51
|
Walters BJ, Mercaldo V, Gillon CJ, Yip M, Neve RL, Boyce FM, Frankland PW, Josselyn SA. The Role of The RNA Demethylase FTO (Fat Mass and Obesity-Associated) and mRNA Methylation in Hippocampal Memory Formation. Neuropsychopharmacology 2017; 42:1502-1510. [PMID: 28205605 PMCID: PMC5436121 DOI: 10.1038/npp.2017.31] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/13/2017] [Accepted: 02/02/2017] [Indexed: 12/17/2022]
Abstract
The formation of long-lasting memories requires coordinated changes in gene expression and protein synthesis. Although many studies implicate DNA modifications (DNA methylation, histone modifications) in memory formation, the contributions of RNA modifications remain largely unexplored. Here we investigated the role of mRNA methylation in hippocampal-dependent memory formation in mice. RNA modifications are highly dynamic and readily reversible. Methyltransferases add a methyl group to mRNA while demethylases remove methyl groups. Here we focused on examining the role of the best characterized RNA demethylase, FTO (fat mass and obesity-associated) in memory. We observed that FTO is expressed in the nuclei, dendrites and near dendritic spines of mouse dorsal hippocampal CA1 neurons. Next, we found that contextual fear conditioning transiently (0.5 h) decreased Fto levels in these neurons, with the largest decrease in FTO observed near synapses. The decrease in FTO observed shortly after contextual fear conditioning suggests that FTO normally constrains memory formation. To directly test this, we artificially decreased FTO levels in dorsal hippocampus of otherwise normal (wild-type) mice by microinjecting before training a single herpes simplex virus (HSV) vector expressing either CRISPR/Cas9 or shRNA targeted against Fto. Decreasing FTO using either method specifically enhanced contextual fear memory. Together, these results show the importance of FTO during memory formation and, furthermore, implicate mRNA modification and epi-transcriptomics as novel regulators of memory formation.
Collapse
Affiliation(s)
- Brandon J Walters
- The Hospital for Sick Children, Department of Neuroscience and Mental Health, Toronto, Ontario, Canada,University of Toronto, Department of Physiology, Toronto, Ontario, Canada,University of Toronto, Department of Psychology, Toronto, Ontario, Canada
| | - Valentina Mercaldo
- The Hospital for Sick Children, Department of Neuroscience and Mental Health, Toronto, Ontario, Canada,University of Toronto, Department of Physiology, Toronto, Ontario, Canada,University of Toronto, Department of Psychology, Toronto, Ontario, Canada
| | - Colleen J Gillon
- The Hospital for Sick Children, Department of Neuroscience and Mental Health, Toronto, Ontario, Canada,University of Toronto, Department of Physiology, Toronto, Ontario, Canada
| | - Matthew Yip
- The Hospital for Sick Children, Department of Neuroscience and Mental Health, Toronto, Ontario, Canada,University of Toronto, Department of Physiology, Toronto, Ontario, Canada,University of Toronto, Department of Psychology, Toronto, Ontario, Canada
| | - Rachael L Neve
- Massachusetts Institute of Technology, Department of Brain and Cognitive Sciences, Cambridge, MA, USA
| | - Frederick M Boyce
- Massachusetts General Hospital, Department of Neurology, Cambridge, MA, USA
| | - Paul W Frankland
- The Hospital for Sick Children, Department of Neuroscience and Mental Health, Toronto, Ontario, Canada,University of Toronto, Department of Physiology, Toronto, Ontario, Canada,University of Toronto, Department of Psychology, Toronto, Ontario, Canada
| | - Sheena A Josselyn
- The Hospital for Sick Children, Department of Neuroscience and Mental Health, Toronto, Ontario, Canada,University of Toronto, Department of Physiology, Toronto, Ontario, Canada,University of Toronto, Department of Psychology, Toronto, Ontario, Canada,Department of Psychology, Physiology, IMS, Hospital for Sick Children, Univerisity of Toronto, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada, Tel: +1 416 813 7654 ext 301824, Fax: +1 416 813 7717, E-mail:
| |
Collapse
|
52
|
Lotfipour S, Mojica C, Nakauchi S, Lipovsek M, Silverstein S, Cushman J, Tirtorahardjo J, Poulos A, Elgoyhen AB, Sumikawa K, Fanselow MS, Boulter J. α2* Nicotinic acetylcholine receptors influence hippocampus-dependent learning and memory in adolescent mice. ACTA ACUST UNITED AC 2017; 24:231-244. [PMID: 28507032 PMCID: PMC5435881 DOI: 10.1101/lm.045369.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/31/2017] [Indexed: 11/24/2022]
Abstract
The absence of α2* nicotinic acetylcholine receptors (nAChRs) in oriens lacunosum moleculare (OLM) GABAergic interneurons ablate the facilitation of nicotine-induced hippocampal CA1 long-term potentiation and impair memory. The current study delineated whether genetic mutations of α2* nAChRs (Chrna2L9′S/L9′S and Chrna2KO) influence hippocampus-dependent learning and memory and CA1 synaptic plasticity. We substituted a serine for a leucine (L9′S) in the α2 subunit (encoded by the Chrna2 gene) to make a hypersensitive nAChR. Using a dorsal hippocampus-dependent task of preexposure-dependent contextual fear conditioning, adolescent hypersensitive Chrna2L9′S/L9′S male mice exhibited impaired learning and memory. The deficit was rescued by low-dose nicotine exposure. Electrophysiological studies demonstrated that hypersensitive α2 nAChRs potentiate acetylcholine-induced ion channel flux in oocytes and acute nicotine-induced facilitation of dorsal/intermediate CA1 hippocampal long-term potentiation in Chrna2L9′S/L9′S mice. Adolescent male mice null for the α2 nAChR subunit exhibited a baseline deficit in learning that was not reversed by an acute dose of nicotine. These effects were not influenced by locomotor, sensory or anxiety-related measures. Our results demonstrated that α2* nAChRs influenced hippocampus-dependent learning and memory, as well as nicotine-facilitated CA1 hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Shahrdad Lotfipour
- Department of Psychiatry, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Celina Mojica
- Department of Psychiatry, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Sakura Nakauchi
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92612, USA
| | - Marcela Lipovsek
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N Torres, CONICET, Buenos Aires, C1428ADN, Argentina
| | - Sarah Silverstein
- Department of Psychiatry, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Jesse Cushman
- Department of Psychology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | | | - Andrew Poulos
- Department of Psychology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N Torres, CONICET, Buenos Aires, C1428ADN, Argentina
| | - Katumi Sumikawa
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92612, USA
| | - Michael S Fanselow
- Department of Psychiatry, University of California, Los Angeles, Los Angeles, California 90095, USA.,Department of Psychology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Jim Boulter
- Department of Psychiatry, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
53
|
Salles A, Krawczyk MDC, Blake M, Romano A, Boccia MM, Freudenthal R. Requirement of NF-kappa B Activation in Different Mice Brain Areas during Long-Term Memory Consolidation in Two Contextual One-Trial Tasks with Opposing Valences. Front Mol Neurosci 2017; 10:104. [PMID: 28439227 PMCID: PMC5383659 DOI: 10.3389/fnmol.2017.00104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/24/2017] [Indexed: 11/13/2022] Open
Abstract
NF-kappa B is a transcription factor whose activation has been shown to be necessary for long-term memory consolidation in several species. NF-kappa B is activated and translocates to the nucleus of cells in a specific temporal window during consolidation. Our work focuses on a one trial learning tasks associated to the inhibitory avoidance (IA) setting. Mice were trained either receiving or not a footshock when entering a dark compartment (aversive vs. appetitive learning). Regardless of training condition (appetitive or aversive), latencies to step-through during testing were significantly different to those measured during training. Additionally, these testing latencies were also different from those of a control group that only received a shock unrelated to context. Moreover, nuclear NF-kappa B DNA-binding activity was augmented in the aversive and the appetitive tasks when compared with control and naïve animals. NF-kappa B inhibition by Sulfasalazine injected either in the Hippocampus, Amygdala or Nucleus accumbens immediately after training was able to impair retention in both training versions. Our results suggest that NF-kappa B is a critical molecular step, in different brain areas on memory consolidation. This was the case for both the IA task and also the modified version of the same task where the footshock was omitted during training. This work aims to further investigate how appetitive and aversive memories are consolidated.
Collapse
Affiliation(s)
- Angeles Salles
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos AiresBuenos Aires, Argentina
| | - Maria Del C Krawczyk
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Fac. Farmacia y Bioquímica, Universidad de Buenos Aires/CONICETBuenos Aires, Argentina
| | - Mariano Blake
- Departamento de Fisiología, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO), Facultad de Medicina, Universidad de Buenos Aires, CONICETBuenos Aires, Argentina
| | - Arturo Romano
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos AiresBuenos Aires, Argentina
| | - Mariano M Boccia
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Fac. Farmacia y Bioquímica, Universidad de Buenos Aires/CONICETBuenos Aires, Argentina
| | - Ramiro Freudenthal
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos AiresBuenos Aires, Argentina
| |
Collapse
|
54
|
Leake J, Zinn R, Corbit L, Vissel B. Dissociation between complete hippocampal context memory formation and context fear acquisition. ACTA ACUST UNITED AC 2017; 24:153-157. [PMID: 28298553 PMCID: PMC5362699 DOI: 10.1101/lm.044578.116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/20/2017] [Indexed: 11/24/2022]
Abstract
Rodents require a minimal time period to explore a context prior to footshock to display plateau-level context fear at test. To investigate whether this rapid fear plateau reflects complete memory formation within that short time-frame, we used the immediate-early gene product Arc as an indicator of hippocampal context memory formation-related activity. We found that hippocampal Arc expression continued to increase well past the minimal time required for plateau-level fear. This raises the possibility that context fear conditioning occurs more rapidly than complete memory formation. Thus, animals may be able to condition robustly to both complete and incomplete contextual representations.
Collapse
Affiliation(s)
- Jessica Leake
- Neurodegenerative Disorders, Neuroscience Department, Garvan Institute of Medical Research, Sydney 2010, Australia.,School of Psychology, The University of Sydney, Sydney 2006, Australia
| | - Raphael Zinn
- Neurodegenerative Disorders, Neuroscience Department, Garvan Institute of Medical Research, Sydney 2010, Australia.,Faculty of Medicine, University of New South Wales, Sydney 2052, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney 2007, Australia
| | - Laura Corbit
- School of Psychology, The University of Sydney, Sydney 2006, Australia
| | - Bryce Vissel
- Neurodegenerative Disorders, Neuroscience Department, Garvan Institute of Medical Research, Sydney 2010, Australia.,Faculty of Medicine, University of New South Wales, Sydney 2052, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney 2007, Australia
| |
Collapse
|
55
|
Keiser AA, Turnbull LM, Darian MA, Feldman DE, Song I, Tronson NC. Sex Differences in Context Fear Generalization and Recruitment of Hippocampus and Amygdala during Retrieval. Neuropsychopharmacology 2017; 42:397-407. [PMID: 27577601 PMCID: PMC5399239 DOI: 10.1038/npp.2016.174] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/21/2016] [Accepted: 08/23/2016] [Indexed: 01/04/2023]
Abstract
Anxiety disorders are commonly associated with increased generalization of fear from a stress- or trauma-associated environment to a neutral context or environment. Differences in context-associated memory in males and females may contribute to increased susceptibility to anxiety disorders in women. Here we examined sex differences in context fear generalization and its neural correlates. We observed stronger context fear conditioning and more generalization of fear to a similar context in females than males. In addition, context preexposure increased fear conditioning in males and decreased generalization in females. Accordingly, males showed stronger cFos activity in dorsal hippocampus during memory retrieval and context generalization, whereas females showed preferential recruitment of basal amygdala. Together, these findings are consistent with previous research showing that hippocampal activity correlates with reduced context fear generalization. Differential competition between hippocampus and amygdala-dependent processes may thus contribute to sex differences in retrieval of context fear and greater generalization of fear-associated memory.
