1
|
Jäger A, Pieper A, Priebe K, Hellweg R, Meyer K, Herrmann S, Wolfarth B, Grummt M, Ströhle A, Schoofs N. Effects of high intensity interval training on serum brain-derived neurotrophic factor in individuals with PTSD. J Psychiatr Res 2024; 180:355-361. [PMID: 39520767 DOI: 10.1016/j.jpsychires.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/25/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is a protein important for synaptic plasticity and formation of memory. It is suggested to play an important role in the development of psychiatric disorders like post-traumatic stress disorder (PTSD). Individuals with PTSD usually show decreased BDNF levels in serum. Physical exercise has shown to be effective in increasing serum BDNF levels. OBJECTIVE As the most beneficial form of exercise to raise serum BDNF levels in individuals with PTSD is yet to be determined, we compared two training protocols and their effects on BDNF release. We expected that a training with higher intensity increases BDNF serum levels more than a low intensity training (LIT). METHOD 40 participants (80% female) diagnosed with PTSD were randomized to either high-intensity interval training (HIIT) or a low intensity training group (LIT). They underwent a 12-day training period. We measured serum BDNF levels pre- and post-exercise on first and last intervention day. BDNF was controlled for platelet counts. RESULTS In the HIIT group there was a significant increase in serum BDNF post-exercise on both days measured, respectively when controlled for platelets. The increase was transient. Both groups did not show an increase in serum BDNF over the course of the 12-day training period. CONCLUSION A single session of HIIT raised serum BDNF levels in individuals with PTSD transiently. Neither HIIT nor LIT raised serum BDNF levels over the course of 12 days.
Collapse
Affiliation(s)
- Annabell Jäger
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Neurosciences, CCM, Charitéplatz 1, 10117, Berlin, Germany.
| | - Anima Pieper
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Neurosciences, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Kathlen Priebe
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Neurosciences, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Rainer Hellweg
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Neurosciences, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Kristina Meyer
- Charité Universitätsmedizin Berlin, Institute of Medical Psychology, Charitéplatz 1, 10117, Berlin, Germany
| | - Sarah Herrmann
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Neurosciences, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Bernd Wolfarth
- Charité Universitätsmedizin Berlin, Department of Sports Medicine, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Maximilian Grummt
- Charité Universitätsmedizin Berlin, Department of Sports Medicine, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Andreas Ströhle
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Neurosciences, CCM, Charitéplatz 1, 10117, Berlin, Germany
| | - Nikola Schoofs
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Neurosciences, CCM, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
2
|
Jaehne EJ, Antolasic EJ, Creutzberg KC, Begni V, Riva MA, van den Buuse M. Impaired fear memory in a rat model of the Brain-Derived Neurotrophic Factor Val66Met polymorphism is reversed by chronic exercise. Neurobiol Learn Mem 2023; 203:107779. [PMID: 37269900 DOI: 10.1016/j.nlm.2023.107779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/08/2023] [Accepted: 05/24/2023] [Indexed: 06/05/2023]
Abstract
The brain-derived neurotrophic factor (BDNF) Val66Met polymorphism is associated with reduced activity-dependent BDNF release in the brain and has been implicated in fear and anxiety disorders, including post-traumatic stress disorder. Exercise has been shown to have benefits in affective disorders but the role of BDNF Val66Met remains unclear. Male and female BDNF Val66Met rats were housed in automated running-wheel cages from weaning while controls were housed in standard cages. During adulthood, all rats underwent standard three-day fear conditioning testing, with three tone/shock pairings on day 1 (acquisition), and extinction learning and memory (40 tones/session) on day 2 and day 3. Expression of BDNF and stress-related genes were measured in the frontal cortex. Extinction testing on day 2 revealed significantly lower freezing in response to initial cue exposure in control Met/Met rats, reflecting impaired fear memory. This deficit was reversed in both male and female Met/Met rats exposed to exercise. There were no genotype effects on acquisition or extinction of fear, however chronic exercise increased freezing in all groups at every stage of testing. Exercise furthermore led to increased expression of Bdnf in the prefrontal cortex of females and its isoforms in both sexes, as well as increased expression of FK506 binding protein 51 (Fkpb5) in females and decreased expression of Serum/glucocorticoid-regulated kinase (Sgk1) in males independent of genotype. These results show that the Met/Met genotype of the Val66Met polymorphism affects fear memory, and that chronic exercise selectively reverses this genotype effect. Chronic exercise also led to an overall increase in freezing in all genotypes which may contribute to results.
Collapse
Affiliation(s)
- Emily J Jaehne
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Emily J Antolasic
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Kerstin C Creutzberg
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Biological Psychiatry Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Maarten van den Buuse
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Australia; Department of Pharmacology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
3
|
Behavioral phenotyping of a rat model of the BDNF Val66Met polymorphism reveals selective impairment of fear memory. Transl Psychiatry 2022; 12:93. [PMID: 35256586 PMCID: PMC8901920 DOI: 10.1038/s41398-022-01858-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 01/02/2023] Open
Abstract
The common brain-derived neurotrophic factor (BDNF) Val66Met polymorphism is associated with reduced activity-dependent BDNF release and increased risk for anxiety disorders and PTSD. Here we behaviorally phenotyped a novel Val66Met rat model with an equivalent valine to methionine substitution in the rat Bdnf gene (Val68Met). In a three-day fear conditioning protocol of fear learning and extinction, adult rats with the Met/Met genotype demonstrated impaired fear memory compared to Val/Met rats and Val/Val controls, with no genotype differences in fear learning or extinction. This deficit in fear memory occurred irrespective of the sex of the animals and was not seen in adolescence (4 weeks of age). There were no changes in open-field locomotor activity or anxiety measured in the elevated plus maze (EPM) nor in other types of memory measured using the novel-object recognition test or Y-maze. BDNF exon VI expression in the dorsal hippocampus was higher and BDNF protein level in the ventral hippocampus was lower in female Val/Met rats than female Val/Val rats, with no other genotype differences, including in total BDNF, BDNF long, or BDNF IV mRNA. These data suggest a specific role for the BDNF Met/Met genotype in fear memory in rats. Further studies are required to investigate gene-environment interactions in this novel animal model.
Collapse
|
4
|
Méndez-Couz M, Krenzek B, Manahan-Vaughan D. Genetic Depletion of BDNF Impairs Extinction Learning of a Spatial Appetitive Task in the Presence or Absence of the Acquisition Context. Front Behav Neurosci 2021; 15:658686. [PMID: 33994970 PMCID: PMC8119774 DOI: 10.3389/fnbeh.2021.658686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Brain derived neurotropic factor (BDNF) supports neuronal survival, growth, and differentiation and is involved in forms of hippocampus-dependent and independent learning, as well as hippocampus-dependent learning. Extinction learning comprises active inhibition of no-longer relevant learned information, in conjunction with a decreased response of a previously learned behavior. It is highly dependent on context, and evidence exists that it requires hippocampal activation. The participation of BDNF in memory processing is experience-dependent. For example, BDNF has been associated with synaptic plasticity needed for spatial learning, and it is involved in acquisition and extinction learning of fear conditioning. However, little is known about its role in spatial appetitive extinction learning. In this study, we evaluated to what extent BDNF contributes to spatial appetitive extinction learning in the presence (ABA) or absence (AAA) of exposure to the acquisition context. Daily training, of BDNF+/--mice or their wildtype (WT) littermates, to reach acquisition criterion in a T-maze, resulted in a similar performance outcome. However, extinction learning was delayed in the AAA, and impaired in the ABA-paradigm compared to performance in WT littermates. Trial-by-trial learning analysis indicated differences in the integration of the context into extinction learning by BDNF+/--mice compared to WT littermates. Taken together, these results support an important role for BDNF in processes that relate to information updating and retrieval that in turn are crucial for effective extinction learning.
