiliation(s)
- Alexander C Partin
- School of Behavioral and Brain Sciences,Department of Molecular & Cell Biology, University of Texas at Dallas, USA
| | | | | | | | | | | |
Collapse
|
49
|
Shvetsov AV, Zachepilo TG, Vaido AI, Kamyshev NG, Lopatina NG. On epigenetic regulation of the process of formation of long-term memory. J EVOL BIOCHEM PHYS+ 2013. [DOI: 10.1134/s0022093013020011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
Effects of continuously enhanced corticotropin releasing factor expression within the bed nucleus of the stria terminalis on conditioned and unconditioned anxiety. Mol Psychiatry 2013; 18:308-19. [PMID: 22290119 PMCID: PMC3578178 DOI: 10.1038/mp.2011.188] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The lateral division of the bed nucleus of the stria terminalis (BNST), which forms part of the circuitry regulating fear and anxiety, contains a large number of neurons expressing corticotropin releasing factor (CRF), a neuropeptide that has a prominent role in the etiology of fear- and anxiety-related psychopathologies. Stress increases CRF expression within BNST neurons, implicating these cells in stress- and anxiety-related behaviors. These experiments examined the effect of chronically enhanced CRF expression within BNST neurons on conditioned and unconditioned anxiety-related behavior by using a lentiviral vector containing a promoter that targets CRF gene overexpression (OE) to CRFergic cells. We found that BNST CRF-OE did not affect unconditioned anxiety-like responses in the elevated plus maze or basal acoustic startle amplitude. CRF-OE induced before training weakened sustained fear (conditioned anxiety); when induced after conditioning, CRF-OE increased expression of the conditioned emotional memory. Increased BNST CRF expression did not affect plasma corticosterone concentration but did decrease CRFR1 receptor density within the BNST and CRFR2 receptor density within the dorsal portion of the caudal dorsal raphe nucleus. These data raise the possibility that the observed behavioral effects may be mediated by enhanced CRF receptor signaling or compensatory changes in CRF receptor density within these structures. Together, these studies demonstrate that CRF neurons within the lateral BNST modulate conditioned anxiety-like behaviors and also suggest that enhanced CRF expression within these neurons may contribute to inappropriate regulation of emotional memories.
Collapse
|