Collapse
Affiliation(s)
- Ashley A Keiser
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Lacie M Turnbull
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Mara A Darian
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Dana E Feldman
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Iris Song
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Natalie C Tronson
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA,Department of Psychology, University of Michigan, 530 Church Street, Ann Arbor, MI 48109, USA, Tel: +1 734 936 1495, E-mail:
| |
Collapse
|
56
|
Li SH, Graham BM. Why are women so vulnerable to anxiety, trauma-related and stress-related disorders? The potential role of sex hormones. Lancet Psychiatry 2017; 4:73-82. [PMID: 27856395 DOI: 10.1016/s2215-0366(16)30358-3] [Citation(s) in RCA: 342] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 02/08/2023]
Abstract
Increased prevalence, severity, and burden of anxiety, trauma-related and stress-related disorders in women compared with men has been well documented. Evidence from a variety of fields has emerged suggesting that sex hormones, particularly oestradiol and progesterone, play a significant part in generation of these sex differences. In this Series paper, we aim to integrate the literature reporting on the effects of sex hormones on biological, behavioural, and cognitive pathways, to propose two broad mechanisms by which oestradiol and progesterone influence sex differences in anxiety disorders: augmentation of vulnerability factors associated with anxiety disorder development; and facilitation of the maintenance of anxious symptoms post-development. The implications for future research, along with novel approaches to psychological and pharmacological treatment of anxiety disorders, are also considered.
Collapse
Affiliation(s)
- Sophie H Li
- School of Psychology, The University of New South Wales, Sydney, NSW, Australia
| | - Bronwyn M Graham
- School of Psychology, The University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
57
|
Huckleberry KA, Ferguson LB, Drew MR. Behavioral mechanisms of context fear generalization in mice. ACTA ACUST UNITED AC 2016; 23:703-709. [PMID: 27918275 PMCID: PMC5110986 DOI: 10.1101/lm.042374.116] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/20/2016] [Indexed: 11/25/2022]
Abstract
There is growing interest in generalization of learned contextual fear, driven in part by the hypothesis that mood and anxiety disorders stem from impaired hippocampal mechanisms of fear generalization and discrimination. However, there has been relatively little investigation of the behavioral and procedural mechanisms that might control generalization of contextual fear. We assessed the relative contribution of different contextual features to context fear generalization and characterized how two common conditioning protocols—foreground (uncued) and background (cued) contextual fear conditioning—affected context fear generalization. In one experiment, mice were fear conditioned in context A, and then tested for contextual fear both in A and in an alternate context created by changing a subset of A's elements. The results suggest that floor configuration and odor are more salient features than chamber shape. A second experiment compared context fear generalization in background and foreground context conditioning. Although foreground conditioning produced more context fear than background conditioning, the two procedures produced equal amounts of generalized fear. Finally, results indicated that the order of context tests (original first versus alternate first) significantly modulates context fear generalization, perhaps because the original and alternate contexts are differentially sensitive to extinction. Overall, results demonstrate that context fear generalization is sensitive to procedural variations and likely reflects the operation of multiple interacting psychological and neural mechanisms.
Collapse
Affiliation(s)
- Kylie A Huckleberry
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712, USA
| | - Laura B Ferguson
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712, USA
| | - Michael R Drew
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712, USA
| |
Collapse
|
58
|
Rashid AJ, Yan C, Mercaldo V, Hsiang HLL, Park S, Cole CJ, De Cristofaro A, Yu J, Ramakrishnan C, Lee SY, Deisseroth K, Frankland PW, Josselyn SA. Competition between engrams influences fear memory formation and recall. Science 2016; 353:383-7. [PMID: 27463673 DOI: 10.1126/science.aaf0594] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 06/17/2016] [Indexed: 12/20/2022]
Abstract
Collections of cells called engrams are thought to represent memories. Although there has been progress in identifying and manipulating single engrams, little is known about how multiple engrams interact to influence memory. In lateral amygdala (LA), neurons with increased excitability during training outcompete their neighbors for allocation to an engram. We examined whether competition based on neuronal excitability also governs the interaction between engrams. Mice received two distinct fear conditioning events separated by different intervals. LA neuron excitability was optogenetically manipulated and revealed a transient competitive process that integrates memories for events occurring closely in time (coallocating overlapping populations of neurons to both engrams) and separates memories for events occurring at distal times (disallocating nonoverlapping populations to each engram).
Collapse
Affiliation(s)
- Asim J Rashid
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada. Department of Psychology, University of Toronto, Toronto, ON M5G 3G3, Canada. Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada. Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Chen Yan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada. Department of Psychology, University of Toronto, Toronto, ON M5G 3G3, Canada. Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada. Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Valentina Mercaldo
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada. Department of Psychology, University of Toronto, Toronto, ON M5G 3G3, Canada. Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada. Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Hwa-Lin Liz Hsiang
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada. Department of Psychology, University of Toronto, Toronto, ON M5G 3G3, Canada. Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada. Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Sungmo Park
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada. Department of Psychology, University of Toronto, Toronto, ON M5G 3G3, Canada. Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada. Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Christina J Cole
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada. Department of Psychology, University of Toronto, Toronto, ON M5G 3G3, Canada. Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada. Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Antonietta De Cristofaro
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Julia Yu
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Charu Ramakrishnan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Soo Yeun Lee
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Paul W Frankland
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada. Department of Psychology, University of Toronto, Toronto, ON M5G 3G3, Canada. Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada. Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada.
| | - Sheena A Josselyn
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada. Department of Psychology, University of Toronto, Toronto, ON M5G 3G3, Canada. Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada. Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
59
|
Schmitz SK, King C, Kortleven C, Huson V, Kroon T, Kevenaar JT, Schut D, Saarloos I, Hoetjes JP, de Wit H, Stiedl O, Spijker S, Li KW, Mansvelder HD, Smit AB, Cornelisse LN, Verhage M, Toonen RF. Presynaptic inhibition upon CB1 or mGlu2/3 receptor activation requires ERK/MAPK phosphorylation of Munc18-1. EMBO J 2016; 35:1236-50. [PMID: 27056679 DOI: 10.15252/embj.201592244] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 03/02/2016] [Indexed: 01/22/2023] Open
Abstract
Presynaptic cannabinoid (CB1R) and metabotropic glutamate receptors (mGluR2/3) regulate synaptic strength by inhibiting secretion. Here, we reveal a presynaptic inhibitory pathway activated by extracellular signal-regulated kinase (ERK) that mediates CB1R- and mGluR2/3-induced secretion inhibition. This pathway is triggered by a variety of events, from foot shock-induced stress to intense neuronal activity, and induces phosphorylation of the presynaptic protein Munc18-1. Mimicking constitutive phosphorylation of Munc18-1 results in a drastic decrease in synaptic transmission. ERK-mediated phosphorylation of Munc18-1 ultimately leads to degradation by the ubiquitin-proteasome system. Conversely, preventing ERK-dependent Munc18-1 phosphorylation increases synaptic strength. CB1R- and mGluR2/3-induced synaptic inhibition and depolarization-induced suppression of excitation (DSE) are reduced upon ERK/MEK pathway inhibition and further reduced when ERK-dependent Munc18-1 phosphorylation is blocked. Thus, ERK-dependent Munc18-1 phosphorylation provides a major negative feedback loop to control synaptic strength upon activation of presynaptic receptors and during intense neuronal activity.
Collapse
Affiliation(s)
- Sabine K Schmitz
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Cillian King
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Christian Kortleven
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Vincent Huson
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Tim Kroon
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Josta T Kevenaar
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Desiree Schut
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Ingrid Saarloos
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Joost P Hoetjes
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Heidi de Wit
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Oliver Stiedl
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands Department of Molecular & Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Sabine Spijker
- Department of Molecular & Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Ka Wan Li
- Department of Molecular & Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular & Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Lennart Niels Cornelisse
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
60
|
Nagaya N, Acca GM, Maren S. Allopregnanolone in the bed nucleus of the stria terminalis modulates contextual fear in rats. Front Behav Neurosci 2015; 9:205. [PMID: 26300750 PMCID: PMC4523814 DOI: 10.3389/fnbeh.2015.00205] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 07/20/2015] [Indexed: 12/20/2022] Open
Abstract
Trauma- and stress-related disorders are among the most common types of mental illness affecting the U.S. population. For many of these disorders, there is a striking sex difference in lifetime prevalence; for instance, women are twice as likely as men to be affected by posttraumatic stress disorder (PTSD). Gonadal steroids and their metabolites have been implicated in sex differences in fear and anxiety. One example, allopregnanolone (ALLO), is a neuroactive metabolite of progesterone that allosterically enhances GABAA receptor activity and has anxiolytic effects. Like other ovarian hormones, it not only occurs at different levels in males and females but also fluctuates over the female reproductive cycle. One brain structure that may be involved in neuroactive steroid regulation of fear and anxiety is the bed nucleus of the stria terminalis (BNST). To explore this question, we examined the consequences of augmenting or reducing ALLO activity in the BNST on the expression of Pavlovian fear conditioning in rats. In Experiment 1, intra-BNST infusions of ALLO in male rats suppressed freezing behavior (a fear response) to the conditioned context, but did not influence freezing to a discrete tone conditioned stimulus (CS). In Experiment 2, intra-BNST infusion of either finasteride (FIN), an inhibitor of ALLO synthesis, or 17-phenyl-(3α,5α)-androst-16-en-3-ol, an ALLO antagonist, in female rats enhanced contextual freezing; neither treatment affected freezing to the tone CS. These findings support a role for ALLO in modulating contextual fear via the BNST and suggest that sex differences in fear and anxiety could arise from differential steroid regulation of BNST function. The susceptibility of women to disorders such as PTSD may be linked to cyclic declines in neuroactive steroid activity within fear circuitry.