Collapse
Affiliation(s)
- Marta Méndez-Couz
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Beate Krenzek
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | | |
Collapse
|
5
|
Notaras M, van den Buuse M. Neurobiology of BDNF in fear memory, sensitivity to stress, and stress-related disorders. Mol Psychiatry 2020; 25:2251-2274. [PMID: 31900428 DOI: 10.1038/s41380-019-0639-2] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/01/2019] [Accepted: 12/12/2019] [Indexed: 01/17/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is widely accepted for its involvement in resilience and antidepressant drug action, is a common genetic locus of risk for mental illnesses, and remains one of the most prominently studied molecules within psychiatry. Stress, which arguably remains the "lowest common denominator" risk factor for several mental illnesses, targets BDNF in disease-implicated brain regions and circuits. Altered stress-related responses have also been observed in animal models of BDNF deficiency in vivo, and BDNF is a common downstream intermediary for environmental factors that potentiate anxiety- and depressive-like behavior. However, BDNF's broad functionality has manifested a heterogeneous literature; likely reflecting that BDNF plays a hitherto under-recognized multifactorial role as both a regulator and target of stress hormone signaling within the brain. The role of BDNF in vulnerability to stress and stress-related disorders, such as posttraumatic stress disorder (PTSD), is a prominent example where inconsistent effects have emerged across numerous models, labs, and disciplines. In the current review we provide a contemporary update on the neurobiology of BDNF including new data from the behavioral neuroscience and neuropsychiatry literature on fear memory consolidation and extinction, stress, and PTSD. First we present an overview of recent advances in knowledge on the role of BDNF within the fear circuitry, as well as address mounting evidence whereby stress hormones interact with endogenous BDNF-TrkB signaling to alter brain homeostasis. Glucocorticoid signaling also acutely recruits BDNF to enhance the expression of fear memory. We then include observations that the functional common BDNF Val66Met polymorphism modulates stress susceptibility as well as stress-related and stress-inducible neuropsychiatric endophenotypes in both man and mouse. We conclude by proposing a BDNF stress-sensitivity hypothesis, which posits that disruption of endogenous BDNF activity by common factors (such as the BDNF Val66Met variant) potentiates sensitivity to stress and, by extension, vulnerability to stress-inducible illnesses. Thus, BDNF may induce plasticity to deleteriously promote the encoding of fear and trauma but, conversely, also enable adaptive plasticity during extinction learning to suppress PTSD-like fear responses. Ergo regulators of BDNF availability, such as the Val66Met polymorphism, may orchestrate sensitivity to stress, trauma, and risk of stress-induced disorders such as PTSD. Given an increasing interest in personalized psychiatry and clinically complex cases, this model provides a framework from which to experimentally disentangle the causal actions of BDNF in stress responses, which likely interact to potentiate, produce, and impair treatment of, stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia. .,College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia. .,Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
6
|
Vaverková Z, Milton AL, Merlo E. Retrieval-Dependent Mechanisms Affecting Emotional Memory Persistence: Reconsolidation, Extinction, and the Space in Between. Front Behav Neurosci 2020; 14:574358. [PMID: 33132861 PMCID: PMC7550798 DOI: 10.3389/fnbeh.2020.574358] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/03/2020] [Indexed: 01/20/2023] Open
Abstract
Maladaptive emotional memories contribute to the persistence of many mental health disorders, and therefore the prospect of disrupting these memories to produce long-term reductions in relapse is of great clinical appeal. Reducing the impact of maladaptive emotional memories on behaviour could be achieved by two retrieval-dependent manipulations that engage separate mnemonic processes: "reconsolidation disruption" and "extinction enhancement." Extinction occurs during a prolonged re-exposure session in the absence of the expected emotional outcome and is widely accepted as reflecting the formation of a new, inhibitory memory that prevents behavioural expression of the original trace. Reconsolidation, by contrast, involves the destabilisation of the original memory, allowing for subsequent updating and restabilisation in specific brain regions, unless the re-stabilization process is prevented through specific pharmacological or behavioural interventions. Both destabilisation of the original memory and memory extinction require that re-exposure induces prediction error-a mismatch between what is expected and what actually occurs-but the parameters that allow reconsolidation and extinction to occur, and control the transition between them, have not been well-characterised. Here, we review what is known about the induction of memory destabilisation and extinction, and the transition period that separates these mnemonic processes, drawing on preclinical and clinical examples. A deeper understanding of the processes that determine the alternative routes to memory persistence or inhibition is critical for designing new and more reliable clinical treatments targeting maladaptive emotional memories.
Collapse
Affiliation(s)
- Zuzana Vaverková
- School of Psychology, University of Sussex, Brighton, United Kingdom
| | - Amy L Milton
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
| | - Emiliano Merlo
- School of Psychology, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
7
|
Rakesh G, Morey RA, Zannas AS, Malik Z, Clausen A, Marx CE, Kritzer MD, Szabo ST. Resilience as a translational endpoint in the treatment of PTSD. Mol Psychiatry 2019; 24:1268-1283. [PMID: 30867558 PMCID: PMC6713904 DOI: 10.1038/s41380-019-0383-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 01/24/2019] [Accepted: 02/14/2019] [Indexed: 12/31/2022]
Abstract
Resilience is a neurobiological entity that shapes an individual's response to trauma. Resilience has been implicated as the principal mediator in the development of mental illness following exposure to trauma. Although animal models have traditionally defined resilience as molecular and behavioral changes in stress responsive circuits following trauma, this concept needs to be further clarified for both research and clinical use. Here, we analyze the construct of resilience from a translational perspective and review optimal measurement methods and models. We also seek to distinguish between resilience, stress vulnerability, and posttraumatic growth. We propose that resilience can be quantified as a multifactorial determinant of physiological parameters, epigenetic modulators, and neurobiological candidate markers. This multifactorial definition can determine PTSD risk before and after trauma exposure. From this perspective, we propose the use of an 'R Factor' analogous to Spearman's g factor for intelligence to denote these multifactorial determinants. In addition, we also propose a novel concept called 'resilience reserve', analogous to Stern's cognitive reserve, to summarize the sum total of physiological processes that protect and compensate for the effect of trauma. We propose the development and application of challenge tasks to measure 'resilience reserve' and guide the assessment and monitoring of 'R Factor' as a biomarker for PTSD.
Collapse
Affiliation(s)
- Gopalkumar Rakesh
- Duke-UNC Brain Imaging and Analysis Center (BIAC), Durham, NC, 27710, USA. .,Durham VA Health Care System, Durham, NC, 27705, USA. .,VISN 6 VA Mid-Atlantic Mental Illness Research Education and Clinical Center (MIRECC), 3022 Croasdaile Drive, Durham, NC, 27705, USA.
| | - Rajendra A Morey
- Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham NC, Duke University School of Medicine, Durham, NC 27710,VISN 6 VA Mid-Atlantic Mental Illness Research Education and Clinical Center (MIRECC), 3022 Croasdaile Drive, Durham, NC 27705
| | | | - Zainab Malik
- Child and Adolescent Psychiatry, University of California, Davis, CA 95616
| | - Ashley Clausen
- Duke-UNC Brain Imaging and Analysis Center (BIAC), Durham VA Health Care System, VISN 6 VA Mid-Atlantic Mental Illness Research Education and Clinical Center, 3022 Croasdaile Drive, Durham, NC 27705
| | - Christine E Marx
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, 27710, USA,Division of Translational Neurosciences, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Michael D Kritzer
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Steven T Szabo
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, 27710, USA,Veterans Affairs Medical Center, Mental Health Service Line, Durham, North Carolina, 27710, USA
| |
Collapse
|
8
|
Trent S, Hall J, Connelly WM, Errington AC. Cyfip1 Haploinsufficiency Does Not Alter GABA A Receptor δ-Subunit Expression and Tonic Inhibition in Dentate Gyrus PV + Interneurons and Granule Cells. eNeuro 2019; 6:ENEURO.0364-18.2019. [PMID: 31209152 PMCID: PMC6635810 DOI: 10.1523/eneuro.0364-18.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 12/01/2022] Open
Abstract
Copy number variation (CNV) at chromosomal region 15q11.2 is linked to increased risk of neurodevelopmental disorders including autism and schizophrenia. A significant gene at this locus is cytoplasmic fragile X mental retardation protein (FMRP) interacting protein 1 (CYFIP1). CYFIP1 protein interacts with FMRP, whose monogenic absence causes fragile X syndrome (FXS). Fmrp knock-out has been shown to reduce tonic GABAergic inhibition by interacting with the δ-subunit of the GABAA receptor (GABAAR). Using in situ hybridization (ISH), qPCR, Western blotting techniques, and patch clamp electrophysiology in brain slices from a Cyfip1 haploinsufficient mouse, we examined δ-subunit mediated tonic inhibition in the dentate gyrus (DG). In wild-type (WT) mice, DG granule cells (DGGCs) responded to the δ-subunit-selective agonist THIP with significantly increased tonic currents. In heterozygous mice, no significant difference was observed in THIP-evoked currents in DGGCs. Phasic GABAergic inhibition in DGGC was also unaltered with no difference in properties of spontaneous IPSCs (sIPSCs). Additionally, we demonstrate that DG granule cell layer (GCL) parvalbumin-positive interneurons (PV+-INs) have functional δ-subunit-mediated tonic GABAergic currents which, unlike DGGC, are also modulated by the α1-selective drug zolpidem. Similar to DGGC, both IPSCs and THIP-evoked currents in PV+-INs were not different between Cyfip1 heterozygous and WT mice. Supporting our electrophysiological data, we found no significant change in hippocampal δ-subunit mRNA expression or protein level and no change in α1/α4-subunit mRNA expression. Thus, Cyfip1 haploinsufficiency, mimicking human 15q11.2 microdeletion syndrome, does not alter hippocampal phasic or tonic GABAergic inhibition, substantially differing from the Fmrp knock-out mouse model.
Collapse
Affiliation(s)
- Simon Trent
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, United Kingdom
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, United Kingdom
| | - William M Connelly
- School of Medicine, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Adam C Errington
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, United Kingdom
| |
Collapse
|
9
|
O'Leary JD, Hoban AE, Cryan JF, O'Leary OF, Nolan YM. Differential effects of adolescent and adult-initiated voluntary exercise on context and cued fear conditioning. Neuropharmacology 2018; 145:49-58. [PMID: 29793890 DOI: 10.1016/j.neuropharm.2018.05.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 04/19/2018] [Accepted: 05/06/2018] [Indexed: 12/20/2022]
Abstract
Adolescence is a critical period for postnatal brain maturation and a time during which there is increased susceptibility to developing emotional and cognitive-related disorders. Exercise during adulthood has been shown to increase hippocampal plasticity and enhance cognition. However, the impact of exercise initiated in adolescence, on brain and behaviour in adulthood is not yet fully explored or understood. The aim of this study was to compare the impact of voluntary exercise that was initiated either during adolescence or early adulthood on cognitive performance in hippocampal and amygdala-dependent fear conditioning tasks in adulthood. Adult (eight weeks old) and adolescent (four weeks old) male Sprague Dawley rats had access to a running wheel (exercise) or were left undisturbed (sedentary control) for seven weeks. Adult-initiated exercise enhanced both contextual and cued fear conditioning, while conversely, exercise that began in adolescence did not affect performance in these tasks. These behaviours were accompanied by differential expression of plasticity-related genes in the hippocampus and amygdala in adulthood. Specifically, adolescent-initiated exercise increased the expression of an array of plasticity related genes in the hippocampus including BDNF, synaptophysin, Creb, PSD-95, Arc, TLX and DCX, while adult-initiated exercise did not affect hippocampal plasticity related genes. Together results show that exercise initiated during adolescence has a differential effect on hippocampal and amygdala-dependent behaviour and neuronal plasticity compared to when exercise was initiated in adulthood. These findings reinforce adolescence as a period during which environmental influences have a distinct impact on neuronal plasticity and cognition. This article is part of the Special Issue entitled "Neurobiology of Environmental Enrichment".
Collapse
Affiliation(s)
- James D O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Alan E Hoban
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
10
|
Alvarez-Ricartes N, Oliveros-Matus P, Mendoza C, Perez-Urrutia N, Echeverria F, Iarkov A, Barreto GE, Echeverria V. Intranasal Cotinine Plus Krill Oil Facilitates Fear Extinction, Decreases Depressive-Like Behavior, and Increases Hippocampal Calcineurin A Levels in Mice. Mol Neurobiol 2018; 55:7949-7960. [PMID: 29488138 DOI: 10.1007/s12035-018-0916-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/18/2018] [Indexed: 02/07/2023]
Abstract
Failure in fear extinction is one of the more troublesome characteristics of posttraumatic stress disorder (PTSD). Cotinine facilitates fear memory extinction and reduces depressive-like behavior when administered 24 h after fear conditioning in mice. In this study, it was investigated the behavioral and molecular effects of cotinine, and other antidepressant preparations infused intranasally. Intranasal (IN) cotinine, IN krill oil, IN cotinine plus krill oil, and oral sertraline were evaluated on depressive-like behavior and fear retention and extinction after fear conditioning in C57BL/6 mice. Since calcineurin A has been involved in facilitating fear extinction in rodents, we also investigated changes of calcineurin in the hippocampus, a region key on contextual fear extinction. Short-term treatment with cotinine formulations was superior to krill oil and oral sertraline in reducing depressive-like behavior and fear consolidation and enhancing contextual fear memory extinction in mice. IN krill oil slowed the extinction of fear. IN cotinine preparations increased the levels of calcineurin A in the hippocampus of conditioned mice. In the light of the results, the future investigation of the use of IN cotinine preparations for the extinction of contextual fear memory and treatment of treatment-resistant depression (TRD) in PTSD is discussed.