Collapse
Affiliation(s)
- Naomi Nagaya
- Texas A&M University College Station, TX, USA ; Institute for Neuroscience, Texas A&M University College Station, TX, USA
| | - Gillian M Acca
- Institute for Neuroscience, Texas A&M University College Station, TX, USA
| | - Stephen Maren
- Texas A&M University College Station, TX, USA ; Institute for Neuroscience, Texas A&M University College Station, TX, USA
| |
Collapse
|
61
|
Matsuda S, Matsuzawa D, Ishii D, Tomizawa H, Sutoh C, Shimizu E. Sex differences in fear extinction and involvements of extracellular signal-regulated kinase (ERK). Neurobiol Learn Mem 2015; 123:117-24. [PMID: 26079214 DOI: 10.1016/j.nlm.2015.05.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 05/02/2015] [Accepted: 05/26/2015] [Indexed: 12/13/2022]
Abstract
Stress-related disorders, such as post-traumatic stress disorder (PTSD) and panic disorders, are disproportionately prevalent in females. However, the biological mechanism underlying these sex differences in the prevalence rate remains unclear. In the present study, we examined sex differences in fear memory, fear extinction, and spontaneous recovery of fear. We investigated the presence of sex differences in recent and remote fear memory in mice using contextual fear conditioning, as well as sex differences in spontaneous recovery of fear memory using a consecutive fear extinction paradigm. We examined the number of fear extinction days required to prevent spontaneous recovery of fear in either sex. We investigated whether ovariectomy affected fear extinction and spontaneous recovery. We also measured the activation of extracellular signal-regulated kinase (ERK) 1 and 2 in the dorsal hippocampus and the medial prefrontal cortex following fear extinction sessions. In our results, we found no sex difference in recent or remote fear memory. However, females required more fear extinction sessions compared to males to prevent spontaneous recovery. Within-extinction freezing also differed between males and females. Moreover, females required more extinction sessions than males to increase ERK2 phosphorylation in the dorsal hippocampus. Our data suggest that contextual fear extinction was unstable in females compared to males and that such sex differences may be related to the ERK2 phosphorylation in the hippocampus.
Collapse
Affiliation(s)
- Shingo Matsuda
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8502, Japan.
| | - Daisuke Matsuzawa
- Department of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba 260-8670, Japan; Research Center for Child Mental Development, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba 260-8670, Japan
| | - Daisuke Ishii
- Department of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba 260-8670, Japan
| | - Haruna Tomizawa
- Department of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba 260-8670, Japan
| | - Chihiro Sutoh
- Department of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba 260-8670, Japan
| | - Eiji Shimizu
- Department of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba 260-8670, Japan; Research Center for Child Mental Development, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba 260-8670, Japan
| |
Collapse
|
62
|
Murthy SRK, Sherrin T, Jansen C, Nijholt I, Robles M, Dolga AM, Andreotti N, Sabatier JM, Knaus HG, Penner R, Todorovic C, Blank T. Small-conductance Ca2+-activated potassium type 2 channels regulate the formation of contextual fear memory. PLoS One 2015; 10:e0127264. [PMID: 25938421 PMCID: PMC4418695 DOI: 10.1371/journal.pone.0127264] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 04/13/2015] [Indexed: 12/28/2022] Open
Abstract
Small-conductance, Ca2+ activated K+ channels (SK channels) are expressed at high levels in brain regions responsible for learning and memory. In the current study we characterized the contribution of SK2 channels to synaptic plasticity and to different phases of hippocampal memory formation. Selective SK2 antisense-treatment facilitated basal synaptic transmission and theta-burst induced LTP in hippocampal brain slices. Using the selective SK2 antagonist Lei-Dab7 or SK2 antisense probes, we found that hippocampal SK2 channels are critical during two different time windows: 1) blockade of SK2 channels before the training impaired fear memory, whereas, 2) blockade of SK2 channels immediately after the training enhanced contextual fear memory. We provided the evidence that the post-training cleavage of the SK2 channels was responsible for the observed bidirectional effect of SK2 channel blockade on memory consolidation. Thus, Lei-Dab7-injection before training impaired the C-terminal cleavage of SK2 channels, while Lei-Dab7 given immediately after training facilitated the C-terminal cleavage. Application of the synthetic peptide comprising a leucine-zipper domain of the C-terminal fragment to Jurkat cells impaired SK2 channel-mediated currents, indicating that the endogenously cleaved fragment might exert its effects on memory formation by blocking SK2 channel-mediated currents. Our present findings suggest that SK2 channel proteins contribute to synaptic plasticity and memory not only as ion channels but also by additionally generating a SK2 C-terminal fragment, involved in both processes. The modulation of fear memory by down-regulating SK2 C-terminal cleavage might have applicability in the treatment of anxiety disorders in which fear conditioning is enhanced.
Collapse
Affiliation(s)
- Saravana R. K. Murthy
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Tessi Sherrin
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Chad Jansen
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Laboratory of Cell and Molecular Signaling, The Queen’s Medical Center, Honolulu, Hawaii, United States of America
| | | | - Michael Robles
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Amalia M. Dolga
- Department of Pharmacology and Toxicology, Philipps-University Marburg, Marburg, Germany
| | - Nicolas Andreotti
- Laboratoire INSERM UMR1097, Parc scientifique et technologique de Luminy, Marseille, cedex 09, France
| | - Jean-Marc Sabatier
- Laboratoire INSERM UMR1097, Parc scientifique et technologique de Luminy, Marseille, cedex 09, France
| | - Hans-Guenther Knaus
- Division for Molecular and Cellular Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Reinhold Penner
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Laboratory of Cell and Molecular Signaling, The Queen’s Medical Center, Honolulu, Hawaii, United States of America
| | - Cedomir Todorovic
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- * E-mail:
| | - Thomas Blank
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Institute for Neuropathology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
63
|
Bernier BE, Lacagnina AF, Drew MR. Potent attenuation of context fear by extinction training contiguous with acquisition. ACTA ACUST UNITED AC 2014; 22:31-8. [PMID: 25512575 PMCID: PMC4274325 DOI: 10.1101/lm.036673.114] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Studies on the behavioral mechanisms underlying contextual fear conditioning (CFC) have demonstrated the importance of preshock context exposure in the formation of aversive context memories. However, there has been comparatively little investigation of the effects of context exposure immediately after the shock. Some models predict that nonreinforced context exposure at the end of the acquisition session will strongly influence the strength of conditioning and/or recruit distinct neural mechanisms relative to extinction after acquisition. Here we investigate the effects of manipulating postshock context exposure on CFC in mice. Prolonging the period of context exposure immediately following the shock caused a significant and durable reduction in conditioned fear. This immediate postshock context exposure was more effective at attenuating conditioned fear than was an equivalent amount of context exposure a day or more after acquisition. The results suggest that nonreinforced exposure to the context influences conditioned fear through distinct mechanisms depending on whether it occurs during acquisition or after it. The superiority of immediate postshock context exposure was specific to single-shock CFC; in two-shock CFC, immediate and delayed postshock context exposure had similar effects. Consistent with previous reports, we hypothesize that the effectiveness of extinction is modulated by emotional state, and procedures engendering higher postshock freezing (such as two-shock CFC) are associated with weaker immediate extinction.
Collapse
Affiliation(s)
- Brian E Bernier
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Anthony F Lacagnina
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Michael R Drew
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas, 78712, USA
| |
Collapse
|
64
|
Chen LS, Tzeng WY, Chuang JY, Cherng CG, Gean PW, Yu L. Roles of testosterone and amygdaloid LTP induction in determining sex differences in fear memory magnitude. Horm Behav 2014; 66:498-508. [PMID: 25066484 DOI: 10.1016/j.yhbeh.2014.07.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 07/17/2014] [Accepted: 07/18/2014] [Indexed: 01/30/2023]
Abstract
Women are thought to form fear memory more robust than men do and testosterone is suspected to play a role in determining such a sex difference. Mouse cued fear freezing was used to study the sex-related susceptibility and the role of testosterone in fear memory in humans. A 75-dB tone was found to provoke weak freezing, while 0.15-mA and 0.20-mA footshock caused strong freezing responses. No sex differences were noticed in the tone- or footshock-induced (naïve fear) freezing. Following the conditionings, female mice exhibited greater tone (cued fear)-induced freezing than did male mice. Nonetheless, female mice demonstrated indistinctive cued fear freezing across the estrous phases and ovariectomy did not affect such freezing in female mice. Orchidectomy enhanced the cued fear freezing in male mice. Systemic testosterone administrations and an intra-lateral nucleus of amygdala (LA) testosterone infusion diminished the cued fear freezing in orchidectomized male mice, while pretreatment with flutamide (Flu) eradicated these effects. Long-term potentiation (LTP) magnitude in LA has been known to correlate with the strength of the cued fear conditioning. We found that LA LTP magnitude was indeed greater in female than male mice. Orchidectomy enhanced LTP magnitude in males' LA, while ovariectomy decreased LTP magnitude in females' LA. Testosterone decreased LTP magnitude in orchidectomized males' LA and estradiol enhanced LTP magnitude in ovariectomized females' LA. Finally, male mice had lower LA GluR1 expression than female mice and orchidectomy enhanced the GluR1 expression in male mice. These findings, taken together, suggest that testosterone plays a critical role in rendering the sex differences in the cued fear freezing and LA LTP. Testosterone is negatively associated with LA LTP and the cued fear memory in male mice. However, ovarian hormones and LA LTP are loosely associated with the cued fear memory in female mice.
Collapse
Affiliation(s)
- Li-Shen Chen
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan 70101, Taiwan, ROC
| | - Wen-Yu Tzeng
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan 70101, Taiwan, ROC
| | - Jia-Ying Chuang
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan 70101, Taiwan, ROC
| | - Chianfang G Cherng
- Department of Health Psychology, Chang Jung Christian University, Tainan 71101, Taiwan, ROC
| | - Po-Wu Gean
- Department of Pharmacology, National Cheng Kung University College of Medicine, Tainan 70101, Taiwan, ROC
| | - Lung Yu
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan 70101, Taiwan, ROC; Institute of Behavioral Medicine, National Cheng Kung University College of Medicine, Tainan 70101, Taiwan, ROC.
| |
Collapse
|
65
|
Fenton GE, Pollard AK, Halliday DM, Mason R, Bredy TW, Stevenson CW. Persistent prelimbic cortex activity contributes to enhanced learned fear expression in females. Learn Mem 2014; 21:55-60. [PMID: 24429423 PMCID: PMC3895223 DOI: 10.1101/lm.033514.113] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Anxiety disorders, such as post-traumatic stress, are more prevalent in women and are characterized by impaired inhibition of learned fear and medial prefrontal cortex (mPFC) dysfunction. Here we examined sex differences in fear extinction and mPFC activity in rats. Females showed more learned fear expression during extinction and its recall, but not fear conditioning. They also showed more spontaneous fear recovery and more contextual fear before extinction and its recall. Moreover, enhanced learned fear expression in females was associated with sustained prelimbic (PL) cortex activity. These results suggest that sex differences in learned fear expression may involve persistent PL activation.