Collapse
Affiliation(s)
- Nathalie Alvarez-Ricartes
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Lientur 1457, 4030000, Concepción, Chile
| | - Patricia Oliveros-Matus
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Lientur 1457, 4030000, Concepción, Chile
| | - Cristhian Mendoza
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Lientur 1457, 4030000, Concepción, Chile
| | - Nelson Perez-Urrutia
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Lientur 1457, 4030000, Concepción, Chile
| | - Florencia Echeverria
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Lientur 1457, 4030000, Concepción, Chile
| | - Alexandre Iarkov
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Lientur 1457, 4030000, Concepción, Chile.
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Valentina Echeverria
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Lientur 1457, 4030000, Concepción, Chile. .,Bay Pines VA Healthcare System, Research and Development, Bay Pines VAHCS, 10,000 Bay Pines Blvd., Bldg. 23, Rm123, Bay Pines, FL, 33744, USA.
| |
Collapse
|
11
|
Does exercise augment operant and Pavlovian extinction: A meta-analysis. J Psychiatr Res 2018; 96:73-93. [PMID: 28987515 DOI: 10.1016/j.jpsychires.2017.09.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/17/2017] [Accepted: 09/14/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Exposure therapy, a behavioral approach to reduce symptomology in fear, anxiety, and drug-related psychiatric disorders, is based on learning and memory principles of extinction, and is subject to relapse. As such, it is important to find ways to enhance outcomes. One such way is through exercise. OBJECTIVES Identify if exercise augments extinction behavior, and whether this depends on the experimental paradigm used (i.e. operant or Pavlovian) and/or stimulus (i.e. appetitive or aversive). Additionally, determine which moderating variables influence the effects of exercise on extinction learning. METHODS A literature search was conducted and a Hedges' g calculation was employed to conduct a meta-analysis (metaSEM) using a structural equation modeling approach. This approach was chosen because of its ability to account for dependencies in effect sizes. RESULTS We found a significant effect of exercise as an augmentation over extinction alone (g = 0.37, p < 0.001), with extinction paradigm (but not stimulus) producing a moderating effect (B = 0.43, p = 0.030). Data were then split by extinction paradigm, with operant extinction models having a significant effect (g = 0.55, p < 0.001), and number of extinction sessions moderating aggregate effects. Pavlovian models did not have significant overall effects (g = 0.11, p = 0.3976), but were moderated by the number of animals housed together and exercise after extinction. CONCLUSIONS The effects of exercise on extinction learning are differentially modulated by the type of paradigm used, the number of extinction sessions, the timing of when exercise treatment was applied (after extinction), and the housing conditions.
Collapse
|
12
|
Hippocampal Regulation of Postsynaptic Density Homer1 by Associative Learning. Neural Plast 2017; 2017:5959182. [PMID: 29238619 PMCID: PMC5697134 DOI: 10.1155/2017/5959182] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/18/2017] [Accepted: 10/10/2017] [Indexed: 11/18/2022] Open
Abstract
Genes involved in synaptic plasticity, particularly genes encoding postsynaptic density proteins, have been recurrently linked to psychiatric disorders including schizophrenia and autism. Postsynaptic density Homer1 proteins contribute to synaptic plasticity through the competing actions of short and long isoforms. The activity-induced expression of short Homer1 isoforms, Homer1a and Ania-3, is thought to be related to processes of learning and memory. However, the precise regulation of Homer1a and Ania-3 with different components of learning has not been investigated. Here, we used in situ hybridization to quantify short and long Homer1 expression in the hippocampus following consolidation, retrieval, and extinction of associative fear memory, using contextual fear conditioning in rats. Homer1a and Ania-3, but not long Homer1, were regulated by contextual fear learning or novelty detection, although their precise patterns of expression in hippocampal subregions were dependent on the isoform. We also show for the first time that the two short Homer1 isoforms are regulated after the retrieval and extinction of contextual fear memory, albeit with distinct temporal and spatial profiles. These findings support a role of activity-induced Homer1 isoforms in learning and memory processes in discrete hippocampal subregions and suggest that Homer1a and Ania-3 may play separable roles in synaptic plasticity.
Collapse
|
13
|
Haubrich J, Machado A, Boos FZ, Crestani AP, Sierra RO, Alvares LDO, Quillfeldt JA. Enhancement of extinction memory by pharmacological and behavioral interventions targeted to its reactivation. Sci Rep 2017; 7:10960. [PMID: 28887561 PMCID: PMC5591313 DOI: 10.1038/s41598-017-11261-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/22/2017] [Indexed: 12/22/2022] Open
Abstract
Extinction is a process that involves new learning that inhibits the expression of previously acquired memories. Although temporarily effective, extinction does not erase an original fear association. Since the extinction trace tends to fade over time, the original memory can resurge. On the other hand, strengthening effects have been described in several reconsolidation studies using different behavioral and pharmacological manipulations. In order to know whether an extinction memory can be strengthened by reactivation-based interventions in the contextual fear conditioning task, we began by replicating the classic phenomenon of spontaneous recovery to show that brief reexposure sessions can prevent the decay of the extinction trace over time in a long-lasting way. This fear attenuation was shown to depend both on L-type calcium channels and protein synthesis, which suggests a reconsolidation process behind the reactivation-induced strengthening effect. The extinction trace was also susceptible to enhancement by a post-reactivation infusion of a memory-enhancing drug (NaB), which was also able to prevent rapid fear reacquisition (savings). These findings point to new reactivation-based approaches able to strengthen an extinction memory to promote its persistence. The constructive interactions between extinction and reconsolidation may represent a promising novel approach in the realm of fear-related disorder treatments.
Collapse
Affiliation(s)
- Josué Haubrich
- Psychobiology and Neurocomputation lab and Neurobiology of Memory lab. Neurosciences Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Adriano Machado
- Psychobiology and Neurocomputation lab and Neurobiology of Memory lab. Neurosciences Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Flávia Zacouteguy Boos
- Psychobiology and Neurocomputation lab and Neurobiology of Memory lab. Neurosciences Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana P Crestani
- Psychobiology and Neurocomputation lab and Neurobiology of Memory lab. Neurosciences Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rodrigo O Sierra
- Psychobiology and Neurocomputation lab and Neurobiology of Memory lab. Neurosciences Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lucas de Oliveira Alvares
- Psychobiology and Neurocomputation lab and Neurobiology of Memory lab. Neurosciences Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jorge A Quillfeldt
- Psychobiology and Neurocomputation lab and Neurobiology of Memory lab. Neurosciences Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
14
|
Epigenetic mechanisms of alcoholism and stress-related disorders. Alcohol 2017; 60:7-18. [PMID: 28477725 DOI: 10.1016/j.alcohol.2017.01.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/30/2016] [Accepted: 01/03/2017] [Indexed: 12/20/2022]
Abstract
Stress-related disorders, such as anxiety, early life stress, and posttraumatic stress disorder appear to be important factors in promoting alcoholism, as alcohol consumption can temporarily attenuate the negative affective symptoms of these disorders. Several molecules involved in signaling pathways may contribute to the neuroadaptation induced during alcohol dependence and stress disorders, and among these, brain-derived neurotrophic factor (BDNF), corticotropin releasing factor (CRF), neuropeptide Y (NPY) and opioid peptides (i.e., nociceptin and dynorphin) are involved in the interaction of stress and alcohol. In fact, alterations in the expression and function of these molecules have been associated with the pathophysiology of stress-related disorders and alcoholism. In recent years, various studies have focused on the epigenetic mechanisms that regulate chromatin architecture, thereby modifying gene expression. Interestingly, epigenetic modifications in specific brain regions have been shown to be associated with the neurobiology of psychiatric disorders, including alcoholism and stress. In particular, the enzymes responsible for chromatin remodeling (i.e., histone deacetylases and methyltransferases, DNA methyltransferases) have been identified as common molecular mechanisms for the interaction of stress and alcohol and have become promising therapeutic targets to treat or prevent alcoholism and associated emotional disorders.
Collapse
|
15
|
Duclot F, Perez-Taboada I, Wright KN, Kabbaj M. Prediction of individual differences in fear response by novelty seeking, and disruption of contextual fear memory reconsolidation by ketamine. Neuropharmacology 2016; 109:293-305. [PMID: 27343386 DOI: 10.1016/j.neuropharm.2016.06.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 05/23/2016] [Accepted: 06/21/2016] [Indexed: 01/07/2023]
Abstract
Only a portion of the population exposed to trauma will develop persistent emotional alterations characteristic of posttraumatic stress disorder (PTSD), which illustrates the necessity for identifying vulnerability factors and novel pharmacotherapeutic alternatives. Interestingly, clinical evidence suggests that novelty seeking is a good predictor for vulnerability to the development of excessive and persistent fear. Here, we first tested this hypothesis by analyzing contextual and cued fear responses of rats selected for their high (high responders, HR) or low (low responders, LR) exploration of a novel environment, indicator of novelty seeking. While HR and LR rats exhibited similar sensitivity to the shock and cued fear memory retention, fewer extinction sessions were required in HR than LR animals to reach extinction, indicating faster contextual and cued memory extinction. In a second part, we found an effective disruption of contextual fear reconsolidation by the N-methyl-d-aspartate receptor antagonist ketamine, associated with a down-regulation of early growth response 1 (Egr1) in the hippocampal CA1 area, and up-regulation of brain-derived neurotrophic factor (Bdnf) mRNA levels in the prelimbic and infralimbic cortices. Altogether, these data demonstrate a link between novelty seeking and conditioned fear extinction, and highlight a promising novel role of ketamine in affecting established fear memory.
Collapse
Affiliation(s)
- Florian Duclot
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, United States; Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States
| | - Iara Perez-Taboada
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, United States; Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States
| | - Katherine N Wright
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, United States; Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, United States; Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States.
| |
Collapse
|
16
|
Elevated paternal glucocorticoid exposure alters the small noncoding RNA profile in sperm and modifies anxiety and depressive phenotypes in the offspring. Transl Psychiatry 2016; 6:e837. [PMID: 27300263 PMCID: PMC4931607 DOI: 10.1038/tp.2016.109] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/25/2016] [Indexed: 12/21/2022] Open
Abstract
Recent studies have suggested that physiological and behavioral traits may be transgenerationally inherited through the paternal lineage, possibly via non-genomic signals derived from the sperm. To investigate how paternal stress might influence offspring behavioral phenotypes, a model of hypothalamic-pituitary-adrenal (HPA) axis dysregulation was used. Male breeders were administered water supplemented with corticosterone (CORT) for 4 weeks before mating with untreated female mice. Female, but not male, F1 offspring of CORT-treated fathers displayed altered fear extinction at 2 weeks of age. Only male F1 offspring exhibited altered patterns of ultrasonic vocalization at postnatal day 3 and, as adults, showed decreased time in open on the elevated-plus maze and time in light on the light-dark apparatus, suggesting a hyperanxiety-like behavioral phenotype due to paternal CORT treatment. Interestingly, expression of the paternally imprinted gene Igf2 was increased in the hippocampus of F1 male offspring but downregulated in female offspring. Male and female F2 offspring displayed increased time spent in the open arm of the elevated-plus maze, suggesting lower levels of anxiety compared with control animals. Only male F2 offspring showed increased immobility time on the forced-swim test and increased latency to feed on the novelty-supressed feeding test, suggesting a depression-like phenotype in these animals. Collectively, these data provide evidence that paternal CORT treatment alters anxiety and depression-related behaviors across multiple generations. Analysis of the small RNA profile in sperm from CORT-treated males revealed marked effects on the expression of small noncoding RNAs. Sperm from CORT-treated males contained elevated levels of three microRNAs, miR-98, miR-144 and miR-190b, which are predicted to interact with multiple growth factors, including Igf2 and Bdnf. Sustained elevation of glucocorticoids is therefore involved in the transmission of paternal stress-induced traits across generations in a process involving small noncoding RNA signals transmitted by the male germline.
Collapse
|
17
|
Scholz B, Doidge AN, Barnes P, Hall J, Wilkinson LS, Thomas KL. The Regulation of Cytokine Networks in Hippocampal CA1 Differentiates Extinction from Those Required for the Maintenance of Contextual Fear Memory after Recall. PLoS One 2016; 11:e0153102. [PMID: 27224427 PMCID: PMC4880201 DOI: 10.1371/journal.pone.0153102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/23/2016] [Indexed: 12/17/2022] Open
Abstract
We investigated the distinctiveness of gene regulatory networks in CA1 associated with the extinction of contextual fear memory (CFM) after recall using Affymetrix GeneChip Rat Genome 230 2.0 Arrays. These data were compared to previously published retrieval and reconsolidation-attributed, and consolidation datasets. A stringent dual normalization and pareto-scaled orthogonal partial least-square discriminant multivariate analysis together with a jack-knifing-based cross-validation approach was used on all datasets to reduce false positives. Consolidation, retrieval and extinction were correlated with distinct patterns of gene expression 2 hours later. Extinction-related gene expression was most distinct from the profile accompanying consolidation. A highly specific feature was the discrete regulation of neuroimmunological gene expression associated with retrieval and extinction. Immunity-associated genes of the tyrosine kinase receptor TGFβ and PDGF, and TNF families' characterized extinction. Cytokines and proinflammatory interleukins of the IL-1 and IL-6 families were enriched with the no-extinction retrieval condition. We used comparative genomics to predict transcription factor binding sites in proximal promoter regions of the retrieval-regulated genes. Retrieval that does not lead to extinction was associated with NF-κB-mediated gene expression. We confirmed differential NF-κBp65 expression, and activity in all of a representative sample of our candidate genes in the no-extinction condition. The differential regulation of cytokine networks after the acquisition and retrieval of CFM identifies the important contribution that neuroimmune signalling plays in normal hippocampal function. Further, targeting cytokine signalling upon retrieval offers a therapeutic strategy to promote extinction mechanisms in human disorders characterised by dysregulation of associative memory.