Collapse
Affiliation(s)
- Georgina E Fenton
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, United Kingdom
| | | | | | | | | | | |
Collapse
|
66
|
Rapid innate defensive responses of mice to looming visual stimuli. Curr Biol 2013; 23:2011-5. [PMID: 24120636 DOI: 10.1016/j.cub.2013.08.015] [Citation(s) in RCA: 385] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 08/07/2013] [Accepted: 08/07/2013] [Indexed: 11/21/2022]
Abstract
Much of brain science is concerned with understanding the neural circuits that underlie specific behaviors. While the mouse has become a favorite experimental subject, the behaviors of this species are still poorly explored. For example, the mouse retina, like that of other mammals, contains ∼20 different circuits that compute distinct features of the visual scene [1, 2]. By comparison, only a handful of innate visual behaviors are known in this species--the pupil reflex [3], phototaxis [4], the optomotor response [5], and the cliff response [6]--two of which are simple reflexes that require little visual processing. We explored the behavior of mice under a visual display that simulates an approaching object, which causes defensive reactions in some other species [7, 8]. We show that mice respond to this stimulus either by initiating escape within a second or by freezing for an extended period. The probability of these defensive behaviors is strongly dependent on the parameters of the visual stimulus. Directed experiments identify candidate retinal circuits underlying the behavior and lead the way into detailed study of these neural pathways. This response is a new addition to the repertoire of innate defensive behaviors in the mouse that allows the detection and avoidance of aerial predators.
Collapse
|
67
|
Germain J, Bruel-Jungerman E, Grannec G, Denis C, Lepousez G, Giros B, Francis F, Nosten-Bertrand M. Doublecortin knockout mice show normal hippocampal-dependent memory despite CA3 lamination defects. PLoS One 2013; 8:e74992. [PMID: 24073232 PMCID: PMC3779246 DOI: 10.1371/journal.pone.0074992] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 08/12/2013] [Indexed: 11/23/2022] Open
Abstract
Mutations in the human X-linked doublecortin gene (DCX) cause major neocortical disorganization associated with severe intellectual disability and intractable epilepsy. Although Dcx knockout (KO) mice exhibit normal isocortical development and architecture, they show lamination defects of the hippocampal pyramidal cell layer largely restricted to the CA3 region. Dcx-KO mice also exhibit interneuron abnormalities. As well as the interest of testing their general neurocognitive profile, Dcx-KO mice also provide a relatively unique model to assess the effects of a disorganized CA3 region on learning and memory. Based on its prominent anatomical and physiological features, the CA3 region is believed to contribute to rapid encoding of novel information, formation and storage of arbitrary associations, novelty detection, and short-term memory. We report here that Dcx-KO adult males exhibit remarkably preserved hippocampal- and CA3-dependant cognitive processes using a large battery of classical hippocampus related tests such as the Barnes maze, contextual fear conditioning, paired associate learning and object recognition. In addition, we show that hippocampal adult neurogenesis, in terms of proliferation, survival and differentiation of granule cells, is also remarkably preserved in Dcx-KO mice. In contrast, following social deprivation, Dcx-KO mice exhibit impaired social interaction and reduced aggressive behaviors. In addition, Dcx-KO mice show reduced behavioral lateralization. The Dcx-KO model thus reinforces the association of neuropsychiatric behavioral impairments with mouse models of intellectual disability.
Collapse
Affiliation(s)
- Johanne Germain
- INSERM UMRS 952, Paris, France
- CNRS UMR 7224, Paris, France
- UPMC, Paris, France
- Université Paris Descartes, Paris, France
| | - Elodie Bruel-Jungerman
- UPMC, Paris, France
- INSERM UMR-S 839, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Gael Grannec
- INSERM UMRS 952, Paris, France
- CNRS UMR 7224, Paris, France
- UPMC, Paris, France
| | - Cécile Denis
- INSERM UMRS 952, Paris, France
- CNRS UMR 7224, Paris, France
- UPMC, Paris, France
| | | | - Bruno Giros
- INSERM UMRS 952, Paris, France
- CNRS UMR 7224, Paris, France
- UPMC, Paris, France
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, Montreal, Canada
| | - Fiona Francis
- UPMC, Paris, France
- INSERM UMR-S 839, Paris, France
- Institut du Fer à Moulin, Paris, France
| | | |
Collapse
|
68
|
Sun N, Qu C, Zhao S, Yu L, Zheng X. Allocation of attention in response to novel neutral stimuli and predictive negative stimuli in men and women: an event-related potentials research study. BIOL RHYTHM RES 2012. [DOI: 10.1080/09291016.2011.605626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
69
|
Ter Horst JP, Carobrez AP, van der Mark MH, de Kloet ER, Oitzl MS. Sex differences in fear memory and extinction of mice with forebrain-specific disruption of the mineralocorticoid receptor. Eur J Neurosci 2012; 36:3096-102. [PMID: 22831399 DOI: 10.1111/j.1460-9568.2012.08237.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Previous studies showed that the mineralocorticoid receptor (MR) is needed for behavioral flexibility in a fear conditioning paradigm. Female mice with forebrain-specific deletion of the MR gene (MR(CaMKCre) ) were unable to show extinction of contextual fear, and could not discriminate between cue and context fear unlike control mice. In the present study, male and female (MR(CaMKCre) ) mice and control littermates were used to study sex-specific fear conditioning, memory performance and extinction. The fear conditioning paradigm assessed both context- and cue-related fear within one experimental procedure. We observed that at the end of the conditioning all mice acquired the fear-motivated response. During the first minutes of the memory test, both male and female MR(CaMKCre) mice remembered and feared the context more than the control mice. Furthermore, female MR(CaMKCre) mice were not able to extinguish this memory even on the second day of memory testing. The female mutants also could not discriminate between cue (more freezing) and context periods (less freezing). In contrast, male MR(CaMKCre) mice and the controls showed extinction and were capable to discriminate, although the MR(CaMKCre) mice needed more time before they started extinction. These findings further support the relevance of MR for behavioral flexibility and extinction of fear-motivated behavior. In conclusion, the loss of MR in the forebrain results in large differences in emotional and cognitive behaviors between female and male mice, which suggests a role of this receptor in the female prevalence of stress- and anxiety-regulated disorders.
Collapse
Affiliation(s)
- J P Ter Horst
- Division of Medical Pharmacology, Leiden Amsterdam Center for Drug Research and Leiden University Medical Center, Leiden University, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
70
|
Pevzner A, Miyashita T, Schiffman AJ, Guzowski JF. Temporal dynamics of Arc gene induction in hippocampus: relationship to context memory formation. Neurobiol Learn Mem 2012; 97:313-20. [PMID: 22390855 DOI: 10.1016/j.nlm.2012.02.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 02/14/2012] [Accepted: 02/16/2012] [Indexed: 11/26/2022]
Abstract
Past studies have proposed a role for the hippocampus in the rapid encoding of context memories. Despite this, there is little data regarding the molecular processes underlying the stable formation of a context representation that occurs in the time window established through such behavioral studies. One task that is useful for investigating the rapid encoding of context is contextual fear conditioning (CFC). Behavioral studies demonstrate that animals require approximately 30 s of exploration prior to a footshock to form a contextual representation supporting CFC. Thus, any potential molecular process required for the stabilization of the cellular representation for context must be activated within this narrow and behaviorally defined time window. Detection of the immediate-early gene Arc presents an ideal method to assess the activation of specific neuronal ensembles, given past studies showing the context specific expression of Arc in CA3 and CA1 subfields and the role of Arc in hippocampal long-term synaptic plasticity. Therefore, we examined the temporal dynamics of Arc induction within the hippocampus after brief context exposure to determine whether experience-dependent Arc expression could be involved in the rapid encoding of incidental context memories. We found that the duration of context exposure differentially activated Arc expression in hippocampal subfields, with CA3 showing rapid engagement within as little as 3 s of exposure. By contrast, Arc induction in CA1 required 30 s of context exposure to reach maximal levels. A parallel behavioral experiment revealed that 30 s, but not 3 s, exposure to a context resulted in strong conditioned freezing 24 h later, consistent with past studies from other laboratories. The current study is the first to examine the rapid temporal dynamics of Arc induction in hippocampus in a well-defined context memory paradigm. These studies demonstrate within 30 s of context exposure Arc is fully activated in CA3 and CA1, suggesting that the engagement of plastic processes requiring Arc function (such as long-term potentiation) occurs within the same temporal domain as that required for behavioral conditioning.
Collapse
Affiliation(s)
- Aleksandr Pevzner
- Center for the Neurobiology of Learning and Memory, Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-3800, USA
| | | | | | | |
Collapse
|
71
|
Lebron-Milad K, Milad MR. Sex differences, gonadal hormones and the fear extinction network: implications for anxiety disorders. BIOLOGY OF MOOD & ANXIETY DISORDERS 2012; 2:3. [PMID: 22738383 PMCID: PMC3384233 DOI: 10.1186/2045-5380-2-3] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 02/07/2012] [Indexed: 01/11/2023]
Abstract
Convergent data from rodents and human studies have led to the development of models describing the neural mechanisms of fear extinction. Key components of the now well-characterized fear extinction network include the amygdala, hippocampus, and medial prefrontal cortical regions. These models are fueling novel hypotheses that are currently being tested with much refined experimental tools to examine the interactions within this network. Lagging far behind, however, is the examination of sex differences in this network and how sex hormones influence the functional activity and reactivity of these brain regions in the context of fear inhibition. Indeed, there is a large body of literature suggesting that sex hormones, such as estrogen, do modulate neural plasticity within the fear extinction network, especially in the hippocampus.After a brief overview of the fear extinction network, we summarize what is currently known about sex differences in fear extinction and the influence of gonadal hormones on the fear extinction network. We then go on to propose possible mechanisms by which sex hormones, such as estrogen, may influence neural plasticity within the fear extinction network. We end with a discussion of how knowledge to be gained from developing this line of research may have significant ramifications towards the etiology, epidemiology and treatment of anxiety disorders.
Collapse
Affiliation(s)
- Kelimer Lebron-Milad
- Department of Psychiatry, Harvard Medical School & Massachusetts General Hospital, Boston, MA, USA.
| | | |
Collapse
|
72
|
Wiltgen BJ, Wood AN, Levy B. The cellular mechanisms of memory are modified by experience. Learn Mem 2011; 18:747-50. [DOI: 10.1101/lm.024026.111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
73
|
Brown KL, Kennard JA, Sherer DJ, Comalli DM, Woodruff-Pak DS. The context preexposure facilitation effect in mice: a dose-response analysis of pretraining scopolamine administration. Behav Brain Res 2011; 225:290-6. [PMID: 21827794 DOI: 10.1016/j.bbr.2011.07.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 07/12/2011] [Accepted: 07/22/2011] [Indexed: 11/29/2022]
Abstract
The context preexposure facilitation effect (CPFE) is an elaboration of contextual fear conditioning and refers to enhanced contextual conditioning resulting from preexposure to the context prior to a separate, brief context-shock episode. A version of the CPFE developed by Rudy and colleagues in rats has demonstrated greater sensitivity to pre-training hippocampal insult relative to standard contextual fear conditioning preparations. Our aim was to adapt the Rudy CPFE procedures to mice. In Experiment 1 we compared performance of young adult male C57BL6/J mice on two versions of the CPFE. One version - not previously used in mice - adapted methods established by Rudy and colleagues, and the other CPFE task replicated procedures previously established in this mouse strain by Gould and colleagues. In Experiment 2 we compared the effects of pre-training intraperitoneal administration of moderate levels of scopolamine or methylscopolamine on contextual conditioning between mice trained using the Rudy CPFE method and a separate group trained using standard contextual fear procedures. Scopolamine is a muscarinic cholinergic receptor antagonist that impairs hippocampal function. Robust freezing to the conditioning context was observed in mice trained using the Rudy CPFE method (Experiment 1), and greater scopolamine-induced impairments in contextual freezing were observed using this CPFE method relative to mice trained using standard contextual fear procedures (Experiment 2). These findings support use of the Rudy CPFE task as a behavioral assay for hippocampal function in mice.