Collapse
Affiliation(s)
- Birger Scholz
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Amie N. Doidge
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Philip Barnes
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Jeremy Hall
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Lawrence S. Wilkinson
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- Schools of Psychology and Medicine, Behavioral Genetics Group, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics and Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Kerrie L. Thomas
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
18
|
Kuipers SD, Trentani A, Tiron A, Mao X, Kuhl D, Bramham CR. BDNF-induced LTP is associated with rapid Arc/Arg3.1-dependent enhancement in adult hippocampal neurogenesis. Sci Rep 2016; 6:21222. [PMID: 26888068 PMCID: PMC4758047 DOI: 10.1038/srep21222] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/12/2016] [Indexed: 11/10/2022] Open
Abstract
Adult neurogenesis in the hippocampus is a remarkable phenomenon involved in various aspects of learning and memory as well as disease pathophysiology. Brain-derived neurotrophic factor (BDNF) represents a major player in the regulation of this unique form of neuroplasticity, yet the mechanisms underlying its pro-neurogenic actions remain unclear. Here, we examined the effects associated with brief (25 min), unilateral infusion of BDNF in the rat dentate gyrus. Acute BDNF infusion induced long-term potentiation (LTP) of medial perforant path-evoked synaptic transmission and, concomitantly, enhanced hippocampal neurogenesis bilaterally, reflected by increased dentate gyrus BrdU + cell numbers. Importantly, inhibition of activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) translation through local, unilateral infusion of anti-sense oligodeoxynucleotides (ArcAS) prior to BDNF infusion blocked both BDNF-LTP induction and the associated pro-neurogenic effects. Notably, basal rates of proliferation and newborn cell survival were unaltered in homozygous Arc/Arg3.1 knockout mice. Taken together these findings link the pro-neurogenic effects of acute BDNF infusion to induction of Arc/Arg3.1-dependent LTP in the adult rodent dentate gyrus.
Collapse
Affiliation(s)
- Sjoukje D Kuipers
- Department of Biomedicine and KG Jebsen Centre for Research on Neuropsychiatric Disorders, Jonas Lies vei 91, University of Bergen, 5009 Bergen, Norway
| | - Andrea Trentani
- Department of Biomedicine and KG Jebsen Centre for Research on Neuropsychiatric Disorders, Jonas Lies vei 91, University of Bergen, 5009 Bergen, Norway
| | - Adrian Tiron
- Department of Biomedicine and KG Jebsen Centre for Research on Neuropsychiatric Disorders, Jonas Lies vei 91, University of Bergen, 5009 Bergen, Norway
| | - Xiaosong Mao
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology (ZMNH) University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Dietmar Kuhl
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology (ZMNH) University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Clive R Bramham
- Department of Biomedicine and KG Jebsen Centre for Research on Neuropsychiatric Disorders, Jonas Lies vei 91, University of Bergen, 5009 Bergen, Norway
| |
Collapse
|
19
|
Rescue of long-term memory after reconsolidation blockade. Nat Commun 2015; 6:7897. [PMID: 26238574 PMCID: PMC4532853 DOI: 10.1038/ncomms8897] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 06/24/2015] [Indexed: 11/08/2022] Open
Abstract
Memory reconsolidation is considered to be the process whereby stored memories become labile on recall, allowing updating. Blocking the restabilization of a memory during reconsolidation is held to result in a permanent amnesia. The targeted knockdown of either Zif268 or Arc levels in the brain, and inhibition of protein synthesis, after a brief recall results in a non-recoverable retrograde amnesia, known as reconsolidation blockade. These experimental manipulations are seen as key proof for the existence of reconsolidation. However, here we demonstrate that despite disrupting the molecular correlates of reconsolidation in the hippocampus, rodents are still able to recover contextual memories. Our results challenge the view that reconsolidation is a separate memory process and instead suggest that the molecular events activated initially at recall act to constrain premature extinction.
Collapse
|
20
|
HDAC inhibitors as cognitive enhancers in fear, anxiety and trauma therapy: where do we stand? Biochem Soc Trans 2015; 42:569-81. [PMID: 24646280 PMCID: PMC3961057 DOI: 10.1042/bst20130233] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A novel strategy to treat anxiety and fear-related disorders such as phobias, panic and PTSD (post-traumatic stress disorder) is combining CBT (cognitive behavioural therapy), including extinction-based exposure therapy, with cognitive enhancers. By targeting and boosting mechanisms underlying learning, drug development in this field aims at designing CBT-augmenting compounds that help to overcome extinction learning deficits, promote long-term fear inhibition and thus support relapse prevention. Progress in revealing the role of epigenetic regulation of specific genes associated with extinction memory generation has opened new avenues in this direction. The present review examines recent evidence from pre-clinical studies showing that increasing histone acetylation, either via genetic or pharmacological inhibition of HDACs (histone deacetylases) by e.g. vorinostat/SAHA (suberoylanilide hydroxamic acid), entinostat/MS-275, sodium butyrate, TSA (trichostatin A) or VPA (valproic acid), or by targeting HATs (histone acetyltransferases), augments fear extinction and, importantly, generates a long-term extinction memory that can protect from return of fear phenomena. The molecular mechanisms and pathways involved including BDNF (brain-derived neurotrophic factor) and NMDA (N-methyl-D-aspartate) receptor signalling are just beginning to be revealed. First studies in healthy humans are in support of extinction-facilitating effects of HDAC inhibitors. Very recent evidence that HDAC inhibitors can rescue deficits in extinction-memory-impaired rodents indicates a potential clinical utility of this approach also for exposure therapy-resistant patients. Important future work includes investigation of the long-term safety aspects of HDAC inhibitor treatment, as well as design of isotype(s)-specific inhibitors. Taken together, HDAC inhibitors display promising potential as pharmacological adjuncts to augment the efficacy of exposure-based approaches in anxiety and trauma therapy.
Collapse
|
21
|
Baldi E, Bucherelli C. Brain sites involved in fear memory reconsolidation and extinction of rodents. Neurosci Biobehav Rev 2015; 53:160-90. [DOI: 10.1016/j.neubiorev.2015.04.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 03/30/2015] [Accepted: 04/06/2015] [Indexed: 12/21/2022]
|
22
|
Stress, trauma and PTSD: translational insights into the core synaptic circuitry and its modulation. Brain Struct Funct 2015; 221:2401-26. [PMID: 25985955 DOI: 10.1007/s00429-015-1056-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 04/30/2015] [Indexed: 12/19/2022]
Abstract
Evidence is considered as to whether behavioral criteria for diagnosis of post-traumatic stress disorder (PTSD) are applicable to that of traumatized animals and whether the phenomena of acquisition, extinction and reactivation of fear behavior in animals are also successfully applicable to humans. This evidence suggests an affirmative answer in both cases. Furthermore, the deficits in gray matter found in PTSD, determined with magnetic resonance imaging, are also observed in traumatized animals, lending neuropsychological support to the use of animals to probe what has gone awry in PTSD. Such animal experiments indicate that the core synaptic circuitry mediating behavior following trauma consists of the amygdala, ventral-medial prefrontal cortex and hippocampus, all of which are modulated by the basal ganglia. It is not clear if this is the case in PTSD as the observations using fMRI are equivocal and open to technical objections. Nevertheless, the effects of the basal ganglia in controlling glutamatergic synaptic transmission through dopaminergic and serotonergic synaptic mechanisms in the core synaptic circuitry provides a ready explanation for why modifying these mechanisms delays extinction in animal models and predisposes towards PTSD. In addition, changes of brain-derived neurotrophic factor (BDNF) in the core synaptic circuitry have significant effects on acquisition and extinction in animal experiments with single nucleotide polymorphisms in the BDNF gene predisposing to PTSD.