Collapse
Affiliation(s)
- Kevin L Brown
- Department of Psychology, Temple University, 1701 North 13th Street/Weiss Hall, Philadelphia, PA 19122, United States
| | | | | | | | | |
Collapse
|
74
|
Villasana LE, Benice TS, Raber J. Long-Term Effects of 56Fe Irradiation on Spatial Memory of Mice: Role of Sex and Apolipoprotein E Isoform. Int J Radiat Oncol Biol Phys 2011; 80:567-73. [DOI: 10.1016/j.ijrobp.2010.12.034] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Revised: 12/09/2010] [Accepted: 12/15/2010] [Indexed: 12/15/2022]
|
75
|
Kennard JA, Woodruff-Pak DS. Age sensitivity of behavioral tests and brain substrates of normal aging in mice. Front Aging Neurosci 2011; 3:9. [PMID: 21647305 PMCID: PMC3103996 DOI: 10.3389/fnagi.2011.00009] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 05/13/2011] [Indexed: 11/21/2022] Open
Abstract
Knowledge of age sensitivity, the capacity of a behavioral test to reliably detect age-related changes, has utility in the design of experiments to elucidate processes of normal aging. We review the application of these tests in studies of normal aging and compare and contrast the age sensitivity of the Barnes maze, eyeblink classical conditioning, fear conditioning, Morris water maze, and rotorod. These tests have all been implemented to assess normal age-related changes in learning and memory in rodents, which generalize in many cases to age-related changes in learning and memory in all mammals, including humans. Behavioral assessments are a valuable means to measure functional outcomes of neuroscientific studies of aging. Highlighted in this review are the attributes and limitations of these measures in mice in the context of age sensitivity and processes of brain aging. Attributes of these tests include reliability and validity as assessments of learning and memory, well-defined neural substrates, and sensitivity to neural and pharmacological manipulations and disruptions. These tests engage the hippocampus and/or the cerebellum, two structures centrally involved in learning and memory that undergo functional and anatomical changes in normal aging. A test that is less well represented in studies of normal aging, the context pre-exposure facilitation effect (CPFE) in fear conditioning, is described as a method to increase sensitivity of contextual fear conditioning to changes in the hippocampus. Recommendations for increasing the age sensitivity of all measures of normal aging in mice are included, as well as a discussion of the potential of the under-studied CPFE to advance understanding of subtle hippocampus-mediated phenomena.
Collapse
Affiliation(s)
- John A. Kennard
- Systems Neuroscience Laboratory, Neuroscience Program and Department of Psychology, Temple UniversityPhiladelphia, PA, USA
| | - Diana S. Woodruff-Pak
- Systems Neuroscience Laboratory, Neuroscience Program and Department of Psychology, Temple UniversityPhiladelphia, PA, USA
| |
Collapse
|
76
|
Tayler KK, Lowry E, Tanaka K, Levy B, Reijmers L, Mayford M, Wiltgen BJ. Characterization of NMDAR-Independent Learning in the Hippocampus. Front Behav Neurosci 2011; 5:28. [PMID: 21629769 PMCID: PMC3099364 DOI: 10.3389/fnbeh.2011.00028] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 05/09/2011] [Indexed: 11/18/2022] Open
Abstract
It is currently thought that memory formation requires the activation of NMDA receptors (NMDARs) in the hippocampus. However, recent studies indicate that these receptors are not necessary for all forms of learning. The current experiments examine this issue using context fear conditioning in mice. First, we show that context fear can be acquired without NMDAR activation in previously trained animals. Mice trained in one environment (context A) are subsequently able to learn about a second environment (context B) in the presence of NMDAR antagonists. Second, we demonstrate that NMDAR-independent learning requires the hippocampus and is dependent on protein synthesis. However, unlike NMDAR-dependent learning, it is not contingent on the expression of activity-regulated cytoskeleton-associated protein (Arc). Lastly, we present data that suggests NMDAR-independent learning is only observed when recently stimulated neurons are reactivated during conditioning. These data suggest that context fear conditioning modifies synaptic plasticity mechanisms in the hippocampus and allows subsequent learning to occur in the absence of NMDAR activation.
Collapse
Affiliation(s)
- Kaycie Kuss Tayler
- Department of Psychology, University of Virginia, Charlottesville VA, USA
| | | | | | | | | | | | | |
Collapse
|
77
|
Wiltgen BJ, Royle GA, Gray EE, Abdipranoto A, Thangthaeng N, Jacobs N, Saab F, Tonegawa S, Heinemann SF, O'Dell TJ, Fanselow MS, Vissel B. A role for calcium-permeable AMPA receptors in synaptic plasticity and learning. PLoS One 2010; 5:e12818. [PMID: 20927382 PMCID: PMC2947514 DOI: 10.1371/journal.pone.0012818] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 07/21/2010] [Indexed: 12/26/2022] Open
Abstract
A central concept in the field of learning and memory is that NMDARs are essential for synaptic plasticity and memory formation. Surprisingly then, multiple studies have found that behavioral experience can reduce or eliminate the contribution of these receptors to learning. The cellular mechanisms that mediate learning in the absence of NMDAR activation are currently unknown. To address this issue, we examined the contribution of Ca(2+)-permeable AMPARs to learning and plasticity in the hippocampus. Mutant mice were engineered with a conditional genetic deletion of GluR2 in the CA1 region of the hippocampus (GluR2-cKO mice). Electrophysiology experiments in these animals revealed a novel form of long-term potentiation (LTP) that was independent of NMDARs and mediated by GluR2-lacking Ca(2+)-permeable AMPARs. Behavioral analyses found that GluR2-cKO mice were impaired on multiple hippocampus-dependent learning tasks that required NMDAR activation. This suggests that AMPAR-mediated LTP interferes with NMDAR-dependent plasticity. In contrast, NMDAR-independent learning was normal in knockout mice and required the activation of Ca(2+)-permeable AMPARs. These results suggest that GluR2-lacking AMPARs play a functional and previously unidentified role in learning; they appear to mediate changes in synaptic strength that occur after plasticity has been established by NMDARs.
Collapse
Affiliation(s)
- Brian J. Wiltgen
- Department of Psychology, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Psychology and the Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Gordon A. Royle
- Neural Plasticity and Regeneration Research Group, Neuroscience Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Erin E. Gray
- Interdepartmental Ph.D. Program for Neuroscience, University of California Los Angeles, Los Angeles, California, United States of America
| | - Andrea Abdipranoto
- Neural Plasticity and Regeneration Research Group, Neuroscience Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Nopporn Thangthaeng
- Neural Plasticity and Regeneration Research Group, Neuroscience Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Nate Jacobs
- Department of Psychology and the Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Faysal Saab
- Department of Psychology and the Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Susumu Tonegawa
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology (MIT), Boston, Massachusetts, United States of America
| | - Stephen F. Heinemann
- Molecular Neurobiology Department, Salk Institute, La Jolla, California, United States of America
| | - Thomas J. O'Dell
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Michael S. Fanselow
- Department of Psychology and the Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Bryce Vissel
- Neural Plasticity and Regeneration Research Group, Neuroscience Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| |
Collapse
|
78
|
Milad MR, Zeidan MA, Contero A, Pitman RK, Klibanski A, Rauch SL, Goldstein JM. The influence of gonadal hormones on conditioned fear extinction in healthy humans. Neuroscience 2010; 168:652-8. [PMID: 20412837 PMCID: PMC2881679 DOI: 10.1016/j.neuroscience.2010.04.030] [Citation(s) in RCA: 247] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 03/05/2010] [Accepted: 04/14/2010] [Indexed: 12/15/2022]
Abstract
Recent rodent studies suggest that gonadal hormones influence extinction of conditioned fear. Here we investigated sex differences in, and the influence of estradiol and progesterone on, fear extinction in healthy humans. Men and women underwent a two-day paradigm in which fear conditioning and extinction learning took place on day 1 and extinction recall was tested on day 2. Visual cues were used as the conditioned stimuli and a mild electric shock was used as the unconditioned stimulus. Skin conductance was recorded throughout the experiment and used to measure conditioned responses (CRs). Blood samples were obtained from all women to measure estradiol and progesterone levels. We found that higher estradiol during extinction learning enhanced subsequent extinction recall but had no effects on fear acquisition or extinction learning itself. Sex differences were only observed during acquisition, with men exhibiting significantly higher CRs. After dividing women into low- and high-estradiol groups, men showed comparable extinction recall to high-estradiol women, and both of these groups showed higher extinction recall than low-estradiol women. Therefore, sex differences in extinction memory emerged only after taking into account women's estradiol levels. Lower estradiol may impair extinction consolidation in women. These findings could have practical applications in the treatment of anxiety disorders through cognitive and behavioral therapies.
Collapse
Affiliation(s)
- M R Milad
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| | | | | | | | | | | | | |
Collapse
|
79
|
Hwang LL, Wang CH, Li TL, Chang SD, Lin LC, Chen CP, Chen CT, Liang KC, Ho IK, Yang WS, Chiou LC. Sex differences in high-fat diet-induced obesity, metabolic alterations and learning, and synaptic plasticity deficits in mice. Obesity (Silver Spring) 2010; 18:463-9. [PMID: 19730425 DOI: 10.1038/oby.2009.273] [Citation(s) in RCA: 318] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Obesity is a potential risk factor for cognitive deficits in the elder humans. Using a high-fat diet (HFD)-induced obese mouse model, we investigated the impacts of HFD on obesity, metabolic and stress hormones, learning performance, and hippocampal synaptic plasticity. Both male and female C57BL/6J mice fed with HFD (3 weeks to 9-12 months) gained significantly more weights than the sex-specific control groups. Compared with the obese female mice, the obese males had similar energy intake but developed more weight gains. The obese male mice developed hyperglycemia, hyperinsulinemia, hypercholesterolemia, and hyperleptinemia, but not hypertriglyceridemia. The obese females had less hyperinsulinemia and hypercholesterolemia than the obese males, and no hyperglycemia and hypertriglyceridemia. In the contextual fear conditioning and step-down passive avoidance tasks, the obese male, but not female, mice showed poorer learning performance than their normal counterparts. These learning deficits were not due to sensorimotor impairment as verified by the open-field and hot-plate tests. Although, basal synaptic transmission characteristics (input-output transfer and paired-pulse facilitation (PPF) ratio) were not significantly different between normal and HFD groups, the magnitudes of synaptic plasticity (long-term potentiation (LTP) and long-term depression (LTD)) were lower at the Schaffer collateral-CA1 synapses of the hippocampal slices isolated from the obese male, but not female, mice, as compared with their sex-specific controls. Our results suggest that male mice are more vulnerable than the females to the impacts of HFD on weight gains, metabolic alterations and deficits of learning, and hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Ling-Ling Hwang
- Department of Physiology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Abstract
Over many years, fear extinction has been conceptualized as one dominant process, new inhibitory learning, which serves to dampen previously acquired fear. Here we present an alternative view, that brain region-specific processing of representations, expectations and emotional attributes of the fear-provoking event, recruits unique mechanisms that interdependently contribute to the conditioning and extinction of fear. The co-occurrence of these mechanisms within the fear circuit can thus be tracked and differentiated at a molecular and cellular level. Among others, the transcriptional regulators cFos, cAMP-dependent response element binding protein (CREB), Zif268, and extracellular signal-regulated kinases (Erk) stand out as hippocampal nuclear markers signaling novelty, arousal, retrieval, and prediction error, respectively. Consistent with evidence from human studies, these findings indicate that, beyond inhibitory learning, fear extinction requires modification of the emotional attributes and expectations that define the threatening context. Given the likely dysregulation of one or more of these processes in anxiety disorders, a key research challenge for the future is the identification and enhancement of individual extinction mechanisms to target the specific components of fear. Environmental stimuli lacking affective properties (conditioned stimuli, CS) rapidly become threatening if presented with stressful events (unconditioned stimuli, US). Consequently, based on a CS-US association, the presentation of the CS triggers species-specific fear responses until the US consistently stops occurring. At that point, new learning takes place and the fear response declines, a phenomenon termed extinction. The view that extinction occurs because a new, inhibitory CS-noUS association gains control over behavior, has remained dominant in the field. The implications of impaired fear regulation in the development of anxiety disorders have stimulated intense research in this area. Rodent studies identified the circuits involved in the conditioning and extinction of fear of salient cues, generating data that were confirmed in humans with brain imaging approaches. Nevertheless, research with experimental animals has not fully taken advantage of human data in order to better interpret extinction mechanisms in the framework of learning, expectation and emotion governing fear-motivated behavior. The present article aims to summarize recent molecular evidence on fear extinction, focusing on hippocampal mechanisms and experimental models of contextual fear, and compare the results with other relevant fear paradigms and human imaging studies. Instead of conceptualizing extinction learning as one process, such as CS-noUS association or inhibitory learning, we propose that fear extinction reflects the behavioral output of several region-specific learning processes that modify different components of the conditioning memory. The significance of these findings is discussed in the framework of fear regulation and anxiety disorders.
Collapse
Affiliation(s)
- Jelena Radulovic
- Department of Psychiatry and Behavioral Sciences, The Asher Center for Study and Treatment of Depressive Disorders, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA.
| | | |
Collapse
|
81
|
Villasana L, Rosenberg J, Raber J. Sex-dependent effects of 56Fe irradiation on contextual fear conditioning in C57BL/6J mice. Hippocampus 2010; 20:19-23. [PMID: 19489001 PMCID: PMC2850563 DOI: 10.1002/hipo.20659] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Effects of irradiation on hippocampal function have been mostly studied in male rodents and relatively little is known about potential effects of irradiation on hippocampal function in female rodents. Moreover, although the long-term effects of clinical radiation on cognitive function have been well established, the effects of other forms of irradiation, such as high charged, high energy radiation (HZE particles) that astronauts encounter during space missions have not been well characterized. In this study we compared the effects of (56)Fe irradiation on fear conditioning in C57BL/6J female and male mice. Hippocampus-dependent contextual fear conditioning was impaired in female mice but improved in male mice following (56)Fe irradiation. Such impairment was not seen for hippocampus-independent cued fear conditioning. Thus, the effects of (56)Fe irradiation on hippocampus-dependent contextual fear conditioning are critically modulated by sex.
Collapse
Affiliation(s)
- Laura Villasana
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | | | | |
Collapse
|
82
|
Moore MD, Cushman J, Chandra D, Homanics GE, Olsen RW, Fanselow MS. Trace and contextual fear conditioning is enhanced in mice lacking the alpha4 subunit of the GABA(A) receptor. Neurobiol Learn Mem 2009; 93:383-7. [PMID: 20018248 DOI: 10.1016/j.nlm.2009.12.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 12/03/2009] [Accepted: 12/10/2009] [Indexed: 11/29/2022]
Abstract
The GABA(A)R alpha4 subunit is highly expressed in the dentate gyrus region of the hippocampus at predominantly extra synaptic locations where, along with the GABA(A)R delta subunit, it forms GABA(A) receptors that mediate a tonic inhibitory current. The present study was designed to test hippocampus-dependent and hippocampus-independent learning and memory in GABA(A)R alpha4 subunit-deficient mice using trace and delay fear conditioning, respectively. Mice were of a mixed C57Bl/6J X 129S1/X1 genetic background from alpha4 heterozygous breeding pairs. The alpha4-knockout mice showed enhanced trace and contextual fear conditioning consistent with an enhancement of hippocampus-dependent learning and memory. These enhancements were sex-dependent, similar to previous studies in GABA(A)R delta knockout mice, but differences were present in both males and females. The convergent findings between alpha4 and delta knockout mice suggests that tonic inhibition mediated by alpha4betadelta GABA(A) receptors negatively modulates learning and memory processes and provides further evidence that tonic inhibition makes important functional contributions to learning and behavior.
Collapse
Affiliation(s)
- M D Moore
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
83
|
McHugh TJ, Tonegawa S. CA3 NMDA receptors are required for the rapid formation of a salient contextual representation. Hippocampus 2009; 19:1153-8. [PMID: 19650121 PMCID: PMC2788054 DOI: 10.1002/hipo.20684] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The acquisition of Pavlovian fear learning engages the hippocampus when the conditioned stimuli are multimodal or temporally isolated from the unconditioned stimuli. By subjecting CA3-NR1 KO mice to conditioning protocols that incorporate time-dependent components, we found that the loss of plasticity at recurrent CA3 synapses resulted in a deficits in contextual conditioning specifically when the exposure to the context was brief or when the unconditioned stimulus was signaled with a competing, predictive unimodal stimulus. Our results suggest CA3 contributes both speed and salience to contextual processing and support the theory of competition between multimodal and unimodal conditioned stimuli for associative learning.
Collapse
Affiliation(s)
- Thomas J McHugh
- The RIKEN-MIT Center for Neural Circuit Genetics, Howard Hughes Medical Institute, The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
| | | |
Collapse
|
84
|
Burman MA, Murawski NJ, Schiffino FL, Rosen JB, Stanton ME. Factors governing single-trial contextual fear conditioning in the weanling rat. Behav Neurosci 2009; 123:1148-52. [PMID: 19824781 PMCID: PMC4075732 DOI: 10.1037/a0016733] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although contextual fear conditioning emerges later in development than explicit-cue fear conditioning, little is known about the stimulus parameters and biological substrates required at early ages. The authors adapted methods for investigating hippocampus function in adult rodents to identify determinants of contextual fear conditioning in developing rats. Experiment 1 examined the duration of exposure required by weanling rats at postnatal day (PND) 23 to demonstrate contextual fear conditioning. This experiment demonstrated that 30 s of context exposure is sufficient to support conditioning. Furthermore, preexposure enhanced conditioning to an immediate footshock, the context preexposure facilitation effect (CPFE), but had no effect on contextual conditioning to a delayed shock. Experiment 2 demonstrated that N-methyl-D-aspartate (NMDA) receptor inactivation during preexposure impairs contextual learning at PND 23. Thus, the conjuctive representations underlying the CPFE are NMDA-dependent as early as PND23 in the rat.
Collapse
Affiliation(s)
- M A Burman
- University of Delaware, Newark, DE 19716, USA.
| | | | | | | | | |
Collapse
|
85
|
Milad MR, Igoe SA, Lebron-Milad K, Novales JE. Estrous cycle phase and gonadal hormones influence conditioned fear extinction. Neuroscience 2009; 164:887-95. [PMID: 19761818 DOI: 10.1016/j.neuroscience.2009.09.011] [Citation(s) in RCA: 318] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 09/02/2009] [Accepted: 09/08/2009] [Indexed: 11/17/2022]
Abstract
Gonadal hormones modulate fear acquisition, but less is known about the influence of gonadal hormones on fear extinction. We assessed sex differences and the influence of gonadal hormone fluctuations and exogenous manipulations of estrogen and progesterone on acquisition, extinction learning and extinction recall in a 3 day auditory fear conditioning and extinction protocol. Experiments were conducted on males and naturally cycling female rats. Regarding female rats, significant differences in fear extinction were observed between subgroups of females, depending on their phase of the estrous cycle. Extinction that took place during the proestrus (high estrogen/progesterone) phase was more fully consolidated, as evidenced by low freezing during a recall test. This suggests that estrogen and/or progesterone facilitate extinction. In support of this, injection of both estrogen and progesterone prior to extinction learning in female rats during the metestrus phase of the cycle (low estrogen/progesterone) facilitated extinction consolidation, and blockade of estrogen and progesterone receptors during the proestrus phase impaired extinction consolidation. When comparing male to female rats without consideration of the estrous cycle phase, no significant sex differences were observed. When accounting for cycle phase in females, sex differences were observed only during extinction recall. Female rats that underwent extinction during the metestrus phase showed significantly higher freezing during the recall test relative to males. Collectively, these data suggest that gonadal hormones influence extinction behavior possibly by influencing the function of brain regions involved in the consolidation of fear extinction. Moreover, the elevated fear observed in female relative to male rats during extinction recall suggests that gonadal hormones may in part play a role in the higher prevalence of anxiety disorders in women.
Collapse
Affiliation(s)
- M R Milad
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| | | | | | | |
Collapse
|
86
|
Laurent V, Westbrook RF. Inactivation of the infralimbic but not the prelimbic cortex impairs consolidation and retrieval of fear extinction. Learn Mem 2009; 16:520-9. [DOI: 10.1101/lm.1474609] [Citation(s) in RCA: 262] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
87
|
Abstract
In essentially every domain of neuroscience, the generally implicit assumption that few, if any, meaningful differences exist between male and female brain function is being challenged. Here we address how this development is influencing studies of the neurobiology of learning and memory. While it has been commonly held that males show an advantage on spatial tasks, and females on verbal tasks, there is increasing evidence that sex differences are more widespread than previously supposed. Differing performance between the sexes have been observed on a number of common learning tasks in both the human and animal literature, many neither purely spatial nor verbal. We review sex differences reported in various areas to date, while attempting to identify common features of sexually dimorphic tasks, and to place these differences in a neurobiological context. This discussion focuses on studies of four classes of memory tasks for which sex differences have been frequently reported: spatial, verbal, autobiographical, and emotional memory. We conclude that the female verbal advantage extends into numerous tasks, including tests of spatial and autobiographical abilities, but that a small but significant advantage may exist for general episodic memory. We further suggest that for some tasks, stress evokes sex differences, which are not normally observed, and that these differences are mediated largely by interactions between stress and sex hormones.