Collapse
|
23
|
Chai N, Liu JF, Xue YX, Yang C, Yan W, Wang HM, Luo YX, Shi HS, Wang JS, Bao YP, Meng SQ, Ding ZB, Wang XY, Lu L. Delayed noradrenergic activation in the dorsal hippocampus promotes the long-term persistence of extinguished fear. Neuropsychopharmacology 2014; 39:1933-45. [PMID: 24553734 PMCID: PMC4059903 DOI: 10.1038/npp.2014.42] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/08/2014] [Accepted: 02/10/2014] [Indexed: 01/18/2023]
Abstract
Fear extinction has been extensively studied, but little is known about the molecular processes that underlie the persistence of extinction long-term memory (LTM). We found that microinfusion of norepinephrine (NE) into the CA1 area of the dorsal hippocampus during the early phase (0 h) after extinction enhanced extinction LTM at 2 and 14 days after extinction. Intra-CA1 infusion of NE during the late phase (12 h) after extinction selectively promoted extinction LTM at 14 days after extinction that was blocked by the β-receptor antagonist propranolol, protein kinase A (PKA) inhibitor Rp-cAMPS, and protein synthesis inhibitors anisomycin and emetine. The phosphorylation levels of PKA, cyclic adenosine monophosphate response element-binding protein (CREB), GluR1, and the membrane GluR1 level were increased by NE during the late phase after extinction that was also blocked by propranolol and Rp-cAMPS. These results suggest that the enhancement of extinction LTM persistence induced by NE requires the activation of the β-receptor/PKA/CREB signaling pathway and membrane GluR1 trafficking. Moreover, extinction increased the phosphorylation levels of Erk1/2, CREB, and GluR1, and the membrane GluR1 level during the late phase, and anisomycin/emetine alone disrupted the persistence of extinction LTM, indicating that the persistence of extinction LTM requires late-phase protein synthesis in the CA1. Propranolol and Rp-cAMPS did not completely disrupt the persistence of extinction LTM, suggesting that another β-receptor/PKA-independent mechanism underlies the persistence of extinction LTM. Altogether, our results showed that enhancing hippocampal noradrenergic activity during the late phase after extinction selectively promotes the persistence of extinction LTM.
Collapse
Affiliation(s)
- Ning Chai
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Beijing, China,Institute of Mental Health and Hebei Brain Ageing and Cognitive Neuroscience Laboratory, Hebei Medical University, Shijiazhuang, China
| | - Jian-Feng Liu
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Beijing, China,National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Chang Yang
- Affiliated Hospital and School of Pharmacy of Guiyang Medical University, Guiyang, China
| | - Wei Yan
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Beijing, China,National Institute on Drug Dependence, Peking University, Beijing, China
| | - Hui-Min Wang
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Beijing, China,National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yi-Xiao Luo
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Beijing, China,National Institute on Drug Dependence, Peking University, Beijing, China
| | - Hai-Shui Shi
- Department of Biochemistry and Molecular Biology, Basic Medical College, Hebei Medical University, Shijiazhuang, China
| | - Ji-Shi Wang
- Affiliated Hospital and School of Pharmacy of Guiyang Medical University, Guiyang, China
| | - Yan-Ping Bao
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Shi-Qiu Meng
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Beijing, China,National Institute on Drug Dependence, Peking University, Beijing, China
| | - Zeng-Bo Ding
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Xue-Yi Wang
- Institute of Mental Health and Hebei Brain Ageing and Cognitive Neuroscience Laboratory, Hebei Medical University, Shijiazhuang, China,Institute of Mental Health Hebei Brain Ageing and Cognitive Neuroscience Laboratory, Hebei Medical University, Shijiazhuang 050031, China, E-mail:
| | - Lin Lu
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Beijing, China,National Institute on Drug Dependence, Peking University, Beijing, China,Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China,Institute of Mental Health and National Institute on Drug Dependence, Peking University, 51 Huayuanbei Road, Beijing 100191, China, Tel: +86 10 82802459, Fax: +86 10 62032624, E-mail:
| |
Collapse
|
24
|
Abstract
Research has identified distinct neuronal circuits within the basolateral amygdala (BLA) that differentially mediate fear expression versus inhibition; however, molecular markers of these populations remain unknown. Here we examine whether optogenetic activation of a cellular subpopulation, which may correlate with the physiologically identified extinction neurons in the BLA, would differentially support fear conditioning versus fear inhibition/extinction. We first molecularly characterized Thy1-channelrhodopsin-2 (Thy1-ChR2-EYFP)-expressing neurons as a subpopulation of glutamatergic pyramidal neurons within the BLA. Optogenetic stimulation of these neurons inhibited a subpopulation of medial central amygdala neurons and shunted excitation from the lateral amygdala. Brief activation of these neurons during fear training disrupted later fear memory in male mice. Optogenetic activation during unreinforced stimulus exposure enhanced extinction retention, but had no effect on fear expression, locomotion, or open-field behavior. Together, these data suggest that the Thy1-expressing subpopulation of BLA pyramidal neurons provide an important molecular and pharmacological target for inhibiting fear and enhancing extinction and for furthering our understanding of the molecular mechanisms of fear processing.
Collapse
|
25
|
Bekinschtein P, Cammarota M, Medina JH. BDNF and memory processing. Neuropharmacology 2013; 76 Pt C:677-83. [PMID: 23688925 DOI: 10.1016/j.neuropharm.2013.04.024] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 03/14/2013] [Accepted: 04/08/2013] [Indexed: 01/19/2023]
Abstract
Brain-derived neurotrophic factor has emerged as one of the most important molecules involved in memory. Its wide role in different types of memories that depend on different structures as well as its involvement in distinct memory stages points at BDNF as one likely target to treat cognitive impairments and anxiety-related memory disorders. However, regulation of BDNF expression is very complex as well as its modes of action. Here we describe the latest research carried out on the function of BDNF in memory to illustrate such complexity. This article is part of the Special Issue entitled 'BDNF Regulation of Synaptic Structure, Function, and Plasticity'.
Collapse
Affiliation(s)
- Pedro Bekinschtein
- Instituto de Biologia Celular y Neurociencias, Facultad de Medicina, UBA, Argentina
| | | | | |
Collapse
|
26
|
MAKINO A, IINUMA M, FUKUMITSU H, SOUMIYA H, FURUKAWA Y, FURUKAWA S. Anxiolytic-like effect of trans-2-decenoic acid ethyl ester in stress-induced anxiety-like model mice. Biomed Res 2013; 34:259-67. [DOI: 10.2220/biomedres.34.259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
27
|
Abstract
It is well established that the activity of chromatin-modifying enzymes is crucial for regulating gene expression associated with hippocampal-dependent memories. However, very little is known about how these epigenetic mechanisms influence the formation of cortically dependent memory, particularly when there is competition between opposing memory traces, such as that which occurs during the acquisition and extinction of conditioned fear. Here we demonstrate, in C57BL/6 mice, that the activity of p300/CBP-associated factor (PCAF) within the infralimbic prefrontal cortex is required for long-term potentiation and is necessary for the formation of memory associated with fear extinction, but not for fear acquisition. Further, systemic administration of the PCAF activator SPV106 enhances memory for fear extinction and prevents fear renewal. The selective influence of PCAF on fear extinction is mediated, in part, by a transient recruitment of the repressive transcription factor ATF4 to the promoter of the immediate early gene zif268, which competitively inhibits its expression. Thus, within the context of fear extinction, PCAF functions as a transcriptional coactivator, which may facilitate the formation of memory for fear extinction by interfering with reconsolidation of the original memory trace.
Collapse
|
28
|
Wang WS, Kang S, Liu WT, Li M, Liu Y, Yu C, Chen J, Chi ZQ, He L, Liu JG. Extinction of aversive memories associated with morphine withdrawal requires ERK-mediated epigenetic regulation of brain-derived neurotrophic factor transcription in the rat ventromedial prefrontal cortex. J Neurosci 2012; 32:13763-75. [PMID: 23035088 PMCID: PMC6704794 DOI: 10.1523/jneurosci.1991-12.2012] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 08/02/2012] [Accepted: 08/10/2012] [Indexed: 12/27/2022] Open
Abstract
Recent evidence suggests that histone deacetylase (HDAC) inhibitors facilitate extinction of rewarding memory of drug taking. However, little is known about the role of chromatin modification in the extinction of aversive memory of drug withdrawal. In this study, we used conditioned place aversion (CPA), a highly sensitive model for measuring aversive memory of drug withdrawal, to investigate the role of epigenetic regulation of brain-derived neurotrophic factor (BDNF) gene expression in extinction of aversive memory. We found that CPA extinction training induced an increase in recruiting cAMP response element-binding protein (CREB) to and acetylation of histone H3 at the promoters of BDNF exon I transcript and increased BDNF mRNA and protein expression in the ventromedial prefrontal cortex (vmPFC) of acute morphine-dependent rats and that such epigenetic regulation of BDNF gene transcription could be facilitated or diminished by intra-vmPFC infusion of HDAC inhibitor trichostatin A or extracellular signal-regulated kinase (ERK) inhibitor U0126 (1,4-diamino-2,3-dicyano-1,4-bis(methylthio)butadiene) before extinction training. Correspondingly, disruption of the epigenetic regulation of BDNF gene transcription with U0126 or suppression of BDNF signaling with Trk receptor antagonist K252a or BDNF scavenger tyrosine kinase receptor B (TrkB)-Fc blocked extinction of CPA behavior. We also found that extinction training-induced activation of ERK and CREB and extinction of CPA behavior could be potentiated or suppressed by intra-vmPFC infusion of d-cycloserine, a NMDA receptor partial agonist or aminophosphonopentanoic acid, a NMDA receptor antagonist. We conclude that extinction of aversive memory of morphine withdrawal requires epigenetic regulation of BDNF gene transcription in the vmPFC through activation of the ERK-CREB signaling pathway perhaps in a NMDA receptor-dependent manner.
Collapse
Affiliation(s)
- Wei-Sheng Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China, and
| | - Shuo Kang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China, and
| | - Wen-Tao Liu
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Mu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China, and
| | - Yao Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China, and
| | - Chuan Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China, and
| | - Jie Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China, and
| | - Zhi-Qiang Chi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China, and
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Jing-Gen Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China, and
| |
Collapse
|
29
|
Mizuno K, Dempster E, Mill J, Giese KP. Long-lasting regulation of hippocampal Bdnf gene transcription after contextual fear conditioning. GENES BRAIN AND BEHAVIOR 2012; 11:651-9. [PMID: 22574690 DOI: 10.1111/j.1601-183x.2012.00805.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Long-term memory formation requires de novo protein synthesis and gene transcription. During contextual long-term memory formation brain-derived neurotrophic factor (BDNF) gene expression changes in conjunction with alterations of DNA methylation in the Bdnf gene. However, little is known about the molecular mechanisms underlying the maintenance and persistence of contextual long-term memory. Here, we examined the transcription of specific Bdnf exons in the hippocampus for long periods after contextual fear conditioning. We found changes in transcription lasting for at least 24 h after contextual fear conditioning, with some sex-specific effects. In addition, hypomethylation at a CpG site in CpG island 2 located at the end of Bdnf exon III sequence was detected at 0.5 h and maintained for up to 24 h after contextual fear conditioning. The identification of these long-lasting changes in transcription and DNA methylation at the Bdnf gene suggests that BDNF might have a role for storage of contextual long-term memory in the hippocampus.
Collapse
Affiliation(s)
- K Mizuno
- King's College London, Centre for the Cellular Basis of Behaviour, Institute of Psychiatry, London, UK.
| | | | | | | |
Collapse
|
30
|
Andero R, Ressler KJ. Fear extinction and BDNF: translating animal models of PTSD to the clinic. GENES BRAIN AND BEHAVIOR 2012; 11:503-12. [PMID: 22530815 DOI: 10.1111/j.1601-183x.2012.00801.x] [Citation(s) in RCA: 197] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is the most studied neurotrophin involved in synaptic plasticity processes that are required for long-term learning and memory. Specifically, BDNF gene expression and activation of its high-affinity tropomyosin-related kinase B (TrkB) receptor are necessary in the amygdala, hippocampus and prefrontal cortex for the formation of emotional memories, including fear memories. Among the psychiatric disorders with altered fear processing, there is post-traumatic stress disorder (PTSD) which is characterized by an inability to extinguish fear memories. Since BDNF appears to enhance extinction of fear, targeting impaired extinction in anxiety disorders such as PTSD via BDNF signalling may be an important and novel way to enhance treatment efficacy. The aim of this review is to provide a translational point of view that stems from findings in the BDNF regulation of synaptic plasticity and fear extinction. In addition, there are different systems that seem to alter fear extinction through BDNF modulation like the endocannabinoid system and the hypothalamic-pituitary adrenal axis. Recent work also finds that the pituitary adenylate cyclase-activating polypeptide and PAC1 receptor, which are upstream of BDNF activation, may be implicated in PTSD. Especially interesting are data that exogenous fear extinction enhancers such as antidepressants, histone deacetylases inhibitors and D-cycloserine, a partial N-methyl d-aspartate agonist, may act through or in concert with the BDNF-TrkB system. Finally, we review studies where recombinant BDNF and a putative TrkB agonist, 7,8-dihydroxyflavone, may enhance extinction of fear. These approaches may lead to novel agents that improve extinction in animal models and eventually humans.
Collapse
Affiliation(s)
- R Andero
- Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
31
|
Tronson NC, Corcoran KA, Jovasevic V, Radulovic J. Fear conditioning and extinction: emotional states encoded by distinct signaling pathways. Trends Neurosci 2011; 35:145-55. [PMID: 22118930 DOI: 10.1016/j.tins.2011.10.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 07/29/2011] [Accepted: 10/21/2011] [Indexed: 10/15/2022]
Abstract
Conditioning and extinction of fear have traditionally been viewed as two independent learning processes for encoding representations of contexts or cues (conditioned stimuli, CS), aversive events (unconditioned stimuli, US), and their relationship. Based on the analysis of protein kinase signaling patterns in neurons of the fear circuit, we propose that fear and extinction are best conceptualized as emotional states triggered by a single CS representation with two opposing values: aversive and non-aversive. These values are conferred by the presence or absence of the US and encoded by distinct sets of kinase signaling pathways and their downstream targets. Modulating specific protein kinases thus has the potential to modify emotional states, and hence, may emerge as a promising treatment for anxiety disorders.
Collapse
Affiliation(s)
- Natalie C Tronson
- Department of Psychiatry and Behavioral Sciences, The Asher Center for Study and Treatment of Depressive Disorders, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|