Collapse
Affiliation(s)
- Joseph M Andreano
- Center for the Neurobiology of Learning and Memory, Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-3800, USA
| | | |
Collapse
|
88
|
Gresack JE, Schafe GE, Orr PT, Frick KM. Sex differences in contextual fear conditioning are associated with differential ventral hippocampal extracellular signal-regulated kinase activation. Neuroscience 2009; 159:451-67. [PMID: 19171181 DOI: 10.1016/j.neuroscience.2009.01.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 12/12/2008] [Accepted: 01/07/2009] [Indexed: 10/21/2022]
Abstract
Although sex differences have been reported in hippocampal-dependent learning and memory, including contextual fear memories, the underlying molecular mechanisms contributing to such differences are not well understood. The present study examined the extent to which sex differences in contextual fear conditioning are related to differential activation of the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK), a protein kinase critically involved in memory formation. We first show that male rats exhibit more long-term retention of contextual fear conditioning than female rats. During a tone test, females spent more time freezing than males, although both sexes exhibited robust retention of auditory fear learning. Using Western blot analysis, we then show that phosphorylated ERK levels in ventral, but not dorsal, hippocampus are higher in males than females, relative to same-sex controls, 60 minutes after fear conditioning. Post-conditioning increases in ERK activation were observed in the amygdala in both males and females, suggesting a selective effect of sex on hippocampal ERK activation. Together, these findings suggest that differential activation of the ERK signal transduction pathway in male and female rats, particularly in the ventral hippocampus, is associated with sex differences in contextual fear.
Collapse
Affiliation(s)
- J E Gresack
- Department of Psychology, Yale University, PO Box 208205, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
89
|
Infusion of the NMDA receptor antagonist, DL-APV, into the basolateral amygdala disrupts learning to fear a novel and a familiar context as well as relearning to fear an extinguished context. Learn Mem 2009; 16:96-105. [PMID: 19141468 DOI: 10.1101/lm.1218709] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Ample evidence suggests that activation of NMDA receptors (NMDAr) in the basolateral complex of the amygdala (BLA) is necessary for context fear conditioning. The present series of experiments examined whether this activation was still required when the to-be-shocked context had a history. We found that BLA infusion of the selective NMDAr antagonist DL-APV impaired the acquisition of fear responses to a novel context, a moderately familiar context, or a highly familiar context. The same treatment also impaired the reacquisition of fear responses to a dangerous context, a context extinguished to criterion, or a context massively extinguished. Importantly, DL-APV persistently suppressed fear responses, suggesting that the NMDAr antagonist disrupted basal synaptic transmission in the BLA. Therefore, we conclude that neuronal activity in the BLA is critical for learning and relearning context-conditioned fear. This finding is consistent with current neural models that attribute context fear conditioning to interactions among several brain regions, most notably the hippocampus and the BLA.
Collapse
|
90
|
Kholodar-Smith DB, Boguszewski P, Brown TH. Auditory trace fear conditioning requires perirhinal cortex. Neurobiol Learn Mem 2008; 90:537-43. [PMID: 18678265 PMCID: PMC2629995 DOI: 10.1016/j.nlm.2008.06.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 06/19/2008] [Accepted: 06/19/2008] [Indexed: 01/13/2023]
Abstract
The hippocampus is well-known to be critical for trace fear conditioning, but nothing is known about the importance of perirhinal cortex (PR), which has reciprocal connections with hippocampus. PR damage severely impairs delay fear conditioning to ultrasonic vocalizations (USVs) and discontinuous tones (pips), but has no effect on delay conditioning to continuous tones. Here we demonstrate that trace auditory fear conditioning also critically depends on PR function. The trace interval between the CS offset and the US onset was 16s. Pre-training neurotoxic lesions were produced through multiple injections of N-methyl-D-aspartate along the full length of PR, which was directly visualized during the injections. Control animals received injections with phosphate-buffered saline. Three-dimensional reconstructions of the lesion volumes demonstrated that the neurotoxic damage was well-localized to PR and included most of its anterior-posterior extent. Automated video analysis quantified freezing behavior, which served as the conditional response. PR-damaged rats were profoundly impaired in trace conditioning to either of three different CSs (a USV, tone pips, and a continuous tone) as well as conditioning to the training context. Within both the lesion and control groups, the type of cue had no effect on the mean CR. The overall PR lesion effect size was 2.7 for cue conditioning and 3.9 for context conditioning. We suggest that the role of PR in trace fear conditioning may be distinct from some of its more perceptual functions. The results further define the essential circuitry underlying trace fear conditioning to auditory cues.
Collapse
Affiliation(s)
- D B Kholodar-Smith
- Departments of Psychology, Yale University, 2 Hillhouse Ave, New Haven, CT 06520, USA
| | | | | |
Collapse
|
91
|
Laurent V, Marchand AR, Westbrook RF. The basolateral amygdala is necessary for learning but not relearning extinction of context conditioned fear. Learn Mem 2008; 15:304-14. [PMID: 18463174 PMCID: PMC2364602 DOI: 10.1101/lm.928208] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 02/21/2008] [Indexed: 12/20/2022]
Abstract
Extinction of conditioned fear involves new learning that inhibits but does not eliminate the original fear memory. This inhibitory learning is thought to require activation of NMDA receptors (NMDAr) within the basolateral amygdala (BLA). However, once extinction has been learned, the role played by the BLA during subsequent extinction procedures remains unknown. The present study examined the role of neuronal activity and NMDAr activation in rats receiving their first or second extinction of context fear. We found that BLA infusion of DL-APV, a competitive antagonist of NMDAr, depressed fear responses at both the first and second extinction. It impaired learning extinction but spared and even facilitated relearning extinction. BLA infusion of muscimol, a GABA(A) agonist, produced a similar outcome, suggesting that DL-APV not only blocked NMDAr-dependent plasticity but also disrupted neuronal activity. In contrast, infusion of ifenprodil, a more selective antagonist of NMDAr containing the NR2B subunit, did not depress fear responses but impaired short- and long-term inhibition of fear at both the first and second extinction. Therefore, we suggest that relearning extinction normally requires NMDAr containing the NR2B subunit in the BLA. However, simultaneous blockade of these receptors and neuronal activity in the BLA results in compensatory learning that is able to promote long-term re-extinction. These data are consistent with a current model that attributes fear extinction to interactions between several neural substrates, including the amygdala and the medial prefrontal cortex.
Collapse
Affiliation(s)
- Vincent Laurent
- School of Psychology, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Centre de Neurosciences Intégratives et Cognitives, UMR 5228, CNRS, F-33405 Talence Cedex, France
| | - Alain R. Marchand
- Centre de Neurosciences Intégratives et Cognitives, UMR 5228, CNRS, F-33405 Talence Cedex, France
| | - R. Frederick Westbrook
- School of Psychology, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
92
|
Perez-Villalba A, Mackintosh NJ, Canales JJ. Influence of massed and distributed context preexposure on contextual fear and Egr-1 expression in the basolateral amygdala. Physiol Behav 2008; 93:206-14. [PMID: 17900634 DOI: 10.1016/j.physbeh.2007.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 08/21/2007] [Accepted: 08/22/2007] [Indexed: 11/23/2022]
Abstract
Preexposure to the conditioning context can influence the expression of context-conditioned fear. We used behavioral and early growth response gene (egr-1) assays in rats to study the effects of massed and distributed context preexposure on context-conditioned fear. The results demonstrated that massed context preexposure impaired acquisition of contextual fear, an effect here referred to as delayed shock deficit. Spaced context preexposure produced similar inhibitory effects. Significantly, the introduction of a brief change of context prior to conditioning completely reversed the deficit induced by massed, but not by distributed, context preexposure. This reversibility was inversely related to the duration of the context shift. The acquisition of context-conditioned fear was associated with enhanced Egr-1 expression in the basolateral amygdala (BLA). No such increase was evident in animals undergoing distributed context preexposure or in those experiencing massed preexposure without change of context. Remarkably, a brief change of context prior to conditioning not only facilitated learning following massed preexposure but also elicited a significant elevation of Egr-1 protein levels in the BLA. The findings shown demonstrated that the inhibitory effects of massed and distributed context preexposure on conditioning could be dissociable both behaviorally and physiologically. We suggest that the delayed shock deficit associated with massed preexposure derives from perceptual fade-out or inattention and its reversal by a brief change of context from attentional recovery.
Collapse
Affiliation(s)
- A Perez-Villalba
- Laboratory of Biopsychology and Comparative Neuroscience, Cavanilles Institute (ICBiBE), University of Valencia, Polígono de la Coma s/n, Paterna-46980 Valencia-Spain.
| | | | | |
Collapse
|
93
|
Abstract
Context memories initially require the hippocampus, but over time become independent of this structure. This shift reflects a consolidation process whereby memories are gradually stored in distributed regions of the cortex. The function of this process is thought to be the extraction of statistical regularities and general knowledge from specific experiences. The current study examined this idea in mice by measuring the specificity of context memories during consolidation. In the first experiment, separate groups of animals were trained with a single shock and tested in the training context or a novel environment 1, 14, 28, or 36 d later. We found a systematic increase in generalization over this period. Initially, mice froze more in the training context, but fear of the novel environment grew over time until animals eventually froze an equivalent amount in both contexts. The second experiment demonstrated that the increase in generalization was due to a loss of detailed information about the context and not fear incubation. In this experiment, mice were exposed to the context and then trained with an immediate shock 1 or 36 d later. Animals trained 1 d after exposure acquired robust context fear that did not generalize across environments. In contrast, mice trained 36 d after exposure froze an equivalent amount in the training context and the novel environment. The same profile was observed in H-ras mutants that exhibit enhanced hippocampal plasticity and learning. These results suggest that context memories are specific early after training when they require the hippocampus, and become more general as they are permanently stored in the cortex.
Collapse
Affiliation(s)
- Brian J Wiltgen
- Departments of Neurobiology, Psychology, Psychiatry and the Brain Research Institute, UCLA, Los Angeles, California 90095, USA.
| | | |
Collapse
|
94
|
Cravens CJ, Vargas-Pinto N, Christian KM, Nakazawa K. CA3 NMDA receptors are crucial for rapid and automatic representation of context memory. Eur J Neurosci 2007; 24:1771-80. [PMID: 17004940 DOI: 10.1111/j.1460-9568.2006.05044.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
It is argued that the hippocampus contributes to acquisition of context-specific memory although neural mechanisms have not been clarified. To evaluate the role of CA3 in context-specific memory, we developed one-trial context discrimination tasks to test acquisition and retrieval of contextual memory in CA3 pyramidal cell-restricted N-methyl-d-aspartate (NMDA) receptor knockout mice. Mutants were unable to discriminate conditioned and no-shock contexts 3 h after one-trial avoidance training. These phenotypes were not evident 24 h after one-trial training or 3 h after multi-trial training. Following one-trial contextual fear conditioning, mutants showed a selective deficit in context discrimination during a retention test 3 h after acquisition, although overall freezing levels were similar to those of the control mice. As in the avoidance task, this context discrimination impairment was not observed 24 h after initial conditioning. Interestingly, extending the post-shock period to 3 min during the one-trial fear conditioning task eliminated the discrimination deficit observed at the 3 h retention interval. These results suggest that: (i) impaired rapid context discrimination during the recall test is dependent on the duration of post-shock period during conditioning; (ii) CA3 NMDA receptors are critically involved in rapid and automatic formation of a unified context memory representation from the sensory information; (iii) CA3 NMDA receptors support contextual pattern separation; (iv) fear memory to foot-shock is acquired without CA3 NMDA receptors. It appears that rapid and automatic context memory representations from one-time experience are mediated, at least in part, by CA3 NMDA receptors.
Collapse
Affiliation(s)
- Catherine J Cravens
- Unit on Genetics of Cognition and Behaviour, Mood and Anxiety Disorders Research Program, National Institute of Mental Health, Bethesda, MD 20892-3710, USA
| | | | | | | |
Collapse
|
95
|
Baarendse PJJ, van Grootheest G, Jansen RF, Pieneman AW, Ogren SO, Verhage M, Stiedl O. Differential involvement of the dorsal hippocampus in passive avoidance in C57bl/6J and DBA/2J mice. Hippocampus 2007; 18:11-9. [PMID: 17696168 DOI: 10.1002/hipo.20356] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The inferior performance of DBA/2 mice when compared to C57BL/6 mice in hippocampus-dependent behavioral tasks including contextual fear conditioning has been attributed to impaired hippocampal function. However, DBA/2J mice have been reported to perform similarly or even better than C57BL/6J mice in the passive avoidance (PA) task that most likely also depends on hippocampal function. The apparent discrepancy in PA versus fear conditioning performance in these two strains of mice was investigated using an automated PA system. The aim was to determine whether these two mouse strains utilize different strategies involving a different contribution of hippocampal mechanisms to encode PA. C57BL/6J mice exhibited significantly longer retention latencies than DBA/2J mice when tested 24 h after training irrespective of the circadian cycle. Dorsohippocampal NMDA receptor inhibition by local injection of the selective antagonist DL-2-amino-5-phosphonovaleric acid (AP5, 3.2 microg/mouse) before training resulted in impaired PA retention in C57BL/6J but not in DBA/2J mice. Furthermore, nonreinforced pre-exposure to the PA system before training caused a latent inhibition-like reduction of retention latencies in C57BL/6J, whereas it improved PA retention in DBA/2J mice. These pre-exposure experiments facilitated the discrimination of hippocampal involvement without local pharmacological intervention. The results indicate differences in PA learning between these two strains based on a different NMDA receptor involvement in the dorsal hippocampus in this emotional learning task. We hypothesize that mouse strains can differ in their PA learning performance based on their relative ability to form associations on the basis of unisensory versus multisensory contextual/spatial cues that involve hippocampal processing.
Collapse
Affiliation(s)
- Petra J J Baarendse
- Center for Neurogenomics and Cognitive Research and Institute for Neurosciences (INW), Vrije Universiteit Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
96
|
Blaeser F, Sanders MJ, Truong N, Ko S, Wu LJ, Wozniak DF, Fanselow MS, Zhuo M, Chatila TA. Long-term memory deficits in Pavlovian fear conditioning in Ca2+/calmodulin kinase kinase alpha-deficient mice. Mol Cell Biol 2006; 26:9105-15. [PMID: 17015467 PMCID: PMC1636841 DOI: 10.1128/mcb.01452-06] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Signaling by the Ca(2+)/calmodulin kinase (CaMK) cascade has been implicated in neuronal gene transcription, synaptic plasticity, and long-term memory consolidation. The CaM kinase kinase alpha (CaMKKalpha) isoform is an upstream component of the CaMK cascade whose function in different behavioral and learning and memory paradigms was analyzed by targeted gene disruption in mice. CaMKKalpha mutants exhibited normal long-term spatial memory formation and cued fear conditioning but showed deficits in context fear during both conditioning and long-term follow-up testing. They also exhibited impaired activation of the downstream kinase CaMKIV/Gr and its substrate, the transcription factor cyclic AMP-responsive element binding protein (CREB) upon fear conditioning. Unlike CaMKIV/Gr-deficient mice, the CaMKKalpha mutants exhibited normal long-term potentiation and normal levels of anxiety-like behavior. These results demonstrate a selective role for CaMKKalpha in contextual fear memory and suggest that different combinations of upstream and downstream components of the CaMK cascade may serve distinct physiological functions.
Collapse
Affiliation(s)
- Frank Blaeser
- Department of Pediatrics, The David Geffen School of Medicine, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1752, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Wiltgen BJ, Sanders MJ, Anagnostaras SG, Sage JR, Fanselow MS. Context fear learning in the absence of the hippocampus. J Neurosci 2006; 26:5484-91. [PMID: 16707800 PMCID: PMC6675287 DOI: 10.1523/jneurosci.2685-05.2006] [Citation(s) in RCA: 276] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Lesions of the rodent hippocampus invariably abolish context fear memories formed in the recent past but do not always prevent new learning. To better understand this discrepancy, we thoroughly examined the acquisition of context fear in rats with pretraining excitotoxic lesions of the dorsal hippocampus. In the first experiment, animals received a shock immediately after placement in the context or after variable delays. Immediate shock produced no context fear learning in lesioned rats or controls. In contrast, delayed shock produced robust context fear learning in both groups. The absence of fear with immediate shock occurs because animals need time to form a representation of the context before shock is presented. The fact that it occurs in both sham and lesioned rats suggests that they learn about the context in a similar manner. However, despite learning about the context in the delay condition, lesioned rats did not acquire as much fear as controls. The second experiment showed that this lesion-induced deficit could be overcome by increasing the number of conditioning trials. Lesioned animals learned normally after multiple shocks, regardless of freezing level or trial spacing. The last experiment showed that animals with complete hippocampus lesions could also learn about the context, although the same lesions produced devastating retrograde amnesia. These results demonstrate that alternative systems can acquire context fear but do so less efficiently than the hippocampus.
Collapse
|
98
|
Sanders MJ, Kieffer BL, Fanselow MS. Deletion of the mu opioid receptor results in impaired acquisition of Pavlovian context fear. Neurobiol Learn Mem 2005; 84:33-41. [PMID: 15936681 DOI: 10.1016/j.nlm.2005.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2005] [Revised: 02/28/2005] [Accepted: 03/03/2005] [Indexed: 11/29/2022]
Abstract
The mu opioid receptor may constitute a critical component of a negative feedback system that regulates Pavlovian fear conditioning. We investigated context fear conditioning acquisition and expression in mu opioid receptor knockout mice (on an inbred, C57 genetic background). We discovered that the mu receptor knockout results in an unexpected and significant deficit in context fear acquisition. Mice lacking the mu receptor showed normal fear acquisition when subjected to a 1-day fear conditioning protocol but evinced deficient fear learning when acquisition was conducted across 5 days. The knockout mice showed normal reactivity to footshock in both fear conditioning protocols. Finally, we confirmed the effectiveness of the receptor deletion in the C57 strain by subjecting the mice to a test of morphine analgesia in the hot-plate assay. As has been seen with mixed genetic background, the receptor deletion resulted in a complete lack of analgesic response to 10 mg/kg morphine. Surprisingly, mice with a single copy of the mu receptor gene (heterozygous knockouts) showed intact sensitivity to morphine but a significant deficit in Pavlovian fear conditioning. The results indicate that deletion of the mu receptor gene impairs fear conditioning and that the conditioning and analgesia effects of heterozygous deletion are dissociable. The conditioning deficit seen in this line of mice may be related to impairment in hippocampus function.
Collapse
|
99
|
Perez-Villalba A, Teruel-Martí V, Ruiz-Torner A, Olucha-Bordonau F. The effect of long context exposure on cued conditioning and c-fos expression in the rat forebrain. Behav Brain Res 2005; 161:263-75. [PMID: 15922053 DOI: 10.1016/j.bbr.2005.02.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2004] [Revised: 02/10/2005] [Accepted: 02/17/2005] [Indexed: 02/05/2023]
Abstract
The c-fos expression was used to study the neural substrates of the cued fear conditioning acquisition, preceded by a short exposure versus a long exposure to the conditioning context. A long-context exposure (either during the night or during the day) prior to conditioning, was associated with low freezing in the learning test. Differences in the c-fos expression of CA1, CA3, BL Amygdala, LS and BNST were found between the short- or long-context groups with a pre-exposure before cued conditioning. Ce Amygdala showed no differences in the c-fos expression labeling. We reported the hippocampal c-fos activation during the cued fear conditioning acquisition. Specifically, the CA1 activation could be related with the context-US processing during the CS-US association acquisition, which might prove that the CS-US associations cannot be made without an integrated context participating. The results showed that a long-context exposure prior to cued conditioning produces an inhibition of the CR (freezing), and this phenomenon is related with a specific c-fos expression in CA1, CA3, BL Amygdala, LS and BNST during the fear acquisition.
Collapse
Affiliation(s)
- Ana Perez-Villalba
- Department of Embryology and Human Anatomy, School of Medicine, Av Blasco Ibanez, 15, University of Valencia, 46010 Valencia, Spain.
| | | | | | | |
Collapse
|
100
|
Wiltgen BJ, Sanders MJ, Ferguson C, Homanics GE, Fanselow MS. Trace fear conditioning is enhanced in mice lacking the delta subunit of the GABAA receptor. Learn Mem 2005; 12:327-33. [PMID: 15897254 PMCID: PMC1142462 DOI: 10.1101/lm.89705] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The delta subunit of the GABA(A) receptor (GABA(A)R) is highly expressed in the dentate gyrus of the hippocampus. Genetic deletion of this subunit reduces synaptic and extrasynaptic inhibition and decreases sensitivity to neurosteroids. This paper examines the effect of these changes on hippocampus-dependent trace fear conditioning. Compared to controls, delta knockout mice exhibited enhanced acquisition of tone and context fear. Hippocampus-independent delay conditioning was normal in these animals. These results suggest that reduced inhibition in the dentate gyrus facilitates the acquisition of trace fear conditioning. However, the enhancement in trace conditioning was only observed in female knockout mice. The sex-specificity of this effect may be a result of neuroactive steroids. These compounds vary during the estrus cycle, can increase GABAergic inhibition, and have been shown to impair hippocampus-dependent learning. We propose that activation of GABA(A)Rs by neuroactive steroids inhibits learning processes in the hippocampus. Knockouts are immune to this effect because of the reduced neurosteroid sensitivity that accompanies deletion of the delta subunit. Relationships between neurosteroids, hippocampal excitability, and memory are discussed.
Collapse
Affiliation(s)
- Brian J Wiltgen
- Psychology Department, and The Brain Research Institute, UCLA, Los Angeles, California 90095, USA
| | | | | | | | | |
Collapse
|